1
|
Mercedes Bigi M, Imperiale B, Soria M, López B, Bigi F, de la Barrera S. Total free lipids from MDR strain of Mycobacterium tuberculosis "M" reduce T cell activation and CTL activity in healthy individuals. Mol Immunol 2025; 183:182-193. [PMID: 40382835 DOI: 10.1016/j.molimm.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/02/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
Increasing evidence highlights the role of cell wall components in the effectiveness of different Mycobacterium tuberculosis (Mtb) strains in modulating host immune response. We previously demonstrated that the outbreak multidrug-resistant strain M displays a distinctive lipid profile in its cell envelope compared to the closely related sporadic strain 410. Both strains markedly differ in their ability to induce fully functional CD8+ T cells because of low CD69 signaling and impaired CD4+ T cell help. In this study, we evaluated the impact of extractable lipids (LP) from M (LP-M) and 410 (LP-410) on the activation and functionality of T cells from healthy individuals. PBMCs were cultured alone or with Mtb in the presence or absence of LP-M, LP-410, or LP from CD1551 mutants in polymorphic genes between M and 410. Then, surface CD69 and intracytoplasmic IL-2 (after 3 days of culture), as well as surface CD107 expression (after 6 days of culture) were determined in T cells by flow cytometry. In contrast to LP-410, LP-M induced low expression of CD69 and IL-2 in CD4+/CD8+ cells and of CD107 in CD8+ cells. Besides, LP from Mtb strains mutated in Rv1861c and Rv3787c genes inhibited H37Rv-induced T cell response without causing cell death. Thus, our results suggest that LP-M likely through mutations in Rv1861 and Rv3787c, inhibits the activation and functionality of T cells from PPD+ healthy human donors and might partially contribute to the development of immune evasion mechanisms in the M strain.
Collapse
Affiliation(s)
- María Mercedes Bigi
- Biomedical Research Institute (UBA-CONICET), School of Medicine, University of Buenos Aires (UBA), Buenos Aires, Argentina.
| | - Belén Imperiale
- Laboratory of Immunology of Physiology of Inflammatory Processes, Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina.
| | - Marcelo Soria
- School of Agronomy, Facultad de Agronomía, Universidad de Buenos Aires (UBA), Facultad de Agronomía, Buenos Aires, Argentina.
| | - Beatriz López
- Laboratory of Mycobacteria, National Institute of Infectious Diseases, ANLIS ''Dr. Carlos G. Malbrán'', Buenos Aires, Argentina.
| | - Fabiana Bigi
- Institute of Biotechnology, National Institute of Agricultural Technology, (INTA)/IABIMO-CONICET, Buenos Aires, Argentina.
| | - Silvia de la Barrera
- Laboratory of Immunology of Physiology of Inflammatory Processes, Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Song HY, Yoo BG, Lee Y, Lim JY, Gu EJ, Jeon J, Byun EB. Isoniazid and nicotinic hydrazide hybrids mitigate trehalose-6,6'-dimycolate-induced inflammatory responses and pulmonary granulomas via Syk/PI3K pathways: A promising host-directed therapy for tuberculosis. Biomed Pharmacother 2025; 183:117798. [PMID: 39764922 DOI: 10.1016/j.biopha.2024.117798] [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: 10/07/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 02/08/2025] Open
Abstract
Granulomas, dense clusters of immune cells and bacteria, are critical barriers in tuberculosis (TB) treatment. Recent advancements in TB management have highlighted granuloma control as a potential host-directed therapy (HDT) strategy. Although isoniazid (INH) is the first-line drug for TB therapy, its efficacy is limited to non-replicating Mycobacterium tuberculosis (Mtb) under granulomatous conditions, necessitating the development of more effective derivatives. In this study, hybrid compounds of isoniazid, designated as INH-D1 and INH-D2, were synthesized and evaluated for their effects on controlling inflammatory responses and pulmonary granuloma lesions induced by trehalose-6,6'-dimycolate (TDM), a glycolipid of Mtb. Both INH-D1 and INH-D2 demonstrated stronger inhibitory effects on inflammatory mediators (TNF-α, interleukin-6, co-stimulatory molecules, and MHC class I) in TDM-stimulated macrophages compared to original INH. These anti-inflammatory effects were mediated by the inhibition of Syk, p38, PI3K, and NF-κB transcription. INH-D1 and INH-D2 exhibited stronger binding energies to Syk and PI3Kα/β than INH, which are known as proximal kinases and key mediator in TDM-mediated inflammatory responses. Oral administration of INH-D2 successfully relieved TDM-induced pulmonary granuloma pathology by reducing innate immune cell infiltration, hypoxic conditions in the lungs, and systemic inflammation by decreasing serum cytokines and chemokines. In contrast, original INH and INH-D1 did not effectively alleviate pulmonary granuloma pathology. These findings demonstrate that the novel molecule INH-D2 is effective in treating pulmonary granulomas owing to its strong anti-inflammatory effects, highlighting it as a promising HDT candidate for the management of pulmonary tuberculosis, thereby providing a strategic alternative to standard anti-TB antibiotics.
Collapse
Affiliation(s)
- Ha-Yeon Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| | - Bo-Gyeong Yoo
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea; Department of Food Science and Technology, Kongju National University, Yesan 32439, Republic of Korea
| | - Yuna Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| | - Jae Yoon Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea; Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eun Ji Gu
- Department of Applied Chemistry, College of Engineering, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jongho Jeon
- Department of Applied Chemistry, College of Engineering, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea.
| |
Collapse
|
3
|
Manjunath P, Ahmad J, Samal J, Rani A, Sheikh JA, Zarin S, Ahuja Y, Alam A, Hasnain SE, Ehtesham NZ. Expression of a unique M. tuberculosis DNA MTase Rv1509 in M. smegmatis alters the gene expression pattern and enhances virulence. Front Microbiol 2024; 15:1344857. [PMID: 38803374 PMCID: PMC11129820 DOI: 10.3389/fmicb.2024.1344857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Mycobacterium tuberculosis (M. tb) genome encompasses 4,173 genes, about a quarter of which remain uncharacterized and hypothetical. Considering the current limitations associated with the diagnosis and treatment of tuberculosis, it is imperative to comprehend the pathomechanism of the disease and host-pathogen interactions to identify new drug targets for intervention strategies. Using in-silico comparative genome analysis, we identified one of the M. tb genes, Rv1509, as a signature protein exclusively present in M. tb. To explore the role of Rv1509, a likely methyl transferase, we constructed a knock-in Mycobacterium smegmatis (M. smegmatis) constitutively expressing Rv1509 (Ms_Rv1509). The Ms_Rv1509 led to differential expression of many transcriptional regulator genes as assessed by RNA-seq analysis. Further, in-vitro and in-vivo studies demonstrated an enhanced survival of Ms_Rv1509 inside the host macrophages. Ms_Rv1509 also promoted phagolysosomal escape inside macrophages to boost bacterial replication and dissemination. In-vivo infection studies revealed that Ms_Rv1509 survives better than BCG and causes pathological manifestations in the pancreas after intraperitoneal infection. Long-time survival of Ms_Rv1509 resulted in lymphocyte migration, increased T regulatory cells, giant cell formation, and likely granuloma formation in the pancreas, pointing toward the role of Rv1509 in M. tb pathogenesis.
Collapse
Affiliation(s)
- P. Manjunath
- Inflammation Biology and Cell Signaling Laboratory, National Institute of Pathology, New Delhi, India
- Department of Biotechnology, Jamia Hamdard, New Delhi, India
| | - Javeed Ahmad
- Molecular Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jasmine Samal
- Inflammation Biology and Cell Signaling Laboratory, National Institute of Pathology, New Delhi, India
| | - Anshu Rani
- Kusuma School of Biological Sciences, Indian Institute of Technology, New Delhi, India
| | | | - Sheeba Zarin
- Department of Biotechnology, Jamia Hamdard, New Delhi, India
- Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Yashika Ahuja
- Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anwar Alam
- Department of Biotechnology, Sharda School of Engineering Sciences and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Seyed E. Hasnain
- Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| | - Nasreen Z. Ehtesham
- Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
4
|
Venkatasubramanian S, Plumlee CR, Dill-McFarland KA, Cohen SB, Gern BH, Rane DA, Meyer MK, Saha A, Hinderstein SA, Pearson GL, Lietzke AC, Pacheco A, Chow YH, Hung CF, Soleimanpour SA, Altman M, Urdahl KB, Shah JA. TOLLIP inhibits lipid accumulation and the integrated stress response in alveolar macrophages to control Mycobacterium tuberculosis infection. Nat Microbiol 2024; 9:949-963. [PMID: 38528148 PMCID: PMC11034867 DOI: 10.1038/s41564-024-01641-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 02/16/2024] [Indexed: 03/27/2024]
Abstract
A polymorphism causing deficiencies in Toll-interacting protein (TOLLIP), an inhibitory adaptor protein affecting endosomal trafficking, is associated with increased tuberculosis (TB) risk. It is, however, unclear how TOLLIP affects TB pathogenesis. Here we show that TB severity is increased in Tollip-/- mice, characterized by macrophage- and T cell-driven inflammation, foam cell formation and lipid accumulation. Tollip-/- alveolar macrophages (AM) specifically accumulated lipid and underwent necrosis. Transcriptional and protein analyses of Mycobacterium tuberculosis (Mtb)-infected, Tollip-/- AM revealed increased EIF2 signalling and downstream upregulation of the integrated stress response (ISR). These phenotypes were linked, as incubation of the Mtb lipid mycolic acid with Mtb-infected Tollip-/- AM activated the ISR and increased Mtb replication. Correspondingly, the ISR inhibitor, ISRIB, reduced Mtb numbers in AM and improved Mtb control, overcoming the inflammatory phenotype. In conclusion, targeting the ISR offers a promising target for host-directed anti-TB therapy towards improved Mtb control and reduced immunopathology.
Collapse
Affiliation(s)
| | | | | | - Sara B Cohen
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Benjamin H Gern
- Seattle Children's Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Divya A Rane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Aparajita Saha
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Gemma L Pearson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Anne C Lietzke
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amanda Pacheco
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yu-Hua Chow
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Chi F Hung
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Scott A Soleimanpour
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Matthew Altman
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kevin B Urdahl
- Seattle Children's Research Institute, Seattle, WA, USA
- Departments of Pediatrics and Immunology, University of Washington, Seattle, WA, USA
| | - Javeed A Shah
- Department of Medicine, University of Washington, Seattle, WA, USA.
- VA Puget Sound Healthcare System, Seattle, WA, USA.
| |
Collapse
|
5
|
Wang H, Liu D, Zhou X. Effect of Mycolic Acids on Host Immunity and Lipid Metabolism. Int J Mol Sci 2023; 25:396. [PMID: 38203570 PMCID: PMC10778799 DOI: 10.3390/ijms25010396] [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: 08/20/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 01/12/2024] Open
Abstract
Mycolic acids constitute pivotal constituents within the cell wall structure of Mycobacterium tuberculosis. Due to their structural diversity, the composition of mycolic acids exhibits substantial variations among different strains, endowing them with the distinctive label of being the 'signature' feature of mycobacterial species. Within Mycobacterium tuberculosis, the primary classes of mycolic acids include α-, keto-, and methoxy-mycolic acids. While these mycolic acids are predominantly esterified to the cell wall components (such as arabinogalactan, alginate, or glucose) of Mycobacterium tuberculosis, a fraction of free mycolic acids are secreted during in vitro growth of the bacterium. Remarkably, different types of mycolic acids possess varying capabilities to induce foamy macro-phages and trigger immune responses. Additionally, mycolic acids play a regulatory role in the lipid metabolism of host cells, thereby exerting influence over the progression of tuberculosis. Consequently, the multifaceted properties of mycolic acids shape the immune evasion strategy employed by Mycobacterium tuberculosis. A comprehensive understanding of mycolic acids is of paramount significance in the pursuit of developing tuberculosis therapeutics and unraveling the intricacies of its pathogenic mechanisms.
Collapse
Affiliation(s)
- Haoran Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100086, China; (H.W.); (D.L.)
- National Key Laboratory of Veterinary Public Health and Safety, Beijing 100086, China
| | - Dingpu Liu
- College of Veterinary Medicine, China Agricultural University, Beijing 100086, China; (H.W.); (D.L.)
- National Key Laboratory of Veterinary Public Health and Safety, Beijing 100086, China
| | - Xiangmei Zhou
- College of Veterinary Medicine, China Agricultural University, Beijing 100086, China; (H.W.); (D.L.)
- National Key Laboratory of Veterinary Public Health and Safety, Beijing 100086, China
| |
Collapse
|
6
|
Sarkar S, Mishra A, Periasamy S, Dyett B, Dogra P, Ball AS, Yeo LY, White JF, Wang Z, Cristini V, Jagannath C, Khan A, Soni SK, Drummond CJ, Conn CE. Prospective Subunit Nanovaccine against Mycobacterium tuberculosis Infection─Cubosome Lipid Nanocarriers of Cord Factor, Trehalose 6,6' Dimycolate. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37262346 DOI: 10.1021/acsami.3c04063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
An improved vaccine is urgently needed to replace the now more than 100-year-old Bacillus Calmette-Guérin (BCG) vaccine against tuberculosis (TB) disease, which represents a significant burden on global public health. Mycolic acid, or cord factor trehalose 6,6' dimycolate (TDM), a lipid component abundant in the cell wall of the pathogen Mycobacterium tuberculosis (MTB), has been shown to have strong immunostimulatory activity but remains underexplored due to its high toxicity and poor solubility. Herein, we employed a novel strategy to encapsulate TDM within a cubosome lipid nanocarrier as a potential subunit nanovaccine candidate against TB. This strategy not only increased the solubility and reduced the toxicity of TDM but also elicited a protective immune response to control MTB growth in macrophages. Both pre-treatment and concurrent treatment of the TDM encapsulated in lipid monoolein (MO) cubosomes (MO-TDM) (1 mol %) induced a strong proinflammatory cytokine response in MTB-infected macrophages, due to epigenetic changes at the promoters of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) in comparison to the untreated control. Furthermore, treatment with MO-TDM (1 mol %) cubosomes significantly improved antigen processing and presentation capabilities of MTB-infected macrophages to CD4 T cells. The ability of MO-TDM (1 mol %) cubosomes to induce a robust innate and adaptive response in vitro was further supported by a mathematical modeling study predicting the vaccine efficacy in vivo. Overall, these results indicate a strong immunostimulatory effect of TDM when delivered through the lipid nanocarrier, suggesting its potential as a novel TB vaccine.
Collapse
Affiliation(s)
- Sampa Sarkar
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Selvakannan Periasamy
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Brendan Dyett
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
| | - Andrew S Ball
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Leslie Y Yeo
- School of Engineering, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Jacinta F White
- The Commonwealth Scientific and Industrial Research Organisation, Clayton 3169, Victoria, Australia
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Neal Cancer Center, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Imaging Physics, University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, United States
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, New York 10021, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Sarvesh K Soni
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| | - Charlotte E Conn
- School of Science, STEM College, RMIT University, Melbourne 3001, Victoria, Australia
| |
Collapse
|
7
|
Hailu E, Cantillon D, Madrazo C, Rose G, Wheeler PR, Golby P, Adnew B, Gagneux S, Aseffa A, Gordon SV, Comas I, Young DB, Waddell SJ, Larrouy-Maumus G, Berg S. Lack of methoxy-mycolates characterizes the geographically restricted lineage 7 of Mycobacterium tuberculosis complex. Microb Genom 2023; 9:mgen001011. [PMID: 37171244 PMCID: PMC10272862 DOI: 10.1099/mgen.0.001011] [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: 09/26/2022] [Accepted: 03/07/2023] [Indexed: 05/13/2023] Open
Abstract
Lineage 7 (L7) emerged in the phylogeny of the Mycobacterium tuberculosis complex (MTBC) subsequent to the branching of 'ancient' lineage 1 and prior to the Eurasian dispersal of 'modern' lineages 2, 3 and 4. In contrast to the major MTBC lineages, the current epidemiology suggests that prevalence of L7 is highly confined to the Ethiopian population, or when identified outside of Ethiopia, it has mainly been in patients of Ethiopian origin. To search for microbiological factors that may contribute to its restricted distribution, we compared the genome of L7 to the genomes of globally dispersed MTBC lineages. The frequency of predicted functional mutations in L7 was similar to that documented in other lineages. These include mutations characteristic of modern lineages - such as constitutive expression of nitrate reductase - as well as mutations in the VirS locus that are commonly found in ancient lineages. We also identified and characterized multiple lineage-specific mutations in L7 in biosynthesis pathways of cell wall lipids, including confirmed deficiency of methoxy-mycolic acids due to a stop-gain mutation in the mmaA3 gene that encodes a methoxy-mycolic acid synthase. We show that the abolished biosynthesis of methoxy-mycolates of L7 alters the cell structure and colony morphology on selected growth media and impacts biofilm formation. The loss of these mycolic acid moieties may change the host-pathogen dynamic for L7 isolates, explaining the limited geographical distribution of L7 and contributing to further understanding the spread of MTBC lineages across the globe.
Collapse
Affiliation(s)
- Elena Hailu
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Daire Cantillon
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Falmer, UK
- Present address: Department of Tropical Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Carlos Madrazo
- Biomedicine Institute of Valencia, Spanish Research Council (IBV-CSIC), Valencia, Spain
| | - Graham Rose
- Francis Crick Institute, London, UK
- Present address: North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children, London, UK
| | | | - Paul Golby
- Animal and Plant Health Agency, Weybridge, UK
| | | | - Sebastien Gagneux
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia
| | - Stephen V. Gordon
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Iñaki Comas
- Biomedicine Institute of Valencia, Spanish Research Council (IBV-CSIC), Valencia, Spain
| | - Douglas B. Young
- Francis Crick Institute, London, UK
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Simon J. Waddell
- Brighton and Sussex Centre for Global Health Research, Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Falmer, UK
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Stefan Berg
- Animal and Plant Health Agency, Weybridge, UK
- Present address: Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
8
|
Zhang C, Shao H, Zhang J, Guo X, Liu Y, Song Z, Liu F, Ling P, Tang L, Wang KN, Chen Q. Long-term live-cell lipid droplet-targeted biosensor development for nanoscopic tracking of lipid droplet-mitochondria contact sites. Theranostics 2021; 11:7767-7778. [PMID: 34335963 PMCID: PMC8315056 DOI: 10.7150/thno.59848] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Background: Lipid droplets (LDs) establish a considerable number of contact sites with mitochondria to enable energy transfer and communication. In this study, we developed a fluorescent biosensor to image LD-mitochondria interactions at the nanoscale and further explored the function of LD-mediated matrix transmission in processes involving multi-organelle interactions. Methods: A fluorescent probe called C-Py (C21H19N3O2, 7-(diethylamino) coumarin-3-vinyl-4-pyridine acetonitrile) was designed and synthesized. Colocalization of C-Py and the commercial LD stain Nile Red was analyzed in HeLa cells. The fluorescence stability and signal to background ratio of C-Py under structured illumination microscopy (SIM) were compared to those of the commercial probe BODIPY493/503. The cytotoxicity of C-Py was assessed using CCK-8 assays. The uptake pattern of C-Py in HeLa cells was then observed under various temperatures, metabolic levels, and endocytosis levels. Contact sites between LDs and various organelles, such as mitochondria, nuclei, and cell membrane, were imaged and quantitated using SIM. Physical changes to the contact sites between LDs and mitochondria were monitored after lipopolysaccharide induction. Results: A LD-targeted fluorescent biosensor, C-Py, with good specificity, low background signal, excellent photostability, low cytotoxicity, and high cellular permeability was developed for tracking LD contact sites with multiple organelles using SIM. Using C-Py, the subcellular distribution and dynamic processes of LDs in living cells were observed under SIM. The formation of contact sites between LDs and multiple organelles was visualized at a resolution below ~200 nm. The number of LD-mitochondria contact sites formed was decreased by lipopolysaccharide treatment inducing an inflammatory environment. Conclusions: C-Py provides strategies for the design of ultra-highly selective biosensors and a new tool for investigating the role and regulation of LDs in living cells at the nanoscale.
Collapse
Affiliation(s)
- Chengying Zhang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250101, PR China
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, PR China
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| | - Huarong Shao
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| | - Jie Zhang
- Advanced Medical Research Institute/Translational Medicine Core Facility of Advanced Medical Research Institute, Shandong University. Jinan 250101, PR China
| | - Xinyan Guo
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| | - Yue Liu
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| | - Zhigang Song
- College of Basic Medicine, Jining Medical University, Jining 272067, PR China
| | - Fei Liu
- School of Pharmaceutical Sciences, Shandong University, Jinan 250101, PR China
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| | - Peixue Ling
- School of Pharmaceutical Sciences, Shandong University, Jinan 250101, PR China
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| | - Longguang Tang
- International Institutes of Medicine, The 4th Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China
| | - Kang-Nan Wang
- Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong 528308, PR China
| | - Qixin Chen
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, PR China
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, PR China
| |
Collapse
|
9
|
McClean CM, Tobin DM. Early cell-autonomous accumulation of neutral lipids during infection promotes mycobacterial growth. PLoS One 2020; 15:e0232251. [PMID: 32407412 PMCID: PMC7224534 DOI: 10.1371/journal.pone.0232251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/12/2020] [Indexed: 11/19/2022] Open
Abstract
Lipids represent an important source of nutrition for infecting mycobacteria, accumulating within the necrotic core of granulomas and present in foamy macrophages associated with mycobacterial infection. In order to better understand the timing, process and importance of lipid accumulation, we developed methods for direct in vivo visualization and quantification of this process using the zebrafish-M. marinum larval model of infection. We find that neutral lipids accumulate cell-autonomously in mycobacterium-infected macrophages in vivo during early infection, with detectable levels of accumulation by two days post-infection. Treatment with ezetimibe, an FDA-approved drug, resulted in decreased levels of free cholesterol and neutral lipids, and a reduction of bacterial growth in vivo. The effect of ezetimibe in reducing bacterial growth was dependent on the mce4 operon, a key bacterial determinant of lipid utilization. Thus, in vivo, lipid accumulation can occur cell-autonomously at early timepoints of mycobacterial infection, and limitation of this process results in decreased bacterial burden.
Collapse
Affiliation(s)
- Colleen M. McClean
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Medical Scientist Training Program, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
10
|
Analyzing the impact of Mycobacterium tuberculosis infection on primary human macrophages by combined exploratory and targeted metabolomics. Sci Rep 2020; 10:7085. [PMID: 32341411 PMCID: PMC7184630 DOI: 10.1038/s41598-020-62911-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/17/2020] [Indexed: 12/15/2022] Open
Abstract
The pathogenic success of Mycobacterium tuberculosis (Mtb) is tightly linked to its ability to recalibrate host metabolic processes in infected host macrophages. Since changes in cellular metabolic intermediates or pathways also affect macrophage function in response to pathogens, we sought to analyse specific metabolic alterations induced by Mtb infection. Stimulation of macrophages with Mtb lysate or lipopolysaccharide (LPS) induced a relative increase in glycolysis versus oxidative phosphorylation. Cellular metabolomics revealed that Mtb infection induced a distinct metabolic profile compared to LPS in both M1 and M2 macrophages. Specifically, Mtb infection resulted in elevated intracellular levels of nicotinamide adenine dinucleotide (NAD+), creatine, creatine phosphate and glutathione compared to uninfected control macrophages. Correspondingly, RNA-sequencing datasets showed altered gene expression of key metabolic enzymes involved in NAD+, creatine, glucose and glutamine metabolism (e.g NAMPT, SLC6A8, HK2) in Mtb-infected M2 macrophages. These findings demonstrate clear modulation of host macrophage metabolic pathways by Mtb infection.
Collapse
|
11
|
Thirunavukkarasu S, Khader SA. Advances in Cardiovascular Disease Lipid Research Can Provide Novel Insights Into Mycobacterial Pathogenesis. Front Cell Infect Microbiol 2019; 9:116. [PMID: 31058102 PMCID: PMC6482252 DOI: 10.3389/fcimb.2019.00116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/02/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in industrialized nations and an emerging health problem in the developing world. Systemic inflammatory processes associated with alterations in lipid metabolism are a major contributing factor that mediates the development of CVDs, especially atherosclerosis. Therefore, the pathways promoting alterations in lipid metabolism and the interplay between varying cellular types, signaling agents, and effector molecules have been well-studied. Mycobacterial species are the causative agents of various infectious diseases in both humans and animals. Modulation of host lipid metabolism by mycobacteria plays a prominent role in its survival strategy within the host as well as in disease pathogenesis. However, there are still several knowledge gaps in the mechanistic understanding of how mycobacteria can alter host lipid metabolism. Considering the in-depth research available in the area of cardiovascular research, this review presents an overview of the parallel areas of research in host lipid-mediated immunological changes that might be extrapolated and explored to understand the underlying basis of mycobacterial pathogenesis.
Collapse
Affiliation(s)
- Shyamala Thirunavukkarasu
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
12
|
Cao T, Lyu L, Jia H, Wang J, Du F, Pan L, Li Z, Xing A, Xiao J, Ma Y, Zhang Z. A Two-Way Proteome Microarray Strategy to Identify Novel Mycobacterium tuberculosis-Human Interactors. Front Cell Infect Microbiol 2019; 9:65. [PMID: 30984625 PMCID: PMC6448480 DOI: 10.3389/fcimb.2019.00065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/01/2019] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB) is still a serious threat to human health which is caused by mycobacterium tuberculosis (Mtb). The main reason for failure to eliminate TB is lack of clearly understanding the molecular mechanism of Mtb pathogenesis. Determining human Mtb-interacting proteins enables us to characterize the mechanism and identify potential molecular targets for TB diagnosis and treatment. However, experimentally systematic Mtb interactors are not readily available. In this study, we performed an unbiased, comprehensive two-way proteome microarray based approach to systematically screen global human Mtb interactors and determine the binding partners of Mtb effectors. Our results, for the first time, screened 84 potential human Mtb interactors. Bioinformatic analysis further highlighted these protein candidates might engage in a wide range of cellular functions such as activation of DNA endogenous promoters, transcription of DNA/RNA and necrosis, as well as immune-related signaling pathways. Then, using Mtb proteome microarray followed His tagged pull-down assay and Co-IP, we identified one interacting partner (Rv0577) for the protein candidate NRF1 and three binding partners (Rv0577, Rv2117, Rv2423) for SMAD2, respectively. This study gives new insights into the profile of global Mtb interactors potentially involved in Mtb pathogenesis and demonstrates a powerful strategy in the discovery of Mtb effectors.
Collapse
Affiliation(s)
- Tingming Cao
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Lingna Lyu
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hongyan Jia
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jinghui Wang
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Fengjiao Du
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Liping Pan
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Zihui Li
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Aiying Xing
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jing Xiao
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Yu Ma
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Zongde Zhang
- Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Groenewald W, Parra-Cruz RA, Jäger CM, Croft AK. Revealing solvent-dependent folding behavior of mycolic acids from Mycobacterium tuberculosis by advanced simulation analysis. J Mol Model 2019; 25:68. [PMID: 30762132 PMCID: PMC7019640 DOI: 10.1007/s00894-019-3943-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/24/2019] [Indexed: 11/09/2022]
Abstract
Mycobacterium tuberculosis remains a persistent pathogen, partly due to its lipid rich cell wall, of which mycolic acids (MAs) are a major component. The fluidity and conformational flexibilities of different MAs in the bacterial cell wall significantly influence its properties, function, and observed pathogenicity; thus, a proper conformational description of different MAs in different environments (e.g., in vacuum, in solution, in monolayers) can inform about their potential role in the complex setup of the bacterial cell wall. Previously, we have shown that molecular dynamics (MD) simulations of MA folding in vacuo can be used to characterize MA conformers in seven groupings relating to bending at the functional groups (W, U and Z-conformations). Providing a new OPLS-based forcefield parameterization for the critical cyclopropyl group of MAs and extensive simulations in explicit solvents (TIP4P water, hexane), we now present a more complete picture of MA folding properties together with improved simulation analysis techniques. We show that the 'WUZ' distance-based analysis can be used to pinpoint conformers with hairpin bends at the functional groups, with these conformers constituting only a fraction of accessible conformations. Applying principle component analysis (PCA) and refinement using free energy landscapes (FELs), we are able to discriminate a complete and unique set of conformational preferences for representative alpha-, methoxy- and keto-MAs, with overall preference for folded conformations. A control backbone-MA without any mero-chain functional groups showed significantly less folding in the mero-chain, confirming the role of functionalization in directing folding. Keto-MA showed the highest percentage of WUZ-type conformations and, in particular, a tendency to fold at its alpha-methyl trans-cyclopropane group, in agreement with results from Villeneuve et al. MAs demonstrate similar folding in vacuum and water, with a majority of folded conformations around the W-conformation, although the molecules are more flexible in vacuum than in water. Exchange between conformations, with a disperse distribution that includes unfolded conformers, is common in hexane for all MAs, although with more organization for Keto-MA. Globular, folded conformations are newly defined and may be specifically relevant in biofilms. Graphical abstract Through advanced simulation analysis, including principle component analysis and free energy landscapes, we reveal detailed physical insights into the solvent-dependant folding behavior of mycolic acids from M. tb.
Collapse
Affiliation(s)
- Wilma Groenewald
- School of Chemistry, Bangor University, Bangor, Gwynedd, LL57 2UW, UK
| | - Ricardo A Parra-Cruz
- Department of Chemical and Environmental Engineering, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Christof M Jäger
- Department of Chemical and Environmental Engineering, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Anna K Croft
- Department of Chemical and Environmental Engineering, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| |
Collapse
|
14
|
Ranchod H, Ndlandla F, Lemmer Y, Beukes M, Niebuhr J, Al-Dulayymi J, Wemmer S, Fehrsen J, Baird M, Verschoor J. The antigenicity and cholesteroid nature of mycolic acids determined by recombinant chicken antibodies. PLoS One 2018; 13:e0200298. [PMID: 30092023 PMCID: PMC6084858 DOI: 10.1371/journal.pone.0200298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/22/2018] [Indexed: 11/24/2022] Open
Abstract
Mycolic acids (MA) are major, species-specific lipid components of Mycobacteria and related genera. In Mycobacterium tuberculosis, it is made up of alpha-, methoxy- and keto-MA, each with specific biological functions and conformational characteristics. Antibodies in tuberculosis (TB) patient sera respond differently towards the three MA classes and were reported to cross-react with cholesterol. To understand the antigenicity and cholesterol cross-reactivity of MA, we generated three different chicken -derived phage-displayed single-chain variable fragments (scFv) that reacted similarly towards the natural mixture of MA, but the first recognized all three classes of chemically synthetic MAs, the second only the two oxygenated types of MAs and the third only methoxy MA. The cholesterol cross-reactivity was investigated after grafting each of the three scFv types onto two configurations of constant chain domains–CH1-4 and CH2-4. Weak but significant cross-reactivity with cholesterol was found only with CH2-4 versions, notably those two that were also able to recognize the trans-keto MA. The cholesteroid nature of mycobacterial mycolic acids therefore seems to be determined by the trans-keto MA subclass. The significantly weaker binding to cholesterol in comparison to MA confirms the potential TB diagnostic application of these antibodies.
Collapse
Affiliation(s)
- Heena Ranchod
- Polymers and Composites, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department Biochemistry, University of Pretoria, Pretoria, South Africa
- * E-mail:
| | - Fortunate Ndlandla
- Polymers and Composites, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department Biochemistry, University of Pretoria, Pretoria, South Africa
| | - Yolandy Lemmer
- Polymers and Composites, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Mervyn Beukes
- Department Biochemistry, University of Pretoria, Pretoria, South Africa
| | - Johann Niebuhr
- Department Biochemistry, University of Pretoria, Pretoria, South Africa
| | | | - Susan Wemmer
- Serology and Immunochemistry, Vaccines and Diagnostics Development Programme, Agricultural Research Council—Onderstepoort Veterinary Institute, Pretoria, South Africa
| | - Jeanni Fehrsen
- Serology and Immunochemistry, Vaccines and Diagnostics Development Programme, Agricultural Research Council—Onderstepoort Veterinary Institute, Pretoria, South Africa
| | - Mark Baird
- School of Chemistry, Bangor University, Wales, United Kingdom
| | - Jan Verschoor
- Department Biochemistry, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
15
|
Walpole GFW, Grinstein S, Westman J. The role of lipids in host-pathogen interactions. IUBMB Life 2018; 70:384-392. [PMID: 29573124 DOI: 10.1002/iub.1737] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 02/27/2018] [Indexed: 11/07/2022]
Abstract
Innate immunity relies on the effective recognition and elimination of pathogenic microorganisms. This entails sequestration of pathogens into phagosomes that promptly acquire microbicidal and degradative properties. This complex series of events, which involve cytoskeletal reorganization, membrane remodeling and the activation of multiple enzymes, is orchestrated by lipid signaling. To overcome this immune response, intracellular pathogens acquired mechanisms to subvert phosphoinositide-mediated signaling and use host lipids, notably cholesterol, as nutrients. We present brief overviews of the role of phosphoinositides in phagosome formation and maturation as well as of cholesterol handling by host cells, and selected Salmonella, Shigella, Chlamydia and Mycobacterium tuberculosis to exemplify the mechanisms whereby intracellular pathogens co-opt lipid metabolism in host cells. © 2018 IUBMB Life, 70(5):384-392, 2018.
Collapse
Affiliation(s)
- Glenn F W Walpole
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Ontario, Canada
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Biochemistry, University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Johannes Westman
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|