1
|
Wu YC, Beets I, Fox BW, Fajardo Palomino D, Chen L, Liao CP, Vandewyer E, Lin LY, He CW, Chen LT, Lin CT, Schroeder FC, Pan CL. Intercellular sphingolipid signaling mediates aversive learning in C. elegans. Curr Biol 2025; 35:2323-2336.e9. [PMID: 40252647 DOI: 10.1016/j.cub.2025.03.082] [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: 12/10/2024] [Revised: 03/02/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025]
Abstract
Physiological stress in non-neural tissues drives aversive learning for sensory cues associated with stress. However, the identities of signals derived from non-neural tissues and the mechanisms by which these signals mediate aversive learning remain elusive. Here, we show that intercellular sphingolipid signaling contributes to aversive learning under mitochondrial stress in C. elegans. We found that stress-induced aversive learning requires sphingosine kinase, SPHK-1, the enzyme that produces sphingosine-1-phosphate (S1P). Genetic and biochemical studies revealed an intercellular signaling pathway in which intestinal or hypodermal SPHK-1 signals through the neuronal G protein-coupled receptor, SPHR-1, and modulates responses of the octopaminergic RIC neuron to promote aversive learning. We further show that SPHK-1-mediated sphingolipid signaling is required for learned aversion of Chryseobacterium indologenes, a bacterial pathogen found in the natural habitats of C. elegans, which causes mitochondrial stress. Taken together, our work reveals a sphingolipid signaling pathway that communicates from intestinal or hypodermal tissues to neurons to promote aversive learning in response to mitochondrial stress and pathogen infection.
Collapse
Affiliation(s)
- Yu-Chun Wu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Isabel Beets
- Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, Isabel Beets, Leuven, Belgium
| | - Bennett William Fox
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, 533 Tower Road, Ithaca, NY 14853, USA
| | - Diana Fajardo Palomino
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, 533 Tower Road, Ithaca, NY 14853, USA
| | - Li Chen
- Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, Isabel Beets, Leuven, Belgium
| | - Chien-Po Liao
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Elke Vandewyer
- Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, Isabel Beets, Leuven, Belgium
| | - Liang-Yi Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chun-Wei He
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Li-Tzu Chen
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chih-Ta Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, 533 Tower Road, Ithaca, NY 14853, USA
| | - Chun-Liang Pan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan; Center for Precision Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan.
| |
Collapse
|
2
|
Inan S, Wilson RP, Tükel Ç. IUPHAR review: From gut to brain: The role of gut dysbiosis, bacterial amyloids, and metabolic disease in Alzheimer's disease. Pharmacol Res 2025; 215:107693. [PMID: 40086611 DOI: 10.1016/j.phrs.2025.107693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Gut microbial dysbiosis, or altered gut microbial communities, in Alzheimer's Disease suggests a pathogenic role for gut inflammation and microbial products in shaping a neuroinflammatory environment. Similarly, metabolic diseases, such as obesity and diabetes, are also associated with an increased risk of Alzheimer's Disease. As the metabolic landscape shifts during gut inflammation, and gut inflammation in turn impacts metabolic processes, we explore how these interconnected pathways may contribute to the progression of Alzheimer's Disease. Additionally, we discuss the role of bacterial amyloids produced by gut microbes, which may exacerbate amyloid aggregation in the brain and contribute to neurodegenerative processes. Furthermore, we highlight potential therapeutic strategies aimed at reducing gut inflammation, improving metabolic health, and decreasing amyloid content as a means to mitigate Alzheimer's Disease progression. These approaches, targeting the gut-brain-metabolic axis, could offer promising avenues for delaying or preventing cognitive decline in affected individuals.
Collapse
Affiliation(s)
- Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| | - R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Nevoit G, Jarusevicius G, Potyazhenko M, Mintser O, Bumblyte IA, Vainoras A. Mitochondrial Dysfunction and Atherosclerosis: The Problem and the Search for Its Solution. Biomedicines 2025; 13:963. [PMID: 40299559 PMCID: PMC12024619 DOI: 10.3390/biomedicines13040963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objectives: This review has been prepared to promote interest in the interdisciplinary study of mitochondrial dysfunction (MD) and atherosclerosis. This review aims to describe the state of this problem and indicate the direction for further implementation of this knowledge in clinical medicine. Methods: Extensive research of the literature was implemented to elucidate the role of the molecular mechanisms of MD in the pathogenesis of atherosclerosis. Results: A view on the pathogenesis of atherosclerosis through the prism of knowledge about MD is presented. MD is the cause and primary mechanism of the onset and progression of atherosclerosis. It is proposed that this problem be considered in the context of a continuum. Conclusions: MD and atherosclerosis are united by common molecular mechanisms of pathogenesis. Knowledge of MD should be used to argue for a healthy lifestyle as the primary way to prevent atherosclerosis. The development of new approaches to diagnosing and treating MD in atherosclerosis is an urgent task and challenge for modern science.
Collapse
Affiliation(s)
- Ganna Nevoit
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Gediminas Jarusevicius
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Maksim Potyazhenko
- Department of Internal Medicine and Emergency Medicine, Poltava State Medical University, 36011 Poltava, Ukraine
| | - Ozar Mintser
- Department of Fundamental Disciplines and Informatics, Shupyk National Healthcare University of Ukraine, 04112 Kyiv, Ukraine
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| | - Alfonsas Vainoras
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
4
|
Scheckhuber CQ, Maciver SK, de Obeso Fernandez del Valle A. Unveiling the molecular architecture of the mitochondrial respiratory chain of Acanthamoeba castellanii. MICROBIAL CELL (GRAZ, AUSTRIA) 2025; 12:65-75. [PMID: 40302930 PMCID: PMC12040293 DOI: 10.15698/mic2025.03.846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 05/02/2025]
Abstract
Acanthamoeba castellanii is a ubiquitous free-living amoeba that can cause severe infections in humans. Unlike most other organisms, A. castellanii possesses a "complete" mitochondrial respiratory chain, meaning it con-tains several additional enzymes that contribute to its metabolic versa-tility and survival in diverse environments. This review provides a com-prehensive overview of the mitochondrial respiratory chain in A. castellanii, focusing on the key alternative components in-volved in oxidative phosphorylation and their roles in energy metabo-lism, stress response, and adaptation to various conditions. The func-tional characterization of the alternative oxidase (AOX), uncoupling pro-tein (UCP), and alternative NAD(P)H dehydrogenases, highlight their roles in reducing oxidative stress, modulating proton gradients, and adapting to changes in temperature and nutrient availability. These pro-teins and systems serve a role in the survival of A. castel-lanii under stressful conditions such as starvation and cold con-ditions. Further knowledge of the respiratory chain of the amoeba has potential implications for understanding the evolution of mitochondrial respiration and developing new therapies for treating Acanthamoeba infections.
Collapse
Affiliation(s)
- Christian Q. Scheckhuber
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, 64849, Monterrey, N.L, Mexico
| | - Sutherland K. Maciver
- Centre for Discovery Brain Sciences, Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, Scotland, UK
| | | |
Collapse
|
5
|
Frandsen JR, Yuan Z, Bedi B, Prasla Z, Choi SR, Narayanasamy P, Sadikot RT. PGC-1α activation to enhance macrophage immune function in mycobacterial infections. PLoS One 2025; 20:e0310908. [PMID: 39913377 PMCID: PMC11801632 DOI: 10.1371/journal.pone.0310908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 09/09/2024] [Indexed: 02/09/2025] Open
Abstract
Nontuberculous Mycobacteria (NTM) are a heterogeneous group of environmental microorganisms with distinct human pathogenesis. Their incidence and prevalence are rising worldwide, due in part to elevated antimicrobial resistance which complicates treatment and potential successful outcomes. Although information exists on the clinical significance of NTMs, little is known about host immune response to infection. NTM infections alter macrophage mitochondrial capacity and decrease ATP production, efficient immune response, and bacterial clearance. Transcription factor peroxisome proliferator activated receptor (PPAR) γ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis, influencing metabolism, mitochondrial pathways, and antioxidant response. Mitochondrial transcription factor A (TFAM) is a protein essential for mitochondrial DNA (mtDNA) genome stability, integrity, and metabolism. Both PGC-1α and TFAM regulate mitochondrial biogenesis and activity, and their disruption is linked to inflammatory signaling and altered macrophage function. We show that NTM causes macrophage mitochondrial damage and disrupted bioenergetics. Mechanistically we show that this is related to attenuation of expression of PGC-1α and TFAM in infected macrophages. Importantly, rescuing expression of PGC-1α and TFAM using pharmacologic approaches restored macrophage immune function. Our results suggest that pharmacologic approaches to enhance mitochondrial function provide a novel approach to target macrophage immune function and means to combat NTM infections.
Collapse
Affiliation(s)
- Joel R. Frandsen
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Zhihong Yuan
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Brahmchetna Bedi
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Zohra Prasla
- Pulmonology and Critical Care Department, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Seoung-Ryoung Choi
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Prabagaran Narayanasamy
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ruxana T. Sadikot
- Division of Pulmonary, Critical Care & Sleep, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
6
|
Nevoit G, Jarusevicius G, Potyazhenko M, Mintser O, Bumblyte IA, Vainoras A. Mitochondrial Dysfunction and Risk Factors for Noncommunicable Diseases: From Basic Concepts to Future Prospective. Diseases 2024; 12:277. [PMID: 39589951 PMCID: PMC11592525 DOI: 10.3390/diseases12110277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Noncommunicable diseases (NCDs) are a very important medical problem. The key role of mitochondrial dysfunction (MD) in the occurrence and progression of NCDs has been proven. However, the etiology and pathogenesis of MD itself in many NCDs has not yet been clarified, which makes it one of the most serious medical problems in the modern world, according to many scientists. METHODS An extensive research in the literature was implemented in order to elucidate the role of MD and NCDs' risk factors in the pathogenesis of NCDs. RESULTS The authors propose to take a broader look at the problem of the pathogenesis of NCDs. It is important to understand exactly how NCD risk factors lead to MD. The review is structured in such a way as to answer this question. Based on a systematic analysis of scientific data, a theoretical concept of modern views on the occurrence of MD under the influence of risk factors for the occurrence of NCDs is presented. This was done in order to update MD issues in clinical medicine. MD and NCDs progress throughout a patient's life. Based on this, the review raised the question of the existence of an NCDs continuum. CONCLUSIONS MD is a universal mechanism that causes organ dysfunction and comorbidity of NCDs. Prevention of MD involves diagnosing and eliminating the factors that cause it. Mitochondria are an important therapeutic target.
Collapse
Affiliation(s)
- Ganna Nevoit
- Laboratory of Population Studies, Cardiology Institute, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Gediminas Jarusevicius
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| | - Maksim Potyazhenko
- Department of Internal Medicine and Emergency Medicine, Poltava State Medical University, 36011 Poltava, Ukraine;
| | - Ozar Mintser
- Department of Fundamental Disciplines and Informatics, Shupyk National Healthcare University of Ukraine, 04112 Kyiv, Ukraine;
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| | - Alfonsas Vainoras
- Laboratory for Automatization of Cardiovascular Investigations, Cardiology Institute, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania;
| |
Collapse
|
7
|
Chatterjee S, Sil PC. Mechanistic Insights into Toxicity of Titanium Dioxide Nanoparticles at the Micro- and Macro-levels. Chem Res Toxicol 2024; 37:1612-1633. [PMID: 39324438 DOI: 10.1021/acs.chemrestox.4c00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Titanium oxide nanoparticles (TiO2 NPs) have been regarded as a legacy nanomaterial due to their widespread usage across multiple fields. The TiO2 NPs have been and are still extensively used as a food and cosmetic additive and in wastewater and sewage treatment, paints, and industrial catalysis as ultrafine TiO2. Recent developments in nanotechnology have catapulted it into a potent antibacterial and anticancer agent due to its excellent photocatalytic potential that generates substantial amounts of highly reactive oxygen radicals. The method of production, surface modifications, and especially size impact its toxicity in biological systems. The anatase form of TiO2 (<30 nm) has been found to exert better and more potent cytotoxicity in bacteria as well as cancer cells than other forms. However, owing to the very small size, anatase particles are able to penetrate deep tissue easily; hence, they have also been implicated in inflammatory reactions and even as a potent oncogenic substance. Additionally, TiO2 NPs have been investigated to assess their toxicity to large-scale ecosystems owing to their excellent reactive oxygen species (ROS)-generating potential compounded with widespread usage over decades. This review discusses in detail the mechanisms by which TiO2 NPs induce toxic effects on microorganisms, including bacteria and fungi, as well as in cancer cells. It also attempts to shed light on how and why it is so prevalent in our lives and by what mechanisms it could potentially affect the environment on a larger scale.
Collapse
Affiliation(s)
- Sharmistha Chatterjee
- Division of Molecular Medicine, Bose Institute, P 1/12, CIT Scheme VIIM, Kankurgachi, Kolkata-700054, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, P 1/12, CIT Scheme VIIM, Kankurgachi, Kolkata-700054, India
| |
Collapse
|
8
|
Rong J, Yuan C, Yin X, Wu X, He F, Wang Y, Leung KSY, Lin S. Co-exposure of polystyrene nanoplastics and copper induces development toxicity and intestinal mitochondrial dysfunction in vivo and in vitro. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 930:172681. [PMID: 38663618 DOI: 10.1016/j.scitotenv.2024.172681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/02/2024]
Abstract
Nanoplastics (NPs) have raised concerns about the combined toxicity to living organisms due to their ability to adsorb heavy metals. There is still uncertainty, however, whether NPs combined with heavy metals exert adverse effects on intestinal microenvironment, especially the intestinal cells and microbiota. Herein, the combined effects of 500 nm spherical-shaped polystyrene nanoplastics (PSNPs) and copper ions (Cu2+) on intestinal cells and gut microbiota were assessed using HCT-116 cells and zebrafish models. The combined exposure of PSNPs (10 mg/L) and Cu2+ (0.5 mg/L) induced more severer hatching interference of zebrafish embryos, deformation, and mortality. In larval stage, PSNPs (10 mg/L) accumulated and carried more Cu2+ in the gastrointestinal tract (GIT) of zebrafish after co-exposure for 5 days. Excessive neutrophil recruitment and oxidative stress in GIT of zebrafish larvae were observed. The mechanism of the combined toxicity was revealed by transmission electron microscopy (TEM) showing the injuries of GIT, transcriptome and 16S rDNA gene sequencing showing the toxicity pathways, including oxidative phosphorylation and respiratory electron transport chain, as well as microbial community analysis showing the induced microbiota dysbiosis. In vitro tests using HCT-116 cells showed that PSNPs (10 mg/L) and Cu2+ (0.5 mg/L) increased cell death while decreasing ATP concentration and mitochondrial membrane potential after 48 h exposure. These findings may provide new insights into the combined toxicity of nanoplastics and heavy metals in the intestinal microenvironment.
Collapse
Affiliation(s)
- Jinyu Rong
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Key Laboratory of Yangtze River Water Environment, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, Shanghai 200092, China
| | - Chenwei Yuan
- Department of Breast Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Xiang Yin
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Key Laboratory of Yangtze River Water Environment, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, Shanghai 200092, China
| | - Xiaohan Wu
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Key Laboratory of Yangtze River Water Environment, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, Shanghai 200092, China
| | - Fei He
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Key Laboratory of Yangtze River Water Environment, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, Shanghai 200092, China
| | - Yixin Wang
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Key Laboratory of Yangtze River Water Environment, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, Shanghai 200092, China
| | - Kelvin Sze-Yin Leung
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong Special Administrative Region, China; HKBU Institute of Research and Continuing Education, Shenzhen Virtual University Park, Shenzhen, China.
| | - Sijie Lin
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China; Key Laboratory of Yangtze River Water Environment, Shanghai Institute of Pollution Control and Ecological Security, Tongji University, Shanghai 200092, China.
| |
Collapse
|
9
|
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
|
10
|
Pereira-Santos AR, Candeias E, Magalhães JD, Empadinhas N, Esteves AR, Cardoso SM. Neuronal control of microglia through the mitochondria. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167167. [PMID: 38626829 DOI: 10.1016/j.bbadis.2024.167167] [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: 02/23/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
The microbial toxin β-N-methylamino-L-alanine (BMAA), which is derived from cyanobacteria, targets neuronal mitochondria, leading to the activation of neuronal innate immunity and, consequently, neurodegeneration. Although known to modulate brain inflammation, the precise role of aberrant microglial function in the neurodegenerative process remains elusive. To determine if neurons signal microglial cells, we treated primary cortical neurons with BMAA and then co-cultured them with the N9 microglial cell line. Our observations indicate that microglial cell activation requires initial neuronal priming. Contrary to what was observed in cortical neurons, BMAA was not able to activate inflammatory pathways in N9 cells. We observed that microglial activation is dependent on mitochondrial dysfunction signaled by BMAA-treated neurons. In this scenario, the NLRP3 pro-inflammatory pathway is activated due to mitochondrial impairment in N9 cells. These results demonstrate that microglia activation in the presence of BMAA is dependent on neuronal signaling. This study provides evidence that neurons may trigger microglia activation and subsequent neuroinflammation. In addition, we demonstrate that microglial activation may have a protective role in ameliorating neuronal innate immune activation, at least in the initial phase. This work challenges the current understanding of neuroinflammation by assigning the primary role to neurons.
Collapse
Affiliation(s)
- A R Pereira-Santos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - J D Magalhães
- CNC-Center for Neuroscience and Cell Biology, 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, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - A Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sandra M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
11
|
Nasser F, Oke MT, Knezevic S, D'Costa VM. Bacterial Pathogenesis: Assessment of Intracellular Positioning of Pathogen-Containing Vacuoles During Infection. Curr Protoc 2024; 4:e1021. [PMID: 38619090 DOI: 10.1002/cpz1.1021] [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: 04/16/2024]
Abstract
Intracellular bacterial pathogens implement a diverse array of strategies to target host cells and establish infection. For vacuolar pathogens, the process of pathogen-containing vacuole movement within host cells, termed intracellular trafficking, is central to both pathogen survival and infection progression. Typically a process mediated by secreted virulence factors that manipulate the host cytoskeletal machinery, internalized pathogen-containing vacuoles traffic to the site of replication to establish a unique replicative niche, and if applicable, traffic back toward the host cell periphery for cell-to-cell spread. As such, the intracellular positioning of pathogen-containing vacuoles represents a fundamental measure of infection progression. Here, we describe a fluorescence microscopy-based method to quantitatively assess bacterial intracellular positioning, using Salmonella enterica serovar Typhimurium infection of epithelial cells as a model. This experimental approach can be modified to study infection in diverse host cell types, and with a broad array of pathogens. The system can also be adapted to examine the kinetics of infection, identify secreted virulence factors that mediate host trafficking, investigate host factors that are targeted by the pathogen for trafficking, and assess functional domains within a virulence factor responsible for mediating the phenotype. Collectively, these tools can provide fundamental insight into the pathogenesis of a diverse array of intracellular bacterial pathogens, and new host factors that are hijacked to mediate infection. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Culture and preparation of host cells Alternate Protocol: Culture and preparation of host cells to assess host factor contribution to bacterial positioning Basic Protocol 2: Infection of epithelial cells with S. Typhimurium Basic Protocol 3: Fluorescence staining for analysis of bacterial positioning Basic Protocol 4: Fluorescence microscopy analysis of bacterial positioning.
Collapse
Affiliation(s)
- Farah Nasser
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
| | - Mosopefoluwa T Oke
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
- These authors contributed equally to this work
| | - Sara Knezevic
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
- These authors contributed equally to this work
| | - Vanessa M D'Costa
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, Canada
| |
Collapse
|
12
|
Mocăniță M, Martz K, D’Costa VM. Bacterial Pathogen-Mediated Suppression of Host Trafficking to Lysosomes: Fluorescence Microscopy-Based DQ-Red BSA Analysis. Bio Protoc 2024; 14:e4951. [PMID: 38464940 PMCID: PMC10917698 DOI: 10.21769/bioprotoc.4951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/04/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Intracellular bacterial pathogens have evolved to be adept at manipulating host cellular function for the benefit of the pathogen, often by means of secreted virulence factors that target host pathways for modulation. The lysosomal pathway is an essential cellular response pathway to intracellular pathogens and, as such, represents a common target for bacterial-mediated evasion. Here, we describe a method to quantitatively assess bacterial pathogen-mediated suppression of host cell trafficking to lysosomes, using Salmonella enterica serovar Typhimurium infection of epithelial cells as a model. This live-cell imaging assay involves the use of a BODIPY TR-X conjugate of BSA (DQ-Red BSA) that traffics to and fluoresces in functional lysosomes. This method can be adapted to study infection with a broad array of pathogens in diverse host cell types. It is capable of being applied to identify secreted virulence factors responsible for a phenotype of interest as well as domains within the bacterial protein that are important for mediating the phenotype. Collectively, these tools can provide invaluable insight into the mechanisms of pathogenesis of a diverse array of pathogenic bacteria, with the potential to uncover virulence factors that may be suitable targets for therapeutic intervention. Key features • Infection-based analysis of bacterial-mediated suppression of host trafficking to lysosomes, using Salmonella enterica serovar Typhimurium infection of human epithelial cells as a model. • Live microscopy-based analysis allows for the visualization of individually infected host cells and is amenable to phenotype quantification. • Assay can be adapted to a broad array of pathogens and diverse host cell types. • Assay can identify virulence factors mediating a phenotype and protein domains that mediate a phenotype.
Collapse
Affiliation(s)
- Mădălina Mocăniță
- Department of Biochemistry, Microbiology and
Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation,
University of Ottawa, Ottawa, ON, Canada
| | - Kailey Martz
- Department of Biochemistry, Microbiology and
Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation,
University of Ottawa, Ottawa, ON, Canada
| | - Vanessa M. D’Costa
- Department of Biochemistry, Microbiology and
Immunology, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation,
University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
13
|
Dodge JD, Browder NJ, Pellegrino MW. Mitochondrial recovery by the UPR mt: Insights from C. elegans. Semin Cell Dev Biol 2024; 154:59-68. [PMID: 36792440 PMCID: PMC11684877 DOI: 10.1016/j.semcdb.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/15/2023]
Abstract
Mitochondria are multifaceted organelles, with such functions as the production of cellular energy to the regulation of cell death. However, mitochondria incur various sources of damage from the accumulation of reactive oxygen species and DNA mutations that can impact the protein folding environment and impair their function. Since mitochondrial dysfunction is often associated with reductions in organismal fitness and possibly disease, cells must have safeguards in place to protect mitochondrial function and promote recovery during times of stress. The mitochondrial unfolded protein response (UPRmt) is a transcriptional adaptation that promotes mitochondrial repair to aid in cell survival during stress. While the earlier discoveries into the regulation of the UPRmt stemmed from studies using mammalian cell culture, much of our understanding about this stress response has been bestowed to us by the model organism Caenorhabditis elegans. Indeed, the facile but powerful genetics of this relatively simple nematode has uncovered multiple regulators of the UPRmt, as well as several physiological roles of this stress response. In this review, we will summarize these major advancements originating from studies using C. elegans.
Collapse
Affiliation(s)
- Joshua D Dodge
- The University of Texas at Arlington, Department of Biology, Arlington, TX 76019, USA
| | - Nicholas J Browder
- The University of Texas at Arlington, Department of Biology, Arlington, TX 76019, USA
| | - Mark W Pellegrino
- The University of Texas at Arlington, Department of Biology, Arlington, TX 76019, USA.
| |
Collapse
|
14
|
Mirza Z, Walhout AJM, Ambros V. A bacterial pathogen induces developmental slowing by high reactive oxygen species and mitochondrial dysfunction in Caenorhabditis elegans. Cell Rep 2023; 42:113189. [PMID: 37801396 PMCID: PMC10929622 DOI: 10.1016/j.celrep.2023.113189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/19/2023] [Accepted: 09/14/2023] [Indexed: 10/08/2023] Open
Abstract
Host-pathogen interactions are complex by nature, and the host developmental stage increases this complexity. By utilizing Caenorhabditis elegans larvae as the host and the bacterium Pseudomonas aeruginosa as the pathogen, we investigated how a developing organism copes with pathogenic stress. By screening 36 P. aeruginosa isolates, we found that the CF18 strain causes a severe but reversible developmental delay via induction of reactive oxygen species (ROS) and mitochondrial dysfunction. While the larvae upregulate mitophagy, antimicrobial, and detoxification genes, mitochondrial unfolded protein response (UPRmt) genes are repressed. Either antioxidant or iron supplementation rescues the phenotypes. We examined the virulence factors of CF18 via transposon mutagenesis and RNA sequencing (RNA-seq). We found that non-phenazine toxins that are regulated by quorum sensing (QS) and the GacA/S system are responsible for developmental slowing. This study highlights the importance of ROS levels and mitochondrial health as determinants of developmental rate and how pathogens can attack these important features.
Collapse
Affiliation(s)
- Zeynep Mirza
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Albertha J M Walhout
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Victor Ambros
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
15
|
Ma L, Zhang L, Li J, Zhang X, Xie Y, Li X, Yang B, Yang H. The potential mechanism of gut microbiota-microbial metabolites-mitochondrial axis in progression of diabetic kidney disease. Mol Med 2023; 29:148. [PMID: 37907885 PMCID: PMC10617243 DOI: 10.1186/s10020-023-00745-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023] Open
Abstract
Diabetic kidney disease (DKD), has become the main cause of end-stage renal disease (ESRD) worldwide. Lately, it has been shown that the onset and advancement of DKD are linked to imbalances of gut microbiota and the abnormal generation of microbial metabolites. Similarly, a body of recent evidence revealed that biological alterations of mitochondria ranging from mitochondrial dysfunction and morphology can also exert significant effects on the occurrence of DKD. Based on the prevailing theory of endosymbiosis, it is believed that human mitochondria originated from microorganisms and share comparable biological characteristics with the microbiota found in the gut. Recent research has shown a strong correlation between the gut microbiome and mitochondrial function in the occurrence and development of metabolic disorders. The gut microbiome's metabolites may play a vital role in this communication. However, the relationship between the gut microbiome and mitochondrial function in the development of DKD is not yet fully understood, and the role of microbial metabolites is still unclear. Recent studies are highlighted in this review to examine the possible mechanism of the gut microbiota-microbial metabolites-mitochondrial axis in the progression of DKD and the new therapeutic approaches for preventing or reducing DKD based on this biological axis in the future.
Collapse
Affiliation(s)
- Leilei Ma
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Li Zhang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Jing Li
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Xiaotian Zhang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Yiran Xie
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Xiaochen Li
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Bo Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China
| | - Hongtao Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese, Medicine Acupuncture and Moxibustion, Tianjin, 300380, China.
| |
Collapse
|
16
|
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
|
17
|
Stefano GB, Büttiker P, Weissenberger S, Esch T, Anders M, Raboch J, Kream RM, Ptacek R. Independent and sensory human mitochondrial functions reflecting symbiotic evolution. Front Cell Infect Microbiol 2023; 13:1130197. [PMID: 37389212 PMCID: PMC10302212 DOI: 10.3389/fcimb.2023.1130197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
The bacterial origin of mitochondria has been a widely accepted as an event that occurred about 1.45 billion years ago and endowed cells with internal energy producing organelle. Thus, mitochondria have traditionally been viewed as subcellular organelle as any other - fully functionally dependent on the cell it is a part of. However, recent studies have given us evidence that mitochondria are more functionally independent than other organelles, as they can function outside the cells, engage in complex "social" interactions, and communicate with each other as well as other cellular components, bacteria and viruses. Furthermore, mitochondria move, assemble and organize upon sensing different environmental cues, using a process akin to bacterial quorum sensing. Therefore, taking all these lines of evidence into account we hypothesize that mitochondria need to be viewed and studied from a perspective of a more functionally independent entity. This view of mitochondria may lead to new insights into their biological function, and inform new strategies for treatment of disease associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- George B. Stefano
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Pascal Büttiker
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | | | - Tobias Esch
- Institute for Integrative Health Care and Health Promotion, School of Medicine, Witten/Herdecke University, Witten, Germany
| | - Martin Anders
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Jiri Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Richard M. Kream
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Radek Ptacek
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| |
Collapse
|
18
|
Zhang J, Cheng H, Di Narzo A, Zhu Y, Xie S, Shao X, Zhang Z, Chung SK, Hao K. Profiling Microbiota from Multiple Sites in the Respiratory Tract to Identify a Biomarker for PM 2.5 Nitrate Exposure-Induced Pulmonary Damages. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:7346-7357. [PMID: 37133311 DOI: 10.1021/acs.est.2c08807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The microbiota present in the respiratory tract (RT) responds to environmental stimuli and engages in a continuous interaction with the host immune system to maintain homeostasis. A total of 40 C57BL/6 mice were divided into four groups and exposed to varying concentrations of PM2.5 nitrate aerosol and clean air. After 10 weeks of exposure, assessments were conducted on the lung and airway microbiome, lung functions, and pulmonary inflammation. Additionally, we analyzed data from both mouse and human respiratory tract (RT) microbiomes to identify possible biomarkers for PM2.5 exposure-induced pulmonary damages. On average, 1.5 and 13.5% inter-individual microbiome variations in the lung and airway were explained by exposure, respectively. In the airway, among the 60 bacterial OTUs (operational taxonomic units) > 0.05% proportion, 40 OTUs were significantly affected by PM2.5 exposure (FDR ≤ 10%). Further, the airway microbiome was associated with peak expiratory flow (PEF) (p = 0.003), pulmonary neutrophil counts (p = 0.01), and alveolar 8-OHdG oxidative lesions (p = 0.0078). The Clostridiales order bacteria showed the strongest signals. For example, the o_Clostridiales;f_;g_ OTU was elevated by PM2.5 nitrate exposure (p = 4.98 × 10-5) and negatively correlated with PEF (r = -0.585 and p = 2.4 × 10-4). It was also associated with the higher pulmonary neutrophil count (p = 8.47 × 10-5) and oxidative lesion (p = 7.17 × 10-3). In human data, we confirmed the association of airway Clostridiales order bacteria with PM2.5 exposure and lung function. For the first time, this study characterizes the impact of PM2.5 exposure on the microbiome of multiple sites in the respiratory tract (RT) and its relevance to airflow obstructive diseases. By analyzing data from both humans and mice, we have identified bacteria belonging to the Clostridiales order as a promising biomarker for PM2.5 exposure-induced decline in pulmonary function and inflammation.
Collapse
Affiliation(s)
- Jushan Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, Tongji University, Shanghai 200072, China
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
- College of Environmental Science and Engineering, Tongji University, Shanghai 200072, China
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| | - Antonio Di Narzo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| | - Yujie Zhu
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Shuanshuan Xie
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xiaowen Shao
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Zhongyang Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| | - Sookja Kim Chung
- Medical Faculty, Macau University of Science and Technology, Taipa, Macau SAR 999078, China
| | - Ke Hao
- College of Environmental Science and Engineering, Tongji University, Shanghai 200072, China
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029-6574, United States
| |
Collapse
|
19
|
Mohareer K, Banerjee S. Mycobacterial infection alters host mitochondrial activity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023. [DOI: 10.1016/bs.ircmb.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
20
|
Oxidative stress in metabolic diseases: current scenario and therapeutic relevance. Mol Cell Biochem 2023; 478:185-196. [PMID: 35764861 DOI: 10.1007/s11010-022-04496-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/01/2022] [Indexed: 01/17/2023]
Abstract
The metabolic syndrome is a clustering condition of increased abdominal obesity in concert with hyperglycemia, insulin resistance, hypertension, and dyslipidemia. It confers higher risk of metabolic diseases such as diabetes and ischemic heart disease and has been observed to be associated with high morbidity and mortality. It is a progressive pathological process for diabetes-induced complications and appears to be multifactorial in origin. Several preclinical, clinical, and epidemiological reports have shown a persistent link between the metabolic syndrome and oxidative stress. There is pronounced imbalance between pro-oxidants and anti-oxidants with increased production of oxidizing molecules, depletion of anti-oxidants, and consequently accumulation of protein and lipid oxidation products in the cell in metabolic syndrome. The increased cellular pro-oxidant activity also results in altered molecular pathways, mitochondrial dysfunction, deregulation in cell cycle control, chromosomal aberrations, inflammation, and overall decreased biological activity as well as impairment of the antioxidant systems. Here, the focus of our review article will be on the formation of oxidative species, the interplay between metabolic syndrome and oxidative stress, and its potential implications in therapeutic approaches.
Collapse
|
21
|
Chlamydia trachomatis Alters Mitochondrial Protein Composition and Secretes Effector Proteins That Target Mitochondria. mSphere 2022; 7:e0042322. [PMID: 36286535 PMCID: PMC9769516 DOI: 10.1128/msphere.00423-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Mitochondria are critical cellular organelles that perform a wide variety of functions, including energy production and immune regulation. To perform these functions, mitochondria contain approximately 1,500 proteins, the majority of which are encoded in the nuclear genome, translated in the cytoplasm, and translocated to the mitochondria using distinct mitochondrial targeting sequences (MTS). Bacterial proteins can also contain MTS and localize to the mitochondria. For the obligate intracellular human pathogen Chlamydia trachomatis, interaction with various host cell organelles promotes intracellular replication. However, the extent and mechanisms through which Chlamydia cells interact directly with mitochondria remain unclear. We investigated the presence of MTS in the C. trachomatis genome and discovered 30 genes encoding proteins with around 70% or greater probability of mitochondrial localization. Five are translocated to the mitochondria upon ectopic expression in HeLa cells. Mass spectrometry of isolated mitochondria from infected cells revealed that two of these proteins localize to the mitochondria during infection. Comparison of mitochondria from infected and uninfected cells suggests that chlamydial infection affects the mitochondrial protein composition. Around 125 host proteins were significantly decreased or absent in mitochondria from infected cells. Among these were proapoptotic factors and those related to mitochondrial fission/fusion dynamics. Conversely, 82 host proteins were increased in or specific to mitochondria of infected cells, many of which act as antiapoptotic factors and upregulators of cellular metabolism. These data support the notion that C. trachomatis specifically targets host mitochondria to manipulate cell fate decisions and metabolic function to support pathogen survival and replication. IMPORTANCE Obligate intracellular bacteria have evolved multiple means to promote their intracellular survival and replication within the otherwise harsh environment of the eukaryotic cell. Nutrient acquisition and avoidance of cellular defense mechanisms are critical to an intracellular lifestyle. Mitochondria are critical organelles that produce energy in the form of ATP and regulate programmed cell death responses to invasive pathogenic microbes. Cell death prior to completion of replication would be detrimental to the pathogen. C. trachomatis produces at least two and possibly more proteins that target the mitochondria. Collectively, C. trachomatis infection modulates the mitochondrial protein composition, favoring a profile suggestive of downregulation of apoptosis.
Collapse
|
22
|
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
|
23
|
Prazdnova EV, Mazanko MS, Chistyakov VA, Bogdanova AA, Refeld AG, Kharchenko EY, Chikindas ML. Antimutagenic Activity as a Criterion of Potential Probiotic Properties. Probiotics Antimicrob Proteins 2022; 14:1094-1109. [PMID: 35028920 DOI: 10.1007/s12602-021-09870-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2021] [Indexed: 12/25/2022]
Abstract
The antimutagenic activity of probiotic strains has been reported over several decades of studying the effects of probiotics. However, this activity is rarely considered an important criterion when choosing strains to produce probiotic preparations and functional food. Meanwhile, the association of antimutagenic activity with the prevention of oncological diseases, as well as with a decrease in the spread of resistant forms in the microbiota, indicates its importance for the selection of probiotics. Besides, an antimutagenic activity can be associated with probiotics' broader systemic effects, such as geroprotective activity. The main mechanisms of such effects are considered to be the binding of mutagens, the transformation of mutagens, and inhibition of the transformation of promutagens into antimutagens. Besides, we should consider the possibility of interaction of the microbiota with regulatory processes in eukaryotic cells, in particular, through the effect on mitochondria. This work aims to systematize data on the antimutagenic activity of probiotics and emphasize antimutagenic activity as a significant criterion for the selection of probiotic strains.
Collapse
Affiliation(s)
- Evgeniya V Prazdnova
- Academy of Biology and Biotechnologies, Southern Federal University, Prospect Stachki, 194/1, Rostov-on-Don, Russia. .,Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia.
| | - Maria S Mazanko
- Academy of Biology and Biotechnologies, Southern Federal University, Prospect Stachki, 194/1, Rostov-on-Don, Russia.,Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | - Vladimir A Chistyakov
- Academy of Biology and Biotechnologies, Southern Federal University, Prospect Stachki, 194/1, Rostov-on-Don, Russia.,Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | - Anna A Bogdanova
- Evolutionary Biomedicine Laboratory, SCAMT Institute, ITMO University, Saint Petersburg, Russia
| | - Aleksandr G Refeld
- Cell Biophysics Laboratory, SCAMT Institute, ITMO University, Saint Petersburg, Russia
| | - Evgeniya Y Kharchenko
- Academy of Biology and Biotechnologies, Southern Federal University, Prospect Stachki, 194/1, Rostov-on-Don, Russia
| | - Michael L Chikindas
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia.,Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA.,I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
24
|
Mahmud SA, Qureshi MA, Pellegrino MW. On the offense and defense: mitochondrial recovery programs amidst targeted pathogenic assault. FEBS J 2022; 289:7014-7037. [PMID: 34270874 PMCID: PMC9192128 DOI: 10.1111/febs.16126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/24/2021] [Accepted: 07/15/2021] [Indexed: 01/13/2023]
Abstract
Bacterial pathogens employ a variety of tactics to persist in their host and promote infection. Pathogens often target host organelles in order to benefit their survival, either through manipulation or subversion of their function. Mitochondria are regularly targeted by bacterial pathogens owing to their diverse cellular roles, including energy production and regulation of programmed cell death. However, disruption of normal mitochondrial function during infection can be detrimental to cell viability because of their essential nature. In response, cells use multiple quality control programs to mitigate mitochondrial dysfunction and promote recovery. In this review, we will provide an overview of mitochondrial recovery programs including mitochondrial dynamics, the mitochondrial unfolded protein response (UPRmt ), and mitophagy. We will then discuss the various approaches used by bacterial pathogens to target mitochondria, which result in mitochondrial dysfunction. Lastly, we will discuss how cells leverage mitochondrial recovery programs beyond their role in organelle repair, to promote host defense against pathogen infection.
Collapse
Affiliation(s)
- Siraje A Mahmud
- Department of Biology, University of Texas Arlington, TX, USA
| | | | | |
Collapse
|
25
|
Kwon JE, Jo SH, Song WS, Lee JS, Jeon HJ, Park JH, Kim YR, Baek JH, Kim MG, Kwon SY, Kim JS, Yang YH, Kim YG. Investigation of metabolic crosstalk between host and pathogenic Clostridioides difficile via multiomics approaches. Front Bioeng Biotechnol 2022; 10:971739. [PMID: 36118584 PMCID: PMC9478559 DOI: 10.3389/fbioe.2022.971739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Clostridioides difficile is a gram-positive anaerobic bacterium that causes antibiotic-associated infections in the gut. C. difficile infection develops in the intestine of a host with an imbalance of the intestinal microbiota and, in severe cases, can lead to toxic megacolon, intestinal perforation, and even death. Despite its severity and importance, however, the lack of a model to understand host-pathogen interactions and the lack of research results on host cell effects and response mechanisms under C. difficile infection remain limited. Here, we developed an in vitro anaerobic-aerobic C. difficile infection model that enables direct interaction between human gut epithelial cells and C. difficile through the Mimetic Intestinal Host–Microbe Interaction Coculture System. Additionally, an integrative multiomics approach was applied to investigate the biological changes and response mechanisms of host cells caused by C. difficile in the early stage of infection. The C. difficile infection model was validated through the induction of disaggregation of the actin filaments and disruption of the intestinal epithelial barrier as the toxin-mediated phenotypes following infection progression. In addition, an upregulation of stress-induced chaperones and an increase in the ubiquitin proteasomal pathway were identified in response to protein stress that occurred in the early stage of infection, and downregulation of proteins contained in the electron transfer chain and ATP synthase was observed. It has been demonstrated that host cell energy metabolism is inhibited through the glycolysis of Caco-2 cells and the reduction of metabolites belonging to the TCA cycle. Taken together, our C. difficile infection model suggests a new biological response pathway in the host cell induced by C. difficile during the early stage of infection at the molecular level under anaerobic-aerobic conditions. Therefore, this study has the potential to be applied to the development of future therapeutics through basic metabolic studies of C. difficile infection.
Collapse
Affiliation(s)
- Ji-Eun Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Won-Suk Song
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Jae-Seung Lee
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Hyo-Jin Jeon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ji-Hyeon Park
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ye-Rim Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ji-Hyun Baek
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Min-Gyu Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Seo-Young Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, South Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University, Seoul, South Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
- *Correspondence: Yun-Gon Kim,
| |
Collapse
|
26
|
Ju S, Chen H, Wang S, Lin J, Ma Y, Aroian RV, Peng D, Sun M. C. elegans monitor energy status via the AMPK pathway to trigger innate immune responses against bacterial pathogens. Commun Biol 2022; 5:643. [PMID: 35773333 PMCID: PMC9246835 DOI: 10.1038/s42003-022-03589-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Pathogen recognition and the triggering of host innate immune system are critical to understanding pathogen-host interaction. Cellular surveillance systems have been identified as an important strategy for the identification of microbial infection. In the present study, using Bacillus thuringiensis-Caenorhabditis elegans as a model, we found an approach for surveillance systems to sense pathogens. We report that Bacillus thuringiensis Cry5Ba, a typical pore-forming toxin, caused mitochondrial damage and energy imbalance by triggering potassium ion leakage, instead of directly targeting mitochondria. Interestingly, we find C. elegans can monitor intracellular energy status to trigger innate immune responses via AMP-activated protein kinase (AMPK), secreting multiple effectors to defend against pathogenic attacks. Our study indicates that the imbalance of energy status is a prevalent side effect of pathogen infection. Furthermore, the AMPK-dependent surveillance system may serve as a practicable strategy for the host to recognize and defense against pathogens. Bacillus thuringiensis toxin Cry5Ba triggers potassium ion leakage, causing mitochondrial damage and energy imbalance. C. elegans can monitor this intracellular energy imbalance via AMP-activated protein kinase to trigger innate immune responses.
Collapse
Affiliation(s)
- Shouyong Ju
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hanqiao Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shaoying Wang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Lin
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yanli Ma
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China
| | - Raffi V Aroian
- Program in Molecular Medicine, University of Massachusetts Chan Medical School Worcester, Worcester, MA, 01605-2377, USA
| | - Donghai Peng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Ming Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, National Engineering Research Center of Microbial Pesticides, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
27
|
Lian WS, Wang FS, Chen YS, Tsai MH, Chao HR, Jahr H, Wu RW, Ko JY. Gut Microbiota Ecosystem Governance of Host Inflammation, Mitochondrial Respiration and Skeletal Homeostasis. Biomedicines 2022; 10:biomedicines10040860. [PMID: 35453611 PMCID: PMC9030723 DOI: 10.3390/biomedicines10040860] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Osteoporosis and osteoarthritis account for the leading causes of musculoskeletal dysfunction in older adults. Senescent chondrocyte overburden, inflammation, oxidative stress, subcellular organelle dysfunction, and genomic instability are prominent features of these age-mediated skeletal diseases. Age-related intestinal disorders and gut dysbiosis contribute to host tissue inflammation and oxidative stress by affecting host immune responses and cell metabolism. Dysregulation of gut microflora correlates with development of osteoarthritis and osteoporosis in humans and rodents. Intestinal microorganisms produce metabolites, including short-chain fatty acids, bile acids, trimethylamine N-oxide, and liposaccharides, affecting mitochondrial function, metabolism, biogenesis, autophagy, and redox reactions in chondrocytes and bone cells to regulate joint and bone tissue homeostasis. Modulating the abundance of Lactobacillus and Bifidobacterium, or the ratio of Firmicutes and Bacteroidetes, in the gut microenvironment by probiotics or fecal microbiota transplantation is advantageous to suppress age-induced chronic inflammation and oxidative damage in musculoskeletal tissue. Supplementation with gut microbiota-derived metabolites potentially slows down development of osteoarthritis and osteoporosis. This review provides latest molecular and cellular insights into the biological significance of gut microorganisms and primary and secondary metabolites important to cartilage and bone integrity. It further highlights treatment options with probiotics or metabolites for modulating the progression of these two common skeletal disorders.
Collapse
Affiliation(s)
- Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostics, Department of Medical Research and Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (F.-S.W.); (Y.-S.C.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Feng-Sheng Wang
- Core Laboratory for Phenomics and Diagnostics, Department of Medical Research and Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (F.-S.W.); (Y.-S.C.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostics, Department of Medical Research and Chang Gung University College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (W.-S.L.); (F.-S.W.); (Y.-S.C.)
- Center for Mitochondrial Research and Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Ming-Hsien Tsai
- Department of Child Care, College of Humanities and Social Sciences, National Pingtung University of Science and Technology, No.1, Shuefu Road, Pingtung 91201, Taiwan;
- Emerging Compounds Research Center, General Research Service Center, National Pingtung University of Science and Technology, No.1, Shuefu Road, Pingtung 91201, Taiwan;
| | - How-Ran Chao
- Emerging Compounds Research Center, General Research Service Center, National Pingtung University of Science and Technology, No.1, Shuefu Road, Pingtung 91201, Taiwan;
- Department of Environmental Science and Engineering, College of Engineering, National Pingtung University of Science and Technology, No.1, Shuefu Road, Pingtung 91201, Taiwan
| | - Holger Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH, 52074 Aachen, Germany;
- Department of Orthopedic Surgery, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| | - Re-Wen Wu
- Department of Orthopedic Surgery, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, College of Medicine, Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
- Correspondence: ; Tel.: +88-67-731-7123
| |
Collapse
|
28
|
Cao C, Zhu H, Yao Y, Zeng R. Gut Dysbiosis and Kidney Diseases. Front Med (Lausanne) 2022; 9:829349. [PMID: 35308555 PMCID: PMC8927813 DOI: 10.3389/fmed.2022.829349] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/09/2022] [Indexed: 12/16/2022] Open
Abstract
Gut dysbiosis is defined as disorders of gut microbiota and loss of barrier integrity, which are ubiquitous on pathological conditions and associated with the development of various diseases. Kidney diseases are accompanied with gut dysbiosis and metabolic disorders, which in turn contribute to the pathogenesis and progression of kidney diseases. Microbial alterations trigger production of harmful metabolites such as uremic toxins and a decrease in the number of beneficial ones such as SCFAs, which is the major mechanism of gut dysbiosis on kidney diseases according to current studies. In addition, the activation of immune responses and mitochondrial dysfunction by gut dysbiosis, also lead to the development of kidney diseases. Based on the molecular mechanisms, modification of gut dysbiosis via probiotics, prebiotics and synbiotics is a potential approach to slow kidney disease progression. Fecal microbiota transplantation (FMT) and genetic manipulation of the gut microbiota are also promising choices. However, the clinical use of probiotics in kidney disease is not supported by the current clinical evidence. Further studies are necessary to explore the causal relationships of gut dysbiosis and kidney diseases, the efficiency and safety of therapeutic strategies targeting gut-kidney axis.
Collapse
Affiliation(s)
- Chujin Cao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Yao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Division of Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zeng
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Nagase N, Ikeda Y, Tsuji A, Kitagishi Y, Matsuda S. Efficacy of probiotics on the modulation of gut microbiota in the treatment of diabetic nephropathy. World J Diabetes 2022; 13:150-160. [PMID: 35432750 PMCID: PMC8984564 DOI: 10.4239/wjd.v13.i3.150] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/21/2021] [Accepted: 02/13/2022] [Indexed: 02/06/2023] Open
Abstract
Diabetic nephropathy (DN) is a major cause of end-stage renal disease, and therapeutic options for preventing its progression are insufficient. The number of patients with DN has been increasing in Asian countries because of westernization of dietary lifestyle, which may be associated with the following changes in gut microbiota. Alterations in the gut microbiota composition can lead to an imbalanced gastrointestinal environment that promotes abnormal production of metabolites and/or inflammatory status. Functional microenvironments of the gut could be changed in the different stages of DN. In particular, altered levels of short chain fatty acids, D-amino acids, and reactive oxygen species biosynthesis in the gut have been shown to be relevant to the pathogenesis of the DN. So far, evidence suggests that the gut microbiota may play a key role in determining networks in the development of DN. Interventions directing the gut microbiota deserve further investigation as a new protective therapy in DN. In this review, we discuss the potential roles of the gut microbiota and future perspectives in the protection and/or treatment of kidneys.
Collapse
Affiliation(s)
- Nozomi Nagase
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Yuka Ikeda
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Ai Tsuji
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| |
Collapse
|
30
|
Zhu Y, Li Y, Zhang Q, Song Y, Wang L, Zhu Z. Interactions Between Intestinal Microbiota and Neural Mitochondria: A New Perspective on Communicating Pathway From Gut to Brain. Front Microbiol 2022; 13:798917. [PMID: 35283843 PMCID: PMC8908256 DOI: 10.3389/fmicb.2022.798917] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
Many studies shown that neurological diseases are associated with neural mitochondrial dysfunctions and microbiome composition alterations. Since mitochondria emerged from bacterial ancestors during endosymbiosis, mitochondria, and bacteria had analogous genomic characteristics, similar bioactive compounds and comparable energy metabolism pathways. Therefore, it is necessary to rationalize the interactions of intestinal microbiota with neural mitochondria. Recent studies have identified neural mitochondrial dysfunction as a critical pathogenic factor for the onset and progress of multiple neurological disorders, in which the non-negligible role of altered gut flora composition was increasingly noticed. Here, we proposed a new perspective of intestinal microbiota – neural mitochondria interaction as a communicating channel from gut to brain, which could help to extend the vision of gut-brain axis regulation and provide additional research directions on treatment and prevention of responsive neurological disorders.
Collapse
Affiliation(s)
- Yao Zhu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Ying Li
- Medical Technology College, Xuzhou Medical University, Xuzhou, China
| | - Qiang Zhang
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Yuanjian Song
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, China
| | - Liang Wang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
- Liang Wang,
| | - Zuobin Zhu
- Xuzhou Engineering Research Center of Medical Genetics and Transformation, Key Laboratory of Genetic Foundation and Clinical Application, Department of Genetics, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Zuobin Zhu,
| |
Collapse
|
31
|
Fu J, Zhou M, Gritsenko MA, Nakayasu ES, Song L, Luo ZQ. Legionella pneumophila modulates host energy metabolism by ADP-ribosylation of ADP/ATP translocases. eLife 2022; 11:73611. [PMID: 35084332 PMCID: PMC8820735 DOI: 10.7554/elife.73611] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 01/26/2022] [Indexed: 11/13/2022] Open
Abstract
The intracellular pathogen Legionella pneumophila delivers more than 330 effectors into host cells by its Dot/Icm secretion system. Those effectors direct the biogenesis of the Legionella-containing vacuole (LCV) that permits its intracellular survival and replication. It has long been documented that the LCV is associated with mitochondria and a number of Dot/Icm effectors have been shown to target to this organelle. Yet, the biochemical function and host cell target of most of these effectors remain unknown. Here, we found that the Dot/Icm substrate Ceg3 (Lpg0080) is a mono-ADP-ribosyltransferase that localizes to the mitochondria in host cells where it attacks ADP/ATP translocases by ADP-ribosylation, and blunts their ADP/ATP exchange activity. The modification occurs on the second arginine residue in the -RRRMMM- element, which is conserved among all known ADP/ATP carriers from different organisms. Our results reveal modulation of host energy metabolism as a virulence mechanism for L. pneumophila.
Collapse
Affiliation(s)
- Jiaqi Fu
- Department of Biological Science, Purdue University, West Lafayette, United States
| | - Mowei Zhou
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, United States
| | - Marina A Gritsenko
- Biological Science Division, Pacific Northwest National Laboratory, Richland, United States
| | - Ernesto S Nakayasu
- Biological Science Division, Pacific Northwest National Laboratory, Richland, United States
| | - Lei Song
- Department of Respiratory Medicine, Jilin University, Changchun, China
| | - Zhao-Qing Luo
- Department of Biological Science, Purdue University, West Lafayette, United States
| |
Collapse
|
32
|
Chen G, Tan Z, Liu Y, Weng T, Xia L, Lu Y. Function and Characterization of an Alanine Dehydrogenase Homolog From Nocardia seriolae. Front Vet Sci 2022; 8:801990. [PMID: 35097049 PMCID: PMC8790279 DOI: 10.3389/fvets.2021.801990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023] Open
Abstract
Fish nocardiosis is a chronic, systemic, granulomatous disease in aquaculture. Nocardia seriolae has been reported to be one of the main pathogenic bacteria of fish nocardiosis. There are few studies on the associated virulence factors and pathogenesis of N. seriolae. Alanine dehydrogenase (ALD), which may be a secreted protein, was discovered by analysis using bioinformatics methods throughout the whole genomic sequence of N. seriolae. Nevertheless, the roles of ALD and its homologs in the pathogenesis of N. seriolae are not demonstrated. In this study, the function of N. seriolae ALD (NsALD) was preliminarily investigated by gene cloning, host cell subcellular localization, secreted protein identification, and cell apoptosis detection. Identification of the extracellular products of N. seriolae via mass spectrometry (MS) analysis revealed that NsALD is a secreted protein. In addition, subcellular localization of NsALD-GFP recombinant protein in fathead minnow (FHM) cells showed that the strong green fluorescence co-localized with the mitochondria. Moreover, apoptosis assays demonstrated that the overexpression of NsALD induces apoptosis in FHM cells. This study may lay the foundation for further exploration of the function of NsALD and facilitate further understanding of the pathogenic mechanism and the associated virulence factors of N. seriolae.
Collapse
Affiliation(s)
- Guoquan Chen
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
| | - Ziyang Tan
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fisheries College of Guangdong Ocean University, Zhanjiang, China
| | - Yansheng Liu
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
| | - Tingting Weng
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
| | - Liqun Xia
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- *Correspondence: Liqun Xia
| | - Yishan Lu
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fisheries College of Guangdong Ocean University, Zhanjiang, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, China
- Yishan Lu
| |
Collapse
|
33
|
Omar M, Marchionni L, Häcker G, Badr MT. Host Blood Gene Signatures Can Detect the Progression to Severe and Cerebral Malaria. Front Cell Infect Microbiol 2021; 11:743616. [PMID: 34746025 PMCID: PMC8569259 DOI: 10.3389/fcimb.2021.743616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
Malaria is a major international public health problem that affects millions of patients worldwide especially in sub-Saharan Africa. Although many tests have been developed to diagnose malaria infections, we still lack reliable diagnostic biomarkers for the identification of disease severity, especially in endemic areas where the diagnosis of cerebral malaria is very difficult and requires the exclusion of all other possible causes. Previous host and pathogen transcriptomic studies have not yielded homogenous results that can be harnessed into a reliable diagnostic tool. Here we utilized a multi-cohort analysis approach using machine-learning algorithms to identify blood gene signatures that can distinguish severe and cerebral malaria from moderate and non-cerebral cases. Using a Regularized Random Forest model, we identified 28-gene and 32-gene signatures that can reliably distinguish severe and cerebral malaria, respectively. We tested the specificity of both signatures against other common infectious diseases to ensure the signatures reliability and suitability as diagnostic markers. The severe and cerebral malaria gene-signatures were further integrated through k-top scoring pairs classifiers into ten and nine gene pairs that could distinguish severe and cerebral malaria, respectively. These signatures have various implications that can be utilized as blood diagnostic tools for malaria severity in endemic countries.
Collapse
Affiliation(s)
- Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Mohamed Tarek Badr
- Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,IMM-PACT-Program, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
34
|
Mitochondrial DNA Heteroplasmy as an Informational Reservoir Dynamically Linked to Metabolic and Immunological Processes Associated with COVID-19 Neurological Disorders. Cell Mol Neurobiol 2021; 42:99-107. [PMID: 34117968 PMCID: PMC8196276 DOI: 10.1007/s10571-021-01117-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Mitochondrial DNA (mtDNA) heteroplasmy is the dynamically determined co-expression of wild type (WT) inherited polymorphisms and collective time-dependent somatic mutations within individual mtDNA genomes. The temporal expression and distribution of cell-specific and tissue-specific mtDNA heteroplasmy in healthy individuals may be functionally associated with intracellular mitochondrial signaling pathways and nuclear DNA gene expression. The maintenance of endogenously regulated tissue-specific copy numbers of heteroplasmic mtDNA may represent a sensitive biomarker of homeostasis of mitochondrial dynamics, metabolic integrity, and immune competence. Myeloid cells, monocytes, macrophages, and antigen-presenting dendritic cells undergo programmed changes in mitochondrial metabolism according to innate and adaptive immunological processes. In the central nervous system (CNS), the polarization of activated microglial cells is dependent on strategically programmed changes in mitochondrial function. Therefore, variations in heteroplasmic mtDNA copy numbers may have functional consequences in metabolically competent mitochondria in innate and adaptive immune processes involving the CNS. Recently, altered mitochondrial function has been demonstrated in the progression of coronavirus disease 2019 (COVID-19) due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Accordingly, our review is organized to present convergent lines of empirical evidence that potentially link expression of mtDNA heteroplasmy by functionally interactive CNS cell types to the extent and severity of acute and chronic post-COVID-19 neurological disorders.
Collapse
|
35
|
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
|
36
|
Silva DF, Candeias E, Esteves AR, Magalhães JD, Ferreira IL, Nunes-Costa D, Rego AC, Empadinhas N, Cardoso SM. Microbial BMAA elicits mitochondrial dysfunction, innate immunity activation, and Alzheimer's disease features in cortical neurons. J Neuroinflammation 2020; 17:332. [PMID: 33153477 PMCID: PMC7643281 DOI: 10.1186/s12974-020-02004-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND After decades of research recognizing it as a complex multifactorial disorder, sporadic Alzheimer's disease (sAD) still has no known etiology. Adding to the myriad of different pathways involved, bacterial neurotoxins are assuming greater importance in the etiology and/or progression of sAD. β-N-Methylamino-L-alanine (BMAA), a neurotoxin produced by some microorganisms namely cyanobacteria, was previously detected in the brains of AD patients. Indeed, the consumption of BMAA-enriched foods has been proposed to induce amyotrophic lateral sclerosis-parkinsonism-dementia complex (ALS-PDC), which implicated this microbial metabolite in neurodegeneration mechanisms. METHODS Freshly isolated mitochondria from C57BL/6 mice were treated with BMAA and O2 consumption rates were determined. O2 consumption and glycolysis rates were also measured in mouse primary cortical neuronal cultures. Further, mitochondrial membrane potential and ROS production were evaluated by fluorimetry and the integrity of mitochondrial network was examined by immunofluorescence. Finally, the ability of BMAA to activate neuronal innate immunity was quantified by addressing TLRs (Toll-like receptors) expression, p65 NF-κB translocation into the nucleus, increased expression of NLRP3 (Nod-like receptor 3), and pro-IL-1β. Caspase-1 activity was evaluated using a colorimetric substrate and mature IL-1β levels were also determined by ELISA. RESULTS Treatment with BMAA reduced O2 consumption rates in both isolated mitochondria and in primary cortical cultures, with additional reduced glycolytic rates, decrease mitochondrial potential and increased ROS production. The mitochondrial network was found to be fragmented, which resulted in cardiolipin exposure that stimulated inflammasome NLRP3, reinforced by decreased mitochondrial turnover, as indicated by increased p62 levels. BMAA treatment also activated neuronal extracellular TLR4 and intracellular TLR3, inducing p65 NF-κB translocation into the nucleus and activating the transcription of NLRP3 and pro-IL-1β. Increased caspase-1 activity resulted in elevated levels of mature IL-1β. These alterations in mitochondrial metabolism and inflammation increased Tau phosphorylation and Aβ peptides production, two hallmarks of AD. CONCLUSIONS Here we propose a unifying mechanism for AD neurodegeneration in which a microbial toxin can induce mitochondrial dysfunction and activate neuronal innate immunity, which ultimately results in Tau and Aβ pathology. Our data show that neurons, alone, can mount inflammatory responses, a role previously attributed exclusively to glial cells.
Collapse
Affiliation(s)
- Diana F Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - A Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - João D Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - I Luísa Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Daniela Nunes-Costa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,Ph.D. Programme in Biomedicine and Experimental Biology (PDBEB), Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - A Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,Institute of Biochemistry, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Nuno Empadinhas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal.,IIIUC-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sandra M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal. .,Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
37
|
Xue Y, Chu J, Li Y, Kong X. The influence of air pollution on respiratory microbiome: A link to respiratory disease. Toxicol Lett 2020; 334:14-20. [DOI: 10.1016/j.toxlet.2020.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/10/2020] [Accepted: 09/12/2020] [Indexed: 01/08/2023]
|
38
|
Sanborn V, Gunstad J. The Potential Mediation of the Effects of Physical Activity on Cognitive Function by the Gut Microbiome. Geriatrics (Basel) 2020; 5:geriatrics5040063. [PMID: 32992812 PMCID: PMC7709629 DOI: 10.3390/geriatrics5040063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
The population of older adults is growing dramatically worldwide. As older adults are at greater risk of developing disorders associated with cognitive dysfunction (i.e., dementia), healthcare costs are expected to double by 2040. Evidence suggests dementia may be slowed or prevented by lifestyle interventions, including physical activity (PA). PA is associated with improved cognitive function and may reduce risk for dementia by mitigating known risk factors (i.e., cardiovascular diseases) and/or by enhancing neurochemical processes. An emerging area of research suggests the gut microbiome may have similar neuroprotective effects. Altering the gut microbiome has been found to target physiological processes associated with dementia risk, and it influences gut-brain-microbiome axis signaling, impacting cognitive functioning. The gut microbiome can be altered by several means (i.e., disease, diet, prebiotics, probiotics), including PA. As PA and the gut microbiome independently influence cognitive function and PA changes the composition of the gut microbiome, cognitive improvement due to PA may be partially mediated by the gut microbiome. The present article provides an overview of the literature regarding the complex associations among PA, cognitive function, and the gut microbiome, as well as their underlying biological mechanisms. A comprehensive, theoretical model integrating evidence for the potential mediation is proposed.
Collapse
Affiliation(s)
- Victoria Sanborn
- Department of Psychological Sciences, Kent State University, Kent, OH 44240, USA;
- Correspondence:
| | - John Gunstad
- Department of Psychological Sciences, Kent State University, Kent, OH 44240, USA;
- Brain Health Research Institute, Kent State University, Kent, OH 44240, USA
| |
Collapse
|
39
|
Vezza T, Abad-Jiménez Z, Marti-Cabrera M, Rocha M, Víctor VM. Microbiota-Mitochondria Inter-Talk: A Potential Therapeutic Strategy in Obesity and Type 2 Diabetes. Antioxidants (Basel) 2020; 9:antiox9090848. [PMID: 32927712 PMCID: PMC7554719 DOI: 10.3390/antiox9090848] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
The rising prevalence of obesity and type 2 diabetes (T2D) is a growing concern worldwide. New discoveries in the field of metagenomics and clinical research have revealed that the gut microbiota plays a key role in these metabolic disorders. The mechanisms regulating microbiota composition are multifactorial and include resistance to stress, presence of pathogens, diet, cultural habits and general health conditions. Recent evidence has shed light on the influence of microbiota quality and diversity on mitochondrial functions. Of note, the gut microbiota has been shown to regulate crucial transcription factors, coactivators, as well as enzymes implicated in mitochondrial biogenesis and metabolism. Moreover, microbiota metabolites seem to interfere with mitochondrial oxidative/nitrosative stress and autophagosome formation, thus regulating the activation of the inflammasome and the production of inflammatory cytokines, key players in chronic metabolic disorders. This review focuses on the association between intestinal microbiota and mitochondrial function and examines the mechanisms that may be the key to their use as potential therapeutic strategies in obesity and T2D management.
Collapse
Affiliation(s)
- Teresa Vezza
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
| | - Zaida Abad-Jiménez
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
| | | | - Milagros Rocha
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
- CIBERehd—Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- Correspondence: (M.R.); (V.M.V.); Tel.: +34-963-189-132 (M.R. & V.M.V.); Fax: +34-961-622-492 (M.R. & V.M.V.)
| | - Víctor Manuel Víctor
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
- CIBERehd—Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
- Correspondence: (M.R.); (V.M.V.); Tel.: +34-963-189-132 (M.R. & V.M.V.); Fax: +34-961-622-492 (M.R. & V.M.V.)
| |
Collapse
|
40
|
Verma V, Kumar P, Gupta S, Yadav S, Dhanda RS, Thorlacius H, Yadav M. α-Hemolysin of uropathogenic E. coli regulates NLRP3 inflammasome activation and mitochondrial dysfunction in THP-1 macrophages. Sci Rep 2020; 10:12653. [PMID: 32724079 PMCID: PMC7387347 DOI: 10.1038/s41598-020-69501-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 07/07/2020] [Indexed: 12/16/2022] Open
Abstract
Hemolysin expressing UPEC strains have been associated with severe advanced kidney pathologies, such as cystitis and pyelonephritis, which are associated with an inflammatory response. Macrophages play an important role in regulating an inflammatory response during a urinary tract infection. We have studied the role of purified recombinant α-hemolysin in inducing inflammatory responses and cell death in macrophages. Acylation at lysine residues through HlyC is known to activate proHlyA into a fully functional pore-forming toxin, HlyA. It was observed that active α-hemolysin (HlyA) induced cleavage of caspase-1 leading to the maturation of IL-1β, while inactive α-hemolysin (proHlyA) failed to do so in THP-1 derived macrophages. HlyA also promotes deubiquitination, oligomerization, and activation of the NLRP3 inflammasome, which was found to be dependent on potassium efflux. We have also observed the co-localization of NLRP3 within mitochondria during HlyA stimulations. Moreover, blocking of potassium efflux improved the mitochondrial health in addition to a decreased inflammatory response. Our study demonstrates that HlyA stimulation caused perturbance in potassium homeostasis, which led to the mitochondrial dysfunction followed by an acute inflammatory response, resulting in cell death. However, the repletion of intracellular potassium stores could avoid HlyA induced macrophage cell death. The findings of this study will help to understand the mechanism of α-hemolysin induced inflammatory response and cell death.
Collapse
Affiliation(s)
- Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India
| | - Parveen Kumar
- Department of Urology, University of Alabama At Birmingham, Hugh Kaul Genetics Building, Birmingham, AL, USA
| | - Surbhi Gupta
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India
| | - Sonal Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India
| | - Rakesh Singh Dhanda
- Stem Cell Laboratory, Longboat Explorers AB, SMiLE Incubator, Scheelevägen 2, Lund, Sweden
| | - Henrik Thorlacius
- Department of Clinical Sciences, Section of Surgery, Malmö, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi (North Campus), Delhi, 110007, India.
- Department of Clinical Sciences, Section of Surgery, Malmö, Skåne University Hospital, Lund University, Malmö, Sweden.
| |
Collapse
|
41
|
Saleh J, Peyssonnaux C, Singh KK, Edeas M. Mitochondria and microbiota dysfunction in COVID-19 pathogenesis. Mitochondrion 2020; 54:1-7. [PMID: 32574708 PMCID: PMC7837003 DOI: 10.1016/j.mito.2020.06.008] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022]
Abstract
Mitochondria are the hub of cellular oxidative homeostasis. Mitochondria are the major source of reactive oxygen species (ROS). Extracellular mitochondria are found in blood, in circulating platelets and vesicles. COVID-19 pathogenesis is aggravated by the hyper- inflammatory state. Inflammation activates events leading to microbiota & mitochondrial oxidative damage. Mitochondrial damage contributes to coagulopathy, ferroptosis & microbial dysbiosis. Blood & platelet mitochondria dysfunction may accelerate systemic coagulopathy events. Targeting mitochondria dysfunction may provide useful therapeutic strategies against COVID-19 pathogenesis.
The COVID-19 pandemic caused by the coronavirus (SARS-CoV-2) has taken the world by surprise into a major crisis of overwhelming morbidity and mortality. This highly infectious disease is associated with respiratory failure unusual in other coronavirus infections. Mounting evidence link the accelerated progression of the disease in COVID-19 patients to the hyper-inflammatory state termed as the “cytokine storm” involving major systemic perturbations. These include iron dysregulation manifested as hyperferritinemia associated with disease severity. Iron dysregulation induces reactive oxygen species (ROS) production and promotes oxidative stress. The mitochondria are the hub of cellular oxidative homeostasis. In addition, the mitochondria may circulate “cell-free” in non-nucleated platelets, in extracellular vesicles and mitochondrial DNA is found in the extracellular space. The heightened inflammatory/oxidative state may lead to mitochondrial dysfunction leading to platelet damage and apoptosis. The interaction of dysfunctional platelets with coagulation cascades aggravates clotting events and thrombus formation. Furthermore, mitochondrial oxidative stress may contribute to microbiota dysbiosis, altering coagulation pathways and fueling the inflammatory/oxidative response leading to the vicious cycle of events. Here, we discuss various cellular and systemic incidents caused by SARS-CoV-2 that may critically impact intra and extracellular mitochondrial function, and contribute to the progression and severity of the disease. It is crucial to understand how these key modulators impact COVID-19 pathogenesis in the quest to identify novel therapeutic targets that may reduce fatal outcomes of the disease.
Collapse
Affiliation(s)
- Jumana Saleh
- College of Medicine, Sultan Qaboos University, Oman
| | - Carole Peyssonnaux
- Université de Paris, INSERM U1016, Institut Cochin, CNRS UMR8104, Faculté de médecine Cochin-Port Royal, Paris, France; Laboratory of Excellence GR-Ex, Paris, France
| | - Keshav K Singh
- Integrated Center for Aging Research, Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Marvin Edeas
- Université de Paris, INSERM U1016, Institut Cochin, CNRS UMR8104, Faculté de médecine Cochin-Port Royal, Paris, France; Laboratory of Excellence GR-Ex, Paris, France.
| |
Collapse
|
42
|
Effect of Borrelia burgdorferi Outer Membrane Vesicles on Host Oxidative Stress Response. Antibiotics (Basel) 2020; 9:antibiotics9050275. [PMID: 32466166 PMCID: PMC7277464 DOI: 10.3390/antibiotics9050275] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Outer membrane vesicles (OMVs) are spherical bodies containing proteins and nucleic acids that are released by Gram-negative bacteria, including Borrelia burgdorferi, the causative agent of Lyme disease. The functional relationship between B. burgdorferi OMVs and host neuron homeostasis is not well understood. The objective of this study was to examine how B. burgdorferi OMVs impact the host cell environment. First, an in vitro model was established by co-culturing human BE2C neuroblastoma cells with B. burgdorferi B31. B. burgdorferi was able to invade BE2C cells within 24 h. Despite internalization, BE2C cell viability and levels of apoptosis remained unchanged, but resulted in dramatically increased production of MCP-1 and MCP-2 cytokines. Elevated secretion of MCP-1 has previously been associated with changes in oxidative stress. BE2C cell mitochondrial superoxides were reduced as early as 30 min after exposure to B. burgdorferi and OMVs. To rule out whether BE2C cell antioxidant response is the cause of decline in superoxides, superoxide dismutase 2 (SOD2) gene expression was assessed. SOD2 expression was reduced upon exposure to B. burgdorferi, suggesting that B. burgdorferi might be responsible for superoxide reduction. These results suggest that B. burgdorferi modulates cell antioxidant defense and immune system reaction in response to the bacterial infection. In summary, these results show that B. burgdorferi OMVs serve to directly counter superoxide production in BE2C neurons, thereby 'priming' the host environment to support B. burgdorferi colonization.
Collapse
|
43
|
Ruiz E, Penrose HM, Heller S, Nakhoul H, Baddoo M, Flemington EF, Kandil E, Savkovic SD. Bacterial TLR4 and NOD2 signaling linked to reduced mitochondrial energy function in active inflammatory bowel disease. Gut Microbes 2020; 11:350-363. [PMID: 31063017 PMCID: PMC7524318 DOI: 10.1080/19490976.2019.1611152] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 02/03/2023] Open
Abstract
Inflammatory bowel disease (IBD) has been linked to active signaling with bacterial components and reduced mitochondrial ATP production; however, synergism between both of these disease characteristics remains unclear. We aimed to determine in human IBD transcriptomes the link between a transcriptional signature unique to intestinal cells (ICs) with reduced mitochondrial ATP production (Mito-0) and bacteria triggered signaling using a bioinformatics approach. We generated an IC Mito-0 panel comprised of 199 differentially expressed (DE) transcripts mediated by reduced mitochondrial ATP function (DEGseq, log2 fold-change > |2|, p < .001). Transcripts from this panel were involved in diverse biological functions including regulation of mitochondrial energy (lower ATP), extracellular matrix, cell-cell contact, cytoskeleton, growth, metabolism, and inflammation. Next, unsupervised hierarchical clustering showed that the Mito-0 panel distinctly separated inflamed IBD from non-inflamed transcriptomes, which was also supported by principal component analysis (PCA) revealing distinct variation between sample types based on presence of the Mito-0 signature (PCA, p = 8.77e-09). Utilizing three independent IBD cohorts, we validated that 60 novel transcripts from the Mito-0 panel were significantly increased in inflamed tissue. Subsequently, KEGG generated bacterial TLR4 and NOD2 transcriptional signatures strongly associated with inflamed IBD transcriptomes and with the Mito-0 signature as determined by Spearman's analysis (coefficient of correlation, r = 0.92, p < .05). Herein, using a comprehensive analysis we demonstrated existence of an axis between bacteria triggered signaling and reduced mitochondrial energy function. Furthermore, we identified and validated novel transcripts within this axis as potential drivers and therapeutic targets for human IBD.
Collapse
Affiliation(s)
- Emmanuelle Ruiz
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, USA
| | - Harrison M. Penrose
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sandra Heller
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Hani Nakhoul
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Erik F. Flemington
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Emad Kandil
- Division of Endocrine and Oncologic Surgery, Department of Surgery, Tulane University, New Orleans, LA, USA
| | - Suzana D. Savkovic
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
44
|
Holley CL, Schroder K. The rOX-stars of inflammation: links between the inflammasome and mitochondrial meltdown. Clin Transl Immunology 2020; 9:e01109. [PMID: 32055400 PMCID: PMC7008497 DOI: 10.1002/cti2.1109] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/26/2022] Open
Abstract
The nod‐like receptor protein 3 (NLRP3) inflammasome drives inflammation in response to mitochondrial dysfunction. As metabolic powerhouses with prokaryotic ancestry, mitochondria are a cache for danger‐associated molecular patterns and pathogen‐associated molecular pattern‐like molecules that elicit potent innate immune responses. Persistent mitochondrial damage caused by infection, or genetic or environmental factors, can lead to inappropriate or sustained inflammasome signalling. Here, we review the features of mitochondria that drive inflammatory signalling, with a particular focus on mitochondrial activation of the NLRP3 inflammasome. Given that mitochondrial network dynamics, metabolic activity and redox state are all intricately linked to each other and to NLRP3 inflammasome activity, we highlight the importance of a holistic approach to investigations of NLRP3 activation by dysfunctional mitochondria.
Collapse
Affiliation(s)
- Caroline L Holley
- Institute for Molecular Bioscience IMB Centre for Inflammation and Disease Research The University of Queensland St. Lucia QLD Australia
| | - Kate Schroder
- Institute for Molecular Bioscience IMB Centre for Inflammation and Disease Research The University of Queensland St. Lucia QLD Australia
| |
Collapse
|
45
|
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
|
46
|
Duan C, Kuang L, Xiang X, Zhang J, Zhu Y, Wu Y, Yan Q, Liu L, Li T. Activated Drp1-mediated mitochondrial ROS influence the gut microbiome and intestinal barrier after hemorrhagic shock. Aging (Albany NY) 2020; 12:1397-1416. [PMID: 31954373 PMCID: PMC7053642 DOI: 10.18632/aging.102690] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
A role of the mitochondrial dynamin-related protein (Drp1) on gut microbiome composition and intestinal barrier function after hemorrhagic shock has not been identified previously and thus addressed in this study. Here, we used a combination of 16S rRNA gene sequencing and mass spectrometry-based metabolomics profiling in WT and Drp1 KO mouse models to examine the functional impact of activated Drp1 on the gut microbiome as well as mitochondrial metabolic regulation after hemorrhagic shock. Our data showed that changes in mitochondrial Drp1 activity participated in the regulation of intestinal barrier function after hemorrhagic shock. Activated Drp1 significantly perturbed gut microbiome composition in the Bacteroidetes phylum. The abundance of short-chain fatty acid (SCFA) producing microbes, such as Bacteroides, Butyricimonas and Odoribacter, was markedly decreased in mice after shock, and was inversely correlated with both the distribution of the tight junction protein ZO1 and intestinal permeability. Together, these data suggest that Drp1 activation perturbs the gut microbiome community and SCFA production in a ROS-specific manner and thereby substantially disturbs tight junctions and intestinal barrier function after hemorrhagic shock. Our findings provide novel insights for targeting Drp1-mediated mitochondrial function as well as the microbiome in the treatment of intestinal barrier dysfunction after shock.
Collapse
Affiliation(s)
- Chenyang Duan
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Lei Kuang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Xinming Xiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yu Zhu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yue Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Qingguang Yan
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
47
|
Husen P, Nielsen C, Martino CF, Solov'yov IA. Molecular Oxygen Binding in the Mitochondrial Electron Transfer Flavoprotein. J Chem Inf Model 2019; 59:4868-4879. [PMID: 31665600 DOI: 10.1021/acs.jcim.9b00702] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Reactive oxygen species such as superoxide are potentially harmful byproducts of the aerobic metabolism in the inner mitochondrial membrane, and complexes I, II, III of the electron transport chain have been identified as primary sources. The mitochondrial fatty acid b-oxidation pathway may also play a yet uncharacterized role in reactive oxygen species generation, apparently at the level of the electron transfer flavoprotein:ubiquinone oxidoreductase (ETF:QO) and/or its redox partner electron-transfer flavoprotein (ETF). These enzymes comprise a key pathway through which electrons are sequentially shuttled from several dehydrogenases to the respiratory chain. The exact mechanisms of superoxide production have not been fully established, but a crucial starting point would be the binding of molecular oxygen within one of the protein complexes. The present investigation offers a comprehensive computational approach for the determination of binding modes and characteristic binding times of small molecules inside proteins, which is then used to reveal several O2 binding sites near the flavin adenine dinucleotide cofactor of the ETF enzyme. The binding sites are further characterized to extract the necessary parameters for further studies of possible electron transfer between flavin and O2 leading to radical pair formation and possible superoxide production.
Collapse
Affiliation(s)
- Peter Husen
- Department of Physics, Chemistry and Pharmacy , University of Southern Denmark , Odense , Denmark
| | - Claus Nielsen
- Department of Physics, Chemistry and Pharmacy , University of Southern Denmark , Odense , Denmark
| | - Carlos F Martino
- Biomedical and Chemical Engineering and Science Department , Florida Institute of Technology , Melbourne , Florida 32901 , United States
| | - Ilia A Solov'yov
- Department of Physics , Carl von Ossietzky Universität Oldenburg , Oldenburg , Germany
| |
Collapse
|
48
|
Chen J, Xia L, Wang W, Wang Z, Hou S, Xie C, Cai J, Lu Y. Identification of a mitochondrial-targeting secretory protein from Nocardia seriolae which induces apoptosis in fathead minnow cells. JOURNAL OF FISH DISEASES 2019; 42:1493-1507. [PMID: 31482589 DOI: 10.1111/jfd.13062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 06/10/2023]
Abstract
Nocardia seriolae is the main pathogen responsible for fish nocardiosis. A mitochondrial-targeting secretory protein (MTSP) 3141 with an N-terminal transit peptide (TP) from N. seriolae was predicted by bioinformatic analysis based on the genomic sequence of the N. seriolae strain ZJ0503. However, the function of the MTSP3141 and its homologs remains totally unknown. In this study, mass spectrometry analysis of the extracellular products from N. seriolae proved that MTSP3141 was a secretory protein, subcellular localization research showed the MTSP3141-GFP fusion protein co-localized with mitochondria in fathead minnow (FHM) cells, the TP played an important role in mitochondria targeting, and only the TP located at N-terminus but not C-terminus can lead to mitochondria directing. Moreover, quantitative assays of mitochondrial membrane potential (ΔΨm) value, caspase-3 activity and apoptosis-related gene (Bcl-2, Bax, Bad, Bid and p53) mRNA expression suggested that cell apoptosis was induced in FHM cells by the overexpression of both MTSP3141 and MTSP3141ΔTP (with the N-terminal TP deleted) proteins. Taken together, the results of this study indicated that the MTSP3141 of N. seriolae was a secretory protein, might target mitochondria, induce apoptosis in host cells and function as a virulence factor.
Collapse
Affiliation(s)
- Jianlin Chen
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Liqun Xia
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Wenji Wang
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Zhiwen Wang
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Suying Hou
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Caixia Xie
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Jia Cai
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| | - Yishan Lu
- Shenzhen Institute of Guangdong Ocean University, Shenzhen City, China
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Fishery College of Guangdong Ocean University, Zhanjiang City, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen City, China
| |
Collapse
|
49
|
Maurice NM, Bedi B, Yuan Z, Goldberg JB, Koval M, Hart CM, Sadikot RT. Pseudomonas aeruginosa Induced Host Epithelial Cell Mitochondrial Dysfunction. Sci Rep 2019; 9:11929. [PMID: 31417101 PMCID: PMC6695387 DOI: 10.1038/s41598-019-47457-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/07/2019] [Indexed: 02/06/2023] Open
Abstract
The pathogenicity of P. aeruginosa is dependent on quorum sensing (QS), an inter-bacterial communication system that can also modulate host biology. The innate immune function of the lung mucosal barrier is dependent on proper mitochondrial function. The purpose of this study was to define the mechanism by which bacterial factors modulate host lung epithelial cell mitochondrial function and to investigate novel therapies that ameliorate this effect. 3-oxo-C12-HSL disrupts mitochondrial morphology, attenuates mitochondrial bioenergetics, and induces mitochondrial DNA oxidative injury. Mechanistically, we show that 3-oxo-C12-HSL attenuates expression of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a master regulator of mitochondrial biogenesis, antioxidant defense, and cellular respiration, and its downstream effectors in both BEAS-2B and primary lung epithelial cells. Overexpression of PGC-1α attenuates the inhibition in cellular respiration caused by 3-oxo-C12-HSL. Pharmacologic activation of PGC-1α restores barrier integrity in cells treated with 3-oxo-C12-HSL. These data demonstrate that the P. aeruginosa QS molecule, 3-oxo-C12-HSL, alters mitochondrial pathways critical for lung mucosal immunity. Genetic and pharmacologic strategies that activate the PGC-1α pathway enhance host epithelial cell mitochondrial function and improve the epithelial innate response to P. aeruginosa. Therapies that rescue PGC-1α function may provide a complementary approach in the treatment of P. aeruginosa 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
| | - Brahmchetna Bedi
- 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
| | - Zhihong Yuan
- 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
| | - Joanna B Goldberg
- Department of Pediatrics, Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis, and Sleep, Emory University, Atlanta, GA, 30322, USA.,Children's Healthcare of Atlanta, Center for CF and Airways Disease Research Atlanta, Atlanta, GA, USA
| | - Michael Koval
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - C Michael Hart
- 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
- 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.
| |
Collapse
|
50
|
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: 217] [Impact Index Per Article: 36.2] [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
|