1
|
Hajra D, Yadav V, Singh A, Chakravortty D. SIRT1 and SIRT3 Impact Host Mitochondrial Function and Host Salmonella pH Balance during Infection. ACS Infect Dis 2025; 11:827-843. [PMID: 40168249 DOI: 10.1021/acsinfecdis.4c00751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Mitochondria are important organelles that regulate energy homeostasis. Mitochondrial health and dynamics are crucial determinants of the outcome of several bacterial infections. SIRT3, a major mitochondrial sirtuin, along with SIRT1 regulates key mitochondrial functions. This led to considerable interest in understanding the role of SIRT1 and SIRT3 in governing mitochondrial functions during Salmonella infection. Here, we show that loss of SIRT1 and SIRT3 function either by shRNA-mediated knockdown or by inhibitor treatment led to increased mitochondrial dysfunction with alteration in mitochondrial bioenergetics alongside increased mitochondrial superoxide generation in Salmonella-infected macrophages. Consistent with dysfunctional mitochondria, mitophagy was induced along with altered mitochondrial fusion-fission dynamics in S. typhimurium-infected macrophages. Additionally, the mitochondrial bioenergetic alteration promotes acidification of the infected macrophage cytosolic pH. This host cytosolic pH imbalance skewed the intraphagosomal and intrabacterial pH in the absence of SIRT1 and SIRT3, resulting in decreased SPI-2 gene expression. Our results suggest a novel role for SIRT1 and SIRT3 in maintaining the intracellular Salmonella niche by modulating the mitochondrial bioenergetics and dynamics in the infected macrophages.
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Vikas Yadav
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Amit Singh
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | - Dipshikha Chakravortty
- Department of Microbiology & Cell Biology, Indian Institute of Science, Bangalore 560012, India
- Adjunct Faculty, School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala 695551, India
| |
Collapse
|
2
|
McGill Percy KC, Liu Z, Qi X. Mitochondrial dysfunction in Alzheimer's disease: Guiding the path to targeted therapies. Neurotherapeutics 2025; 22:e00525. [PMID: 39827052 PMCID: PMC12047401 DOI: 10.1016/j.neurot.2025.e00525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration, marked by the accumulation of amyloid-β (Aβ) plaques and tau tangles. Emerging evidence suggests that mitochondrial dysfunction plays a pivotal role in AD pathogenesis, driven by impairments in mitochondrial quality control (MQC) mechanisms. MQC is crucial for maintaining mitochondrial integrity through processes such as proteostasis, mitochondrial dynamics, mitophagy, and precise communication with other subcellular organelles. In AD, disruptions in these processes lead to bioenergetic failure, gene dysregulation, the accumulation of damaged mitochondria, neuroinflammation, and lipid homeostasis impairment, further exacerbating neurodegeneration. This review elucidates the molecular pathways involved in MQC and their pathological relevance in AD, highlighting recent discoveries related to mitochondrial mechanisms underlying neurodegeneration. Furthermore, we explore potential therapeutic strategies targeting mitochondrial dysfunction, including gene therapy and pharmacological interventions, offering new avenues for slowing AD progression. The complex interplay between mitochondrial health and neurodegeneration underscores the need for innovative approaches to restore mitochondrial function and mitigate the onset and progression of AD.
Collapse
Affiliation(s)
- Kyle C McGill Percy
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Zunren Liu
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xin Qi
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Center for Mitochondrial Research and Therapeutics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
3
|
Son YS, Kwon YH, Lee MS, Kwon O, Jeong YJ, Mun SJ, Jeon S, Park JH, Han MH, Bae JS, Hur K, Jang AR, Park JH, Cho HS, Jung CR, Ryu CM, Son MJ, Park DS, Son MY. Helicobacter pylori VacA-induced mitochondrial damage in the gastric pit cells of the antrum and therapeutic rescue. Biomaterials 2025; 314:122842. [PMID: 39383778 DOI: 10.1016/j.biomaterials.2024.122842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/06/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
Exploring host cell specificity, pathogenicity, and molecular mechanisms of the vacuolating cytotoxin A (VacA), secreted by Helicobacter pylori (Hp) is crucial for developing novel treatment strategies. VacA affects subcellular events, particularly mitochondria, at a cell-type-specific level. However, the lack of reliable models that mimic VacA-induced subcellular damages and enable novel drug screening linked to the human stomach clinically limits our understanding of the mitochondrial networks in vivo. Here, human antrum gastric organoids (hAGOs) and tissue samples from Hp-infected patients were used to show the toxic effects of VacA-induced mitochondrial damage mainly in mucus-producing gastric pit cells by employing transcriptional, translational, and functional analyses. In VacA-intoxicated or Hp-infected hAGOs, robust mitochondrial fragmentation in gastric pit cells reduced ATP production during respiration, and loss of mucosal barrier integrity was first demonstrated experimentally. Using hAGOs, clinically relevant small molecules were screened for efficacy, and MLN8054, an Aurora kinase A inhibitor, reversed VacA-induced mitochondrial damage and loss of gastric epithelium integrity. MLN8054 was effective in VacA-treated and Hp-infected hAGOs and mice, highlighting hAGOs as a promising drug-screening model. These findings suggest that mitochondrial quality control may serve as a promising therapeutic target for Hp VacA-mediated toxicity and disease progression.
Collapse
Affiliation(s)
- Ye Seul Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Yong Hwan Kwon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Moo-Seung Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Ohman Kwon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Yu-Jin Jeong
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Seon Ju Mun
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Sojeong Jeon
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Ji Hye Park
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Man-Hoon Han
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Jae-Sung Bae
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Keun Hur
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Ah-Ra Jang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Soo Cho
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Cho-Rok Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Choong-Min Ryu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Myung Jin Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Doo-Sang Park
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea.
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea; KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, 34113, Republic of Korea; Department of Biological Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
4
|
Sun L, Huang J, Dou X, Dong Z, Li Y, Tan S, Yu R, Li C, Zhao W. Resveratrol Alleviates NEFA-Induced Oxidative Damage in Bovine Mammary Epithelial Cells by Restoring Mitochondrial Function. Animals (Basel) 2025; 15:118. [PMID: 39858118 PMCID: PMC11758345 DOI: 10.3390/ani15020118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
In periparturient dairy cows, high non-esterified fatty acids (NEFAs) caused by a severe negative energy balance induce oxidative stress and metabolic dysfunction, which pose a severe challenge to the dairy industry. Resveratrol (RES) is a polyphenolic compound with antioxidant, anti-inflammatory and multiple other physiological effects. However, its effect on oxidative damage triggered by NEFAs in bovine mammary epithelial cells is rarely reported. This study aimed to investigate the antioxidant effects and underlying molecular mechanisms of RES in NEFA-challenged BMECs. The results showed that RES ameliorated NEFA-induced oxidative damage by upregulating antioxidant enzyme expression and reducing malondialdehyde (MDA) and reactive oxygen species (ROS). Furthermore, exogenous NEFAs resulted in a decrease in mitochondrial membrane potential (MMP), cellular adenosine triphosphate (ATP) production, energy metabolism (NAD+/NADH ratio), abnormal mitochondrial structure and an increase in apoptosis levels. RES treatment restored mitochondrial function in NEFA-stressed BMECs, as evidenced by the increase in MMP, ATP generation and NAD+/NADH ratio accompanying the decline in mitochondrial structural abnormalities and cell apoptosis. In addition, in vivo studies in a mouse model of oxidative damage induced by high-fat diet (HFD) demonstrated that RES alleviated oxidative damage (decreased MDA content) and mitochondrial dysfunction (decreased expression of Drp1 and Fis1 and increased levels of Mfn2, Cyt C mRNA and ATP production) in mammary gland tissue. Overall, these findings suggested that RES could alleviate NEFA-induced oxidative damage in BMECs by modulating mitochondrial function, thereby contributing to the prevention and treatment of oxidative damage in perinatal dairy cows with a negative energy balance.
Collapse
Affiliation(s)
- Longwei Sun
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Junpeng Huang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Xiangyang Dou
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Zhenyu Dong
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yuan Li
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Shujing Tan
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Ran Yu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Chengmin Li
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Weiguo Zhao
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (L.S.); (J.H.); (X.D.); (Z.D.); (Y.L.); (S.T.); (R.Y.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
5
|
Mohareer K, Medikonda J, Yandrapally S, Agarwal A, Banerjee S. Monitoring the mitochondrial localization of mycobacterial proteins. Methods Cell Biol 2024; 194:135-149. [PMID: 40058957 DOI: 10.1016/bs.mcb.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Mitochondrion apart from being the energy hub of the cell, is also the center of various signaling pathways. During intracellular infection, either bacterial or viral, several pathogen proteins, metabolites, and possibly, lipids interact with the host mitochondria. These interactions allow the pathogens, such as Mtb, to reprogram the host mitochondrial functions, screwing the host immune responses and resulting in the persistence of the bacteria. Therefore, mitochondria offer a critical target organelle for various therapeutic interventions. This chapter deals with methods to demonstrate and establish the mitochondrial localization of Mtb proteins by confocal microscopy and mitochondrial enrichment. Transient transfection of mammalian constructs or infection Mycolicibacterium smegmatis infection could be used to overexpress the candidate Mtb protein in host cells, allowing the study of changes in the mitochondria's composition and function with regard to localization studies, mitochondrial DNA and RNA, proteomics, metabolomics, lipidomics, and bioenergetics.
Collapse
Affiliation(s)
- Krishnaveni Mohareer
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, India.
| | | | - Sriram Yandrapally
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, India
| | - Anushka Agarwal
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, India
| | - Sharmistha Banerjee
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, India.
| |
Collapse
|
6
|
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
|
7
|
Ramachandran RA, Abdallah JT, Rehman M, Baniasadi H, Blanton AM, Vizcaino S, Robertson DM. Pseudomonas aeruginosa impairs mitochondrial function and metabolism during infection of corneal epithelial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600521. [PMID: 38979356 PMCID: PMC11230238 DOI: 10.1101/2024.06.24.600521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Pseudomonas aeruginosa (PA) is a gram-negative opportunistic pathogen that can infect the cornea as a result of trauma or contact lens wear. In addition to their known energy producing role, mitochondria are important mediators of immune signaling and host defense. While certain pathogens have developed strategies to evade host defenses by modulating host mitochondrial dynamics and metabolism, the ability of PA to harness host cell mitochondria during corneal infection is unknown. Using a combination of biochemical and imaging techniques, we show that PA infection of corneal epithelial cells induced mitochondrial fission in a DRP1-dependent manner that preceded PINK1/Parkin and FUNDC1-mediated mitophagy. PA also impaired NADH-linked respiration through a reduction in complex 1. This corresponded to a decrease in metabolic pathways related to glycolysis and the TCA cycle. Metabolomics analysis further demonstrated an upregulation of the pentose phosphate pathway, arginine, purine, and pyrimidine metabolism in PA infected cells. These pathways may provide a key source of nucleotides, amino acids, and nitrogen for both the host cell and PA, in addition to antioxidant functions. Following treatment with gentamicin to kill all extracellular bacteria, metabolic flux analysis showed that corneal epithelial cells were able to restore mitochondrial function despite the continued presence of intracellular PA. Taken together, these data demonstrate that mitochondrial dysfunction and metabolic rewiring in host cells is triggered by extracellular PA, but once inside, PA requires healthy mitochondria to ensure host cell survival.
Collapse
|
8
|
Achi SC, McGrosso D, Tocci S, Ibeawuchi SR, Sayed IM, Gonzalez DJ, Das S. Proteome profiling identifies a link between the mitochondrial pathways and host-microbial sensor ELMO1 following Salmonella infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592405. [PMID: 38746404 PMCID: PMC11092768 DOI: 10.1101/2024.05.03.592405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The host EnguLfment and cell MOtility protein 1 (ELMO1) is a cytosolic microbial sensor that facilitates bacterial sensing, internalization, clearance, and inflammatory responses. We have shown previously that ELMO1 binds bacterial effector proteins, including pathogenic effectors from Salmonella and controls host innate immune signaling. To understand the ELMO1-regulated host pathways, we have performed liquid chromatography Multinotch MS3-Tandem Mass Tag (TMT) multiplexed proteomics to determine the global quantification of proteins regulated by ELMO1 in macrophages during Salmonella infection. Comparative proteome analysis of control and ELMO1-depleted murine J774 macrophages after Salmonella infection quantified more than 7000 proteins with a notable enrichment in mitochondrial-related proteins. Gene ontology enrichment analysis revealed 19 upregulated and 11 downregulated proteins exclusive to ELMO1-depleted cells during infection, belonging to mitochondrial functions, metabolism, vesicle transport, and the immune system. By assessing the cellular energetics via Seahorse analysis, we found that Salmonella infection alters mitochondrial metabolism, shifting it from oxidative phosphorylation to glycolysis. Importantly, these metabolic changes are significantly influenced by the depletion of ELMO1. Furthermore, ELMO1 depletion resulted in a decreased ATP rate index following Salmonella infection, indicating its importance in counteracting the effects of Salmonella on immunometabolism. Among the proteins involved in mitochondrial pathways, mitochondrial fission protein DRP1 was significantly upregulated in ELMO1-depleted cells and in ELMO1-KO mice intestine following Salmonella infection. Pharmacological Inhibition of DRP1 revealed the link of the ELMO1-DRP1 pathway in regulating the pro-inflammatory cytokine TNF-α following infection. The role of ELMO1 has been further characterized by a proteome profile of ELMO1-depleted macrophage infected with SifA mutant and showed the involvement of ELMO1-SifA on mitochondrial function, metabolism and host immune/defense responses. Collectively, these findings unveil a novel role for ELMO1 in modulating mitochondrial functions, potentially pivotal in modulating inflammatory responses. Significance Statement Host microbial sensing is critical in infection and inflammation. Among these sensors, ELMO1 has emerged as a key regulator, finely tuning innate immune signaling and discriminating between pathogenic and non-pathogenic bacteria through interactions with microbial effectors like SifA of Salmonella . In this study, we employed Multinotch MS3-Tandem Mass Tag (TMT) multiplexed proteomics to determine the proteome alterations mediated by ELMO1 in macrophages following WT and SifA mutant Salmonella infection. Our findings highlight a substantial enrichment of host proteins associated with metabolic pathways and mitochondrial functions. Notably, we validated the mitochondrial fission protein DRP1 that is upregulated in ELMO1-depleted macrophages and in ELMO1 knockout mice intestine after infection. Furthermore, we demonstrated that Salmonella -induced changes in cellular energetics are influenced by the presence of ELMO1. This work shed light on a possible novel link between mitochondrial dynamics and microbial sensing in modulating immune responses.
Collapse
|
9
|
Candeias E, Pereira-Santos AR, Empadinhas N, Cardoso SM, Esteves ARF. The Gut-Brain Axis in Alzheimer's and Parkinson's Diseases: The Catalytic Role of Mitochondria. J Alzheimers Dis 2024; 100:413-429. [PMID: 38875045 DOI: 10.3233/jad-240524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Accumulating evidence suggests that gut inflammation is implicated in neuroinflammation in Alzheimer's and Parkinson's diseases. Despite the numerous connections it remains unclear how the gut and the brain communicate and whether gut dysbiosis is the cause or consequence of these pathologies. Importantly, several reports highlight the importance of mitochondria in the gut-brain axis, as well as in mechanisms like gut epithelium self-renewal, differentiation, and homeostasis. Herein we comprehensively address the important role of mitochondria as a cellular hub in infection and inflammation and as a link between inflammation and neurodegeneration in the gut-brain axis. The role of mitochondria in gut homeostasis and as well the crosstalk between mitochondria and gut microbiota is discussed. Significantly, we also review studies highlighting how gut microbiota can ultimately affect the central nervous system. Overall, this review summarizes novel findings regarding this cross-talk where the mitochondria has a main role in the pathophysiology of both Alzheimer's and Parkinson's disease strengthen by cellular, animal and clinical studies.
Collapse
Affiliation(s)
- Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Pereira-Santos
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Fernandes Esteves
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
10
|
Kumar M, Sharma S, Mazumder S. Role of UPR mt and mitochondrial dynamics in host immunity: it takes two to tango. Front Cell Infect Microbiol 2023; 13:1135203. [PMID: 37260703 PMCID: PMC10227438 DOI: 10.3389/fcimb.2023.1135203] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/24/2023] [Indexed: 06/02/2023] Open
Abstract
The immune system of a host contains a group of heterogeneous cells with the prime aim of restraining pathogenic infection and maintaining homeostasis. Recent reports have proved that the various subtypes of immune cells exploit distinct metabolic programs for their functioning. Mitochondria are central signaling organelles regulating a range of cellular activities including metabolic reprogramming and immune homeostasis which eventually decree the immunological fate of the host under pathogenic stress. Emerging evidence suggests that following bacterial infection, innate immune cells undergo profound metabolic switching to restrain and countervail the bacterial pathogens, promote inflammation and restore tissue homeostasis. On the other hand, bacterial pathogens affect mitochondrial structure and functions to evade host immunity and influence their intracellular survival. Mitochondria employ several mechanisms to overcome bacterial stress of which mitochondrial UPR (UPRmt) and mitochondrial dynamics are critical. This review discusses the latest advances in our understanding of the immune functions of mitochondria against bacterial infection, particularly the mechanisms of mitochondrial UPRmt and mitochondrial dynamics and their involvement in host immunity.
Collapse
Affiliation(s)
- Manmohan Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, India
| |
Collapse
|
11
|
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
|
12
|
Rubio-Tomás T, Sotiriou A, Tavernarakis N. The interplay between selective types of (macro)autophagy: Mitophagy and xenophagy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:129-157. [PMID: 36858654 DOI: 10.1016/bs.ircmb.2022.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Autophagy is a physiological response, activated by a myriad of endogenous and exogenous cues, including DNA damage, perturbation of proteostasis, depletion of nutrients or oxygen and pathogen infection. Upon sensing those stimuli, cells employ multiple non-selective and selective autophagy pathways to promote fitness and survival. Importantly, there are a variety of selective types of autophagy. In this review we will focus on autophagy of bacteria (xenophagy) and autophagy of mitochondria (mitophagy). We provide a brief introduction to bulk autophagy, as well as xenophagy and mitophagy, highlighting their common molecular factors. We also describe the role of xenophagy and mitophagy in the detection and elimination of pathogens by the immune system and the adaptive mechanisms that some pathogens have developed through evolution to escape the host autophagic response. Finally, we summarize the recent articles (from the last five years) linking bulk autophagy with xenophagy and/or mitophagy in the context on developmental biology, cancer and metabolism.
Collapse
Affiliation(s)
- Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Aggeliki Sotiriou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece; Division of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece; Division of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete, Greece.
| |
Collapse
|
13
|
Mitochondrial dynamics in macrophages: divide to conquer or unite to survive? Biochem Soc Trans 2023; 51:41-56. [PMID: 36815717 PMCID: PMC9988003 DOI: 10.1042/bst20220014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/24/2023]
Abstract
Mitochondria have long been appreciated as the metabolic hub of cells. Emerging evidence also posits these organelles as hubs for innate immune signalling and activation, particularly in macrophages. Macrophages are front-line cellular defenders against endogenous and exogenous threats in mammals. These cells use an array of receptors and downstream signalling molecules to respond to a diverse range of stimuli, with mitochondrial biology implicated in many of these responses. Mitochondria have the capacity to both divide through mitochondrial fission and coalesce through mitochondrial fusion. Mitochondrial dynamics, the balance between fission and fusion, regulate many cellular functions, including innate immune pathways in macrophages. In these cells, mitochondrial fission has primarily been associated with pro-inflammatory responses and metabolic adaptation, so can be considered as a combative strategy utilised by immune cells. In contrast, mitochondrial fusion has a more protective role in limiting cell death under conditions of nutrient starvation. Hence, fusion can be viewed as a cellular survival strategy. Here we broadly review the role of mitochondria in macrophage functions, with a focus on how regulated mitochondrial dynamics control different functional responses in these cells.
Collapse
|
14
|
Roxas JL, Ramamurthy S, Cocchi K, Rutins I, Harishankar A, Agellon A, Wilbur JS, Sylejmani G, Vedantam G, Viswanathan V. Enteropathogenic Escherichia coli regulates host-cell mitochondrial morphology. Gut Microbes 2022; 14:2143224. [PMID: 36476073 PMCID: PMC9733699 DOI: 10.1080/19490976.2022.2143224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The diarrheagenic pathogen enteropathogenic Escherichia coli is responsible for significant childhood mortality and morbidity. EPEC and related attaching-and-effacing (A/E) pathogens use a type III secretion system to hierarchically deliver effector proteins into host cells and manipulate epithelial structure and function. Subversion of host mitochondrial biology is a key aspect of A/E pathogen virulence strategy, but the mechanisms remain poorly defined. We demonstrate that the early-secreted effector EspZ and the late-secreted effector EspH have contrasting effects on host mitochondrial structure and function. EspZ interacts with FIS1, a protein that induces mitochondrial fragmentation and mitophagy. Infection of epithelial cells with either wildtype EPEC or an isogenic espZ deletion mutant (ΔespZ) robustly upregulated FIS1 abundance, but a marked increase in mitochondrial fragmentation and mitophagy was seen only in ΔespZ-infected cells. FIS1-depleted cells were protected against ΔespZ-induced fission, and EspZ-expressing transfected epithelial cells were protected against pharmacologically induced mitochondrial fission and membrane potential disruption. Thus, EspZ interacts with FIS1 and blocks mitochondrial fragmentation and mitophagy. In contrast to WT EPEC, ΔespH-infected epithelial cells had minimal FIS1 upregulation and exhibited hyperfused mitochondria. Consistent with the contrasting impacts on organelle shape, mitochondrial membrane potential was preserved in ΔespH-infected cells, but profoundly disrupted in ΔespZ-infected cells. Collectively, our studies reveal hitherto unappreciated roles for two essential EPEC virulence factors in the temporal and dynamic regulation of host mitochondrial biology.
Collapse
Affiliation(s)
- Jennifer Lising Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Shylaja Ramamurthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Katie Cocchi
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Ilga Rutins
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Anusha Harishankar
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Al Agellon
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - John Scott Wilbur
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Gresa Sylejmani
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Gayatri Vedantam
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA,Department of Immunobiology, University of Arizona, Tucson, AZ, USA,BIO5 Institute for Collaborative Research, University of Arizona, Tucson, AZ, USA,Research Service, Southern Arizona VA Healthcare System, Tucson, AZ, USA
| | - V.K. Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA,Department of Immunobiology, University of Arizona, Tucson, AZ, USA,BIO5 Institute for Collaborative Research, University of Arizona, Tucson, AZ, USA,CONTACT V.K. Viswanathan School of Animal & Comparative Biomedical Sciences, the University of Arizona, Room 227, 1117 E. Lowell Street, Tucson, AZ85721, USA
| |
Collapse
|
15
|
Paredes-González IS, Aparicio-Trejo OE, Ramos-Espinosa O, López-Torres MO, Maya-Hoyos M, Mendoza-Trujillo M, Barrera-Rosales A, Mata-Espinosa D, León-Contreras JC, Pedraza-Chaverri J, Espitia C, Hernández-Pando R. Effect of mycobacterial proteins that target mitochondria on the alveolar macrophages activation during Mycobacterium tuberculosis infection. Exp Lung Res 2022; 48:251-265. [PMID: 36102603 DOI: 10.1080/01902148.2022.2120649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Purpose of the study: During the early and progressive (late) stages of murine experimental pulmonary tuberculosis, the differential activation of macrophages contributes to disease development by controlling bacterial growth and immune regulation. Mycobacterial proteins P27 and PE_PGRS33 can target the mitochondria of macrophages. This study aims to evaluate the effect of both proteins on macrophage activation during mycobacterial infection. Materials and methods: We assess both proteins for mitochondrial oxygen consumption, and morphological changes, as well as bactericide activity, production of metabolites, cytokines, and activation markers in infected MQs. The cell line MH-S was used for all the experiments. Results: We show that P27 and PE_PGRS33 proteins modified mitochondrial dynamics, oxygen consumption, bacilli growth, cytokine production, and some genes that contribute to macrophage alternative activation and mycobacterial intracellular survival. Conclusions: Our findings showed that these bacterial proteins partially contribute to promoting M2 differentiation by altering mitochondrial metabolic activity.
Collapse
Affiliation(s)
- Iris Selene Paredes-González
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City, Mexico
| | - Octavio Ramos-Espinosa
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Manuel Othoniel López-Torres
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Milena Maya-Hoyos
- Departamento de Química, Universidad Nacional de Colombia, Ciudad Universitaria, Bogota, Colombia
| | - Monserrat Mendoza-Trujillo
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Alejandra Barrera-Rosales
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Dulce Mata-Espinosa
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Juan Carlos León-Contreras
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Clara Espitia
- Departamento de Inmunología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rogelio Hernández-Pando
- División de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
16
|
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: 182] [Impact Index Per Article: 45.5] [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
|
17
|
Cervantes‐Silva MP, Cox SL, Curtis AM. Alterations in mitochondrial morphology as a key driver of immunity and host defence. EMBO Rep 2021; 22:e53086. [PMID: 34337844 PMCID: PMC8447557 DOI: 10.15252/embr.202153086] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are dynamic organelles whose architecture changes depending on the cell's energy requirements and other signalling events. These structural changes are collectively known as mitochondrial dynamics. Mitochondrial dynamics are crucial for cellular functions such as differentiation, energy production and cell death. Importantly, it has become clear in recent years that mitochondrial dynamics are a critical control point for immune cell function. Mitochondrial remodelling allows quiescent immune cells to rapidly change their metabolism and become activated, producing mediators, such as cytokines, chemokines and even metabolites to execute an effective immune response. The importance of mitochondrial dynamics in immunity is evident, as numerous pathogens have evolved mechanisms to manipulate host cell mitochondrial remodelling in order to promote their own survival. In this review, we comprehensively address the roles of mitochondrial dynamics in immune cell function, along with modulation of host cell mitochondrial morphology during viral and bacterial infections to facilitate either pathogen survival or host immunity. We also speculate on what the future may hold in terms of therapies targeting mitochondrial morphology for bacterial and viral control.
Collapse
Affiliation(s)
- Mariana P Cervantes‐Silva
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| | - Shannon L Cox
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| | - Annie M Curtis
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
18
|
Pei G, Dorhoi A. NOD-Like Receptors: Guards of Cellular Homeostasis Perturbation during Infection. Int J Mol Sci 2021; 22:ijms22136714. [PMID: 34201509 PMCID: PMC8268748 DOI: 10.3390/ijms22136714] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 12/30/2022] Open
Abstract
The innate immune system relies on families of pattern recognition receptors (PRRs) that detect distinct conserved molecular motifs from microbes to initiate antimicrobial responses. Activation of PRRs triggers a series of signaling cascades, leading to the release of pro-inflammatory cytokines, chemokines and antimicrobials, thereby contributing to the early host defense against microbes and regulating adaptive immunity. Additionally, PRRs can detect perturbation of cellular homeostasis caused by pathogens and fine-tune the immune responses. Among PRRs, nucleotide binding oligomerization domain (NOD)-like receptors (NLRs) have attracted particular interest in the context of cellular stress-induced inflammation during infection. Recently, mechanistic insights into the monitoring of cellular homeostasis perturbation by NLRs have been provided. We summarize the current knowledge about the disruption of cellular homeostasis by pathogens and focus on NLRs as innate immune sensors for its detection. We highlight the mechanisms employed by various pathogens to elicit cytoskeleton disruption, organelle stress as well as protein translation block, point out exemplary NLRs that guard cellular homeostasis during infection and introduce the concept of stress-associated molecular patterns (SAMPs). We postulate that integration of information about microbial patterns, danger signals, and SAMPs enables the innate immune system with adequate plasticity and precision in elaborating responses to microbes of variable virulence.
Collapse
Affiliation(s)
- Gang Pei
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany
- Correspondence: (G.P.); (A.D.)
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, 17489 Greifswald, Germany
- Correspondence: (G.P.); (A.D.)
| |
Collapse
|
19
|
Nandi I, Aroeti L, Ramachandran RP, Kassa EG, Zlotkin-Rivkin E, Aroeti B. Type III secreted effectors that target mitochondria. Cell Microbiol 2021; 23:e13352. [PMID: 33960116 DOI: 10.1111/cmi.13352] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 01/05/2023]
Abstract
A type III secretion system (T3SS) is used by Gram-negative bacterial pathogens to secrete and translocate a battery of proteins, termed effectors, from the bacteria directly into the host cells. These effectors, which are thought to play a key role in bacterial virulence, hijack and modify the activity of diverse host cell organelles, including mitochondria. Mitochondria-the energy powerhouse of the cell-are important cell organelles that play role in numerous critical cellular processes, including the initiation of apoptosis and the induction of innate immunity. Therefore, it is not surprising that pathogenic bacteria use mitochondrially targeted effectors to control host cell death and immunity pathways. Surprisingly, however, we found that despite their importance, only a limited number of type III secreted effectors have been characterised to target host mitochondria, and the mechanisms underlying their mitochondrial activity have not been sufficiently analysed. These include effectors secreted by the enteric attaching and effacing (A/E), Salmonella and Shigella bacterial pathogens. Here we give an overview of key findings, present gaps in knowledge and hypotheses concerning the mode by which these type III secreted effectors control the host and the bacterial cell life (and death) through targeting mitochondria.
Collapse
Affiliation(s)
- Ipsita Nandi
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lior Aroeti
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachana Pattani Ramachandran
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ephrem G Kassa
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Efrat Zlotkin-Rivkin
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Benjamin Aroeti
- Department of Cell and Developmental Biology, The Alexander Silberman Institute of Life Sciences, Safra Campus Givat Ram, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
20
|
Mancini NL, Rajeev S, Jayme TS, Wang A, Keita ÅV, Workentine ML, Hamed S, Söderholm JD, Lopes F, Shutt TE, Shearer J, McKay DM. Crohn's Disease Pathobiont Adherent-Invasive E coli Disrupts Epithelial Mitochondrial Networks With Implications for Gut Permeability. Cell Mol Gastroenterol Hepatol 2020; 11:551-571. [PMID: 32992049 PMCID: PMC7797367 DOI: 10.1016/j.jcmgh.2020.09.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Adherent-invasive Escherichia coli are implicated in inflammatory bowel disease, and mitochondrial dysfunction has been observed in biopsy specimens from patients with inflammatory bowel disease. As a novel aspect of adherent-invasive E coli-epithelial interaction, we hypothesized that E coli (strain LF82) would elicit substantial disruption of epithelial mitochondrial form and function. METHODS Monolayers of human colon-derived epithelial cell lines were exposed to E coli-LF82 or commensal E coli and RNA sequence analysis, mitochondrial function (adenosine triphosphate synthesis) and dynamics (mitochondrial network imaging, immunoblotting for fission and fusion proteins), and epithelial permeability (transepithelial resistance, flux of fluorescein isothiocyanate-dextran and bacteria) were assessed. RESULTS E coli-LF82 significantly affected epithelial expression of ∼8600 genes, many relating to mitochondrial function. E coli-LF82-infected epithelia showed swollen mitochondria, reduced mitochondrial membrane potential and adenosine triphosphate, and fragmentation of the mitochondrial network: events not observed with dead E coli-LF82, medium from bacterial cultures, or control E coli. Treatment with Mitochondrial Division Inhibitor 1 (Mdivi1, inhibits dynamin-related peptide 1, guanosine triphosphatase principally responsible for mitochondrial fission) or P110 (prevents dynamin-related peptide 1 binding to mitochondrial fission 1 protein) partially reduced E coli-LF82-induced mitochondrial fragmentation in the short term. E coli-LF82-infected epithelia showed loss of the long isoform of optic atrophy factor 1, which mediates mitochondrial fusion. Mitochondrial Division Inhibitor 1 reduced the magnitude of E coli-LF82-induced increased transepithelial flux of fluorescein isothiocyanate dextran. By 8 hours after infection, increased cytosolic cytochrome C and DNA fragmentation were apparent without evidence of caspase-3 or apoptosis inducing factor activation. CONCLUSIONS Epithelial mitochondrial fragmentation caused by E coli-LF82 could be targeted to maintain cellular homeostasis and mitigate infection-induced loss of epithelial barrier function. Data have been deposited in NCBI's Gene Expression Omnibus and are accessible through GEO series accession numbers GSE154121 and GSE154122 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE154121).
Collapse
Affiliation(s)
- Nicole L Mancini
- Gastrointestinal Research Group 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, Alberta, Canada
| | - Sruthi Rajeev
- Gastrointestinal Research Group 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, Alberta, Canada
| | - Timothy S Jayme
- Gastrointestinal Research Group 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, Alberta, Canada
| | - Arthur Wang
- Gastrointestinal Research Group 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, Alberta, Canada
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Samira Hamed
- Gastrointestinal Research Group 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, Alberta, Canada
| | - Johan D Söderholm
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Department of Surgery, County Council of Östergötland, Linköping, Sweden
| | - Fernando Lopes
- Institute of Parasitology, Faculty of Agriculture and Environmental Sciences, Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Timothy E Shutt
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, Alberta, Canada
| | - Jane Shearer
- Department of Biochemistry and Molecular Biology, Faculty of Kinesiology, University of Calgary, Alberta, Canada
| | - Derek M McKay
- Gastrointestinal Research Group 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, Alberta, Canada.
| |
Collapse
|
21
|
Khan S, Raj D, Jaiswal K, Lahiri A. Modulation of host mitochondrial dynamics during bacterial infection. Mitochondrion 2020; 53:140-149. [PMID: 32470613 DOI: 10.1016/j.mito.2020.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria is a dynamic organelle of the cell that can regulate and maintain cellular ATP level, ROS production, calcium signaling and immune response. In order to retain their shape and distribution, mitochondria go through coordinated cycles of fission and fusion. Further, dysfunctional mitochondria are selectively eliminated from the cell via mitophagy to synchronize mitochondrial quality control and cellular homeostasis. In addition, mitochondria when in close proximity with the endoplasmic reticulum can alter the signaling pathways and some recent findings also reveal a direct correlation between the mitochondrial localization in the cell to the immune response elicited against the invading pathogen. These modulations in the mitochondrial network are collectively termed as 'mitochondrial dynamics'. Diverse bacteria, virus and parasitic pathogens upon infecting a cell can alter the host mitochondrial dynamics in favor of their multiplication and this in turn can be a major determinant of the disease outcome. Pharmacological perturbations in these pathways thus could lead to generation of additional therapeutic opportunities. This review will focus on the pathogenic modulation of the host mitochondrial dynamics, specifically during the bacterial infections and describes how dysregulated mitochondrial dynamics facilitates the pathogen's ability to establish efficient infection.
Collapse
Affiliation(s)
- Shaziya Khan
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Desh Raj
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Kritika Jaiswal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amit Lahiri
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India.
| |
Collapse
|
22
|
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
|
23
|
Tiku V, Tan MW, Dikic I. Mitochondrial Functions in Infection and Immunity. Trends Cell Biol 2020; 30:263-275. [PMID: 32200805 PMCID: PMC7126537 DOI: 10.1016/j.tcb.2020.01.006] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/21/2022]
Abstract
Mitochondria have a central role in regulating a range of cellular activities and host responses upon bacterial infection. Multiple pathogens affect mitochondria dynamics and functions to influence their intracellular survival or evade host immunity. On the other side, major host responses elicited against infections are directly dependent on mitochondrial functions, thus placing mitochondria centrally in maintaining homeostasis upon infection. In this review, we summarize how different bacteria and viruses impact morphological and functional changes in host mitochondria and how this manipulation can influence microbial pathogenesis as well as the host cell metabolism and immune responses. Bacteria and viruses have evolved specific ways of targeting mitochondria to perturb mitochondrial function that can prove to be beneficial for these microbes. Many bacteria and viruses use specific virulence mechanisms to modulate mitochondrial dynamics, leading to either mitochondrial fusion or fission. Mitochondrial metabolism can also be impacted by bacterial and viral infections. While in some cases bacteria and viruses induce the mitochondrial cell death pathway, in others cell death is inhibited promoting intracellular bacterial and viral proliferation. Mitochondria regulate different innate immune signaling pathways induced upon bacterial or viral infections.
Collapse
Affiliation(s)
- Varnesh Tiku
- Department of Infectious Diseases, Genentech Inc, South San Francisco, USA
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech Inc, South San Francisco, USA.
| | - Ivan Dikic
- Department of Infectious Diseases, Genentech Inc, South San Francisco, USA; Institute for Biochemistry II. Goethe University Clinic, Frankfurt, Germany.
| |
Collapse
|
24
|
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
|
25
|
Bussi C, Gutierrez MG. Mycobacterium tuberculosis infection of host cells in space and time. FEMS Microbiol Rev 2019; 43:341-361. [PMID: 30916769 PMCID: PMC6606852 DOI: 10.1093/femsre/fuz006] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/26/2019] [Indexed: 12/16/2022] Open
Abstract
Tuberculosis (TB) caused by the bacterial pathogen Mycobacterium tuberculosis (Mtb) remains one of the deadliest infectious diseases with over a billion deaths in the past 200 years (Paulson 2013). TB causes more deaths worldwide than any other single infectious agent, with 10.4 million new cases and close to 1.7 million deaths in 2017. The obstacles that make TB hard to treat and eradicate are intrinsically linked to the intracellular lifestyle of Mtb. Mtb needs to replicate within human cells to disseminate to other individuals and cause disease. However, we still do not completely understand how Mtb manages to survive within eukaryotic cells and why some cells are able to eradicate this lethal pathogen. Here, we summarise the current knowledge of the complex host cell-pathogen interactions in TB and review the cellular mechanisms operating at the interface between Mtb and the human host cell, highlighting the technical and methodological challenges to investigating the cell biology of human host cell-Mtb interactions.
Collapse
Affiliation(s)
- Claudio Bussi
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Maximiliano G Gutierrez
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Two-in-one: UV radiation simultaneously induces apoptosis and NETosis. Cell Death Discov 2018; 4:51. [PMID: 29736268 PMCID: PMC5919968 DOI: 10.1038/s41420-018-0048-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/03/2018] [Accepted: 03/08/2018] [Indexed: 01/10/2023] Open
Abstract
NETosis is a unique form of neutrophil death that differs from apoptosis and necrosis. However, whether NETosis and apoptosis can occur simultaneously in the same neutrophil is unknown. In this paper, we show that increasing doses of ultraviolet (UV) irradiation increases NETosis, which is confirmed by myeloperoxidase colocalisation to neutrophil extracellular DNA. Increasing UV irradiation increases caspase 3 activation, mitochondrial reactive oxygen species (ROS) generation and p38, but not ERK, phosphorylation. Inhibition of mitochondrial ROS production and p38 activation, but not NADPH oxidase (NOX) activity, suppresses UV-induced NETosis, indicating that UV induces NOX-independent NETosis. Like classical NOX-dependent and -independent NETosis, UV-induced NETosis requires transcriptional firing for chromatin decondensation. Cell death-specific inhibitor studies indicate that UV-mediated NETosis is not apoptosis, necrosis or necroptosis. Collectively, these studies indicate that increasing doses of UV irradiation induce both apoptosis and NETosis simultaneously, but the ultimate outcome is the induction of a novel form of NOX-independent NETosis, or “ApoNETosis”.
Collapse
|
29
|
Kozjak-Pavlovic V, Chowdhury SR, Rudel T. Fragment and Conquer. Cell Host Microbe 2018; 22:255-257. [PMID: 28910630 DOI: 10.1016/j.chom.2017.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The replication vacuole of Legionella pneumophila makes contact with host mitochondria. In this issue of Cell Host & Microbe, Escoll et al. (2017) dissect the mechanisms of this interaction, the effect of the T4SS effector MitF on mitochondrial function, and the resultant metabolic reprogramming of infected cells to benefit the bacteria.
Collapse
Affiliation(s)
- Vera Kozjak-Pavlovic
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, D-97074 Wuerzburg, Germany
| | - Suvagata Roy Chowdhury
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, D-97074 Wuerzburg, Germany
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, D-97074 Wuerzburg, Germany.
| |
Collapse
|
30
|
Kozjak-Pavlovic V, Herweg JA, Rudel T. The role of host cell organelles in the development of Simkania negevensis. Int J Med Microbiol 2018; 308:155-160. [DOI: 10.1016/j.ijmm.2017.10.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/17/2017] [Accepted: 10/24/2017] [Indexed: 01/22/2023] Open
|
31
|
Torraca V, Mostowy S. Septins and Bacterial Infection. Front Cell Dev Biol 2016; 4:127. [PMID: 27891501 PMCID: PMC5104955 DOI: 10.3389/fcell.2016.00127] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/26/2016] [Indexed: 12/04/2022] Open
Abstract
Septins, a unique cytoskeletal component associated with cellular membranes, are increasingly recognized as having important roles in host defense against bacterial infection. A role for septins during invasion of Listeria monocytogenes into host cells was first proposed in 2002. Since then, work has shown that septins assemble in response to a wide variety of invasive bacterial pathogens, and septin assemblies can have different roles during the bacterial infection process. Here we review the interplay between septins and bacterial pathogens, highlighting septins as a structural determinant of host defense. We also discuss how investigation of septin assembly in response to bacterial infection can yield insight into basic cellular processes including phagocytosis, autophagy, and mitochondrial dynamics.
Collapse
Affiliation(s)
- Vincenzo Torraca
- Department of Medicine, MRC Centre of Molecular Bacteriology and Infection, Imperial College London London, UK
| | - Serge Mostowy
- Department of Medicine, MRC Centre of Molecular Bacteriology and Infection, Imperial College London London, UK
| |
Collapse
|
32
|
Dengue virus induces mitochondrial elongation through impairment of Drp1-triggered mitochondrial fission. Virology 2016; 500:149-160. [PMID: 27816895 DOI: 10.1016/j.virol.2016.10.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 02/08/2023]
Abstract
Mitochondria are highly dynamic organelles that undergo continuous cycles of fission and fusion to maintain essential cellular functions. An imbalance between these two processes can result in many pathophysiological outcomes. Dengue virus (DENV) interacts with cellular organelles, including mitochondria, to successfully replicate in cells. This study used live-cell imaging and found an increase in mitochondrial length and respiration during DENV infection. The level of mitochondrial fission protein, Dynamin-related protein 1 (Drp1), was decreased on mitochondria during DENV infection, as well as Drp1 phosphorylated on serine 616, which is important for mitochondrial fission. DENV proteins NS4b and NS3 were also associated with subcellular fractions of mitochondria. Induction of fission through uncoupling of mitochondria or overexpression of Drp1 wild-type and Drp1 with a phosphomimetic mutation (S616D) significantly reduced viral replication. These results demonstrate that DENV infection causes an imbalance in mitochondrial dynamics by inhibiting Drp1-triggered mitochondrial fission, which promotes viral replication.
Collapse
|
33
|
Sirianni A, Krokowski S, Lobato-Márquez D, Buranyi S, Pfanzelter J, Galea D, Willis A, Culley S, Henriques R, Larrouy-Maumus G, Hollinshead M, Sancho-Shimizu V, Way M, Mostowy S. Mitochondria mediate septin cage assembly to promote autophagy of Shigella. EMBO Rep 2016; 17:1029-43. [PMID: 27259462 PMCID: PMC4931556 DOI: 10.15252/embr.201541832] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 05/04/2016] [Indexed: 11/30/2022] Open
Abstract
Septins, cytoskeletal proteins with well‐characterised roles in cytokinesis, form cage‐like structures around cytosolic Shigella flexneri and promote their targeting to autophagosomes. However, the processes underlying septin cage assembly, and whether they influence S. flexneri proliferation, remain to be established. Using single‐cell analysis, we show that the septin cages inhibit S. flexneri proliferation. To study mechanisms of septin cage assembly, we used proteomics and found mitochondrial proteins associate with septins in S. flexneri‐infected cells. Strikingly, mitochondria associated with S. flexneri promote septin assembly into cages that entrap bacteria for autophagy. We demonstrate that the cytosolic GTPase dynamin‐related protein 1 (Drp1) interacts with septins to enhance mitochondrial fission. To avoid autophagy, actin‐polymerising Shigella fragment mitochondria to escape from septin caging. Our results demonstrate a role for mitochondria in anti‐Shigella autophagy and uncover a fundamental link between septin assembly and mitochondria.
Collapse
Affiliation(s)
- Andrea Sirianni
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Sina Krokowski
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Damián Lobato-Márquez
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Stephen Buranyi
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Julia Pfanzelter
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK
| | - Dieter Galea
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Alexandra Willis
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Siân Culley
- Quantitative Imaging and NanoBiophysics Group, MRC Laboratory for Molecular Cell Biology, Department of Cell and Developmental Biology, University College London, London, UK
| | - Ricardo Henriques
- Quantitative Imaging and NanoBiophysics Group, MRC Laboratory for Molecular Cell Biology, Department of Cell and Developmental Biology, University College London, London, UK
| | - Gerald Larrouy-Maumus
- Faculty of Natural Sciences, Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | | | - Vanessa Sancho-Shimizu
- Section of Virology, St. Mary's Medical School, Imperial College London, London, UK Section of Paediatrics, St. Mary's Medical School, Imperial College London, London, UK
| | - Michael Way
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London, UK Section of Virology, St. Mary's Medical School, Imperial College London, London, UK
| | - Serge Mostowy
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| |
Collapse
|
34
|
Pernas L, Scorrano L. Mito-Morphosis: Mitochondrial Fusion, Fission, and Cristae Remodeling as Key Mediators of Cellular Function. Annu Rev Physiol 2015; 78:505-31. [PMID: 26667075 DOI: 10.1146/annurev-physiol-021115-105011] [Citation(s) in RCA: 568] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Permanent residency in the eukaryotic cell pressured the prokaryotic mitochondrial ancestor to strategize for intracellular living. Mitochondria are able to autonomously integrate and respond to cellular cues and demands by remodeling their morphology. These processes define mitochondrial dynamics and inextricably link the fate of the mitochondrion and that of the host eukaryote, as exemplified by the human diseases that result from mutations in mitochondrial dynamics proteins. In this review, we delineate the architecture of mitochondria and define the mechanisms by which they modify their shape. Key players in these mechanisms are discussed, along with their role in manipulating mitochondrial morphology during cellular action and development. Throughout, we highlight the evolutionary context in which mitochondrial dynamics emerged and consider unanswered questions whose dissection might lead to mitochondrial morphology-based therapies.
Collapse
Affiliation(s)
- Lena Pernas
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; ,
| | - Luca Scorrano
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; ,
| |
Collapse
|