1
|
Zhang X, Ding G, Yang X, Lu H, Xu Y, Hu Y, Liu S, Zhang H, Huang K, Deng G, Ye T, Yu Q, Cai Y, Xie S, Wang W, Chen X. Myriocin enhances the clearance of M. tuberculosis by macrophages through the activation of PLIN2. mSphere 2024; 9:e0025724. [PMID: 38920406 PMCID: PMC11288015 DOI: 10.1128/msphere.00257-24] [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: 03/27/2024] [Accepted: 05/19/2024] [Indexed: 06/27/2024] Open
Abstract
Myriocin is an inhibitor of de novo synthesis of sphingolipids and ceramides. In this research, we showed myriocin could significantly reduce Mtb burden and histopathological inflammation in mice. However, the underlying mechanism remains unclear. RNA-seq analysis revealed a significant increase in gene expression of PLIN2/CD36/CERT1 after myriocin treatment. The reduced bactericidal burden was only reversed after silencing the lipid droplets (LDs) surface protein PLIN2. This suggests that myriocin enhances the ability of macrophages to clear Mtb depending on the PLIN2 gene, which is part of the PPARγ pathway. Indeed, we observed a significant increase in the number of LDs following myriocin treatment.IMPORTANCEMycobacterium tuberculosis has the ability to reprogram host cell lipid metabolism and alter the antimicrobial functions of infected macrophages. The sphingolipids, such as ceramides, are the primary host lipids utilized by the bacteria, making the sphingomyelinase/ceramide system critical in Mtb infections. Surprisingly, the antimicrobial effect of myriocin was found to be independent of its role in reducing ceramides, but instead, it depends on the lipid droplets surface protein PLIN2. Our findings provide a novel mechanism for how myriocin enhances Mtb clearance in macrophages.
Collapse
Affiliation(s)
- Ximeng Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Guanggui Ding
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xirui Yang
- Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California, USA
| | - Hailin Lu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yuzhong Xu
- Department of Clinical Laboratory, Shenzhen Baoan Hospital, Shenzhen, China
| | - Yunlong Hu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Song Liu
- Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Huihua Zhang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Kaisong Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Guofang Deng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Taosheng Ye
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qing Yu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Shuixiang Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Wenfei Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
2
|
Buratta S, Urbanelli L, Pellegrino RM, Alabed HBR, Latella R, Cerrotti G, Emiliani C, Bassotti G, Spaterna A, Marconi P, Fettucciari K. PhosphoLipidome Alteration Induced by Clostridioides difficile Toxin B in Enteric Glial Cells. Cells 2024; 13:1103. [PMID: 38994956 PMCID: PMC11240607 DOI: 10.3390/cells13131103] [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: 05/29/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Clostridioides difficile (C. difficile) is responsible for a spectrum of nosocomial/antibiotic-associated gastrointestinal diseases that are increasing in global incidence and mortality rates. The C. difficile pathogenesis is due to toxin A and B (TcdA/TcdB), both causing cytopathic and cytotoxic effects and inflammation. Recently, we demonstrated that TcdB induces cytopathic and cytotoxic (apoptosis and necrosis) effects in enteric glial cells (EGCs) in a dose/time-dependent manner and described the underlying signaling. Despite the role played by lipids in host processes activated by pathogens, to counter infection and/or induce cell death, to date no studies have investigated lipid changes induced by TcdB/TcdA. Here, we evaluated the modification of lipid composition in our in vitro model of TcdB infection. Apoptosis, cell cycle, cell viability, and lipidomic profiles were evaluated in EGCs treated for 24 h with two concentrations of TcdB (0.1 ng/mL; 10 ng/mL). In EGCs treated with the highest concentration of TcdB, not only an increased content of total lipids was observed, but also lipidome changes, allowing the separation of TcdB-treated cells and controls into different clusters. The statistical analyses also allowed us to ascertain which lipid classes and lipid molecular species determine the clusterization. Changes in lipid species containing inositol as polar head and plasmalogen phosphatidylethanolamine emerged as key indicators of altered lipid metabolism in TcdB-treated EGCs. These results not only provide a picture of the phospholipid profile changes but also give information regarding the lipid metabolism pathways altered by TcdB, and this might represent an important step for developing strategies against C. difficile infection.
Collapse
Affiliation(s)
- Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Raffaella Latella
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy; (S.B.); (L.U.); (R.M.P.); (H.B.R.A.); (R.L.); (G.C.); (C.E.)
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Gabrio Bassotti
- Department of Medicine and Surgery, Gastroenterology, Hepatology & Digestive Endoscopy Section, University of Perugia, Piazzale Lucio Severi 1, 06132 Perugia, Italy;
- Santa Maria Della Misericordia Hospital, Gastroenterology & Hepatology Unit, Piazzale Menghini 1, 06129 Perugia, Italy
| | - Andrea Spaterna
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Macerata, Italy
| | - Pierfrancesco Marconi
- Department of Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Piazzale Lucio Severi 1, 06132 Perugia, Italy;
| | - Katia Fettucciari
- Department of Medicine and Surgery, Biosciences & Medical Embryology Section, University of Perugia, Piazzale Lucio Severi 1, 06132 Perugia, Italy;
| |
Collapse
|
3
|
Tan YJ, Jin Y, Zhou J, Yang YF. Lipid droplets in pathogen infection and host immunity. Acta Pharmacol Sin 2024; 45:449-464. [PMID: 37993536 PMCID: PMC10834987 DOI: 10.1038/s41401-023-01189-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023] Open
Abstract
As the hub of cellular lipid metabolism, lipid droplets (LDs) have been linked to a variety of biological processes. During pathogen infection, the biogenesis, composition, and functions of LDs are tightly regulated. The accumulation of LDs has been described as a hallmark of pathogen infection and is thought to be driven by pathogens for their own benefit. Recent studies have revealed that LDs and their subsequent lipid mediators contribute to effective immunological responses to pathogen infection by promoting host stress tolerance and reducing toxicity. In this comprehensive review, we delve into the intricate roles of LDs in governing the replication and assembly of a wide spectrum of pathogens within host cells. We also discuss the regulatory function of LDs in host immunity and highlight the potential for targeting LDs for the diagnosis and treatment of infectious diseases.
Collapse
Affiliation(s)
- Yan-Jie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Yi Jin
- Research Center of Translational Medicine, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yun-Fan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
4
|
Wang X, Wu H, Fang C, Li Z. Insights into innate immune cell evasion by Chlamydia trachomatis. Front Immunol 2024; 15:1289644. [PMID: 38333214 PMCID: PMC10850350 DOI: 10.3389/fimmu.2024.1289644] [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: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Chlamydia trachomatis, is a kind of obligate intracellular pathogen. The removal of C. trachomatis relies primarily on specific cellular immunity. It is currently considered that CD4+ Th1 cytokine responses are the major protective immunity against C. trachomatis infection and reinfection rather than CD8+ T cells. The non-specific immunity (innate immunity) also plays an important role in the infection process. To survive inside the cells, the first process that C. trachomatis faces is the innate immune response. As the "sentry" of the body, mast cells attempt to engulf and remove C. trachomatis. Dendritic cells present antigen of C. trachomatis to the "commanders" (T cells) through MHC-I and MHC-II. IFN-γ produced by activated T cells and natural killer cells (NK) further activates macrophages. They form the body's "combat troops" and produce immunity against C. trachomatis in the tissues and blood. In addition, the role of eosinophils, basophils, innate lymphoid cells (ILCs), natural killer T (NKT) cells, γδT cells and B-1 cells should not be underestimated in the infection of C. trachomatis. The protective role of innate immunity is insufficient, and sexually transmitted diseases (STDs) caused by C. trachomatis infections tend to be insidious and recalcitrant. As a consequence, C. trachomatis has developed a unique evasion mechanism that triggers inflammatory immunopathology and acts as a bridge to protective to pathological adaptive immunity. This review focuses on the recent advances in how C. trachomatis evades various innate immune cells, which contributes to vaccine development and our understanding of the pathophysiologic consequences of C. trachomatis infection.
Collapse
Affiliation(s)
| | | | | | - Zhongyu Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, School of Nursing, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
5
|
Rodríguez-Fernández P, Botella L, Cavet JS, Domínguez J, Gutierrez MG, Suckling CJ, Scott FJ, Tabernero L. MptpB Inhibitor Improves the Action of Antibiotics against Mycobacterium tuberculosis and Nontuberculous Mycobacterium avium Infections. ACS Infect Dis 2024; 10:170-183. [PMID: 38085851 PMCID: PMC10788870 DOI: 10.1021/acsinfecdis.3c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024]
Abstract
Treatment of Mycobacterium tuberculosis and Mycobacterium avium infections requires multiple drugs for long time periods. Mycobacterium protein-tyrosine-phosphatase B (MptpB) is a key M. tuberculosis virulence factor that subverts host antimicrobial activity to promote intracellular survival. Inhibition of MptpB reduces the infection burden in vivo and offers new opportunities to improve current treatments. Here, we demonstrate that M. avium produces an MptpB orthologue and that the MptpB inhibitor C13 reduces the M. avium infection burden in macrophages. Combining C13 with the antibiotics rifampicin or bedaquiline showed an additive effect, reducing intracellular infection of both M. tuberculosis and M. avium by 50%, compared to monotreatment with antibiotics alone. This additive effect was not observed with pretomanid. Combining C13 with the minor groove-binding compounds S-MGB-362 and S-MGB-363 also reduced the M. tuberculosis intracellular burden. Similar additive effects of C13 and antibiotics were confirmed in vivo using Galleria mellonella infections. We demonstrate that the reduced mycobacterial burden in macrophages observed with C13 treatments is due to the increased trafficking to lysosomes.
Collapse
Affiliation(s)
- Pablo Rodríguez-Fernández
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health
Science Centre, M13 9PT Manchester, U.K.
| | - Laure Botella
- Host
Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, NW1 1AT London, U.K.
| | - Jennifer S. Cavet
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health
Science Centre, M13 9PT Manchester, U.K.
- Lydia
Becker Institute for Immunology and Inflammation, University of Manchester, M13 9PT Manchester, U.K.
| | - Jose Domínguez
- Institut
d’Investigació Germans Trias i Pujol, CIBER Enfermedades
Respiratorias (CIBERES), Universitat Autònoma
de Barcelona, 08916 Barcelona, Spain
| | - Maximiliano G. Gutierrez
- Host
Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, NW1 1AT London, U.K.
| | - Colin J. Suckling
- Department
of Pure and Applied Chemistry, University
of Strathclyde, 295 Cathedral Street, G1 1XL Glasgow, U.K.
| | - Fraser J. Scott
- Department
of Pure and Applied Chemistry, University
of Strathclyde, 295 Cathedral Street, G1 1XL Glasgow, U.K.
| | - Lydia Tabernero
- School
of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health
Science Centre, M13 9PT Manchester, U.K.
- Lydia
Becker Institute for Immunology and Inflammation, University of Manchester, M13 9PT Manchester, U.K.
| |
Collapse
|
6
|
Araújo M, Moreira D, Mesquita I, Ferreira C, Mendes-Frias A, Barros-Carvalho S, Dinis-Oliveira RJ, Duarte-Oliveira C, Cunha C, Carvalho A, Saha B, Cordeiro-da-Silva A, Estaquier J, Silvestre R. Intramacrophage lipid accumulation compromises T cell responses and is associated with impaired drug therapy against visceral leishmaniasis. Immunology 2023; 170:510-526. [PMID: 37635289 DOI: 10.1111/imm.13686] [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: 02/03/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Under perturbing conditions such as infection with Leishmania, a protozoan parasite living within the phagosomes in mammalian macrophages, cellular and organellar structures, and metabolism are dynamically regulated for neutralizing the pressure of parasitism. However, how modulations of the host cell metabolic pathways support Leishmania infection remains unknown. Herein, we report that lipid accumulation heightens the susceptibility of mice to L. donovani infection and promotes resistance to first-line anti-leishmanial drugs. Despite being pro-inflammatory, the in vitro generated uninfected lipid-laden macrophages (LLMs) or adipose-tissue macrophages (ATMs) display lower levels of reactive oxygen and nitrogen species. Upon infection, LLMs secrete higher IL-10 and lower IL-12p70 cytokines, inhibiting CD4+ T cell activation and Th1 response suggesting a key modulatory role for intramacrophage lipid accumulation in anti-leishmanial host defence. We, therefore, examined this causal relationship between lipids and immunomodulation using an in vivo high-fat diet (HFD) mouse model. HFD increased the susceptibility to L. donovani infection accompanied by a defective CD4+ Th1 and CD8+ T cell response. The white adipose tissue of HFD mice displays increased susceptibility to L. donovani infection with the preferential infection of F4/80+ CD11b+ CD11c+ macrophages with higher levels of neutral lipids reserve. The HFD increased resistance to a first-line anti-leishmanial drug associated with a defective adaptive immune response. These data demonstrate that the accumulation of neutral lipids contributes to susceptibility to visceral leishmaniasis hindering host-protective immune response and reducing the efficacy of antiparasitic drug therapies.
Collapse
Affiliation(s)
- Marta Araújo
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diana Moreira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Inês Mesquita
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carolina Ferreira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Mendes-Frias
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sónia Barros-Carvalho
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- TOXRUN - Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, Porto, Portugal
- MTG Research and Development Lab, Porto, Portugal
| | - Cláudio Duarte-Oliveira
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | - Anabela Cordeiro-da-Silva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Parasite Disease Group, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Biológicas, Faculdade de Farmácia da Universidade do Porto (FFUP), Porto, Portugal
| | - Jérôme Estaquier
- INSERM U1124, Université Paris Cité, Paris, France
- Pathophysiology of Cell Death in Host-Pathogen Interactions, CHU de Québec - Université Laval Research Center, Québec City, Québec, Canada
| | - Ricardo Silvestre
- Immunobiology of Inflammatory and Infectious Diseases (i3D), Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
7
|
Prado LG, Camara NOS, Barbosa AS. Cell lipid biology in infections: an overview. Front Cell Infect Microbiol 2023; 13:1148383. [PMID: 37868347 PMCID: PMC10587689 DOI: 10.3389/fcimb.2023.1148383] [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: 01/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Lipids are a big family of molecules with a vast number of functions in the cell membranes, within the cytoplasm, and extracellularly. Lipid droplets (LDs) are the most common storage organelles and are present in almost every tissue type in the body. They also have structural functions serving as building blocks of cellular membranes and may be precursors of other molecules such as hormones, and lipoproteins, and as messengers in signal transduction. Fatty acids (FAs), such as sterol esters and triacylglycerols, are stored in LDs and are used in β-oxidation as fuel for tricarboxylic acid cycle (TCA) and adenosine triphosphate (ATP) generation. FA uptake and entrance in the cytoplasm are mediated by membrane receptors. After a cytoplasmic round of α- and β-oxidation, FAs are guided into the mitochondrial matrix by the L-carnitine shuttle system, where they are fully metabolized, and enter the TCA cycle. Pathogen infections may lead to impaired lipid metabolism, usage of membrane phospholipids, and LD accumulation in the cytoplasm of infected cells. Otherwise, bacterial pathogens may use lipid metabolism as a carbon source, thus altering the reactions and leading to cellular and organelles malfunctioning. This review aims to describe cellular lipid metabolism and alterations that occur upon infections.
Collapse
Affiliation(s)
- Luan Gavião Prado
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
8
|
Petsana M, Roumia AF, Bagos PG, Boleti H, Braliou GG. In Silico Identification and Analysis of Proteins Containing the Phox Homology Phosphoinositide-Binding Domain in Kinetoplastea Protists: Evolutionary Conservation and Uniqueness of Phox-Homology-Domain-Containing Protein Architectures. Int J Mol Sci 2023; 24:11521. [PMID: 37511280 PMCID: PMC10380299 DOI: 10.3390/ijms241411521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/27/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Kinetoplastea are free living and parasitic protists with unique features among Eukaryota. Pathogenic Kinetoplastea parasites (i.e., Trypanosoma and Leishmania spp.) undergo several developmental transitions essential for survival in their hosts. These transitions require membrane and cytoskeleton reorganizations that involve phosphoinositides (PIs). Phospholipids like PIs are key regulators of vital functions in all eukaryotes including signal transduction, protein transport and sorting, membrane trafficking, and cytoskeleton and membrane remodeling. A large repertoire of PI-metabolizing enzymes and PI-binding proteins/effectors carrying distinct PI-binding modules like the PX (phox homology) module could play significant roles in the life and virulence of pathogenic Kinetoplastea. The aim of this study was to retrieve the entire spectrum of Kinetoplastea protein sequences containing the PX module (PX-proteins), predict their structures, and identify in them evolutionary conserved and unique traits. Using a large array of bioinformatics tools, protein IDs from two searches (based on PFam's pHMM for PX domain (PF00787)) were combined, aligned, and utilized for the construction of a new Kinetoplastea_PX pHMM. This three-step search retrieved 170 PX-protein sequences. Structural domain configuration analysis identified PX, Pkinase, Lipocalin_5, and Vps5/BAR3-WASP domains and clustered them into five distinct subfamilies. Phylogenetic tree and domain architecture analysis showed that some domain architectures exist in proteomes of all Kinetoplastea spp., while others are genus-specific. Finally, amino acid conservation logos of the Kinetoplastea spp. and Homo sapiens PX domains revealed high evolutionary conservation in residues forming the critical structural motifs for PtdIns3P recognition. This study highlights the PX-Pkinase domain architecture as unique within Trypanosoma spp. and forms the basis for a targeted functional analysis of Kinetoplastea PX-proteins as putative targets for a rational design of anti-parasitic drugs.
Collapse
Affiliation(s)
- Marina Petsana
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Ahmed F Roumia
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
- Department of Agricultural Biochemistry, Faculty of Agriculture, Menoufia University, Shibin El-Kom 32514, Egypt
| | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
| | - Haralabia Boleti
- Intracellular Parasitism Laboratory, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Georgia G Braliou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 2-4 Papasiopoulou Str., 35131 Lamia, Greece
| |
Collapse
|
9
|
González-Escalada A, Rebollo MJ, Barrios Payan J, Hernández-Pando R, García MJ. Detection of Mycobacterial DNA in Human Bone Marrow. Microorganisms 2023; 11:1788. [PMID: 37512960 PMCID: PMC10384717 DOI: 10.3390/microorganisms11071788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Bone marrow is a cell-rich tissue of the reticuloendothelial system essential in the homeostasis and accurate functioning of hematopoiesis and of the immune system; moreover, it is also rich in lipids because it contains marrow adipocytes. This work aimed to evaluate the detection of mycobacterial DNA in human bone marrow as a tool to understand the complex pathology caused by the main pathogen Mycobacterium tuberculosis (Mtb). Formalin-fixed paraffin-embedded human bone marrow samples were studied using both conventional PCR + hybridization and in situ PCR to figure out the cell distribution of the targeted DNA. Samples were retrospectively collected from HIV+ patients with microbiologically proved mycobacterial infection and from subjects without evidence of infection. Mycobacterium avium (Mav) as well as Mtb DNA was detected in both settings, including tissues with and without granulomas. We detected DNA from both mycobacterial species, using in situ PCR, inside bone marrow macrophages. Other cell types, including adipocytes, showed positive signals only for Mtb DNA. This result suggested, for the first time, that marrow adipocytes could constitute an ideal reservoir for the persistence of Mtb, allowing the bacilli to establish long-lasting latent infection within a suitable lipid environment. This fact might differentiate pathogenic behavior of non-specialized pathogens such as Mav from that of specialized pathogens such as Mtb.
Collapse
Affiliation(s)
- Alba González-Escalada
- Facultad de Ciencias de la Salud, Area of Medical Microbiology, Rey Juan Carlos University, 28922 Alcorcon, Spain
| | - María José Rebollo
- Department of Preventive Medicine and Public Health and Microbiology, School of Medicine, Autonoma University of Madrid, 28029 Madrid, Spain
| | - Jorge Barrios Payan
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubirán, México City 14080, Mexico
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, Department of Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubirán, México City 14080, Mexico
| | - María Jesús García
- Department of Preventive Medicine and Public Health and Microbiology, School of Medicine, Autonoma University of Madrid, 28029 Madrid, Spain
| |
Collapse
|
10
|
Zhang QA, Ma S, Li P, Xie J. The dynamics of Mycobacterium tuberculosis phagosome and the fate of infection. Cell Signal 2023; 108:110715. [PMID: 37192679 DOI: 10.1016/j.cellsig.2023.110715] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/25/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023]
Abstract
Phagosomes are vesicles produced by phagocytosis of phagocytes, which are crucial in immunity against Mycobacterium tuberculosis (Mtb) infection. After the phagocyte ingests the pathogen, it activates the phagosomes to recruit a series of components and process proteins, to phagocytose, degrade and kill Mtb. Meanwhile, Mtb can resist acid and oxidative stress, block phagosome maturation, and manipulate host immune response. The interaction between Mtb and phagocytes leads to the outcome of infection. The dynamic of this process can affect the cell fate. This article mainly reviews the development and maturation of phagosomes, as well as the dynamics and modifications of Mtb effectors and phagosomes components, and new diagnostic and therapeutic markers involved in phagosomes.
Collapse
Affiliation(s)
- Qi-Ao Zhang
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Shaying Ma
- Chongqing Emergency Medical Center, Chongqing the Fourth Hospital, Jiankang Road, Yuzhong, Chongqing 400014, China
| | - Peibo Li
- Chongqing Public Health Medical Center, Chongqing, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China; Chongqing Public Health Medical Center, Chongqing, China.
| |
Collapse
|
11
|
Aguilera-Herce J, Panadero-Medianero C, Sánchez-Romero MA, Balbontín R, Bernal-Bayard J, Ramos-Morales F. Salmonella Type III Secretion Effector SrfJ: A Glucosylceramidase Affecting the Lipidome and the Transcriptome of Mammalian Host Cells. Int J Mol Sci 2023; 24:ijms24098403. [PMID: 37176110 PMCID: PMC10179164 DOI: 10.3390/ijms24098403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Type III secretion systems are found in many Gram-negative pathogens and symbionts of animals and plants. Salmonella enterica has two type III secretion systems associated with virulence, one involved in the invasion of host cells and another involved in maintaining an appropriate intracellular niche. SrfJ is an effector of the second type III secretion system. In this study, we explored the biochemical function of SrfJ and the consequences for mammalian host cells of the expression of this S. enterica effector. Our experiments suggest that SrfJ is a glucosylceramidase that alters the lipidome and the transcriptome of host cells, both when expressed alone in epithelial cells and when translocated into macrophages in the context of Salmonella infection. We were able to identify seventeen lipids with higher levels and six lipids with lower levels in the presence of SrfJ. Analysis of the forty-five genes, the expression of which is significantly altered by SrfJ with a fold-change threshold of two, suggests that this effector may be involved in protecting Salmonella from host immune defenses.
Collapse
Affiliation(s)
- Julia Aguilera-Herce
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Avda Reina Mercedes, 6, 41012 Sevilla, Spain
| | - Concepción Panadero-Medianero
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Avda Reina Mercedes, 6, 41012 Sevilla, Spain
| | - María Antonia Sánchez-Romero
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, Avda Reina Mercedes, 6, 41012 Sevilla, Spain
| | - Roberto Balbontín
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Avda Reina Mercedes, 6, 41012 Sevilla, Spain
| | - Joaquín Bernal-Bayard
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Avda Reina Mercedes, 6, 41012 Sevilla, Spain
| | - Francisco Ramos-Morales
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Avda Reina Mercedes, 6, 41012 Sevilla, Spain
| |
Collapse
|
12
|
Choudhury A, Saha S, Maiti NC, Datta S. Exploring structural features and potential lipid interactions of Pseudomonas aeruginosa type three secretion effector PemB by spectroscopic and calorimetric experiments. Protein Sci 2023; 32:e4627. [PMID: 36916835 PMCID: PMC10044109 DOI: 10.1002/pro.4627] [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: 10/21/2022] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/15/2023]
Abstract
Type Three Secretion System (T3SS) is a sophisticated nano-scale weapon utilized by several gram negative bacteria under stringent spatio-temporal regulation to manipulate and evade host immune systems in order to cause infection. To the best of our knowledge, this present study is the first report where we embark upon characterizing inherent features of native type three secretion effector protein PemB through biophysical techniques. Herein, first, we demonstrate binding affinity of PemB for phosphoinositides through isothermal calorimetric titrations. Second, we shed light on its strong homo-oligomerization propensity in aqueous solution through multiple biophysical methods. Third, we also employ several spectroscopic techniques to delineate its disordered and helical conformation. Lastly, we perform a phylogenetic analysis of this new effector to elucidate evolutionary relationship with other organisms. Taken together, our results shall surely contribute to our existing knowledge of Pseudomonas aeruginosa secretome.
Collapse
Affiliation(s)
- Arkaprabha Choudhury
- Department of Structural Biology and BioinformaticsCSIR‐Indian Institute of Chemical Biology (CSIR‐IICB)Kolkata700032India
- Biological SciencesAcademy of Scientific and Innovative Research (AcSIR)201002GhaziabadIndia
| | - Saumen Saha
- Department of Structural Biology and BioinformaticsCSIR‐Indian Institute of Chemical Biology (CSIR‐IICB)Kolkata700032India
| | - Nakul Chandra Maiti
- Department of Structural Biology and BioinformaticsCSIR‐Indian Institute of Chemical Biology (CSIR‐IICB)Kolkata700032India
- Biological SciencesAcademy of Scientific and Innovative Research (AcSIR)201002GhaziabadIndia
| | - Saumen Datta
- Department of Structural Biology and BioinformaticsCSIR‐Indian Institute of Chemical Biology (CSIR‐IICB)Kolkata700032India
- Biological SciencesAcademy of Scientific and Innovative Research (AcSIR)201002GhaziabadIndia
| |
Collapse
|
13
|
Ji H, Li T, Li X, Li J, Yu J, Zhang X, Liu D. XopZ and ORP1C cooperate to regulate the virulence of Xanthomonas oryzae pv. oryzae on Nipponbare. PLANT SIGNALING & BEHAVIOR 2022; 17:2035126. [PMID: 35184695 PMCID: PMC8959505 DOI: 10.1080/15592324.2022.2035126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 06/14/2023]
Abstract
Bacterial leaf blight caused by Xanthomonas oryzae pv. oryzae (Xoo) has always been considered to be one of the most severe worldwide diseases in rice. Xoo strains usually use the highly conserved type III secretion system (T3SS) to deliver virulence effectors into rice cells and further suppress the host's immunity. Previous studies reported that different Xanthomonas outer protein (Xop) effectors include XopZ from one strain appear to share functional redundancies on suppressing rice PAMP-triggered immunity (PTI). But only xopZ, except other xop genes, could significantly impaire Xoo virulence when individually deleting in PXO99 strains. Thus, the XopZ effector should not only suppress rice PTI pathway, but also has other unknown indispensable pathological functions in PXO99-rice interactions. Here, we also found that ∆xopZ mutant strains displayed lower virulence on Nipponbare leaves compared with PXO99 strains. We identified an oxysterol-binding related protein, ORP1C, as a XopZ-interacting protein in rice. Further studies found that rice ORP1C preliminarily played a positive role in regulating the resistance to PXO99 strains, and XopZ-ORP1C interactions cooperated to regulate the compatible interactions of PXO99-Nipponbare rice. The reactive oxygen species (ROS) burst and PTI marker gene expression data indicated that ORP1C were not directly relevant to the PTI pathway in rice. The deeper mechanisms underlying XopZ-ORP1C interaction and how XopZ and ORP1C cooperate for regulating the PXO99-rice interactions require further exploration.
Collapse
Affiliation(s)
- Hongtao Ji
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Taoran Li
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Xiaochen Li
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Jiangyu Li
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Jiayi Yu
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Xin Zhang
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Delong Liu
- Institute of Integrative Plant Biology, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| |
Collapse
|
14
|
Campo-Pérez V, Guallar-Garrido S, Luquin M, Sánchez-Chardi A, Julián E. The High Plasticity of Nonpathogenic Mycobacterium brumae Induces Rapid Changes in Its Lipid Profile during Pellicle Maturation: The Potential of This Bacterium as a Versatile Cell Factory for Lipid Compounds of Therapeutic Interest. Int J Mol Sci 2022; 23:13609. [PMID: 36362396 PMCID: PMC9655737 DOI: 10.3390/ijms232113609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 09/08/2024] Open
Abstract
The immunomodulatory potential of mycobacteria to be used for therapeutic purposes varies by species and culture conditions and is closely related to mycobacterial lipid composition. Although the lipids present in the mycobacterial cell wall are relevant, lipids are mainly stored in intracellular lipid inclusions (ILIs), which have emerged as a crucial structure in understanding mycobacteria-host interaction. Little is known about ILI ultrastructure, production, and composition in nonpathogenic species. In this study, we compared the lipid profiles of the nonpathogenic immunomodulatory agent Mycobacterium brumae during pellicle maturation under different culture conditions with qualitative and quantitative approaches by using high-resolution imaging and biochemical and composition analyses to understand ILI dynamics. The results showed wax esters, mainly in early stages of development, and acylglycerols in mature ILI composition, revealing changes in dynamics, amount, and morphometry, depending on pellicle maturation and the culture media used. Low-glycerol cultures induced ILIs with lower molecular weights which were smaller in size in comparison with the ILIs produced in glycerol-enriched media. The data also indicate the simple metabolic plasticity of lipid synthesis in M. brumae, as well as its high versatility in generating different lipid profiles. These findings provide an interesting way to enhance the production of key lipid structures via the simple modulation of cell culture conditions.
Collapse
Affiliation(s)
- Víctor Campo-Pérez
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Bacterial Infections and Antimicrobial Therapy Group, Institute for Bioengineering of Catalonia (IBEC), Baldiri Reixac 15-21, 08028 Barcelona, Spain
| | - Sandra Guallar-Garrido
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Marina Luquin
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Alejandro Sánchez-Chardi
- Servei de Microscòpia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Esther Julián
- Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
15
|
Tsujikawa R, Thapa J, Okubo T, Nakamura S, Zhang S, Furuta Y, Higashi H, Yamaguchi H. Chlamydia trachomatis L2/434/Bu Favors Hypoxia for its Growth in Human Lymphoid Jurkat Cells While Maintaining Production of Proinflammatory Cytokines. Curr Microbiol 2022; 79:265. [PMID: 35859064 DOI: 10.1007/s00284-022-02961-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/27/2022] [Indexed: 11/03/2022]
Abstract
The role of lymphocytes as a cornerstone of the inflammatory response in the invasive pathogenesis of Chlamydia trachomatis (Ct) LGV (L1-3) infection is unclear. Therefore, we assessed whether the adaptation of CtL2 to immortal lymphoid Jurkat cells under hypoxic conditions occurred through proinflammatory cytokine profile modification. The quantities of inclusion-forming units with chlamydial 16S rDNA confirmed that CtL2 grew well under hypoxic rather than normoxic conditions in the cells. Confocal microscopic imaging and transmission electron microscopy revealed the presence of bacterial progeny in the inclusions and showed that the inclusions were larger under hypoxic rather than normoxic conditions; this was supported by the results of 3D image construction. Furthermore, PCR-based analysis of proinflammatory cytokines revealed that the gene expression levels under hypoxic conditions were significantly higher than those under normoxic conditions. In particular, the expression of two genes (CXCL8 and CXCR3) was significantly diminished under normoxic conditions. Taken together, the results indicated that hypoxia promoted CtL2 growth in Jurkat cells while maintaining the levels of proinflammatory cytokines. Thus, Ct LGV infection in lymphocytes under hypoxic conditions might be crucial to a complete understanding of the invasive pathogenesis.
Collapse
Affiliation(s)
- Ryoya Tsujikawa
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan
| | - Jeewan Thapa
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo, 001-0020, Japan
| | - Torahiko Okubo
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan
| | - Shinji Nakamura
- Division of Biomedical Imaging Research, and Division of Ultrastructural Research, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Saicheng Zhang
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan
| | - Yoshikazu Furuta
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo, 001-0020, Japan
| | - Hideaki Higashi
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo, 001-0020, Japan
| | - Hiroyuki Yamaguchi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
16
|
Chen X, Ye J, Lei H, Wang C. Novel Potential Diagnostic Serum Biomarkers of Metabolomics in Osteoarticular Tuberculosis Patients: A Preliminary Study. Front Cell Infect Microbiol 2022; 12:827528. [PMID: 35402287 PMCID: PMC8992656 DOI: 10.3389/fcimb.2022.827528] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/21/2022] [Indexed: 11/17/2022] Open
Abstract
Osteoarticular tuberculosis is one of the extrapulmonary tuberculosis, which is mainly caused by direct infection of Mycobacterium tuberculosis or secondary infection of tuberculosis in other parts. Due to the low specificity of the current detection method, it is leading to a high misdiagnosis rate and subsequently affecting the follow-up treatment and prognosis. Metabolomics is mainly used to study the changes of the body’s metabolites in different states, so it can serve as an important means in the discovery of disease-related metabolic biomarkers and the corresponding mechanism research. Liquid chromatography tandem mass spectrometry (LC-MS/MS) was used to detect and analyze metabolites in the serum with osteoarticular tuberculosis patients, disease controls, and healthy controls to find novel metabolic biomarkers that could be used in the diagnosis of osteoarticular tuberculosis. Our results showed that 68 differential metabolites (p<0.05, fold change>1.0) were obtained in osteoarticular tuberculosis serum after statistical analysis. Then, through the evaluation of diagnostic efficacy, PC[o-16:1(9Z)/18:0], PC[20:4(8Z,11Z,14Z,17Z)/18:0], PC[18:0/22:5(4Z,7Z,10Z,13Z,16Z)], SM(d18:1/20:0), and SM[d18:1/18:1(11Z)] were found as potential biomarkers with high diagnostic efficacy. Using bioinformatics analysis, we further found that these metabolites share many lipid metabolic signaling pathways, such as choline metabolism, sphingolipid signaling, retrograde endocannabinoid signaling, and sphingolipid and glycerophospholipid metabolism; these results suggest that lipid metabolism plays an important role in the pathological process of tuberculosis. This study can provide certain reference value for the study of metabolic biomarkers of osteoarticular tuberculosis and the mechanism of lipid metabolism in osteoarticular tuberculosis and even other tuberculosis diseases.
Collapse
Affiliation(s)
- Ximeng Chen
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Clinical Laboratory Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Jingyun Ye
- Department of Clinical Laboratory Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hong Lei
- Department of Clinical Laboratory Medicine, The Eighth Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Chengbin Wang, ; Hong Lei,
| | - Chengbin Wang
- Department of Clinical Laboratory Medicine, The First Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Chengbin Wang, ; Hong Lei,
| |
Collapse
|
17
|
Abstract
Phosphoinositides are signalling lipids derived from phosphatidylinositol, a ubiquitous phospholipid in the cytoplasmic leaflet of eukaryotic membranes. Initially discovered for their roles in cell signalling, phosphoinositides are now widely recognized as key integrators of membrane dynamics that broadly impact on all aspects of cell physiology and on disease. The past decade has witnessed a vast expansion of our knowledge of phosphoinositide biology. On the endocytic and exocytic routes, phosphoinositides direct the inward and outward flow of membrane as vesicular traffic is coupled to the conversion of phosphoinositides. Moreover, recent findings on the roles of phosphoinositides in autophagy and the endolysosomal system challenge our view of lysosome biology. The non-vesicular exchange of lipids, ions and metabolites at membrane contact sites in between organelles has also been found to depend on phosphoinositides. Here we review our current understanding of how phosphoinositides shape and direct membrane dynamics to impact on cell physiology, and provide an overview of emerging concepts in phosphoinositide regulation.
Collapse
|
18
|
Vaughn B, Abu Kwaik Y. Idiosyncratic Biogenesis of Intracellular Pathogens-Containing Vacuoles. Front Cell Infect Microbiol 2021; 11:722433. [PMID: 34858868 PMCID: PMC8632064 DOI: 10.3389/fcimb.2021.722433] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
While most bacterial species taken up by macrophages are degraded through processing of the bacteria-containing vacuole through the endosomal-lysosomal degradation pathway, intravacuolar pathogens have evolved to evade degradation through the endosomal-lysosomal pathway. All intra-vacuolar pathogens possess specialized secretion systems (T3SS-T7SS) that inject effector proteins into the host cell cytosol to modulate myriad of host cell processes and remodel their vacuoles into proliferative niches. Although intravacuolar pathogens utilize similar secretion systems to interfere with their vacuole biogenesis, each pathogen has evolved a unique toolbox of protein effectors injected into the host cell to interact with, and modulate, distinct host cell targets. Thus, intravacuolar pathogens have evolved clear idiosyncrasies in their interference with their vacuole biogenesis to generate a unique intravacuolar niche suitable for their own proliferation. While there has been a quantum leap in our knowledge of modulation of phagosome biogenesis by intravacuolar pathogens, the detailed biochemical and cellular processes affected remain to be deciphered. Here we discuss how the intravacuolar bacterial pathogens Salmonella, Chlamydia, Mycobacteria, Legionella, Brucella, Coxiella, and Anaplasma utilize their unique set of effectors injected into the host cell to interfere with endocytic, exocytic, and ER-to-Golgi vesicle traffic. However, Coxiella is the main exception for a bacterial pathogen that proliferates within the hydrolytic lysosomal compartment, but its T4SS is essential for adaptation and proliferation within the lysosomal-like vacuole.
Collapse
Affiliation(s)
- Bethany Vaughn
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States.,Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
19
|
Wang L, Lin J, Yu J, Yang K, Sun L, Tang H, Pan L. Downregulation of Perilipin1 by the Immune Deficiency Pathway Leads to Lipid Droplet Reconfiguration and Adaptation to Bacterial Infection in Drosophila. THE JOURNAL OF IMMUNOLOGY 2021; 207:2347-2358. [PMID: 34588219 DOI: 10.4049/jimmunol.2100343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/26/2021] [Indexed: 11/19/2022]
Abstract
Lipid droplets (LDs), the highly dynamic intracellular organelles, are critical for lipid metabolism. Dynamic alterations in the configurations and functions of LDs during innate immune responses to bacterial infections and the underlying mechanisms, however, remain largely unknown. In this study, we trace the time-course morphology of LDs in fat bodies of Drosophila after transient bacterial infection. Detailed analysis shows that perilipin1 (plin1), a core gene involved in the regulation of LDs, is suppressed by the immune deficiency signaling, one major innate immune pathway in Drosophila During immune activation, downregulated plin1 promotes the enlargement of LDs, which in turn alleviates immune reaction-associated reactive oxygen species stress. Thus, the growth of LDs is likely an active adaptation to maintain redox homeostasis in response to immune deficiency activation. Therefore, our study provides evidence that plin1 serves as a modulator on LDs' reconfiguration in regulating infection-induced pathogenesis, and plin1 might be a potential therapeutic target for coordinating inflammation resolution and lipid metabolism.
Collapse
Affiliation(s)
- Lei Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.,The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiaxin Lin
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Junjing Yu
- Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China; and
| | - Kaiyan Yang
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Li Sun
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Hong Tang
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China;
| | - Lei Pan
- The Center for Microbes, Development, and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China; .,University of Chinese Academy of Sciences, Beijing, China.,Chinese Academy of Sciences Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
20
|
Fountain A, Inpanathan S, Alves P, Verdawala MB, Botelho RJ. Phagosome maturation in macrophages: Eat, digest, adapt, and repeat. Adv Biol Regul 2021; 82:100832. [PMID: 34717137 DOI: 10.1016/j.jbior.2021.100832] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 11/30/2022]
Abstract
Phagocytosis is a dynamic process that requires an intricate interplay between phagocytic receptors, membrane lipids, and numerous signalling proteins and their effectors, to coordinate the engulfment of a bound particle. These particles are diverse in their physico-chemical properties such as size and shape and include bacteria, fungi, apoptotic cells, living tumour cells, and abiotic particles. Once engulfed, these particles are enclosed within a phagosome, which undergoes a striking transformation referred to as phagosome maturation, which will ultimately lead to the processing and degradation of the enclosed particulate. In this review, we focus on recent advancements in phagosome maturation in macrophages, highlighting new discoveries and emerging themes. Such advancements include identification of new GTPases and their effectors and the intricate spatio-temporal dynamics of phosphoinositides in governing phagosome maturation. We then explore phagosome fission and recycling, the emerging role of membrane contact sites, and delve into mechanisms of phagosome resolution to recycle and reform lysosomes. We further illustrate how phagosome maturation is context-dependent, subject to the type of particle, phagocytic receptors, the phagocytes and their state of activation during phagocytosis. Lastly, we discuss how phagosomes serve as signalling platforms to help phagocytes adapt to their environmental conditions. Overall, this review aims to cover recent findings, identify emerging themes, and highlight current challenges and directions to improve our understanding of phagosome maturation in macrophages.
Collapse
Affiliation(s)
- Aaron Fountain
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Subothan Inpanathan
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Patris Alves
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Munira B Verdawala
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada
| | - Roberto J Botelho
- Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada; Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario, M5B2K3, Canada.
| |
Collapse
|
21
|
Sirisin J, Kamnate A, Polsan Y, Somintara S, Chomphoo S, Sakagami H, Kondo H, Hipkaeo W. Localization of phosphatidylinositol 4-phosphate 5-kinase (PIP5K) α confined to the surface of lipid droplets and adjacent narrow cytoplasm in progesterone-producing cells of in situ ovaries of adult mice. Acta Histochem 2021; 123:151794. [PMID: 34624591 DOI: 10.1016/j.acthis.2021.151794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/15/2022]
Abstract
Phosphatidylinositol(4,5)bisphosphate (PI(4,5)P2) produced by phosphatidylinositol phosphate 5 kinase (PIP5K) plays not only as a precursor of second messengers in the phosphoinositide signal transduction, but also multiple roles influencing a variety of cellular activities. From this viewpoint, the present study attempted to localize PIP5Kα in the ovaries in situ of adult mice. PIP5Kα-immunoreactivity was confined to the surfaces of lipid droplets (LDs) and their adjacent cytoplasm in progesterone-producing cells of the interstitial glands, corpora lutea and theca interna. The LDs often contained membranous tubules/lamellae along their surfaces and within their interior whose membranes were continuous with those delineating LDs composed of a monolayer of phospholipids and were partially PIP5Kα-immunoreactive. Although granulosa cells of healthy-looking follicles were immunonegative, as the atresia progressed, PIP5Kα-immunoreactivity first appeared in sparsely dispersed dot forms in mural cells of the follicular epithelia, and then were dominant in almost all mural cells that remained after desquamation of the antral cells. The present study provides evidence suggesting that PI(4,5)P2 locally synthesized by PIP5K in LDs is involved in the lipid transfer between lipid droplets (LDs) and the endoplasmic reticulum, which eventually regulates ovarian progesterone production through control of multiple dynamic activities of LDs. It is also suggested that PIP5Kα and PI(4,5)P2 are implicated in the modulation of programmed cell death and/or acquiring the ability of progesterone production in some follicular cells surviving atresia.
Collapse
Affiliation(s)
- Juthathip Sirisin
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Anussara Kamnate
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Yada Polsan
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Somsuda Somintara
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Surang Chomphoo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hiroyuki Sakagami
- Department of Anatomy, School of Medicine, Kitasato University, Sagamihara, Japan
| | - Hisatake Kondo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand; Department of Anatomy, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Wiphawi Hipkaeo
- Electron Microscopy Unit, Department of Anatomy, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
22
|
Allen PE, Noland RC, Martinez JJ. Rickettsia conorii survival in THP-1 macrophages involves host lipid droplet alterations and active rickettsial protein production. Cell Microbiol 2021; 23:e13390. [PMID: 34464019 DOI: 10.1111/cmi.13390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/29/2022]
Abstract
Rickettsia conorii is a Gram-negative, cytosolic intracellular bacterium that has classically been investigated in terms of endothelial cell infection. However, R. conorii and other human pathogenic Rickettsia species have evolved mechanisms to grow in various cell types, including macrophages, during mammalian infection. During infection of these phagocytes, R. conorii shifts the host cell's overall metabolism towards an anti-inflammatory M2 response, metabolically defined by an increase in host lipid metabolism and oxidative phosphorylation. Lipid metabolism has more recently been identified as a key regulator of host homeostasis through modulation of immune signalling and metabolism. Intracellular pathogens have adapted mechanisms of hijacking host metabolic pathways including host lipid catabolic pathways for various functions required for growth and survival. In the present study, we hypothesised that alterations of host lipid droplets initiated by lipid catabolic pathways during R. conorii infection is important for bacterial survival in macrophages. Herein, we determined that host lipid droplet modulation is initiated early during R. conorii infection, and these alterations rely on active bacteria and lipid catabolic pathways. We also find that these lipid catabolic pathways are essential for efficient bacterial survival. Unlike the mechanisms used by other intracellular pathogens, the catabolism of lipid droplets induced by R. conorii infection is independent of upstream host peroxisome proliferator-activated receptor-alpha (PPARα) signalling. Inhibition of PPARɣ signalling and lipid droplet accumulation in host cells cause a significant decrease in R. conorii survival suggesting a negative correlation with lipid droplet production and R. conorii survival. Together, these results strongly suggest that the modulation of lipid droplets in macrophage cells infected by R. conorii is an important and underappreciated aspect of the infection process. TAKE AWAYS: Host lipid droplets are differentially altered in early and replicative stages of THP-1 macrophage infection with R. conorii. Lipid droplet alterations are initiated in a bacterial-dependent manner and do not require host peroxisome proliferator-activated receptors α or ɣ activation. Pharmacological inhibition of host lipid catabolic processes during R. conorii infection indicates a requirement of lipid catabolism for bacterial survival and initiation of lipid droplet modulation. A significant increase in host lipid droplets during infection has a negative impact on R. conorii survival in THP-1 macrophages.
Collapse
Affiliation(s)
- Paige E Allen
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Robert C Noland
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Juan J Martinez
- Vector Borne Disease Laboratories, Department of Pathobiological Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| |
Collapse
|
23
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
24
|
Unique Attributes of the Laurel Wilt Fungal Pathogen, Raffaelea lauricola, as Revealed by Metabolic Profiling. Pathogens 2021; 10:pathogens10050528. [PMID: 33925553 PMCID: PMC8146198 DOI: 10.3390/pathogens10050528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 11/18/2022] Open
Abstract
Raffaelea lauricola is the causative agent of laurel wilt, a devastating disease of lauraceous trees. R. lauricola is also an obligate nutritional symbiont of several ambrosia beetle species who act as vectors for the pathogen. Here, we sought to establish the baseline “phenome” of R. lauricola with knowledge concerning its metabolic capability, expanding our understanding of how these processes are impacted by environmental and host nutrients. Phenotypic screening using a microarray of over one thousand compounds was used to generate a detailed profile of R. lauricola substrate utilization and chemical sensitivity. These data revealed (i) relatively restricted carbon utilization, (ii) broad sulfur and phosphate utilization, and (iii) pH and osmotic sensitivities that could be rescued by specific compounds. Additional growth profiling on fatty acids revealed toxicity on C10 substrates and lower, with robust growth on C12–C18 fatty acids. Conditions for lipid droplet (LD) visualization and LD dynamics were examined using a series of lipid dyes. These data provide unique insights regarding R. lauricola metabolism and physiology, and identify distinct patterns of substrate usage and sensitivity which likely reflect important aspects of the host-microbe interface and can be exploited for the development of strategies for mitigating the spread of laurel wilt.
Collapse
|
25
|
Voss OH, Rahman MS. Rickettsia-host interaction: strategies of intracytosolic host colonization. Pathog Dis 2021; 79:ftab015. [PMID: 33705517 PMCID: PMC8023194 DOI: 10.1093/femspd/ftab015] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/09/2021] [Indexed: 12/29/2022] Open
Abstract
Bacterial infection is a highly complex biological process involving a dynamic interaction between the invading microorganism and the host. Specifically, intracellular pathogens seize control over the host cellular processes including membrane dynamics, actin cytoskeleton, phosphoinositide metabolism, intracellular trafficking and immune defense mechanisms to promote their host colonization. To accomplish such challenging tasks, virulent bacteria deploy unique species-specific secreted effectors to evade and/or subvert cellular defense surveillance mechanisms to establish a replication niche. However, despite superficially similar infection strategies, diverse Rickettsia species utilize different effector repertoires to promote host colonization. This review will discuss our current understandings on how different Rickettsia species deploy their effector arsenal to manipulate host cellular processes to promote their intracytosolic life within the mammalian host.
Collapse
Affiliation(s)
- Oliver H Voss
- Department of Microbiology and Immunology, University of Maryland School of Medicine, HSF2, room 416, 20 Penn St, Baltimore, MD 21201, USA
| | - M Sayeedur Rahman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, HSF2, room 416, 20 Penn St, Baltimore, MD 21201, USA
| |
Collapse
|
26
|
Martins CA, Santos MCBD, Gonçalves-de-Albuquerque CF, Castro-Faria-Neto HC, Castro-Faria MV, Burth P, Younes-Ibrahim M. The relationship of oleic acid/albumin molar ratio and clinical outcomes in leptospirosis. Heliyon 2021; 7:e06420. [PMID: 33732938 PMCID: PMC7944043 DOI: 10.1016/j.heliyon.2021.e06420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 01/12/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
Human leptospirosis is an acute infectious zoonosis presenting specific lipid disorders. Previous in vitro studies showed both leptospira glycolipoprotein endotoxin, and high oleic acid levels were associated with Na/K-ATPase inhibition that is amplified by the reduction of circulating albumin levels. In this study, we aimed to investigate the relationship of oleic acid/albumin (OA/A) molar ratio and clinical outcomes in Leptospirosis. Through a prospective observational cohort study employing high-performance liquid chromatography (HPLC) we sequentially determined serum concentrations of nonesterified fatty acids (NEFA) and albumin in twenty-eight patients with severe leptospirosis since their hospital admission. Twenty patients recovered, and eight died. Data was distributed in two groups according to clinical outcomes. Oleic acid/albumin molar ratios (OA/A), initial samples, were higher than those in healthy donors. The ratio OA/A, however, persisted high in dying patients, whereas patients who survived had a reduction matching to healthy donors. Biochemical alterations suggest that cure is correlated to the reestablishment of the OA/A molar ratio, while fatal outcomes related to persisting OA/A imbalances. Analysis by receiver operating characteristic (ROC) showed the area under the curve of 0.864 and the cutoff value of 0.715 being associated with a high odds ratio. Lipid analysis from patients with leptospirosis had an acute high serum OA/A molar ratio, and sustained imbalance has a high odds ratio and strong correlation with mortality.
Collapse
Affiliation(s)
- Caroline Azevedo Martins
- Laboratório Integrado de Nefrologia, Department of Internal Medicine, Medical Sciences School, State University of Rio de Janeiro, Brazil
| | - Maria Conceição B dos Santos
- Laboratório Integrado de Nefrologia, Department of Internal Medicine, Medical Sciences School, State University of Rio de Janeiro, Brazil
| | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Departamento de Bioquímica, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Mauro Velho Castro-Faria
- Laboratório Integrado de Nefrologia, Department of Internal Medicine, Medical Sciences School, State University of Rio de Janeiro, Brazil
| | - Patricia Burth
- Laboratório de Enzimologia e Sinalização Celular, Department of Cellular and Molecular Biology, Federal Fluminense University, Niteroi, Brazil
| | - Mauricio Younes-Ibrahim
- Laboratório Integrado de Nefrologia, Department of Internal Medicine, Medical Sciences School, State University of Rio de Janeiro, Brazil
- Departamento de Medicina, Pontifícia Universidade Católica, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Westman J, Grinstein S. Determinants of Phagosomal pH During Host-Pathogen Interactions. Front Cell Dev Biol 2021. [PMID: 33505976 DOI: 10.3389/fcell.2020.624958/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The ability of phagosomes to halt microbial growth is intimately linked to their ability to acidify their luminal pH. Establishment and maintenance of an acidic lumen requires precise co-ordination of H+ pumping and counter-ion permeation to offset the countervailing H+ leakage. Despite the best efforts of professional phagocytes, however, a number of specialized pathogens survive and even replicate inside phagosomes. In such instances, pathogens target the pH-regulatory machinery of the host cell in an effort to survive inside or escape from phagosomes. This review aims to describe how phagosomal pH is regulated during phagocytosis, why it varies in different types of professional phagocytes and the strategies developed by prototypical intracellular pathogens to manipulate phagosomal pH to survive, replicate, and eventually escape from the phagocyte.
Collapse
Affiliation(s)
- Johannes Westman
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
28
|
Westman J, Grinstein S. Determinants of Phagosomal pH During Host-Pathogen Interactions. Front Cell Dev Biol 2021; 8:624958. [PMID: 33505976 PMCID: PMC7829662 DOI: 10.3389/fcell.2020.624958] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
The ability of phagosomes to halt microbial growth is intimately linked to their ability to acidify their luminal pH. Establishment and maintenance of an acidic lumen requires precise co-ordination of H+ pumping and counter-ion permeation to offset the countervailing H+ leakage. Despite the best efforts of professional phagocytes, however, a number of specialized pathogens survive and even replicate inside phagosomes. In such instances, pathogens target the pH-regulatory machinery of the host cell in an effort to survive inside or escape from phagosomes. This review aims to describe how phagosomal pH is regulated during phagocytosis, why it varies in different types of professional phagocytes and the strategies developed by prototypical intracellular pathogens to manipulate phagosomal pH to survive, replicate, and eventually escape from the phagocyte.
Collapse
Affiliation(s)
- Johannes Westman
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
29
|
Bruno M, Dewi IM, Matzaraki V, ter Horst R, Pekmezovic M, Rösler B, Groh L, Röring RJ, Kumar V, Li Y, Carvalho A, Netea MG, Latgé JP, Gresnigt MS, van de Veerdonk FL. Comparative host transcriptome in response to pathogenic fungi identifies common and species-specific transcriptional antifungal host response pathways. Comput Struct Biotechnol J 2020; 19:647-663. [PMID: 33510868 PMCID: PMC7817431 DOI: 10.1016/j.csbj.2020.12.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
Candidiasis, aspergillosis, and mucormycosis cause the majority of nosocomial fungal infections in immunocompromised patients. Using an unbiased transcriptional profiling in PBMCs exposed to the fungal species causing these infections, we found a core host response in healthy individuals that may govern effective fungal clearance: it consists of 156 transcripts, involving canonical and non-canonical immune pathways. Systematic investigation of key steps in antifungal host defense revealed fungal-specific signatures. As previously demonstrated, Candida albicans induced type I and Type II interferon-related pathways. In contrast, central pattern recognition receptor, reactive oxygen species production, and host glycolytic pathways were down-regulated in response to Rhizopus oryzae, which was associated with an ER-stress response. TLR5 was identified to be uniquely regulated by Aspergillus fumigatus and to control cytokine release in response to this fungus. In conclusion, our data reveals the transcriptional profiles induced by C. albicans, A. fumigatus, and R. oryzae, and describes both the common and specific antifungal host responses that could be exploited for novel therapeutic strategies.
Collapse
Affiliation(s)
- Mariolina Bruno
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Intan M.W. Dewi
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vicky Matzaraki
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob ter Horst
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marina Pekmezovic
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a 07745, Jena, Germany
| | - Berenice Rösler
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Laszlo Groh
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rutger J. Röring
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Vinod Kumar
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yang Li
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- Centre for Individualised Infection Medicine (CiiM) and TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Mihai G. Netea
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | | | - Mark S. Gresnigt
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a 07745, Jena, Germany
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Austermeier S, Kasper L, Westman J, Gresnigt MS. I want to break free – macrophage strategies to recognize and kill Candida albicans, and fungal counter-strategies to escape. Curr Opin Microbiol 2020; 58:15-23. [DOI: 10.1016/j.mib.2020.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 12/31/2022]
|
31
|
EPEC Recruits a Cdc42-Specific GEF, Frabin, To Facilitate PAK Activation and Host Cell Colonization. mBio 2020; 11:mBio.01423-20. [PMID: 33144373 PMCID: PMC7642674 DOI: 10.1128/mbio.01423-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) is a leading cause of diarrhea in children, especially in the developing world. EPEC initiates infection by attaching to cells in the host intestine, triggering the formation of actin-rich “pedestal” structures directly beneath the adherent pathogen. These bacteria inject their own receptor into host cells, which upon binding to a protein on the pathogen surface triggers pedestal formation. Multiple other proteins are also delivered into the cells of the host intestine, which work together to hijack host signaling pathways to drive pedestal production. Here we show how EPEC hijacks a host protein, Frabin, which creates the conditions in the cell necessary for the pathogen to manipulate a specific pathway that promotes pedestal formation. This provides new insights into this essential early stage in disease caused by EPEC. Enteropathogenic Escherichia coli (EPEC) is an extracellular pathogen that tightly adheres to host cells by forming “actin pedestals” beneath the bacteria, a critical step in pathogenesis. EPEC injects effector proteins that manipulate host cell signaling cascades to trigger pedestal assembly. We have recently shown that one such effector, EspG, hijacks p21-activated kinase (PAK) and sustains its activated state to drive the cytoskeletal changes necessary for attachment of the pathogen to target cells. This EspG subversion of PAK required active Rho family small GTPases in the host cell. Here we show that EPEC itself promotes the activation of Rho GTPases by recruiting Frabin, a host guanine nucleotide exchange factor (GEF) for the Rho GTPase Cdc42. Cells devoid of Frabin showed significantly lower EPEC-induced PAK activation, pedestal formation, and bacterial attachment. Frabin recruitment to sites of EPEC attachment was driven by EspG and required localized enrichment of phosphatidylinositol 4,5-bisphosphate (PIP2) and host Arf6. Our findings identify Frabin as a key target for EPEC to ensure the activation status of cellular GTPases required for actin pedestal formation.
Collapse
|
32
|
Westman J, Walpole GFW, Kasper L, Xue BY, Elshafee O, Hube B, Grinstein S. Lysosome Fusion Maintains Phagosome Integrity during Fungal Infection. Cell Host Microbe 2020; 28:798-812.e6. [PMID: 33022213 DOI: 10.1016/j.chom.2020.09.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 08/10/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022]
Abstract
Phagosomes must maintain membrane integrity to exert their microbicidal function. Some microorganisms, however, survive and grow within phagosomes. In such instances, phagosomes must expand to avoid rupture and microbial escape. We studied whether phagosomes regulate their size to preserve integrity during infection with the fungal pathogen Candida albicans. Phagosomes release calcium as C. albicans hyphae elongate, inducing lysosome recruitment and insertion, thereby increasing the phagosomal surface area. As hyphae grow, the expanding phagosome consumes the majority of free lysosomes. Simultaneously, lysosome biosynthesis is stimulated by activation of TFEB, a transcriptional regulator of lysosomal biogenesis. Preventing lysosomal insertion causes phagosomal rupture, NLRP3 inflammasome activation, IL-1β secretion and host-cell death. Whole-genome transcriptomic analysis demonstrate that stress responses elicited in C. albicans upon engulfment are reversed if phagosome expansion is prevented. Our findings reveal a mechanism whereby phagosomes maintain integrity while expanding, ensuring that growing pathogens remain entrapped within this microbicidal compartment.
Collapse
Affiliation(s)
- Johannes Westman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Glenn F W Walpole
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lydia Kasper
- Department Microbial Pathogenicity Mechanisms, Hans Knoell Institute, 07745 Jena, Germany
| | - Bessie Y Xue
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Osama Elshafee
- Department Microbial Pathogenicity Mechanisms, Hans Knoell Institute, 07745 Jena, Germany
| | - Bernhard Hube
- Department Microbial Pathogenicity Mechanisms, Hans Knoell Institute, 07745 Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, 07743 Jena, Germany
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, the Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON M5C 1N8, Canada.
| |
Collapse
|
33
|
Formation and Maturation of the Phagosome: A Key Mechanism in Innate Immunity against Intracellular Bacterial Infection. Microorganisms 2020; 8:microorganisms8091298. [PMID: 32854338 PMCID: PMC7564318 DOI: 10.3390/microorganisms8091298] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Phagocytosis is an essential mechanism in innate immune defense, and in maintaining homeostasis to eliminate apoptotic cells or microbes, such as Mycobacterium tuberculosis, Salmonella enterica, Streptococcus pyogenes and Legionella pneumophila. After internalizing microbial pathogens via phagocytosis, phagosomes undergo a series of ‘maturation’ steps, to form an increasingly acidified compartment and subsequently fuse with the lysosome to develop into phagolysosomes and effectively eliminate the invading pathogens. Through this mechanism, phagocytes, including macrophages, neutrophils and dendritic cells, are involved in the processing of microbial pathogens and antigen presentation to T cells to initiate adaptive immune responses. Therefore, phagocytosis plays a role in the bridge between innate and adaptive immunity. However, intracellular bacteria have evolved diverse strategies to survive and replicate within hosts. In this review, we describe the sequential stages in the phagocytosis process. We also discuss the immune evasion strategies used by pathogens to regulate phagosome maturation during intracellular bacterial infection, and indicate that these might be used for the development of potential therapeutic strategies for infectious diseases.
Collapse
|
34
|
Mingione A, Ottaviano E, Barcella M, Merelli I, Rosso L, Armeni T, Cirilli N, Ghidoni R, Borghi E, Signorelli P. Cystic Fibrosis Defective Response to Infection Involves Autophagy and Lipid Metabolism. Cells 2020; 9:cells9081845. [PMID: 32781626 PMCID: PMC7463682 DOI: 10.3390/cells9081845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is a hereditary disease, with 70% of patients developing a proteinopathy related to the deletion of phenylalanine 508. CF is associated with multiple organ dysfunction, chronic inflammation, and recurrent lung infections. CF is characterized by defective autophagy, lipid metabolism, and immune response. Intracellular lipid accumulation favors microbial infection, and autophagy deficiency impairs internalized pathogen clearance. Myriocin, an inhibitor of sphingolipid synthesis, significantly reduces inflammation, promotes microbial clearance in the lungs, and induces autophagy and lipid oxidation. RNA-seq was performed in Aspergillusfumigatus-infected and myriocin-treated CF patients’ derived monocytes and in a CF bronchial epithelial cell line. Fungal clearance was also evaluated in CF monocytes. Myriocin enhanced CF patients’ monocytes killing of A. fumigatus. CF patients’ monocytes and cell line responded to infection with a profound transcriptional change; myriocin regulates genes that are involved in inflammation, autophagy, lipid storage, and metabolism, including histones and heat shock proteins whose activity is related to the response to infection. We conclude that the regulation of sphingolipid synthesis induces a metabolism drift by promoting autophagy and lipid consumption. This process is driven by a transcriptional program that corrects part of the differences between CF and control samples, therefore ameliorating the infection response and pathogen clearance in the CF cell line and in CF peripheral blood monocytes.
Collapse
Affiliation(s)
- Alessandra Mingione
- Biochemistry and Molecular Biology Laboratory, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (A.M.); (R.G.)
| | - Emerenziana Ottaviano
- Laboratory of Clinical Microbiology, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (E.O.); (M.B.); (E.B.)
| | - Matteo Barcella
- Laboratory of Clinical Microbiology, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (E.O.); (M.B.); (E.B.)
| | - Ivan Merelli
- National Research Council of Italy, Institute for Biomedical Technologies, Via Fratelli Cervi 93, 20090 Milan, Italy;
| | - Lorenzo Rosso
- Health Sciences Department, University of Milan, Thoracic surgery and transplantation Unit, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Polytechnic University of Marche, 60131 Ancona, Italy;
| | - Natalia Cirilli
- Cystic Fibrosis Referral Care Center, Mother-Child Department, United Hospitals Le Torrette, 60126 Ancona, Italy;
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Laboratory, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (A.M.); (R.G.)
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Elisa Borghi
- Laboratory of Clinical Microbiology, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (E.O.); (M.B.); (E.B.)
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (A.M.); (R.G.)
- Correspondence:
| |
Collapse
|
35
|
König A, Hube B, Kasper L. The Dual Function of the Fungal Toxin Candidalysin during Candida albicans-Macrophage Interaction and Virulence. Toxins (Basel) 2020; 12:toxins12080469. [PMID: 32722029 PMCID: PMC7471981 DOI: 10.3390/toxins12080469] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022] Open
Abstract
The dimorphic fungus Candida albicans is both a harmless commensal organism on mucosal surfaces and an opportunistic pathogen. Under certain predisposing conditions, the fungus can overgrow the mucosal microbiome and cause both superficial and life-threatening systemic infections after gaining access to the bloodstream. As the first line of defense of the innate immune response, infecting C. albicans cells face macrophages, which mediate the clearance of invading fungi by intracellular killing. However, the fungus has evolved sophisticated strategies to counteract macrophage antimicrobial activities and thus evade immune surveillance. The cytolytic peptide toxin, candidalysin, contributes to this fungal defense machinery by damaging immune cell membranes, providing an escape route from the hostile phagosome environment. Nevertheless, candidalysin also induces NLRP3 inflammasome activation, leading to an increased host-protective pro-inflammatory response in mononuclear phagocytes. Therefore, candidalysin facilitates immune evasion by acting as a classical virulence factor but also contributes to an antifungal immune response, serving as an avirulence factor. In this review, we discuss the role of candidalysin during C. albicans infections, focusing on its implications during C. albicans-macrophage interactions.
Collapse
Affiliation(s)
- Annika König
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knoell Institute, 07745 Jena, Germany;
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knoell Institute, 07745 Jena, Germany;
- Center for Sepsis Control and Care, University Hospital Jena, 07747 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, 07743 Jena, Germany
- Correspondence: (B.H.); (L.K.)
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology–Hans Knoell Institute, 07745 Jena, Germany;
- Correspondence: (B.H.); (L.K.)
| |
Collapse
|
36
|
Walpole GFW, Grinstein S. Endocytosis and the internalization of pathogenic organisms: focus on phosphoinositides. F1000Res 2020; 9. [PMID: 32494357 PMCID: PMC7233180 DOI: 10.12688/f1000research.22393.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Despite their comparatively low abundance in biological membranes, phosphoinositides are key to the regulation of a diverse array of signaling pathways and direct membrane traffic. The role of phosphoinositides in the initiation and progression of endocytic pathways has been studied in considerable depth. Recent advances have revealed that distinct phosphoinositide species feature prominently in clathrin-dependent and -independent endocytosis as well as in phagocytosis and macropinocytosis. Moreover, a variety of intracellular and cell-associated pathogens have developed strategies to commandeer host cell phosphoinositide metabolism to gain entry and/or metabolic advantage, thereby promoting their survival and proliferation. Here, we briefly survey the current knowledge on the involvement of phosphoinositides in endocytosis, phagocytosis, and macropinocytosis and highlight several examples of molecular mimicry employed by pathogens to either “hitch a ride” on endocytic pathways endogenous to the host or create an entry path of their own.
Collapse
Affiliation(s)
- Glenn F W Walpole
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
37
|
Choudhary E, Bullen CK, Goel R, Singh AK, Praharaj M, Thakur P, Dhiman R, Bishai WR, Agarwal N. Relative and Quantitative Phosphoproteome Analysis of Macrophages in Response to Infection by Virulent and Avirulent Mycobacteria Reveals a Distinct Role of the Cytosolic RNA Sensor RIG-I in Mycobacterium tuberculosis Pathogenesis. J Proteome Res 2020; 19:2316-2336. [PMID: 32407090 DOI: 10.1021/acs.jproteome.9b00895] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Comparative phosphoproteomics of Mycobacterium tuberculosis (Mtb)- and Mycobacterium bovis BCG (BCG)-infected macrophages could be instrumental in understanding the characteristic post-translational modifications of host proteins and their subsequent involvement in determining Mtb pathogenesis. To identify proteins acquiring a distinct phosphorylation status, herein, we compared the phosphorylation profile of macrophages upon exposure to Mtb and BCG. We observed a significant dephosphorylation of proteins following Mtb infection relative to those with uninfected or BCG-infected cells. A comprehensive tandem mass tag mass spectrometry (MS) approach detected ∼10% phosphosites on a variety of host proteins that are modulated in response to infection. Interestingly, the innate immune-enhancing interferon (IFN)-stimulated genes were identified as a class of proteins differentially phosphorylated during infection, including the cytosolic RNA sensor RIG-I, which has been implicated in the immune response to bacterial infection. We show that Mtb infection results in the activation of RIG-I in primary human macrophages. Studies using RIG-I knockout macrophages reveal that the Mtb-mediated activation of RIG-I promotes IFN-β, IL-1α, and IL-1β levels, dampens autophagy, and facilitates intracellular Mtb survival. To our knowledge, this is the first study providing exhaustive information on relative and quantitative changes in the global phosphoproteome profile of host macrophages that can be further explored in designing novel anti-TB drug targets. The peptide identification and MS/MS spectra have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD013171.
Collapse
Affiliation(s)
- Eira Choudhary
- Laboratory of Mycobacterial Genetics, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India.,Symbiosis School of Biomedical Sciences, Symbiosis International (Deemed University), Pune 412115, Maharashtra, India
| | - C Korin Bullen
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Renu Goel
- Laboratory of Mycobacterial Genetics, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| | - Alok Kumar Singh
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Monali Praharaj
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Preeti Thakur
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - William R Bishai
- Center for Tuberculosis Research, Johns Hopkins University, Baltimore, Maryland 21287, United States
| | - Nisheeth Agarwal
- Laboratory of Mycobacterial Genetics, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, Haryana, India
| |
Collapse
|
38
|
The Role of Membrane Surface Charge in Phagocytosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1246:43-54. [DOI: 10.1007/978-3-030-40406-2_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
39
|
Christensen S, Camacho M, Sharmin Z, Momtaz AJMZ, Perez L, Navarro G, Triana J, Samarah H, Turelli M, Serbus LR. Quantitative methods for assessing local and bodywide contributions to Wolbachia titer in maternal germline cells of Drosophila. BMC Microbiol 2019; 19:206. [PMID: 31481018 PMCID: PMC6724367 DOI: 10.1186/s12866-019-1579-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 08/25/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Little is known about how bacterial endosymbionts colonize host tissues. Because many insect endosymbionts are maternally transmitted, egg colonization is critical for endosymbiont success. Wolbachia bacteria, carried by approximately half of all insect species, provide an excellent model for characterizing endosymbiont infection dynamics. To date, technical limitations have precluded stepwise analysis of germline colonization by Wolbachia. It is not clear to what extent titer-altering effects are primarily mediated by growth rates of Wolbachia within cell lineages or migration of Wolbachia between cells. RESULTS The objective of this work is to inform mechanisms of germline colonization through use of optimized methodology. The approaches are framed in terms of nutritional impacts on Wolbachia. Yeast-rich diets in particular have been shown to suppress Wolbachia titer in the Drosophila melanogaster germline. To determine the extent of Wolbachia sensitivity to diet, we optimized 3-dimensional, multi-stage quantification of Wolbachia titer in maternal germline cells. Technical and statistical validation confirmed the identity of Wolbachia in vivo, the reproducibility of Wolbachia quantification and the statistical power to detect these effects. The data from adult feeding experiments demonstrated that germline Wolbachia titer is distinctly sensitive to yeast-rich host diets in late oogenesis. To investigate the physiological basis for these nutritional impacts, we optimized methodology for absolute Wolbachia quantification by real-time qPCR. We found that yeast-rich diets exerted no significant effect on bodywide Wolbachia titer, although ovarian titers were significantly reduced. This suggests that host diets affects Wolbachia distribution between the soma and late stage germline cells. Notably, relative qPCR methods distorted apparent wsp abundance, due to altered host DNA copy number in yeast-rich conditions. This highlights the importance of absolute quantification data for testing mechanistic hypotheses. CONCLUSIONS We demonstrate that absolute quantification of Wolbachia, using well-controlled cytological and qPCR-based methods, creates new opportunities to determine how bacterial abundance within the germline relates to bacterial distribution within the body. This methodology can be applied to further test germline infection dynamics in response to chemical treatments, genetic conditions, new host/endosymbiont combinations, or potentially adapted to analyze other cell and tissue types.
Collapse
Affiliation(s)
- Steen Christensen
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Moises Camacho
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Zinat Sharmin
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - A. J. M. Zehadee Momtaz
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Laura Perez
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Giselle Navarro
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Jairo Triana
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Hani Samarah
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| | - Michael Turelli
- Department of Evolution and Ecology, University of California, Davis, Davis, CA 95616 USA
| | - Laura R. Serbus
- Department of Biological Sciences, Florida International University, Miami, FL 33199 USA
- Biomolecular Sciences Institute, Florida International University, Miami, FL 33199 USA
| |
Collapse
|
40
|
Mechanism of catalysis and inhibition of Mycobacterium tuberculosis SapM, implications for the development of novel antivirulence drugs. Sci Rep 2019; 9:10315. [PMID: 31312014 PMCID: PMC6635428 DOI: 10.1038/s41598-019-46731-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/10/2019] [Indexed: 01/02/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) SapM is a secreted virulence factor critical for intracellular survival of the pathogen. The role of SapM in phagosome maturation arrest in host macrophages suggests its potential as a drug target to assist in the clearance of tuberculosis infection. However, the mechanism of action of SapM at the molecular level remains unknown. In this study, we provide new insights into the mechanism of catalysis, substrate specificity and inhibition of SapM, and we identify the critical residues for catalysis and substrate binding. Our findings demonstrate that SapM is an atypical monoester alkaline phosphatase, with a serine-based mechanism of catalysis probably metal-dependent. Particularly relevant to SapM function and pathogenesis, is its activity towards PI(4,5)P2 and PI3P, two phosphoinositides that function at the early stages of microbial phagocytosis and phagosome formation. This suggests that SapM may have a pleiotropic role with a wider importance on Mtb infection than initially thought. Finally, we have identified two inhibitors of SapM, L-ascorbic acid and 2-phospho-L-ascorbic, which define two different mechanisms by which the catalytic activity of this phosphatase could be regulated. Critically, we demonstrate that 2-phospho-L-ascorbic reduces mycobacterial survival in macrophage infections, hence confirming the potential of SapM as a therapeutic drug target.
Collapse
|
41
|
Westman J, Grinstein S, Maxson ME. Revisiting the role of calcium in phagosome formation and maturation. J Leukoc Biol 2019; 106:837-851. [DOI: 10.1002/jlb.mr1118-444r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Johannes Westman
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| | - Sergio Grinstein
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
- Department of BiochemistryUniversity of Toronto Toronto Ontario Canada
- Keenan Research Centre of the Li Ka Shing Knowledge InstituteSt. Michael's Hospital Toronto Ontario Canada
| | - Michelle E. Maxson
- Program in Cell BiologyHospital for Sick Children Toronto Ontario Canada
| |
Collapse
|
42
|
Lipid Droplets: A Significant but Understudied Contributor of Host⁻Bacterial Interactions. Cells 2019; 8:cells8040354. [PMID: 30991653 PMCID: PMC6523240 DOI: 10.3390/cells8040354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 04/05/2019] [Accepted: 04/12/2019] [Indexed: 12/13/2022] Open
Abstract
Lipid droplets (LDs) are cytosolic lipid storage organelles that are important for cellular lipid metabolism, energy homeostasis, cell signaling, and inflammation. Several bacterial, viral and protozoal pathogens exploit host LDs to promote infection, thus emphasizing the importance of LDs at the host–pathogen interface. In this review, we discuss the thus far reported relation between host LDs and bacterial pathogens including obligate and facultative intracellular bacteria, and extracellular bacteria. Although there is less evidence for a LD–extracellular bacterial interaction compared to interactions with intracellular bacteria, in this review, we attempt to compare the bacterial mechanisms that target LDs, the host signaling pathways involved and the utilization of LDs by these bacteria. Many intracellular bacteria employ unique mechanisms to target host LDs and potentially obtain nutrients and lipids for vacuolar biogenesis and/or immune evasion. However, extracellular bacteria utilize LDs to either promote host tissue damage or induce host death. We also identify several areas that require further investigation. Along with identifying LD interactions with bacteria besides the ones reported, the precise mechanisms of LD targeting and how LDs benefit pathogens should be explored for the bacteria discussed in the review. Elucidating LD–bacterial interactions promises critical insight into a novel host–pathogen interaction.
Collapse
|
43
|
The Mycobacterium tuberculosis CRISPR-Associated Cas1 Involves Persistence and Tolerance to Anti-Tubercular Drugs. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7861695. [PMID: 31061828 PMCID: PMC6466960 DOI: 10.1155/2019/7861695] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/13/2019] [Accepted: 03/13/2019] [Indexed: 11/29/2022]
Abstract
Tuberculosis remains one of the leading causes of death worldwide. Even if new antitubercular drugs are currently being developed, the rapid emergence and spread of drug-resistant strain remain a severe challenge. The CRISPR associated proteins 1 (Cas1), a most conserved endonuclease which is responsible for spacer integration into CRISPR arrays, was found deleted in many specific drug-resistant strains. The function of Cas1 is still unknown in Mycobacterium type III-A CRISPR family. In this study, the Cas1 (Rv2817c) defect was found in 57.14% of clinical isolates. To investigate the function of Cas1 in new spacer acquisition, we challenged Bacillus Calmette–Guérin (BCG) with a mycobacteriophage D29. Newly acquired spacer sequence matches D29 genome was not found by spacer deep-sequencing. We further expressed Cas1 in recombinant Mycobacterium smegmatis. We found that Cas1 increased the sensitivity to multiple anti-tuberculosis drugs by reducing the persistence during drug treatment. We also showed that Cas1 impaired the repair of DNA damage and changed the stress response of Mycobacterium smegmatis. This study provides a further understanding of Cas1 in Mycobacterium tuberculosis complex (MTBC) drug-resistance evolution and a new sight for the tuberculosis treatment.
Collapse
|
44
|
Cockburn CL, Green RS, Damle SR, Martin RK, Ghahrai NN, Colonne PM, Fullerton MS, Conrad DH, Chalfant CE, Voth DE, Rucks EA, Gilk SD, Carlyon JA. Functional inhibition of acid sphingomyelinase disrupts infection by intracellular bacterial pathogens. Life Sci Alliance 2019; 2:e201800292. [PMID: 30902833 PMCID: PMC6431796 DOI: 10.26508/lsa.201800292] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Intracellular bacteria that live in host cell-derived vacuoles are significant causes of human disease. Parasitism of low-density lipoprotein (LDL) cholesterol is essential for many vacuole-adapted bacteria. Acid sphingomyelinase (ASM) influences LDL cholesterol egress from the lysosome. Using functional inhibitors of ASM (FIASMAs), we show that ASM activity is key for infection cycles of vacuole-adapted bacteria that target cholesterol trafficking-Anaplasma phagocytophilum, Coxiella burnetii, Chlamydia trachomatis, and Chlamydia pneumoniae. Vacuole maturation, replication, and infectious progeny generation by A. phagocytophilum, which exclusively hijacks LDL cholesterol, are halted and C. burnetii, for which lysosomal cholesterol accumulation is bactericidal, is killed by FIASMAs. Infection cycles of Chlamydiae, which hijack LDL cholesterol and other lipid sources, are suppressed but less so than A. phagocytophilum or C. burnetii A. phagocytophilum fails to productively infect ASM-/- or FIASMA-treated mice. These findings establish the importance of ASM for infection by intracellular bacteria and identify FIASMAs as potential host-directed therapies for diseases caused by pathogens that manipulate LDL cholesterol.
Collapse
Affiliation(s)
- Chelsea L Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Ryan S Green
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Sheela R Damle
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Naomi N Ghahrai
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Punsiri M Colonne
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Marissa S Fullerton
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Daniel E Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Elizabeth A Rucks
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stacey D Gilk
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| |
Collapse
|
45
|
Westman J, Hube B, Fairn GD. Integrity under stress: Host membrane remodelling and damage by fungal pathogens. Cell Microbiol 2019; 21:e13016. [DOI: 10.1111/cmi.13016] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Johannes Westman
- Program in Cell Biology The Hospital for Sick Children Toronto Ontario Canada
| | - Bernhard Hube
- Department Microbial Pathogenicity Mechanisms Hans Knoell Institute Jena Germany
- Institute of Microbiology Microbial Pathogenicity Friedrich Schiller University Jena Germany
| | - Gregory D. Fairn
- Keenan Research Centre for Biomedical Sciences St. Michael's Hospital Toronto Ontario Canada
- Department of Surgery University of Toronto Toronto Ontario Canada
| |
Collapse
|
46
|
Kasper L, König A, Koenig PA, Gresnigt MS, Westman J, Drummond RA, Lionakis MS, Groß O, Ruland J, Naglik JR, Hube B. The fungal peptide toxin Candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes. Nat Commun 2018; 9:4260. [PMID: 30323213 PMCID: PMC6189146 DOI: 10.1038/s41467-018-06607-1] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/13/2018] [Indexed: 01/03/2023] Open
Abstract
Clearance of invading microbes requires phagocytes of the innate immune system. However, successful pathogens have evolved sophisticated strategies to evade immune killing. The opportunistic human fungal pathogen Candida albicans is efficiently phagocytosed by macrophages, but causes inflammasome activation, host cytolysis, and escapes after hypha formation. Previous studies suggest that macrophage lysis by C. albicans results from early inflammasome-dependent cell death (pyroptosis), late damage due to glucose depletion and membrane piercing by growing hyphae. Here we show that Candidalysin, a cytolytic peptide toxin encoded by the hypha-associated gene ECE1, is both a central trigger for NLRP3 inflammasome-dependent caspase-1 activation via potassium efflux and a key driver of inflammasome-independent cytolysis of macrophages and dendritic cells upon infection with C. albicans. This suggests that Candidalysin-induced cell damage is a third mechanism of C. albicans-mediated mononuclear phagocyte cell death in addition to damage caused by pyroptosis and the growth of glucose-consuming hyphae.
Collapse
Affiliation(s)
- Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Beutenbergstrasse 11a, Jena, 07745, Germany
| | - Annika König
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Beutenbergstrasse 11a, Jena, 07745, Germany
| | - Paul-Albert Koenig
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, München, 81675, Germany
| | - Mark S Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Beutenbergstrasse 11a, Jena, 07745, Germany
| | - Johannes Westman
- Program in Cell Biology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1×8, Canada
| | - Rebecca A Drummond
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, 9000 Rockville Pike, Bldg 10 / Rm 11C102, Bethesda, MD, 20892, USA.,Institute of Immunology and Immunotherapy, Institute of Microbiology and Infection, University of Birmingham, Birmingham, B15 2TT, UK
| | - Michail S Lionakis
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fungal Pathogenesis Section, Laboratory of Clinical Immunology & Microbiology, 9000 Rockville Pike, Bldg 10 / Rm 11C102, Bethesda, MD, 20892, USA
| | - Olaf Groß
- Institute of Neuropathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, Freiburg, 79106, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, München, 81675, Germany.,TranslaTUM, Center for Translational Cancer Research, Technische Universität München, München, 81675, Germany.,German Cancer Consortium (DKTK), Heidelberg, 69120, Germany.,German Center for Infection Research (DZIF), Munich, 81675, Germany
| | - Julian R Naglik
- Mucosal and Salivary Biology Division, King's College London Dental Institute, London, SE1 1UL, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Beutenbergstrasse 11a, Jena, 07745, Germany. .,Friedrich Schiller University, Fürstengraben 1, Jena, 07743, Germany.
| |
Collapse
|
47
|
Abstract
C. albicans is the most common cause of nosocomial fungal infection, and over 3 million people acquire life-threatening invasive fungal infections every year. Even if antifungal drugs exist, almost half of these patients will die. Despite this, fungi remain underestimated as pathogens. Our study uses quantitative biophysical approaches to demonstrate that yeast-to-hypha transition occurs within the nutrient-deprived, acidic phagosome and that alkalinization is a consequence, as opposed to the cause, of hyphal growth. Macrophages rely on phagosomal acidity to destroy engulfed microorganisms. To survive this hostile response, opportunistic fungi such as Candida albicans developed strategies to evade the acidic environment. C. albicans is polymorphic and able to convert from yeast to hyphae, and this transition is required to subvert the microbicidal activity of the phagosome. However, the phagosomal lumen, which is acidic and nutrient deprived, is believed to inhibit the yeast-to-hypha transition. To account for this apparent paradox, it was recently proposed that C. albicans produces ammonia that alkalinizes the phagosome, thus facilitating yeast-to-hypha transition. We reexamined the mechanism underlying phagosomal alkalinization by applying dual-wavelength ratiometric pH measurements. The phagosomal membrane was found to be highly permeable to ammonia, which is therefore unlikely to account for the pH elevation. Instead, we find that yeast-to-hypha transition begins within acidic phagosomes and that alkalinization is a consequence of proton leakage induced by excessive membrane distension caused by the expanding hypha.
Collapse
|