1
|
Krueger G, Faisal S, Dorhoi A. Microenvironments of tuberculous granuloma: advances and opportunities for therapy. Front Immunol 2025; 16:1575133. [PMID: 40196129 PMCID: PMC11973276 DOI: 10.3389/fimmu.2025.1575133] [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: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
The hallmark tissue lesions of tuberculosis (TB) are granulomas. These multicellular structures exhibit varying degrees of cellular complexity, are dynamic, and show considerable diversity within and between hosts. Categorization based on gross pathologic features, particularly caseation and necrosis, was historically coined prior to the identification of mycobacteria as the causative agent of TB. More recently, granuloma zonation based on immune cell composition, metabolite abundance, and physical characteristics has gained attention. With the advent of single-cell analyses, distinct microenvironments and cellular ecosystems within TB granulomas have been identified. We summarize the architecture of TB granulomas and highlight their cellular heterogeneity, including cell niches as well as physical factors such as oxygen gradients that modulate lesion fate. We discuss opportunities for therapy, highlighting new models and the power of in silico modeling to unravel granuloma features and trajectories. Understanding the relevance of the granuloma microenvironment to disease pathophysiology will facilitate the development of more effective interventions, such as host-directed therapies for TB.
Collapse
Affiliation(s)
- Gesa Krueger
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Shah Faisal
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
2
|
Xie L, Wu H, Li Y, Shi L, Liu Y. Recent Development of Nanozymes for Combating Bacterial Drug Resistance: A Review. Adv Healthc Mater 2025; 14:e2402659. [PMID: 39388414 DOI: 10.1002/adhm.202402659] [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: 07/19/2024] [Revised: 08/31/2024] [Indexed: 10/12/2024]
Abstract
The World Health Organization has warned that without effective action, deaths from drug-resistant bacteria can exceed 10 million annually, making it the leading cause of death. Conventional antibiotics are becoming less effective due to rapid bacterial drug resistance and slowed new antibiotic development, necessitating new strategies. Recently, materials with catalytic/enzymatic properties, known as nanozymes, have been developed, inspired by natural enzymes essential for bacterial eradication. Unlike recent literature reviews that broadly cover nanozyme design and biomedical applications, this review focuses on the latest advancements in nanozymes for combating bacterial drug resistance, emphasizing their design, structural characteristics, applications in combination therapy, and future prospects. This approach aims to promote nanozyme development for combating bacterial drug resistance, especially towards clinical translation.
Collapse
Affiliation(s)
- Lingping Xie
- The People's Hospital of Yuhuan, Taizhou, Zhejiang, 317600, China
| | - Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Yuanfeng Li
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- The People's Hospital of Yuhuan, Taizhou, Zhejiang, 317600, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
3
|
Venkateswaran SV, Kreuzaler P, Maclachlan C, McMahon G, Greenidge G, Collinson L, Bunch J, Yuneva M. A multimodal imaging pipeline to decipher cell-specific metabolic functions and tissue microenvironment dynamics. Nat Protoc 2025:10.1038/s41596-024-01118-4. [PMID: 39880930 PMCID: PMC7617660 DOI: 10.1038/s41596-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/15/2024] [Indexed: 01/31/2025]
Abstract
Tissue microenvironments are extremely complex and heterogeneous. It is challenging to study metabolic interaction between the different cell types in a tissue with the techniques that are currently available. Here we describe a multimodal imaging pipeline that allows cell type identification and nanoscale tracing of stable isotope-labeled compounds. This pipeline extends upon the principles of correlative light, electron and ion microscopy, by combining confocal microscopy reporter or probe-based fluorescence, electron microscopy, stable isotope labeling and nanoscale secondary ion mass spectrometry. We apply this method to murine models of hepatocellular and mammary gland carcinomas to study uptake of glucose derived carbon (13C) and glutamine derived nitrogen (15N) by tumor-associated immune cells. In vivo labeling with fluorescent-tagged antibodies (B220, CD3, CD8a, CD68) in tandem with confocal microscopy allows for the identification of specific cell types (B cells, T cells and macrophages) in the tumor microenvironment. Subsequent image correlation with electron microscopy offers the contrast and resolution to image membranes and organelles. Nanoscale secondary ion mass spectrometry tracks the enrichment of stable isotopes within these intracellular compartments. The whole protocol described here would take ~6 weeks to perform from start to finish. Our pipeline caters to a broad spectrum of applications as it can easily be adapted to trace the uptake and utilization of any stable isotope-labeled nutrient, drug or a probe by defined cellular populations in any tissue in situ.
Collapse
Affiliation(s)
| | - Peter Kreuzaler
- The Francis Crick Institute, London, UK.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany.
| | | | - Greg McMahon
- The National Physical Laboratory, Teddington, UK
| | | | | | | | | |
Collapse
|
4
|
Kim J, Spears I, Erice C, Kim HYH, Porter NA, Tressler C, Tucker EW. Spatially heterogeneous lipid dysregulation in tuberculous meningitis. Neurobiol Dis 2024; 202:106721. [PMID: 39489454 DOI: 10.1016/j.nbd.2024.106721] [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: 06/21/2024] [Revised: 10/03/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024] Open
Abstract
Tuberculous (TB) meningitis is the deadliest form of extrapulmonary TB which disproportionately affects children and immunocompromised individuals. Studies in pulmonary TB have shown that Mycobacterium tuberculosis can alter host lipid metabolism to evade the immune system. Cholesterol lowering drugs (i.e., statins) reduce the risk of infection, making them a promising host-directed therapy in pulmonary TB. However, the effect of M. tuberculosis infection on the young or adult brain lipidome has not been studied. The brain is the second-most lipid-rich organ, after adipose tissue, with a temporally and spatially heterogeneous lipidome that changes from infancy to adulthood. The young, developing brain in children may be uniquely vulnerable to alterations in lipid composition and homeostasis, as perturbations in cholesterol metabolism can cause developmental disorders leading to intellectual disabilities. To begin to understand the alterations to the brain lipidome in pediatric TB meningitis, we utilized our previously published young rabbit model of TB meningitis and applied mass spectrometry (MS) techniques to elucidate spatial differences. We used matrix assisted laser desorption/ionization-MS imaging (MALDI-MSI) and complemented it with region-specific liquid chromatography (LC)-MS/MS developed to identify and quantify sterols and oxysterols difficult to identify by MALDI-MSI. MALDI-MSI revealed several sphingolipids, glycerolipids and glycerophospholipids that were downregulated in brain lesions. LC-MS/MS revealed the downregulation of cholesterol, several sterol intermediates along the cholesterol biosynthesis pathway and enzymatically produced oxysterols as a direct result of M. tuberculosis infection. However, oxysterols produced by oxidative stress were increased in brain lesions. Together, these results demonstrate significant spatially regulated brain lipidome dysregulation in pediatric TB meningitis.
Collapse
Affiliation(s)
- John Kim
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ian Spears
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Clara Erice
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Hye-Young H Kim
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Ned A Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37235, USA
| | - Caitlin Tressler
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer, Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Elizabeth W Tucker
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
5
|
Sherry J, Rego EH. Phenotypic Heterogeneity in Pathogens. Annu Rev Genet 2024; 58:183-209. [PMID: 39083846 DOI: 10.1146/annurev-genet-111523-102459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Pathogen diversity within an infected organism has traditionally been explored through the lens of genetic heterogeneity. Hallmark studies have characterized how genetic diversity within pathogen subpopulations contributes to treatment escape and infectious disease progression. However, recent studies have begun to reveal the mechanisms by which phenotypic heterogeneity is established within genetically identical populations of invading pathogens. Furthermore, exciting new work highlights how these phenotypically heterogeneous subpopulations contribute to a pathogen population better equipped to handle the complex and fluctuating environment of a host organism. In this review, we focus on how bacterial pathogens, including Staphylococcus aureus, Salmonella typhimurium, Pseudomonas aeruginosa, and Mycobacterium tuberculosis, establish and maintain phenotypic heterogeneity, and we explore recent work demonstrating causative links between this heterogeneity and infection outcome.
Collapse
Affiliation(s)
- Jessica Sherry
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA; ,
| | - E Hesper Rego
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA; ,
| |
Collapse
|
6
|
Nitschke J, Huber R, Vossio S, Moreau D, Marcourt L, Gindro K, Queiroz EF, Soldati T, Hanna N. Discovery of anti-infective compounds against Mycobacterium marinum after biotransformation of simple natural stilbenes by a fungal secretome. Front Microbiol 2024; 15:1439814. [PMID: 39355425 PMCID: PMC11443511 DOI: 10.3389/fmicb.2024.1439814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, remains a serious threat to human health worldwide and the quest for new anti-tubercular drugs is an enduring and demanding journey. Natural products (NPs) have played a significant role in advancing drug therapy of infectious diseases. Methods This study evaluated the suitability of a high-throughput infection system composed of the host amoeba Dictyostelium discoideum (Dd) and Mycobacterium marinum (Mm), a close relative of Mtb, to identify anti-infective compounds. Growth of Dd and intracellular Mm were quantified by using luminescence and fluorescence readouts in phenotypic assays. The system was first benchmarked with a set of therapeutic anti-Mtb antibiotics and then used to screen a library of biotransformed stilbenes. Results The study confirmed both efficacy of established antibiotics such as rifampicin and bedaquiline, with activities below defined anti-mycobacterium susceptibility breakpoints, and the lack of activity of pyrazinamide against Mm. The screening revealed the promising anti-infective activities of trans-δ-viniferins and in particular of two compounds 17 and 19 with an IC50 of 18.1 μM, 9 μM, respectively. Both compounds had no activity on Mm in broth. Subsequent exploration via halogenation and structure-activity relationship studies led to the identification of derivatives with improved selectivity and potency. The modes of action of the anti-infective compounds may involve inhibition of mycobacterial virulence factors or boosting of host defense. Discussion The study highlights the potential of biotransformation and NP-inspired derivatization approaches for drug discovery and underscores the utility of the Dd-Mm infection system in identifying novel anti-infective compounds.
Collapse
Affiliation(s)
- Jahn Nitschke
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Robin Huber
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Stefania Vossio
- ACCESS Screening Platform, NCCR Chemical Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Dimitri Moreau
- ACCESS Screening Platform, NCCR Chemical Biology, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Katia Gindro
- Mycology Group, Research Department Plant Protection, Agroscope, Nyon, Switzerland
| | - Emerson F. Queiroz
- School of Pharmaceutical Sciences, University of Geneva, CMU, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, Geneva, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| | - Nabil Hanna
- Department of Biochemistry, Faculty of Science, University of Geneva, Geneva, Switzerland
| |
Collapse
|
7
|
King JJ, Chen K, Evans CW, Norret M, Almasri R, Pavlos NJ, Hui HY, Lin Q, Bhatt U, Young SG, Smith NM, Nikan M, Prestidge CA, Jiang H, Iyer KS. High-resolution visualisation of antisense oligonucleotide release from polymers in cells. Chem Sci 2024:d3sc06773d. [PMID: 39246363 PMCID: PMC11378015 DOI: 10.1039/d3sc06773d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Antisense oligonucleotides (ASOs) are a well-established therapeutic modality based on RNA interference, but low cellular uptake, limited ability to direct ASO trafficking, and a range of intracellular barriers to successful activity compromise both gene silencing outcomes and clinical translations. Herein, we demonstrate that polymers can increase ASO internalisation via intracellular trafficking pathways that are distinct from lipid-based delivery reagents. For the first time, we spatially define internalisation and dissociation stages in the polymer-mediated cytosolic delivery of ASOs using Nanoscale Secondary Ion Mass Spectrometry (NanoSIMS), which enables visualisation of ASO localisation at the organelle level. We find that polymer-ASO complexes are imported into cells, from which free ASO enters the cytosol following complex dissociation. This information enables a better understanding of the intracellular trafficking pathways of nucleic acid therapeutics and may be exploited for therapeutic delivery to enhance the effectiveness of nucleic acid therapeutics in the future.
Collapse
Affiliation(s)
- Jessica J King
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Kai Chen
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Cameron W Evans
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Marck Norret
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Ruba Almasri
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
- UniSA Clinical & Health Sciences, University of South Australia Adelaide SA Australia
| | - Nathan J Pavlos
- School of Biomedical Sciences, The University of Western Australia Perth WA 6009 Australia
| | - Henry Yl Hui
- Translational Cancer Pathology Laboratory, School of Biomedical Sciences, The University of Western Australia Perth WA 6009 Australia
| | - Qiongxiang Lin
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Uditi Bhatt
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Stephen G Young
- Department of Medicine, University of California Los Angeles CA 90095 USA
| | - Nicole M Smith
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| | - Mehran Nikan
- Ionis Pharmaceuticals, Inc. Carlsbad CA 92010 USA
| | - Clive A Prestidge
- UniSA Clinical & Health Sciences, University of South Australia Adelaide SA Australia
| | - Haibo Jiang
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
- Department of Chemistry, Faculty of Science, University of Hong Kong Pok Fu Lam Hong Kong
| | - K Swaminathan Iyer
- School of Molecular Sciences, The University of Western Australia Perth WA 6009 Australia
- ARC Training Centre for Next-Generation Biomedical Analysis, The University of Western Australia Perth WA 6009 Australia
| |
Collapse
|
8
|
Bussi C, Lai R, Athanasiadi N, Gutierrez MG. Physiologic medium renders human iPSC-derived macrophages permissive for M. tuberculosis by rewiring organelle function and metabolism. mBio 2024; 15:e0035324. [PMID: 38984828 PMCID: PMC11323749 DOI: 10.1128/mbio.00353-24] [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: 02/05/2024] [Accepted: 06/04/2024] [Indexed: 07/11/2024] Open
Abstract
In vitro studies are crucial for our understanding of the human macrophage immune functions. However, traditional in vitro culture media poorly reflect the metabolic composition of blood, potentially affecting the outcomes of these studies. Here, we analyzed the impact of a physiological medium on human induced pluripotent stem cell (iPSC)-derived macrophages (iPSDM) function. Macrophages cultured in a human plasma-like medium (HPLM) were more permissive to Mycobacterium tuberculosis (Mtb) replication and showed decreased lipid metabolism with increased metabolic polarization. Functionally, we discovered that HPLM-differentiated macrophages showed different metabolic organelle content and activity. Specifically, HPLM-differentiated macrophages displayed reduced lipid droplet and peroxisome content, increased lysosomal proteolytic activity, and increased mitochondrial activity and dynamics. Inhibiting or inducing lipid droplet formation revealed that lipid droplet content is a key factor influencing macrophage permissiveness to Mtb. These findings underscore the importance of using physiologically relevant media in vitro for accurately studying human macrophage function. IMPORTANCE This work compellingly demonstrates that the choice of culture medium significantly influences M. tuberculosis replication outcomes, thus emphasizing the importance of employing physiologically relevant media for accurate in vitro host-pathogen interaction studies. We anticipate that our work will set a precedent for future research with clinical relevance, particularly in evaluating antibiotic efficacy and resistance in cellulo.
Collapse
Affiliation(s)
- Claudio Bussi
- The Francis Crick Institute, London, United Kingdom
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Rachel Lai
- The Francis Crick Institute, London, United Kingdom
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
9
|
Lu C, Chen G, Song W, Chen K, Hee C, Nikan M, Guagliardo P, Bennett CF, Seth P, Iyer KS, Young SG, Qi X, Jiang H. Tool to Resolve Distortions in Elemental and Isotopic Imaging. J Am Chem Soc 2024; 146:20221-20229. [PMID: 38985464 DOI: 10.1021/jacs.4c05384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Nanoscale secondary ion mass spectrometry (NanoSIMS) makes it possible to visualize elements and isotopes in a wide range of samples at a high resolution. However, the fidelity and quality of NanoSIMS images often suffer from distortions because of a requirement to acquire and integrate multiple image frames. We developed an optical flow-based algorithm tool, NanoSIMS Stabilizer, for all-channel postacquisition registration of images. The NanoSIMS Stabilizer effectively deals with the distortions and artifacts, resulting in a high-resolution visualization of isotope and element distribution. It is open source with an easy-to-use ImageJ plugin and is accompanied by a Python version with GPU acceleration.
Collapse
Affiliation(s)
- Chixiang Lu
- Department of Chemistry, The University of Hong Kong, Pok Fu Lam, Hong Kong 999077, P. R. China
| | - Gu Chen
- Department of Chemistry, The University of Hong Kong, Pok Fu Lam, Hong Kong 999077, P. R. China
| | - Wenxin Song
- Departments of Medicine, University of California, Los Angeles, California 90095, United States
| | - Kai Chen
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | - Charmaine Hee
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | - Mehran Nikan
- Ionis Pharmaceuticals, Inc., Carlsbad, California 92010, United States
| | - Paul Guagliardo
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - C Frank Bennett
- Ionis Pharmaceuticals, Inc., Carlsbad, California 92010, United States
| | - Punit Seth
- Ionis Pharmaceuticals, Inc., Carlsbad, California 92010, United States
| | | | - Stephen G Young
- Departments of Medicine, University of California, Los Angeles, California 90095, United States
- Human Genetics, University of California, Los Angeles, California 90095, United States
| | - Xiaojuan Qi
- Electrical and Electronic Engineering, The University of Hong Kong, Pok Fu Lam, Hong Kong 999077, P. R. China
| | - Haibo Jiang
- Department of Chemistry, The University of Hong Kong, Pok Fu Lam, Hong Kong 999077, P. R. China
| |
Collapse
|
10
|
Parkhill SL, Johnson EO. Integrating bacterial molecular genetics with chemical biology for renewed antibacterial drug discovery. Biochem J 2024; 481:839-864. [PMID: 38958473 PMCID: PMC11346456 DOI: 10.1042/bcj20220062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
The application of dyes to understanding the aetiology of infection inspired antimicrobial chemotherapy and the first wave of antibacterial drugs. The second wave of antibacterial drug discovery was driven by rapid discovery of natural products, now making up 69% of current antibacterial drugs. But now with the most prevalent natural products already discovered, ∼107 new soil-dwelling bacterial species must be screened to discover one new class of natural product. Therefore, instead of a third wave of antibacterial drug discovery, there is now a discovery bottleneck. Unlike natural products which are curated by billions of years of microbial antagonism, the vast synthetic chemical space still requires artificial curation through the therapeutics science of antibacterial drugs - a systematic understanding of how small molecules interact with bacterial physiology, effect desired phenotypes, and benefit the host. Bacterial molecular genetics can elucidate pathogen biology relevant to therapeutics development, but it can also be applied directly to understanding mechanisms and liabilities of new chemical agents with new mechanisms of action. Therefore, the next phase of antibacterial drug discovery could be enabled by integrating chemical expertise with systematic dissection of bacterial infection biology. Facing the ambitious endeavour to find new molecules from nature or new-to-nature which cure bacterial infections, the capabilities furnished by modern chemical biology and molecular genetics can be applied to prospecting for chemical modulators of new targets which circumvent prevalent resistance mechanisms.
Collapse
Affiliation(s)
- Susannah L. Parkhill
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
| | - Eachan O. Johnson
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
- Department of Chemistry, Imperial College, London, U.K
- Department of Chemistry, King's College London, London, U.K
| |
Collapse
|
11
|
Fu L, Zhang J, Wang Y, Wu H, Xu X, Li C, Li J, Liu J, Wang H, Jiang X, Li Z, He Y, Liu P, Wu Y, Zou X, Liang B. LET-767 determines lipid droplet protein targeting and lipid homeostasis. J Cell Biol 2024; 223:e202311024. [PMID: 38551495 PMCID: PMC10982117 DOI: 10.1083/jcb.202311024] [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: 11/06/2023] [Revised: 02/22/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024] Open
Abstract
Lipid droplets (LDs) are composed of a core of neutral lipids wrapped by a phospholipid (PL) monolayer containing several hundred proteins that vary between different cells or organisms. How LD proteins target to LDs is still largely unknown. Here, we show that RNAi knockdown or gene mutation of let-767, encoding a member of hydroxysteroid dehydrogenase (HSD), displaced the LD localization of three well-known LD proteins: DHS-3 (dehydrogenase/reductase), PLIN-1 (perilipin), and DGAT-2 (diacylglycerol O-acyltransferase 2), and also prevented LD growth in Caenorhabditis elegans. LET-767 interacts with ARF-1 (ADP-ribosylation factor 1) to prevent ARF-1 LD translocation for appropriate LD protein targeting and lipid homeostasis. Deficiency of LET-767 leads to the release of ARF-1, which further recruits and promotes translocation of ATGL-1 (adipose triglyceride lipase) to LDs for lipolysis. The displacement of LD proteins caused by LET-767 deficiency could be reversed by inhibition of either ARF-1 or ATGL-1. Our work uncovers a unique LET-767 for determining LD protein targeting and maintaining lipid homeostasis.
Collapse
Affiliation(s)
- Lin Fu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Jingjing Zhang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Yanli Wang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Huiyin Wu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Xiumei Xu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Chunxia Li
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Jirong Li
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Jing Liu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Haizhen Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Xue Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan province, Kunming Institute of Zoology, Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Zhihao Li
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Yaomei He
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yingjie Wu
- School of Laboratory Animal and Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models Dalian Medical University, Dalian, China
| | - Xiaoju Zou
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Bin Liang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming, China
- Southwest United Graduate School, Kunming, China
| |
Collapse
|
12
|
Maure A, Lawarée E, Fiorentino F, Pawlik A, Gona S, Giraud-Gatineau A, Eldridge MJG, Danckaert A, Hardy D, Frigui W, Keck C, Gutierrez C, Neyrolles O, Aulner N, Mai A, Hamon M, Barreiro LB, Brodin P, Brosch R, Rotili D, Tailleux L. A host-directed oxadiazole compound potentiates antituberculosis treatment via zinc poisoning in human macrophages and in a mouse model of infection. PLoS Biol 2024; 22:e3002259. [PMID: 38683873 PMCID: PMC11081512 DOI: 10.1371/journal.pbio.3002259] [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: 07/05/2023] [Revised: 05/09/2024] [Accepted: 03/13/2024] [Indexed: 05/02/2024] Open
Abstract
Antituberculosis drugs, mostly developed over 60 years ago, combined with a poorly effective vaccine, have failed to eradicate tuberculosis. More worryingly, multiresistant strains of Mycobacterium tuberculosis (MTB) are constantly emerging. Innovative strategies are thus urgently needed to improve tuberculosis treatment. Recently, host-directed therapy has emerged as a promising strategy to be used in adjunct with existing or future antibiotics, by improving innate immunity or limiting immunopathology. Here, using high-content imaging, we identified novel 1,2,4-oxadiazole-based compounds, which allow human macrophages to control MTB replication. Genome-wide gene expression analysis revealed that these molecules induced zinc remobilization inside cells, resulting in bacterial zinc intoxication. More importantly, we also demonstrated that, upon treatment with these novel compounds, MTB became even more sensitive to antituberculosis drugs, in vitro and in vivo, in a mouse model of tuberculosis. Manipulation of heavy metal homeostasis holds thus great promise to be exploited to develop host-directed therapeutic interventions.
Collapse
Affiliation(s)
- Alexandra Maure
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Emeline Lawarée
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Alexandre Pawlik
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Saideep Gona
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | | | | | - Anne Danckaert
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - David Hardy
- Institut Pasteur, Université Paris Cité, Histopathology Platform, Paris, France
| | - Wafa Frigui
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Camille Keck
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Claude Gutierrez
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nathalie Aulner
- Institut Pasteur, Université Paris Cité, UTechS BioImaging-C2RT, Paris, France
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
- Pasteur Institute, Cenci-bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - Mélanie Hamon
- Institut Pasteur, Université Paris Cité, Chromatine et Infection unit, Paris, France
| | - Luis B. Barreiro
- Department of Genetic Medicine, University of Chicago, Chicago, Illinois, United States of America
| | - Priscille Brodin
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Roland Brosch
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Ludovic Tailleux
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Unit for Integrated Mycobacterial Pathogenomics, Paris, France
| |
Collapse
|
13
|
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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
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
|
14
|
Day NJ, Santucci P, Gutierrez MG. Host cell environments and antibiotic efficacy in tuberculosis. Trends Microbiol 2024; 32:270-279. [PMID: 37709598 DOI: 10.1016/j.tim.2023.08.009] [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: 05/08/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
The aetiologic agent of tuberculosis (TB), Mycobacterium tuberculosis (Mtb), can survive, persist, and proliferate in a variety of heterogeneous subcellular compartments. Therefore, TB chemotherapy requires antibiotics crossing multiple biological membranes to reach distinct subcellular compartments and target these bacterial populations. These compartments are also dynamic, and our understanding of intracellular pharmacokinetics (PK) often represents a challenge for antitubercular drug development. In recent years, the development of high-resolution imaging approaches in the context of host-pathogen interactions has revealed the intracellular distribution of antibiotics at a new level, yielding discoveries with important clinical implications. In this review, we describe the current knowledge regarding cellular PK of antibiotics and the complexity of drug distribution within the context of TB. We also discuss the recent advances in quantitative imaging and highlight their applications for drug development in the context of how intracellular environments and microbial localisation affect TB treatment efficacy.
Collapse
Affiliation(s)
- Nathan J Day
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Pierre Santucci
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
15
|
Kamińska D, Skrzycki M. Lipid droplets, autophagy, and ER stress as key (survival) pathways during ischemia-reperfusion of transplanted grafts. Cell Biol Int 2024; 48:253-279. [PMID: 38178581 DOI: 10.1002/cbin.12114] [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: 07/13/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 01/06/2024]
Abstract
Ischemia-reperfusion injury is an event concerning any organ under a procedure of transplantation. The early result of ischemia is hypoxia, which causes malfunction of mitochondria and decrease in cellular ATP. This leads to disruption of cellular metabolism. Reperfusion also results in cell damage due to reoxygenation and increased production of reactive oxygen species, and later by induced inflammation. In damaged and hypoxic cells, the endoplasmic reticulum (ER) stress pathway is activated by increased amount of damaged or misfolded proteins, accumulation of free fatty acids and other lipids due to inability of their oxidation (lipotoxicity). ER stress is an adaptive response and a survival pathway, however, its prolonged activity eventually lead to induction of apoptosis. Sustaining cell functionality in stress conditions is a great challenge for transplant surgeons as it is crucial for maintaining a desired level of graft vitality. Pathways counteracting negative consequences of ischemia-reperfusion are autophagy and lipid droplets (LD) metabolism. Autophagy remove damaged organelles and molecules driving them to lysosomes, digested simpler compounds are energy source for the cell. Mitophagy and ER-phagy results in improvement of cell energetic balance and alleviation of ER stress. This is important in sustaining metabolic homeostasis and thus cell survival. LD metabolism is connected with autophagy as LD are degraded by lipophagy, a source of free fatty acids and glycerol-thus autophagy and LD can readily remove lipotoxic compounds in the cell. In conclusion, monitoring and pharmaceutic regulation of those pathways during transplantation procedure might result in increased/improved vitality of transplanted organ.
Collapse
Affiliation(s)
- Daria Kamińska
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, Poland
| | - Michał Skrzycki
- Chair and Department of Biochemistry, Medical University of Warsaw, Warszawa, Poland
| |
Collapse
|
16
|
Chen Y, Jiang Y, Xue T, Cheng J. Strategies for the eradication of intracellular bacterial pathogens. Biomater Sci 2024; 12:1115-1130. [PMID: 38284808 DOI: 10.1039/d3bm01498c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Intracellular pathogens affect a significant portion of world population and cause millions of deaths each year. They can invade host cells and survive inside them and are extremely resistant to immune systems and antibiotics. Current treatments have limitations, and therefore, new effective therapies are needed to combat this ongoing health challenge. Active research efforts have been made to develop many new strategies to eradicate these intracellular pathogens. In this review, we focus on the intracellular bacterial pathogens and first introduce several representative intracellular bacteria and the diseases they cause. We then discuss the challenges in eradicating these bacteria and summarize the current therapeutics for intracellular bacteria. Finally, recent advances in intracellular bacteria eradication are highlighted.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | - Yunjiang Jiang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- BayRay Innovation Center, Shenzhen Bay Laboratory, Shenzhen, 518071, China
| | - Tianrui Xue
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Jianjun Cheng
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
- Biomaterials and Drug Delivery Laboratory, School of Engineering, Westlake University, Hangzhou 310024, China
| |
Collapse
|
17
|
Angara RK, Sladek MF, Gilk SD. ER-LD Membrane Contact Sites: A Budding Area in the Pathogen Survival Strategy. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241304196. [PMID: 39697586 PMCID: PMC11653285 DOI: 10.1177/25152564241304196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024]
Abstract
The endoplasmic reticulum (ER) and lipid droplets (LDs) are essential organelles involved in lipid synthesis, storage, and transport. Physical membrane contacts between the ER and LDs facilitate lipid and protein exchange and thus play a critical role in regulating cellular lipid homeostasis. Recent research has revealed that ER-LD membrane contact sites are targeted by pathogens seeking to exploit host lipid metabolic processes. Both viruses and bacteria manipulate ER-LD membrane contact sites to enhance their replication and survival within the host. This review discusses the research advancements elucidating the mechanisms by which pathogens manipulate the ER-LD contacts through protein molecular mimicry and host cell protein manipulation, thereby hijacking host lipid metabolic processes to facilitate pathogenesis. Understanding the crosstalk between ER and LDs during infection provides deeper insight into host lipid regulation and uncovers potential therapeutic targets for treating infectious diseases.
Collapse
Affiliation(s)
- Rajendra Kumar Angara
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Margaret F. Sladek
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Stacey D. Gilk
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
18
|
Toniolo C, Sage D, McKinney JD, Dhar N. Quantification of Mycobacterium tuberculosis Growth in Cell-Based Infection Assays by Time-Lapse Fluorescence Microscopy. Methods Mol Biol 2024; 2813:167-188. [PMID: 38888778 DOI: 10.1007/978-1-0716-3890-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Quantification of Mycobacterium tuberculosis (Mtb) growth dynamics in cell-based in vitro infection models is traditionally carried out by measurement of colony forming units (CFU). However, Mtb being an extremely slow growing organism (16-24 h doubling time), this approach requires at least 3 weeks of incubation to obtain measurable readouts. In this chapter, we describe an alternative approach based on time-lapse microscopy and quantitative image analysis that allows faster quantification of Mtb growth dynamics in host cells. In addition, this approach provides the capability to capture other readouts from the same experimental setup, such as host cell viability, bacterial localization as well as the dynamics of propagation of infection between the host cells.
Collapse
Affiliation(s)
- Chiara Toniolo
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Daniel Sage
- Biomedical Imaging Group, School of Engineering, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - John D McKinney
- School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland
| | - Neeraj Dhar
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada.
- School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
19
|
Boyang H, Yangyanqiu W, Wenting R, Chenxin Y, Jian C, Zhanbo Q, Yanjun Y, Qiang Y, Shuwen H. Application and progress of highcontent imaging in molecular biology. Biotechnol J 2023; 18:e2300170. [PMID: 37639283 DOI: 10.1002/biot.202300170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Humans have adopted many different methods to explore matter imaging, among which high content imaging (HCI) could conduct automated imaging analysis of cells while maintaining its structural and functional integrity. Meanwhile, as one of the most important research tools for diagnosing human diseases, HCI is widely used in the frontier of medical research, and its future application has attracted researchers' great interests. Here, the meaning of HCI was briefly explained, the history of optical imaging and the birth of HCI were described, and the experimental methods of HCI were described. Furthermore, the directions of the application of HCI were highlighted in five aspects: protein localization changes, gene identification, chemical and genetic analysis, microbiology, and drug discovery. Most importantly, some challenges and future directions of HCI were discussed, and the application and optimization of HCI were expected to be further explored.
Collapse
Affiliation(s)
- Hu Boyang
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Wang Yangyanqiu
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Rui Wenting
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Yan Chenxin
- Shulan International Medical School, Zhejiang Shuren University, Hangzhou, China
| | - Chu Jian
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
| | - Qu Zhanbo
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
| | - Yao Yanjun
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Yan Qiang
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Han Shuwen
- Huzhou Hospital of Zhejiang University, Affiliated Central Hospital Huzhou University, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
| |
Collapse
|
20
|
Mishra R, Hannebelle M, Patil VP, Dubois A, Garcia-Mouton C, Kirsch GM, Jan M, Sharma K, Guex N, Sordet-Dessimoz J, Perez-Gil J, Prakash M, Knott GW, Dhar N, McKinney JD, Thacker VV. Mechanopathology of biofilm-like Mycobacterium tuberculosis cords. Cell 2023; 186:5135-5150.e28. [PMID: 37865090 PMCID: PMC10642369 DOI: 10.1016/j.cell.2023.09.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/26/2023] [Accepted: 09/14/2023] [Indexed: 10/23/2023]
Abstract
Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC) and mouse models, cords in alveolar cells contribute to suppression of innate immune signaling via nuclear compression. Thereafter, extracellular cords cause contact-dependent phagocyte death but grow intercellularly between epithelial cells. The absence of these mechanopathological mechanisms explains the greater proportion of alveolar lesions with increased immune infiltration and dissemination defects in cording-deficient Mtb infections. Compression of Mtb lipid monolayers induces a phase transition that enables mechanical energy storage. Agent-based simulations demonstrate that the increased energy storage capacity is sufficient for the formation of cords that maintain structural integrity despite mechanical perturbation. Bacteria in cords remain translationally active despite antibiotic exposure and regrow rapidly upon cessation of treatment. This study provides a conceptual framework for the biophysics and function in tuberculosis infection and therapy of cord architectures independent of mechanisms ascribed to single bacteria.
Collapse
Affiliation(s)
- Richa Mishra
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Melanie Hannebelle
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Vishal P Patil
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Anaëlle Dubois
- BioElectron Microscopy Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Gabriela M Kirsch
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Maxime Jan
- Bioinformatics Competence Centre, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Centre, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Kunal Sharma
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Nicolas Guex
- Bioinformatics Competence Centre, University of Lausanne, 1015 Lausanne, Switzerland; Bioinformatics Competence Centre, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jessica Sordet-Dessimoz
- Histology Core Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jesus Perez-Gil
- Department of Biochemistry, University Complutense Madrid, 28040 Madrid, Spain
| | - Manu Prakash
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Graham W Knott
- BioElectron Microscopy Facility, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Neeraj Dhar
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - John D McKinney
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Vivek V Thacker
- Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
21
|
Zhao P, Zhao Z, Yu Z, Chen L, Jin Y, Wu J, Ren Z. Application of synthetic lipid droplets in metabolic diseases. Clin Transl Med 2023; 13:e1441. [PMID: 37997538 PMCID: PMC10668006 DOI: 10.1002/ctm2.1441] [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: 05/15/2023] [Revised: 09/16/2023] [Accepted: 10/01/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The study and synthesis of membrane organelles are becoming increasingly important, not only as simplified cellular models for corresponding molecular and metabolic studies but also for applications in synthetic biology of artificial cells and drug delivery vehicles. Lipid droplets (LDs) are central organelles in cellular lipid metabolism and are involved in almost all metabolic processes. Multiple studies have also demonstrated a high correlation between LDs and metabolic diseases. During these processes, LDs reveal a highly dynamic character, with their lipid fraction, protein composition and subcellular localisation constantly changing in response to metabolic demands. However, the molecular mechanisms underlying these functions have not been fully understood due to the limitations of cell biology approaches. Fortunately, developments in synthetic biology have provided a huge breakthrough for metabolism research, and methods for in vitro synthesis of LDs have been successfully established, with great advances in protein binding, lipid function, membrane dynamics and enzymatic reactions. AIMS AND METHODS In this review, we provide a comprehensive overview of the assembly and function of endogenous LDs, from the generation of lipid molecules to how they are assembled into LDs in the endoplasmic reticulum. In particular, we highlight two major classes of synthetic LD models for fabrication techniques and their recent advances in biology and explore their roles and challenges in achieving real applications of artificial LDs in the future.
Collapse
Affiliation(s)
- Pengxiang Zhao
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
- College of Animal Science and TechnologyShandong Agricultural UniversityTaianShandongP. R. China
| | - Zichen Zhao
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
| | - Ziwei Yu
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
| | - Lupeng Chen
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
| | - Yi Jin
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
| | - Jian Wu
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionWuhanHubeiP. R. China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal GeneticsBreeding and Reproduction of the Ministry of Education, College of Animal ScienceHuazhong Agricultural UniversityWuhanHubeiP. R. China
- Frontiers Science Center for Animal Breeding and Sustainable ProductionWuhanHubeiP. R. China
- Hubei Hongshan LaboratoryWuhanHubeiP. R. China
| |
Collapse
|
22
|
Kreuzaler P, Inglese P, Ghanate A, Gjelaj E, Wu V, Panina Y, Mendez-Lucas A, MacLachlan C, Patani N, Hubert CB, Huang H, Greenidge G, Rueda OM, Taylor AJ, Karali E, Kazanc E, Spicer A, Dexter A, Lin W, Thompson D, Silva Dos Santos M, Calvani E, Legrave N, Ellis JK, Greenwood W, Green M, Nye E, Still E, Barry S, Goodwin RJA, Bruna A, Caldas C, MacRae J, de Carvalho LPS, Poulogiannis G, McMahon G, Takats Z, Bunch J, Yuneva M. Vitamin B 5 supports MYC oncogenic metabolism and tumor progression in breast cancer. Nat Metab 2023; 5:1870-1886. [PMID: 37946084 PMCID: PMC10663155 DOI: 10.1038/s42255-023-00915-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/28/2023] [Indexed: 11/12/2023]
Abstract
Tumors are intrinsically heterogeneous and it is well established that this directs their evolution, hinders their classification and frustrates therapy1-3. Consequently, spatially resolved omics-level analyses are gaining traction4-9. Despite considerable therapeutic interest, tumor metabolism has been lagging behind this development and there is a paucity of data regarding its spatial organization. To address this shortcoming, we set out to study the local metabolic effects of the oncogene c-MYC, a pleiotropic transcription factor that accumulates with tumor progression and influences metabolism10,11. Through correlative mass spectrometry imaging, we show that pantothenic acid (vitamin B5) associates with MYC-high areas within both human and murine mammary tumors, where its conversion to coenzyme A fuels Krebs cycle activity. Mechanistically, we show that this is accomplished by MYC-mediated upregulation of its multivitamin transporter SLC5A6. Notably, we show that SLC5A6 over-expression alone can induce increased cell growth and a shift toward biosynthesis, whereas conversely, dietary restriction of pantothenic acid leads to a reversal of many MYC-mediated metabolic changes and results in hampered tumor growth. Our work thus establishes the availability of vitamins and cofactors as a potential bottleneck in tumor progression, which can be exploited therapeutically. Overall, we show that a spatial understanding of local metabolism facilitates the identification of clinically relevant, tractable metabolic targets.
Collapse
Affiliation(s)
- Peter Kreuzaler
- The Francis Crick Institute, London, UK.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany.
| | - Paolo Inglese
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, UK
| | | | | | - Vincen Wu
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, UK
| | | | - Andres Mendez-Lucas
- The Francis Crick Institute, London, UK
- Department of Physiological Sciences, University of Barcelona, Barcelona, Spain
| | | | | | | | - Helen Huang
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, UK
| | | | - Oscar M Rueda
- University of Cambridge, MRC Biostatistics Unit, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Evdoxia Karali
- Signalling and Cancer Metabolism Team, Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Emine Kazanc
- Signalling and Cancer Metabolism Team, Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | | | - Alex Dexter
- The National Physical Laboratory, Teddington, UK
| | - Wei Lin
- The Francis Crick Institute, London, UK
| | | | | | | | | | | | - Wendy Greenwood
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | | | - Emma Nye
- The Francis Crick Institute, London, UK
| | | | - Simon Barry
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Richard J A Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Alejandra Bruna
- Modelling of Paediatric Cancer Evolution, Centre for Paediatric Oncology, Experimental Medicine, Centre for Cancer Evolution: Molecular Pathology Division, The Institute of Cancer Research, Belmont, Sutton, London, UK
| | - Carlos Caldas
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge, UK
| | | | | | - George Poulogiannis
- Signalling and Cancer Metabolism Team, Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Greg McMahon
- The National Physical Laboratory, Teddington, UK
| | - Zoltan Takats
- Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, UK
| | - Josephine Bunch
- The National Physical Laboratory, Teddington, UK
- The Rosalind Franklin Institute, Harwell Campus, Didcot, UK
| | | |
Collapse
|
23
|
Beatty KE, López CS. Characteristics of genetic tags for correlative light and electron microscopy. Curr Opin Chem Biol 2023; 76:102369. [PMID: 37453163 DOI: 10.1016/j.cbpa.2023.102369] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023]
Abstract
Fluorescence microscopy is indispensable in live cell studies of fluorescently-labeled proteins, but has limited resolution and context. Electron microscopy offers high-resolution imaging of cellular ultrastructure, including membranes, organelles, and other nanoscale features. However, identifying proteins by EM remains a substantial challenge. There is potential to combine the strengths of both FM and EM through correlative light and EM (CLEM), and bridging the two modalities enables new discoveries and biological insights. CLEM enables cellular proteins to be observed dynamically, across size scales, and in relationship to sub-cellular structures. A central limitation to using CLEM is the scarcity of methods for labeling proteins with CLEM reporters. This review will describe the characteristics of genetic tags for CLEM that are available today, including fixation-resistant fluorescent proteins, 3,3'-diaminobenzidine (DAB)-based tags, metal-chelating tags, DNA origami tags, and VIP tags.
Collapse
Affiliation(s)
- Kimberly E Beatty
- Department of Chemical Physiology and Biochemistry Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | - Claudia S López
- Department of Biomedical Engineering Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
24
|
Ran H, Sun W, Wang L, Wang X, Yu H, Chen J, Liu F, Chao Z, Pu Q, Liu Y, Zeng Y, Li Z, Wan Y, Yuan J. Proteomics coupled transcriptomics reveals lipopolysaccharide inhibiting peroxisome proliferator-activated receptors signalling pathway to reduce lipid droplets accumulation in mouse liver. Proteomics 2023; 23:e2300043. [PMID: 37269196 DOI: 10.1002/pmic.202300043] [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: 01/30/2023] [Revised: 05/04/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023]
Abstract
Lipid droplets (LDs) are multifunctional organelles consisting of a central compartment of non-polar lipids shielded from the cytoplasm by a phospholipid monolayer. The excessive accumulation of LDs in cells is closely related to the development and progression of many diseases in humans and animals, such as liver-related and cardiovascular diseases. Thus, regulating the LDs size and abundance is necessary to maintain metabolic homeostasis. This study found that lipopolysaccharide (LPS) stimulation reduced the LDs content in the mouse liver. We tried to explain the possible molecular mechanisms at the broad protein and mRNA levels, finding that inhibition of the peroxisome proliferator-activated receptors (PPAR) signalling pathway by LPS may be a critical factor in reducing LDs content.
Collapse
Affiliation(s)
- Haiying Ran
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Xiaoyang Wang
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Haili Yu
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Jiajia Chen
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Fang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Zhiyin Chao
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Qi Pu
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Yang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Youlong Zeng
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Zhangfu Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China
| | - Ying Wan
- Biomedical Analysis Center, Army Medical University, Chongqing, China
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong Province, China
| |
Collapse
|
25
|
Kim H, Shin SJ. Revolutionizing control strategies against Mycobacterium tuberculosis infection through selected targeting of lipid metabolism. Cell Mol Life Sci 2023; 80:291. [PMID: 37704889 PMCID: PMC11072447 DOI: 10.1007/s00018-023-04914-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/12/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023]
Abstract
Lipid species play a critical role in the growth and virulence expression of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). During Mtb infection, foamy macrophages accumulate lipids in granulomas, providing metabolic adaptation and survival strategies for Mtb against multiple stresses. Host-derived lipid species, including triacylglycerol and cholesterol, can also contribute to the development of drug-tolerant Mtb, leading to reduced efficacy of antibiotics targeting the bacterial cell wall or transcription. Transcriptional and metabolic analyses indicate that lipid metabolism-associated factors of Mtb are highly regulated by antibiotics and ultimately affect treatment outcomes. Despite the well-known association between major antibiotics and lipid metabolites in TB treatment, a comprehensive understanding of how altered lipid metabolites in both host and Mtb influence treatment outcomes in a drug-specific manner is necessary to overcome drug tolerance. The current review explores the controversies and correlations between lipids and drug efficacy in various Mtb infection models and proposes novel approaches to enhance the efficacy of anti-TB drugs. Moreover, the review provides insights into the efficacious control of Mtb infection by elucidating the impact of lipids on drug efficacy. This review aims to improve the effectiveness of current anti-TB drugs and facilitate the development of innovative therapeutic strategies against Mtb infection by making reverse use of Mtb-favoring lipid species.
Collapse
Affiliation(s)
- Hagyu Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
26
|
Safi R, Sánchez-Álvarez M, Bosch M, Demangel C, Parton RG, Pol A. Defensive-lipid droplets: Cellular organelles designed for antimicrobial immunity. Immunol Rev 2023; 317:113-136. [PMID: 36960679 DOI: 10.1111/imr.13199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Microbes have developed many strategies to subvert host organisms, which, in turn, evolved several innate immune responses. As major lipid storage organelles of eukaryotes, lipid droplets (LDs) are an attractive source of nutrients for invaders. Intracellular viruses, bacteria, and protozoan parasites induce and physically interact with LDs, and the current view is that they "hijack" LDs to draw on substrates for host colonization. This dogma has been challenged by the recent demonstration that LDs are endowed with a protein-mediated antibiotic activity, which is upregulated in response to danger signals and sepsis. Dependence on host nutrients could be a generic "Achilles' heel" of intracellular pathogens and LDs a suitable chokepoint harnessed by innate immunity to organize a front-line defense. Here, we will provide a brief overview of the state of the conflict and discuss potential mechanisms driving the formation of the 'defensive-LDs' functioning as hubs of innate immunity.
Collapse
Affiliation(s)
- Rémi Safi
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Miguel Sánchez-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Instituto de Investigaciones Biomédicas Alberto Sols (IIB), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Caroline Demangel
- Immunobiology and Therapy Unit, Institut Pasteur, Université Paris Cité, INSERM U1224, Paris, France
| | - Robert G Parton
- Institute for Molecular Bioscience (IMB), Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis (CMM), University of Queensland, Brisbane, Queensland, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
27
|
Hüsler D, Stauffer P, Hilbi H. Tapping lipid droplets: A rich fat diet of intracellular bacterial pathogens. Mol Microbiol 2023; 120:194-209. [PMID: 37429596 DOI: 10.1111/mmi.15120] [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: 05/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Lipid droplets (LDs) are dynamic and versatile organelles present in most eukaryotic cells. LDs consist of a hydrophobic core of neutral lipids, a phospholipid monolayer coat, and a variety of associated proteins. LDs are formed at the endoplasmic reticulum and have diverse roles in lipid storage, energy metabolism, membrane trafficking, and cellular signaling. In addition to their physiological cellular functions, LDs have been implicated in the pathogenesis of several diseases, including metabolic disorders, cancer, and infections. A number of intracellular bacterial pathogens modulate and/or interact with LDs during host cell infection. Members of the genera Mycobacterium, Legionella, Coxiella, Chlamydia, and Salmonella exploit LDs as a source of intracellular nutrients and membrane components to establish their distinct intracellular replicative niches. In this review, we focus on the biogenesis, interactions, and functions of LDs, as well as on their role in lipid metabolism of intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Dario Hüsler
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Pia Stauffer
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Jin Y, Tan Y, Wu J, Ren Z. Lipid droplets: a cellular organelle vital in cancer cells. Cell Death Discov 2023; 9:254. [PMID: 37474495 PMCID: PMC10359296 DOI: 10.1038/s41420-023-01493-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
Lipid droplets (LDs) are cellular organelles comprising a core of neutral lipids (glycerides, sterols) encased within a single phospholipid membrane, responsible for storing surplus lipids and furnishing cellular energy. LDs engage in lipid synthesis, catabolism, and transport processes by interacting with other organelles (e.g., endoplasmic reticulum, mitochondria), and they play critical roles in regulating cellular stress and immunity. Recent research has uncovered that an elevated number of LDs is a hallmark of cancer cells, attributable to their enhanced lipid uptake and synthesis capacity, with lipids stored as LDs. Depletion of LDs in cancer cells induces apoptosis, prompting the emergence of small molecule antitumor drugs targeting LDs or key factors (e.g., FASN, SCD1) within the lipid synthesis pathway. Advancements in LD isolation and artificial synthesis have demonstrated their potential applicability in antitumor research. LDs extracted from murine adipose tissue and incubated with lipophilic antitumor drugs yield drug-coated LDs, which promote apoptosis in cancer cells. Furthermore, LDs have been employed as biological lenses to augment the resolution of subcellular structures (microfilaments, microtubules), facilitating the observation of intricate structures within thicker cells, including cancer cells. This review delineates the functional and metabolic mechanisms of LDs in cancer cells and encapsulates recent progress in LD-centered antitumor research, offering novel insights for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, P. R. China
| | - Yanjie Tan
- Institute of Biomedical Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, P. R. China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, P. R. China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, P. R. China.
- Hubei Hongshan Laboratory, Wuhan, P. R. China.
| |
Collapse
|
29
|
Lensen A, Gomes MC, López-Jiménez AT, Mostowy S. An automated microscopy workflow to study Shigella-neutrophil interactions and antibiotic efficacy in vivo. Dis Model Mech 2023; 16:dmm049908. [PMID: 37161932 PMCID: PMC10184671 DOI: 10.1242/dmm.049908] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/04/2023] [Indexed: 05/11/2023] Open
Abstract
Shigella are Gram-negative bacterial pathogens responsible for bacillary dysentery (also called shigellosis). The absence of a licensed vaccine and widespread emergence of antibiotic resistance has led the World Health Organisation (WHO) to highlight Shigella as a priority pathogen requiring urgent attention. Several infection models have been useful to explore the Shigella infection process; yet, we still lack information regarding events taking place in vivo. Here, using a Shigella-zebrafish infection model and high-content microscopy, we developed an automated microscopy workflow to non-invasively study fluorescently labelled bacteria and neutrophils in vivo. We applied our workflow to antibiotic-treated zebrafish, and demonstrate that antibiotics reduce bacterial burden and not neutrophil recruitment to the hindbrain ventricle. We discovered that nalidixic acid (a bactericidal antibiotic) can work with leukocytes in an additive manner to control Shigella flexneri infection and can also restrict dissemination of Shigella sonnei from the hindbrain ventricle. We envision that our automated microscopy workflow, applied here to study the interactions between Shigella and neutrophils as well as antibiotic efficacy in zebrafish, can be useful to innovate treatments for infection control in humans.
Collapse
Affiliation(s)
- Arthur Lensen
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
- Département de Biologie, École Normale Supérieure, PSL Université Paris, F-75005, Paris, France
| | - Margarida C. Gomes
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Ana Teresa López-Jiménez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| |
Collapse
|
30
|
Ness T, Van LH, Petermane I, Duarte R, Lange C, Menzies D, Cirillo DM. Rolling out new anti-tuberculosis drugs without diagnostic capacity. Breathe (Sheff) 2023; 19:230084. [PMID: 37492347 PMCID: PMC10365078 DOI: 10.1183/20734735.0084-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/25/2023] [Indexed: 07/27/2023] Open
Abstract
Deaths from tuberculosis (TB) reached over 1.6 million in 2021 with 10.6 million people becoming ill. Multidrug-resistant TB, defined as the Mycobacterium tuberculosis organism having resistance to at least isoniazid and rifampicin, represented 3.9% of new TB cases and 18% of previously treated cases. While new drug regimens continue to be developed and introduced to improve treatment of drug-resistant forms of TB, diagnostic capability to identify drug resistance lags woefully behind. While significant mortality benefits exist for these newer drug regimens, implementing them without proper drug resistance diagnostic capacity could lead to development of more drug resistances and exhaust these new therapeutic tools. Moving forward, the roll-out of new TB drugs and regimens must be paired with implementation of diagnostics to ensure judicious use of resources and the best chance for improving TB worldwide.
Collapse
Affiliation(s)
- Tara Ness
- Baylor College of Medicine, Department of Pediatrics, Global TB Program, Houston, TX, USA
- Contributed equally as first authors
| | - Le Hong Van
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
- Contributed equally as first authors
| | - Ilze Petermane
- Riga East Clinical University Hospital Centre of Tuberculosis and Lung Diseases, Riga, Latvia
| | - Raquel Duarte
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
- EPI Unit, Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- Unidade de Investigação Clínica da Administração Regional de Saúde do Norte, Porto, Portugal
- Serviço de Pneumologia, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Christoph Lange
- Baylor College of Medicine, Department of Pediatrics, Global TB Program, Houston, TX, USA
- Division of Clinical Infectious Diseases, Research Center Borstel, Borstel, Germany
- German Center for Infection Research (DZIF) Partner Site Borstel-Hamburg-Lübeck-Riems, Borstel, Germany
- Respiratory Medicine and International Health, University of Lübeck, Lübeck, Germany
| | - Dick Menzies
- McGill International TB Centre, Montreal Chest Institute, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | | |
Collapse
|
31
|
Finin P, Khan RMN, Oh S, Boshoff HIM, Barry CE. Chemical approaches to unraveling the biology of mycobacteria. Cell Chem Biol 2023; 30:420-435. [PMID: 37207631 PMCID: PMC10201459 DOI: 10.1016/j.chembiol.2023.04.014] [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: 02/06/2023] [Revised: 04/07/2023] [Accepted: 04/27/2023] [Indexed: 05/21/2023]
Abstract
Mycobacterium tuberculosis (Mtb), perhaps more than any other organism, is intrinsically appealing to chemical biologists. Not only does the cell envelope feature one of the most complex heteropolymers found in nature1 but many of the interactions between Mtb and its primary host (we humans) rely on lipid and not protein mediators.2,3 Many of the complex lipids, glycolipids, and carbohydrates biosynthesized by the bacterium still have unknown functions, and the complexity of the pathological processes by which tuberculosis (TB) disease progress offers many opportunities for these molecules to influence the human response. Because of the importance of TB in global public health, chemical biologists have applied a wide-ranging array of techniques to better understand the disease and improve interventions.
Collapse
Affiliation(s)
- Peter Finin
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - R M Naseer Khan
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Sangmi Oh
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Helena I M Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA
| | - Clifton E Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD, USA.
| |
Collapse
|
32
|
Omollo C, Singh V, Kigondu E, Wasuna A, Agarwal P, Moosa A, Ioerger TR, Mizrahi V, Chibale K, Warner DF. Developing synergistic drug combinations to restore antibiotic sensitivity in drug-resistant Mycobacterium tuberculosis. Antimicrob Agents Chemother 2023; 65:AAC.02554-20. [PMID: 33619062 PMCID: PMC8092878 DOI: 10.1128/aac.02554-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/14/2021] [Indexed: 12/17/2022] Open
Abstract
Tuberculosis (TB) is a leading global cause of mortality owing to an infectious agent, accounting for almost one-third of antimicrobial resistance (AMR) deaths annually. We aimed to identify synergistic anti-TB drug combinations with the capacity to restore therapeutic efficacy against drug-resistant mutants of the causative agent, Mycobacterium tuberculosis We investigated combinations containing the known translational inhibitors, spectinomycin (SPT) and fusidic acid (FA), or the phenothiazine, chlorpromazine (CPZ), which disrupts mycobacterial energy metabolism. Potentiation of whole-cell drug efficacy was observed in SPT-CPZ combinations. This effect was lost against an M. tuberculosis mutant lacking the major facilitator superfamily (MFS) efflux pump, Rv1258c. Notably, the SPT-CPZ combination partially restored SPT efficacy against an SPT-resistant mutant carrying a g1379t point mutation in rrs, encoding the mycobacterial 16S ribosomal RNA. Combinations of SPT with FA, which targets the mycobacterial elongation factor G, exhibited potentiating activity against wild-type M. tuberculosis Moreover, this combination produced a modest potentiating effect against both FA-monoresistant and SPT-monoresistant mutants. Finally, combining SPT with the frontline anti-TB agents, rifampicin (RIF) and isoniazid, resulted in enhanced activity in vitro and ex vivo against both drug-susceptible M. tuberculosis and a RIF-monoresistant rpoB S531L mutant.These results support the utility of novel potentiating drug combinations in restoring antibiotic susceptibility of M. tuberculosis strains carrying genetic resistance to any one of the partner compounds.
Collapse
Affiliation(s)
- Charles Omollo
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Vinayak Singh
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Elizabeth Kigondu
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| | - Antonina Wasuna
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
| | - Pooja Agarwal
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Atica Moosa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Thomas R Ioerger
- Texas A&M University, Department of Computer Science, College Station, TX, 77843, USA
| | - Valerie Mizrahi
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DSI/NRF Centre of Excellence for Biomedical Tuberculosis Research, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
33
|
Zhu J, Liu YJ, Fortune SM. Spatiotemporal perspectives on tuberculosis chemotherapy. Curr Opin Microbiol 2023; 72:102266. [PMID: 36745965 PMCID: PMC10023397 DOI: 10.1016/j.mib.2023.102266] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/04/2023] [Indexed: 02/05/2023]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), accounts for over ten million infections and over 1.5 million deaths every year [1]. Upon infection, the seesaw between Mtb and our immune systems creates microenvironments that are compositionally distinctive and changing over time. While the field has begun to better understand the spatial complexity of TB disease, our understanding and experimental dissection of the temporal dynamics of TB and TB drug treatment is much more rudimentary. However, it is the combined spatiotemporal heterogeneity of TB disease that creates niches and time windows within which the pathogen can survive and thrive during treatment. Here, we review the emerging data on the interactions of spatial and temporal dynamics as they relate to TB disease and treatment. A better understanding of the interactions of Mtb, host, and antibiotics through space and time will elucidate treatment failure and potentially identify opportunities for new TB treatment regimens.
Collapse
Affiliation(s)
- Junhao Zhu
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, USA
| | - Yue J Liu
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, USA
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
34
|
Bedard M, van der Niet S, Bernard EM, Babunovic G, Cheng TY, Aylan B, Grootemaat AE, Raman S, Botella L, Ishikawa E, O'Sullivan MP, O'Leary S, Mayfield JA, Buter J, Minnaard AJ, Fortune SM, Murphy LO, Ory DS, Keane J, Yamasaki S, Gutierrez MG, van der Wel N, Moody DB. A terpene nucleoside from M. tuberculosis induces lysosomal lipid storage in foamy macrophages. J Clin Invest 2023; 133:161944. [PMID: 36757797 PMCID: PMC10014106 DOI: 10.1172/jci161944] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Induction of lipid-laden foamy macrophages is a cellular hallmark of tuberculosis (TB) disease, which involves the transformation of infected phagolysosomes from a site of killing into a nutrient-rich replicative niche. Here, we show that a terpenyl nucleoside shed from Mycobacterium tuberculosis, 1-tuberculosinyladenosine (1-TbAd), caused lysosomal maturation arrest and autophagy blockade, leading to lipid storage in M1 macrophages. Pure 1-TbAd, or infection with terpenyl nucleoside-producing M. tuberculosis, caused intralysosomal and peribacillary lipid storage patterns that matched both the molecules and subcellular locations known in foamy macrophages. Lipidomics showed that 1-TbAd induced storage of triacylglycerides and cholesterylesters and that 1-TbAd increased M. tuberculosis growth under conditions of restricted lipid access in macrophages. Furthermore, lipidomics identified 1-TbAd-induced lipid substrates that define Gaucher's disease, Wolman's disease, and other inborn lysosomal storage diseases. These data identify genetic and molecular causes of M. tuberculosis-induced lysosomal failure, leading to successful testing of an agonist of TRPML1 calcium channels that reverses lipid storage in cells. These data establish the host-directed cellular functions of an orphan effector molecule that promotes survival in macrophages, providing both an upstream cause and detailed picture of lysosome failure in foamy macrophages.
Collapse
Affiliation(s)
- Melissa Bedard
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sanne van der Niet
- Electron Microscopy Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Elliott M Bernard
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Gregory Babunovic
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Tan-Yun Cheng
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Beren Aylan
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Anita E Grootemaat
- Electron Microscopy Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Sahadevan Raman
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laure Botella
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Mary P O'Sullivan
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College, Dublin, Ireland
| | - Seónadh O'Leary
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College, Dublin, Ireland
| | - Jacob A Mayfield
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey Buter
- Department of Chemical Biology, Stratingh Institute for Chemistry, Groningen, Netherlands
| | - Adriaan J Minnaard
- Department of Chemical Biology, Stratingh Institute for Chemistry, Groningen, Netherlands
| | - Sarah M Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Daniel S Ory
- Casma Therapeutics, Cambridge, Massachusetts, USA
| | - Joseph Keane
- Department of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College, Dublin, Ireland
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nicole van der Wel
- Electron Microscopy Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - D Branch Moody
- Division of Rheumatology, Immunity and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Rombouts Y, Neyrolles O. The fat is in the lysosome: how Mycobacterium tuberculosis tricks macrophages into storing lipids. J Clin Invest 2023; 133:168366. [PMID: 36919697 PMCID: PMC10014092 DOI: 10.1172/jci168366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), infects primarily macrophages, causing them to differentiate into lipid-laden foamy macrophages that are a primary source of tissue destruction in patients with TB. In this issue of the JCI, Bedard et al. demonstrate that 1-tuberculosinyladenosine, a virulence factor produced by M. tuberculosis, caused lysosomal dysfunction associated with lipid storage in the phagolysosome of macrophages in a manner that mimicked lysosomal storage diseases. This work sheds light on how M. tuberculosis manipulates host lipid metabolism for its survival and opens avenues toward host-directed therapy against TB.
Collapse
|
36
|
Lanni F, Wijnant GJ, Xie M, Osiecki P, Dartois V, Sarathy JP. Adaptation to the intracellular environment of primary human macrophages influences drug susceptibility of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2023; 139:102318. [PMID: 36889104 DOI: 10.1016/j.tube.2023.102318] [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: 08/29/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 01/24/2023]
Abstract
As a facultative intracellular pathogen, M. tuberculosis (Mtb) is highly adapted to evading antibacterial mechanisms in phagocytic cells. Both the macrophage and pathogen experience transcriptional and metabolic changes from the onset of phagocytosis. To account for this interaction in the assessment of intracellular drug susceptibility, we allowed a 3-day preadaptation phase post-macrophage infection prior to drug treatment. We found that intracellular Mtb in human monocyte-derived macrophages (MDM) presents dramatic alterations in susceptibility to isoniazid, sutezolid, rifampicin and rifapentine when compared to axenic culture. Infected MDM gradually accumulate lipid bodies, adopting a characteristic appearance reminiscent of foamy macrophages in granulomas. Furthermore, TB granulomas in vivo develop hypoxic cores with decreasing oxygen tension gradients across their radii. Accordingly, we evaluated the effects of hypoxia on preadapted intracellular Mtb in our MDM model. We observed that hypoxia induced greater lipid body formation and no additional shifts in drug tolerance, suggesting that the adaptation of intracellular Mtb to baseline host cell conditions under normoxia dominates changes to intracellular drug susceptibility. Using unbound plasma concentrations in patients as surrogates for free drug concentrations in lung interstitial fluid, we estimate that intramacrophage Mtb in granulomas are exposed to bacteriostatic concentrations of most study drugs.
Collapse
Affiliation(s)
- Faye Lanni
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Gert-Jan Wijnant
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Min Xie
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Paulina Osiecki
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States; Hackensack School of Medicine, Department of Medical Sciences, 123, Metro Boulevard, Nutley, NJ, 07110, United States
| | - Jansy P Sarathy
- Center for Discovery and Innovation, 111 Ideation Way, Nutley, NJ, 07110, United States.
| |
Collapse
|
37
|
Greenstein T, Aldridge BB. Tools to develop antibiotic combinations that target drug tolerance in Mycobacterium tuberculosis. Front Cell Infect Microbiol 2023; 12:1085946. [PMID: 36733851 PMCID: PMC9888313 DOI: 10.3389/fcimb.2022.1085946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 01/08/2023] Open
Abstract
Combination therapy is necessary to treat tuberculosis to decrease the rate of disease relapse and prevent the acquisition of drug resistance, and shorter regimens are urgently needed. The adaptation of Mycobacterium tuberculosis to various lesion microenvironments in infection induces various states of slow replication and non-replication and subsequent antibiotic tolerance. This non-heritable tolerance to treatment necessitates lengthy combination therapy. Therefore, it is critical to develop combination therapies that specifically target the different types of drug-tolerant cells in infection. As new tools to study drug combinations earlier in the drug development pipeline are being actively developed, we must consider how to best model the drug-tolerant cells to use these tools to design the best antibiotic combinations that target those cells and shorten tuberculosis therapy. In this review, we discuss the factors underlying types of drug tolerance, how combination therapy targets these populations of bacteria, and how drug tolerance is currently modeled for the development of tuberculosis multidrug therapy. We highlight areas for future studies to develop new tools that better model drug tolerance in tuberculosis infection specifically for combination therapy testing to bring the best drug regimens forward to the clinic.
Collapse
Affiliation(s)
- Talia Greenstein
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Bree B Aldridge
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, Boston, MA, United States
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, United States
| |
Collapse
|
38
|
Larkins-Ford J, Aldridge BB. Advances in the design of combination therapies for the treatment of tuberculosis. Expert Opin Drug Discov 2023; 18:83-97. [PMID: 36538813 PMCID: PMC9892364 DOI: 10.1080/17460441.2023.2157811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Tuberculosis requires lengthy multi-drug therapy. Mycobacterium tuberculosis occupies different tissue compartments during infection, making drug access and susceptibility patterns variable. Antibiotic combinations are needed to ensure each compartment of infection is reached with effective drug treatment. Despite drug combinations' role in treating tuberculosis, the design of such combinations has been tackled relatively late in the drug development process, limiting the number of drug combinations tested. In recent years, there has been significant progress using in vitro, in vivo, and computational methodologies to interrogate combination drug effects. AREAS COVERED This review discusses the advances in these methodologies and how they may be used in conjunction with new successful clinical trials of novel drug combinations to design optimized combination therapies for tuberculosis. Literature searches for approaches and experimental models used to evaluate drug combination effects were undertaken. EXPERT OPINION We are entering an era richer in combination drug effect and pharmacokinetic/pharmacodynamic data, genetic tools, and outcome measurement types. Application of computational modeling approaches that integrate these data and produce predictive models of clinical outcomes may enable the field to generate novel, effective multidrug therapies using existing and new drug combination backbones.
Collapse
Affiliation(s)
- Jonah Larkins-Ford
- Department of Molecular Biology and Microbiology and Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (CIMAR), Tufts University, Boston, MA, USA
- Current address: MarvelBiome Inc, Woburn, MA, USA
| | - Bree B. Aldridge
- Department of Molecular Biology and Microbiology and Tufts University School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (CIMAR), Tufts University, Boston, MA, USA
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, USA
| |
Collapse
|
39
|
de Almeida PE, Pereira de Sousa NM, Rampinelli PG, Silva RVDS, Correa JR, D’Avila H. Lipid droplets as multifunctional organelles related to the mechanism of evasion during mycobacterial infection. Front Cell Infect Microbiol 2023; 13:1102643. [PMID: 36909724 PMCID: PMC9996354 DOI: 10.3389/fcimb.2023.1102643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Tuberculosis (TB) is an infectious disease caused by the bacteria of the Mycobaterium tuberculosis (Mtb) complex. The modulation of the lipid metabolism has been implicated in the immune response regulation, including the formation of lipid droplets (LD)s, LD-phagosome association and eicosanoid synthesis. Mtb, M. bovis BCG and other pathogenic mycobacteria, as well as wall components, such as LAM, can induce LDs formation in a mechanism involving surface receptors, for instance TLRs, CD36, CD14, CD11b/CD18 and others. In addition, the activation of the lipid-activated nuclear receptor PPARγ is involved in the mechanisms of LD biogenesis, as well as in the modulation of the synthesis of lipid mediators. In infected cells, LDs are sites of compartmentalized prostaglandin E2 synthesis involved in macrophage deactivation, bacterial replication and regulation of the host cytokine profile. LDs also have a function in vesicle traffic during infection. Rab7 and RILP, but not Rab5, are located on LDs of infected macrophages, suggesting that LDs and phagosomes could exchange essential proteins for phagosomal maturation, interfering in mycobacterial survival. The pharmacological inhibition of LDs biogenesis affects the bacterial replication and the synthesis of lipid mediators and cytokines, suggesting that LDs may be new targets for antimicrobial therapies. However, it is still controversial if the accumulation of LDs favors the mycobacterial survival acting as an escape mechanism, or promotes the host resistance to infection. Thus, in this mini-review we discuss recent advances in understanding the important role of LDs in the course of infections and the implications for the pathophysiology of mycobacteriosis.
Collapse
Affiliation(s)
- Patrícia Elaine de Almeida
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
- *Correspondence: Heloisa D’Avila, ; Patrícia Elaine de Almeida, ; José Raimundo Correa,
| | - Núbia Maria Pereira de Sousa
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil
| | - Pollianne Garbero Rampinelli
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - Renata Vieira de Sousa Silva
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - José Raimundo Correa
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, University of Brasilia, Brasilia, DF, Brazil
- *Correspondence: Heloisa D’Avila, ; Patrícia Elaine de Almeida, ; José Raimundo Correa,
| | - Heloisa D’Avila
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
- *Correspondence: Heloisa D’Avila, ; Patrícia Elaine de Almeida, ; José Raimundo Correa,
| |
Collapse
|
40
|
Ramaprasad A, Burda PC, Calvani E, Sait AJ, Palma-Duran SA, Withers-Martinez C, Hackett F, Macrae J, Collinson L, Gilberger TW, Blackman MJ. A choline-releasing glycerophosphodiesterase essential for phosphatidylcholine biosynthesis and blood stage development in the malaria parasite. eLife 2022; 11:e82207. [PMID: 36576255 PMCID: PMC9886279 DOI: 10.7554/elife.82207] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
The malaria parasite Plasmodium falciparum synthesizes significant amounts of phospholipids to meet the demands of replication within red blood cells. De novo phosphatidylcholine (PC) biosynthesis via the Kennedy pathway is essential, requiring choline that is primarily sourced from host serum lysophosphatidylcholine (lysoPC). LysoPC also acts as an environmental sensor to regulate parasite sexual differentiation. Despite these critical roles for host lysoPC, the enzyme(s) involved in its breakdown to free choline for PC synthesis are unknown. Here, we show that a parasite glycerophosphodiesterase (PfGDPD) is indispensable for blood stage parasite proliferation. Exogenous choline rescues growth of PfGDPD-null parasites, directly linking PfGDPD function to choline incorporation. Genetic ablation of PfGDPD reduces choline uptake from lysoPC, resulting in depletion of several PC species in the parasite, whilst purified PfGDPD releases choline from glycerophosphocholine in vitro. Our results identify PfGDPD as a choline-releasing glycerophosphodiesterase that mediates a critical step in PC biosynthesis and parasite survival.
Collapse
Affiliation(s)
- Abhinay Ramaprasad
- Malaria Biochemistry Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Paul-Christian Burda
- Centre for Structural Systems BiologyHamburgGermany
- Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- University of HamburgHamburgGermany
| | - Enrica Calvani
- Metabolomics Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Aaron J Sait
- Electron Microscopy Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | | | | | - Fiona Hackett
- Malaria Biochemistry Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - James Macrae
- Metabolomics Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Lucy Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Tim Wolf Gilberger
- Centre for Structural Systems BiologyHamburgGermany
- Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- University of HamburgHamburgGermany
| | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick InstituteLondonUnited Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical MedicineLondonUnited Kingdom
| |
Collapse
|
41
|
Sarrazin M, Martin BP, Avellan R, Gnawali GR, Poncin I, Le Guenno H, Spilling CD, Cavalier JF, Canaan S. Synthesis and Biological Characterization of Fluorescent Cyclipostins and Cyclophostin Analogues: New Insights for the Diagnosis of Mycobacterial-Related Diseases. ACS Infect Dis 2022; 8:2564-2578. [PMID: 36379042 DOI: 10.1021/acsinfecdis.2c00448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Patients with cystic fibrosis (CF) have a significantly higher risk of acquiring nontuberculous mycobacteria infections, predominantly due to Mycobacterium abscessus, than the healthy population. Because M. abscessus infections are a major cause of clinical decline and morbidity in CF patients, improving treatment and the detection of this mycobacterium in the context of a polymicrobial culture represents a critical component to better manage patient care. We report here the synthesis of fluorescent Dansyl derivatives of four active cyclipostins and cyclophostin analogues (CyCs) and provide new insights regarding the CyC's lack of activity against Gram-negative and Gram-positive bacteria, and above all into their mode of action against intramacrophagic M. abscessus cells. Our results pointed out that the intracellularly active CyC accumulate in acidic compartments within macrophage cells, that this accumulation appears to be essential for their delivery to mycobacteria-containing phagosomes, and consequently, for their antimicrobial effect against intracellular replicating M. abscessus, and that modification of such intracellular localization via disruption of endolysosomal pH strongly affects the CyC accumulation and efficacy. Moreover, we discovered that these fluorescent compounds could become efficient probes to specifically label mycobacterial species with high sensitivity, including M. abscessus in the presence several other pathogens like Pseudomonas aeruginosa and Staphylococcus aureus. Collectively, all present and previous data emphasized the therapeutic potential of unlabeled CyCs and the attractiveness of the fluorescent CyC as a potential new efficient diagnostic tool to be exploited in future diagnostic developments against mycobacterial-related infections, especially against M. abscessus.
Collapse
Affiliation(s)
- Morgane Sarrazin
- CNRS, LISM, IMM FR3479, Aix-Marseille Univ, Marseille 13009, France
| | - Benjamin P Martin
- Department of Chemistry & Biochemistry, University of Missouri St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| | - Romain Avellan
- CNRS, LISM, IMM FR3479, Aix-Marseille Univ, Marseille 13009, France
| | - Giri Raj Gnawali
- Department of Chemistry & Biochemistry, University of Missouri St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| | - Isabelle Poncin
- CNRS, LISM, IMM FR3479, Aix-Marseille Univ, Marseille 13009, France
| | - Hugo Le Guenno
- Microscopy Core Facility, IMM FR3479, CNRS, Aix-Marseille Univ, Marseille 13009, France
| | - Christopher D Spilling
- Department of Chemistry & Biochemistry, University of Missouri St. Louis, One University Boulevard, St. Louis, Missouri 63121, United States
| | | | - Stéphane Canaan
- CNRS, LISM, IMM FR3479, Aix-Marseille Univ, Marseille 13009, France
| |
Collapse
|
42
|
Huang S, Chen K, Leung JK, Guagliardo P, Chen W, Song W, Clode P, Xu J, Young SG, Jiang H. Subcellular Partitioning of Arsenic Trioxide Revealed by Label-Free Imaging. Anal Chem 2022; 94:13889-13896. [PMID: 36189785 DOI: 10.1021/acs.analchem.2c02770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Subcellular partitioning of therapeutic agents is highly relevant to their interactions with target molecules and drug efficacy, but studying subcellular partitioning is an enormous challenge. Here, we describe the application of nanoscale secondary ion mass spectrometry (NanoSIMS) analysis to define the subcellular pharmacokinetics of a cytotoxic chemotherapy drug, arsenic trioxide (ATO). We reasoned that defining the partitioning of ATO would yield valuable insights into the mechanisms underlying ATO efficacy. NanoSIMS imaging made it possible to define the intracellular fate of ATO in a label-free manner─and with high resolution and high sensitivity. Our studies of ATO-treated cells revealed that arsenic accumulates in the nucleolus. After prolonged ATO exposure, ∼40 nm arsenic- and sulfur-rich protein aggregates appeared in the cell nucleolus, nucleus, and membrane-free compartments in the cytoplasm, and our studies suggested that the partitioning of nanoscale aggregates could be relevant to cell survival. All-trans retinoic acid increased intracellular ATO levels and accelerated the nanoscale aggregate formation in the nucleolus. This study yielded fresh insights into the subcellular pharmacokinetics of an important cancer therapeutic agent and the potential impact of drug partitioning and pharmacokinetics on drug activity.
Collapse
Affiliation(s)
- Song Huang
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia
| | - Kai Chen
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia.,Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Jong-Kai Leung
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Paul Guagliardo
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia
| | - Weihua Chen
- Department of Chemistry, The University of Hong Kong, Hong Kong, China
| | - Wenxin Song
- Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Peta Clode
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Perth 6009, Australia.,School of Biological Sciences, University of Western Australia, Perth 6009, Australia
| | - Jiake Xu
- School of Biological Sciences, University of Western Australia, Perth 6009, Australia
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, California 90095, United States.,School of Biomedical Sciences, University of Western Australia, Perth 6009, Australia.,Department of Human Genetics, University of California, Los Angeles, California 90095, United States
| | - Haibo Jiang
- School of Molecular Sciences, University of Western Australia, Perth 6009, Australia.,Department of Chemistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
43
|
Singh J, Sanghavi P, Mallik R. Microtubule motor driven interactions of lipid droplets: Specificities and opportunities. Front Cell Dev Biol 2022; 10:893375. [PMID: 36200039 PMCID: PMC9527339 DOI: 10.3389/fcell.2022.893375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Lipid Droplets (LDs) are evolutionarily conserved cellular organelles that store neutral lipids such as triacylglycerol and cholesterol-esters. Neutral lipids are enclosed within the limiting membrane of the LD, which is a monolayer of phospholipids and is therefore fundamentally different from the bilayer membrane enclosing most other organelles. LDs have long been viewed as a storehouse of lipids needed on demand for generating energy and membranes inside cells. Outside this classical view, we are now realizing that LDs have significant roles in protein sequestration, supply of signalling lipids, viral replication, lipoprotein production and many other functions of important physiological consequence. To execute such functions, LDs must often exchange lipids and proteins with other organelles (e.g., the ER, lysosomes, mitochondria) via physical contacts. But before such exchanges can occur, how does a micron-sized LD with limited ability to diffuse around find its cognate organelle? There is growing evidence that motor protein driven motion of LDs along microtubules may facilitate such LD-organelle interactions. We will summarize some aspects of LD motion leading to LD-organelle contacts, how these change with metabolic state and pathogen infections, and also ask how these pathways could perhaps be targeted selectively in the context of disease and drug delivery. Such a possibility arises because the binding of motor proteins to the monolayer membrane on LDs could be different from motor binding to the membrane on other cellular organelles.
Collapse
Affiliation(s)
- Jagjeet Singh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- *Correspondence: Roop Mallik, ; Jagjeet Singh,
| | - Paulomi Sanghavi
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Roop Mallik
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- *Correspondence: Roop Mallik, ; Jagjeet Singh,
| |
Collapse
|
44
|
Lewis HM, Costa C, Dartois V, Kaya F, Chambers M, de Jesus J, Palitsin V, Webb R, Bailey MJ. Colocation of Lipids, Drugs, and Metal Biomarkers Using Spatially Resolved Lipidomics with Elemental Mapping. Anal Chem 2022; 94:11798-11806. [PMID: 35981335 PMCID: PMC9434551 DOI: 10.1021/acs.analchem.2c01940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
![]()
Elemental imaging is widely used for imaging cells and
tissues
but rarely in combination with organic mass spectrometry, which can
be used to profile lipids and measure drug concentrations. Here, we
demonstrate how elemental imaging and a new method for spatially resolved
lipidomics (DAPNe-LC-MS, based on capillary microsampling and liquid
chromatography mass spectrometry) can be used in combination to probe
the relationship between metals, drugs, and lipids in discrete areas
of tissues. This new method for spatial lipidomics, reported here
for the first time, has been applied to rabbit lung tissues containing
a lesion (caseous granuloma) caused by tuberculosis infection. We
demonstrate how elemental imaging with spatially resolved lipidomics
can be used to probe the association between ion accumulation and
lipid profiles and verify local drug distribution.
Collapse
Affiliation(s)
- Holly-May Lewis
- Department of Chemistry, University of Surrey, Guildford, Surrey GU2 7XH, U.K
| | - Catia Costa
- University of Surrey Ion Beam Centre, Guildford, Surrey GU2 7XH, U.K
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, 123 Metro Boulevard, Nutley, New Jersey 07110, United States
| | - Firat Kaya
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, 123 Metro Boulevard, Nutley, New Jersey 07110, United States
| | - Mark Chambers
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey GU2 7XH, U.K
| | - Janella de Jesus
- Department of Chemistry, University of Surrey, Guildford, Surrey GU2 7XH, U.K
| | - Vladimir Palitsin
- University of Surrey Ion Beam Centre, Guildford, Surrey GU2 7XH, U.K
| | - Roger Webb
- University of Surrey Ion Beam Centre, Guildford, Surrey GU2 7XH, U.K
| | - Melanie J Bailey
- Department of Chemistry, University of Surrey, Guildford, Surrey GU2 7XH, U.K
| |
Collapse
|
45
|
Peddie CJ, Genoud C, Kreshuk A, Meechan K, Micheva KD, Narayan K, Pape C, Parton RG, Schieber NL, Schwab Y, Titze B, Verkade P, Aubrey A, Collinson LM. Volume electron microscopy. NATURE REVIEWS. METHODS PRIMERS 2022; 2:51. [PMID: 37409324 PMCID: PMC7614724 DOI: 10.1038/s43586-022-00131-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 07/07/2023]
Abstract
Life exists in three dimensions, but until the turn of the century most electron microscopy methods provided only 2D image data. Recently, electron microscopy techniques capable of delving deep into the structure of cells and tissues have emerged, collectively called volume electron microscopy (vEM). Developments in vEM have been dubbed a quiet revolution as the field evolved from established transmission and scanning electron microscopy techniques, so early publications largely focused on the bioscience applications rather than the underlying technological breakthroughs. However, with an explosion in the uptake of vEM across the biosciences and fast-paced advances in volume, resolution, throughput and ease of use, it is timely to introduce the field to new audiences. In this Primer, we introduce the different vEM imaging modalities, the specialized sample processing and image analysis pipelines that accompany each modality and the types of information revealed in the data. We showcase key applications in the biosciences where vEM has helped make breakthrough discoveries and consider limitations and future directions. We aim to show new users how vEM can support discovery science in their own research fields and inspire broader uptake of the technology, finally allowing its full adoption into mainstream biological imaging.
Collapse
Affiliation(s)
- Christopher J. Peddie
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Christel Genoud
- Electron Microscopy Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Kimberly Meechan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Present address: Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA, USA
| | - Kedar Narayan
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Constantin Pape
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Robert G. Parton
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicole L. Schieber
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Yannick Schwab
- Cell Biology and Biophysics Unit/ Electron Microscopy Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Paul Verkade
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Aubrey Aubrey
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| |
Collapse
|
46
|
Methods combine to decode the biology of tuberculosis. Nature 2022; 605:385-386. [DOI: 10.1038/d41586-022-01260-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Gutierrez MG, Enninga J. Intracellular niche switching as host subversion strategy of bacterial pathogens. Curr Opin Cell Biol 2022; 76:102081. [PMID: 35487154 DOI: 10.1016/j.ceb.2022.102081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/08/2022] [Accepted: 03/13/2022] [Indexed: 11/03/2022]
Abstract
Numerous bacterial pathogens "confine" themselves within host cells with an intracellular localization as main or exclusive niche. Many of them switch dynamically between a membrane-bound or cytosolic lifestyle. This requires either membrane damage and/or repair of the bacterial-containing compartment. Niche switching has profound consequences on how the host cell recognizes the pathogens in time and space for elimination. Moreover, niche switching impacts how bacteria communicate with host cells to obtain nutrients, and it affects the accessibility to antibiotics. Understanding the local environments and cellular phenotypes that lead to niche switching is critical for developing new host-targeted antimicrobial strategies, and has the potential to shed light into fundamental cellular processes.
Collapse
Affiliation(s)
- Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| | - Jost Enninga
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France; Université de Paris, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
48
|
Santucci P, Aylan B, Botella L, Bernard EM, Bussi C, Pellegrino E, Athanasiadi N, Gutierrez MG. Visualizing Pyrazinamide Action by Live Single-Cell Imaging of Phagosome Acidification and Mycobacterium tuberculosis pH Homeostasis. mBio 2022; 13:e0011722. [PMID: 35323041 PMCID: PMC9040869 DOI: 10.1128/mbio.00117-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/24/2022] [Indexed: 01/28/2023] Open
Abstract
Mycobacterium tuberculosis segregates within multiple subcellular niches with different biochemical and biophysical properties that, upon treatment, may impact antibiotic distribution, accumulation, and efficacy. However, it remains unclear whether fluctuating intracellular microenvironments alter mycobacterial homeostasis and contribute to antibiotic enrichment and efficacy. Here, we describe a live dual-imaging approach to monitor host subcellular acidification and M. tuberculosis intrabacterial pH. By combining this approach with pharmacological and genetic perturbations, we show that M. tuberculosis can maintain its intracellular pH independently of the surrounding pH in human macrophages. Importantly, unlike bedaquiline (BDQ), isoniazid (INH), or rifampicin (RIF), the drug pyrazinamide (PZA) displays antibacterial efficacy by disrupting M. tuberculosis intrabacterial pH homeostasis in cellulo. By using M. tuberculosis mutants, we confirmed that intracellular acidification is a prerequisite for PZA efficacy in cellulo. We anticipate this imaging approach will be useful to identify host cellular environments that affect antibiotic efficacy against intracellular pathogens. IMPORTANCE We still do not completely understand why tuberculosis (TB) treatment requires the combination of several antibiotics for up to 6 months. M. tuberculosis is a facultative intracellular pathogen, and it is still unknown whether heterogenous and dynamic intracellular populations of bacteria in different cellular environments affect antibiotic efficacy. By developing a dual live imaging approach to monitor mycobacterial pH homeostasis, host cell environment, and antibiotic action, we show here that intracellular localization of M. tuberculosis affects the efficacy of one first-line anti-TB drug. Our observations can be applicable to the treatment of other intracellular pathogens and help to inform the development of more effective combined therapies for tuberculosis that target heterogenous bacterial populations within the host.
Collapse
Affiliation(s)
- Pierre Santucci
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Beren Aylan
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Laure Botella
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Elliott M. Bernard
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Claudio Bussi
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Enrica Pellegrino
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Natalia Athanasiadi
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Maximiliano G. Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
49
|
Sholeye AR, Williams AA, Loots DT, Tutu van Furth AM, van der Kuip M, Mason S. Tuberculous Granuloma: Emerging Insights From Proteomics and Metabolomics. Front Neurol 2022; 13:804838. [PMID: 35386409 PMCID: PMC8978302 DOI: 10.3389/fneur.2022.804838] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium tuberculosis infection, which claims hundreds of thousands of lives each year, is typically characterized by the formation of tuberculous granulomas — the histopathological hallmark of tuberculosis (TB). Our knowledge of granulomas, which comprise a biologically diverse body of pro- and anti-inflammatory cells from the host immune responses, is based mainly upon examination of lungs, in both human and animal studies, but little on their counterparts from other organs of the TB patient such as the brain. The biological heterogeneity of TB granulomas has led to their diverse, relatively uncoordinated, categorization, which is summarized here. However, there is a pressing need to elucidate more fully the phenotype of the granulomas from infected patients. Newly emerging studies at the protein (proteomics) and metabolite (metabolomics) levels have the potential to achieve this. In this review we summarize the diverse nature of TB granulomas based upon the literature, and amplify these accounts by reporting on the relatively few, emerging proteomics and metabolomics studies on TB granulomas. Metabolites (for example, trimethylamine-oxide) and proteins (such as the peptide PKAp) associated with TB granulomas, and knowledge of their localizations, help us to understand the resultant phenotype. Nevertheless, more multidisciplinary ‘omics studies, especially in human subjects, are required to contribute toward ushering in a new era of understanding of TB granulomas – both at the site of infection, and on a systemic level.
Collapse
Affiliation(s)
- Abisola Regina Sholeye
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Aurelia A. Williams
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - A. Marceline Tutu van Furth
- Department of Pediatric Infectious Diseases and Immunology, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, Netherlands
| | - Martijn van der Kuip
- Department of Pediatric Infectious Diseases and Immunology, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, Netherlands
| | - Shayne Mason
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
- *Correspondence: Shayne Mason
| |
Collapse
|
50
|
A Novel Fluoro-Pyrazine-Bridged Donor-Accepter-Donor Fluorescent Probe for Lipid Droplet-Specific Imaging in Diverse Cells and Superoxide Anion Generation. Pharm Res 2022; 39:1205-1214. [PMID: 35237921 DOI: 10.1007/s11095-022-03216-y] [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: 10/19/2021] [Accepted: 02/23/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE Lipid droplets (LDs) are dynamic organelles which associated with many metabolic processes. Reliable long-term imaging of LD is of great importance in LD-based therapy and research. Conventional fluorescent probes suffer from poor photostability and difficulty of preparation, which compromise their LD imaging ability. In this study, we aim to provide a novel and universal fluorescent probe for LD-specific imaging in both eukaryotic and prokaryotic cells. The versatile and potential applications of the probe were also evaluated. METHODS We used one-step Suzuki coupling reaction to synthesize a fluoro-pyrazine-bridged donor-acceptor-donor fluorescent probe (T-FP-T). The fluorescent properties and stability of T-FP-T were detected. Then, LD-specific imaging and dynamic movement tracking capabilities of T-FP-T were studied in fungus, bacteria, plant and animal tissues. The biosafety and photodynamic toxicity of the probe under different light irradiation were characterized. RESULTS T-FP-T showed large Stokes shift, superior brightness, excellent photostability, low toxicity. T-FP-T exhibited significant overlaps with adipophilin antibody or the commercial LD probe (LipidSpot™) in the cytoplasm, but not with Mitotracker red, Lysotracker red and Peroxisome Labeling dye. Moreover, T-FP-T also showed efficient superoxide anion generation capability under white LED light irradiation. The viability of Hela cells co-treated with T-FP-T and 1-h white LED light irradiation decreased to 62%. CONCLUSIONS All these outstanding capabilities make T-FP-T a new efficient LD-specific imaging probe. The generated superoxide anion from T-FP-T under white LED light irradiation could cause obvious cell death, which will inspire broad study in LD-targeted photodynamic therapy.
Collapse
|