1
|
Bhargava M, Crouser ED. Application of laboratory models for sarcoidosis research. J Autoimmun 2024; 149:103184. [PMID: 38443221 DOI: 10.1016/j.jaut.2024.103184] [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: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 03/07/2024]
Abstract
This manuscript will review the implications and applications of sarcoidosis models towards advancing our understanding of sarcoidosis disease mechanisms, identification of biomarkers, and preclinical testing of novel therapies. Emerging disease models and innovative research tools will also be considered.
Collapse
Affiliation(s)
- Maneesh Bhargava
- University of Minnesota Medical Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, 420 Delaware Street SE, MMC 276. Minneapolis, MN 55455, USA
| | - Elliott D Crouser
- Ohio State University Wexner Medicine Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, 241 W. 11th Street, Suite 5000, Columbus, OH 43201, USA.
| |
Collapse
|
2
|
Martínez-López MF, de Almeida CR, Fontes M, Mendes RV, Kaufmann SHE, Fior R. Macrophages directly kill bladder cancer cells through TNF signaling as an early response to BCG therapy. Dis Model Mech 2024; 17:dmm050693. [PMID: 39114912 PMCID: PMC11554267 DOI: 10.1242/dmm.050693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/09/2024] [Indexed: 11/13/2024] Open
Abstract
The Bacillus Calmette-Guérin (BCG) vaccine is the oldest cancer immunotherapeutic agent in use. Despite its effectiveness, its initial mechanisms of action remain largely unknown. Here, we elucidate the earliest cellular mechanisms involved in BCG-induced tumor clearance. We developed a fast preclinical in vivo assay to visualize in real time and at single-cell resolution the initial interactions among bladder cancer cells, BCG and innate immunity using the zebrafish xenograft model. We show that BCG induced the recruitment and polarization of macrophages towards a pro-inflammatory phenotype, accompanied by induction of the inflammatory cytokines tnfa, il1b and il6 in the tumor microenvironment. Macrophages directly induced apoptosis of human cancer cells through zebrafish TNF signaling. Macrophages were crucial for this response as their depletion completely abrogated the BCG-induced phenotype. Contrary to the general concept that macrophage anti-tumoral activities mostly rely on stimulating an effective adaptive response, we demonstrate that macrophages alone can induce tumor apoptosis and clearance. Thus, our results revealed an additional step to the BCG-induced tumor immunity model, while providing proof-of-concept experiments demonstrating the potential of this unique model to test innate immunomodulators.
Collapse
Affiliation(s)
| | | | - Márcia Fontes
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon 1400-038, Portugal
| | - Raquel Valente Mendes
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon 1400-038, Portugal
| | - Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin 10117, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX 77843, USA
| | - Rita Fior
- Champalimaud Research, Champalimaud Foundation, Av. Brasilia, Lisbon 1400-038, Portugal
| |
Collapse
|
3
|
Johansen MD, Spaink HP, Oehlers SH, Kremer L. Modeling nontuberculous mycobacterial infections in zebrafish. Trends Microbiol 2024; 32:663-677. [PMID: 38135617 DOI: 10.1016/j.tim.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
The incidence of infections due to nontuberculous mycobacteria (NTM) has increased rapidly in recent years, surpassing tuberculosis in developed countries. Due to inherent antimicrobial resistance, NTM infections are particularly difficult to treat with low cure rates. There is an urgent need to understand NTM pathogenesis and to develop novel therapeutic approaches for the treatment of NTM diseases. Zebrafish have emerged as an excellent animal model due to genetic amenability and optical transparency during embryonic development, allowing spatiotemporal visualization of host-pathogen interactions. Furthermore, adult zebrafish possess fully functional innate and adaptive immunity and recapitulate important pathophysiological hallmarks of mycobacterial infection. Here, we report recent breakthroughs in understanding the hallmarks of NTM infections using the zebrafish model.
Collapse
Affiliation(s)
- Matt D Johansen
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW, Australia
| | - Herman P Spaink
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | - Stefan H Oehlers
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Laurent Kremer
- Centre National de la Recherche Scientifique, UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 Route de Mende, 34293, Montpellier, France; INSERM, IRIM, 34293 Montpellier, France.
| |
Collapse
|
4
|
Gupta VK, Vaishnavi VV, Arrieta-Ortiz ML, P S A, K M J, Jeyasankar S, Raghunathan V, Baliga NS, Agarwal R. 3D Hydrogel Culture System Recapitulates Key Tuberculosis Phenotypes and Demonstrates Pyrazinamide Efficacy. Adv Healthc Mater 2024:e2304299. [PMID: 38655817 PMCID: PMC7616495 DOI: 10.1002/adhm.202304299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/29/2024] [Indexed: 04/26/2024]
Abstract
The mortality caused by tuberculosis (TB) infections is a global concern, and there is a need to improve understanding of the disease. Current in vitro infection models to study the disease have limitations such as short investigation durations and divergent transcriptional signatures. This study aims to overcome these limitations by developing a 3D collagen culture system that mimics the biomechanical and extracellular matrix (ECM) of lung microenvironment (collagen fibers, stiffness comparable to in vivo conditions) as the infection primarily manifests in the lungs. The system incorporates Mycobacterium tuberculosis (Mtb) infected human THP-1 or primary monocytes/macrophages. Dual RNA sequencing reveals higher mammalian gene expression similarity with patient samples than 2D macrophage infections. Similarly, bacterial gene expression more accurately recapitulates in vivo gene expression patterns compared to bacteria in 2D infection models. Key phenotypes observed in humans, such as foamy macrophages and mycobacterial cords, are reproduced in the model. This biomaterial system overcomes challenges associated with traditional platforms by modulating immune cells and closely mimicking in vivo infection conditions, including showing efficacy with clinically relevant concentrations of anti-TB drug pyrazinamide, not seen in any other in vitro infection model, making it reliable and readily adoptable for tuberculosis studies and drug screening.
Collapse
Affiliation(s)
- Vishal K Gupta
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Vijaya V Vaishnavi
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | | | - Abhirami P S
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Jyothsna K M
- Department of Electrical Communication Engineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Sharumathi Jeyasankar
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Varun Raghunathan
- Department of Electrical Communication Engineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| | - Nitin S Baliga
- Institute of Systems Biology, 401 Terry Ave N, Seattle, WA, 98109, USA
| | - Rachit Agarwal
- Department of Bioengineering, Indian Institute of Science, CV Raman Road, Bengaluru, Karnataka, 560012, India
| |
Collapse
|
5
|
Petrucciani A, Hoerter A, Kotze L, Du Plessis N, Pienaar E. In silico agent-based modeling approach to characterize multiple in vitro tuberculosis infection models. PLoS One 2024; 19:e0299107. [PMID: 38517920 PMCID: PMC10959380 DOI: 10.1371/journal.pone.0299107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/05/2024] [Indexed: 03/24/2024] Open
Abstract
In vitro models of Mycobacterium tuberculosis (Mtb) infection are a valuable tool for examining host-pathogen interactions and screening drugs. With the development of more complex in vitro models, there is a need for tools to help analyze and integrate data from these models. To this end, we introduce an agent-based model (ABM) representation of the interactions between immune cells and bacteria in an in vitro setting. This in silico model was used to simulate both traditional and spheroid cell culture models by changing the movement rules and initial spatial layout of the cells in accordance with the respective in vitro models. The traditional and spheroid simulations were calibrated to published experimental data in a paired manner, by using the same parameters in both simulations. Within the calibrated simulations, heterogeneous outputs are seen for bacterial count and T cell infiltration into the macrophage core of the spheroid. The simulations also predict that equivalent numbers of activated macrophages do not necessarily result in similar bacterial reductions; that host immune responses can control bacterial growth in both spheroid structure dependent and independent manners; that STAT1 activation is the limiting step in macrophage activation in spheroids; and that drug screening and macrophage activation studies could have different outcomes depending on the in vitro culture used. Future model iterations will be guided by the limitations of the current model, specifically which parts of the output space were harder to reach. This ABM can be used to represent more in vitro Mtb infection models due to its flexible structure, thereby accelerating in vitro discoveries.
Collapse
Affiliation(s)
- Alexa Petrucciani
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Alexis Hoerter
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
| | - Leigh Kotze
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nelita Du Plessis
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medical and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States of America
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
6
|
Luo G, Zeng D, Liu J, Li D, Takiff HE, Song S, Gao Q, Yan B. Temporal and cellular analysis of granuloma development in mycobacterial infected adult zebrafish. J Leukoc Biol 2024; 115:525-535. [PMID: 37982587 DOI: 10.1093/jleuko/qiad145] [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: 11/09/2022] [Revised: 09/25/2023] [Accepted: 11/01/2023] [Indexed: 11/21/2023] Open
Abstract
Because granulomas are a hallmark of tuberculosis pathogenesis, the study of the dynamic changes in their cellular composition and morphological character can facilitate our understanding of tuberculosis pathogenicity. Adult zebrafish infected with Mycobacterium marinum form granulomas that are similar to the granulomas in human patients with tuberculosis and therefore have been used to study host-mycobacterium interactions. Most studies of zebrafish granulomas, however, have focused on necrotic granulomas, while a systematic description of the different stages of granuloma formation in the zebrafish model is lacking. Here, we characterized the stages of granulomas in M. marinum-infected zebrafish, including early immune cell infiltration, nonnecrotizing granulomas, and necrotizing granulomas, using corresponding samples from patients with pulmonary tuberculosis as references. We combined hematoxylin and eosin staining and in situ hybridization to identify the different immune cell types and follow their spatial distribution in the different stages of granuloma development. The macrophages in zebrafish granulomas were shown to belong to distinct subtypes: epithelioid macrophages, foamy macrophages, and multinucleated giant cells. By defining the developmental stages of zebrafish granulomas and the spatial distribution of the different immune cells they contain, this work provides a reference for future studies of mycobacterial granulomas and their immune microenvironments.
Collapse
Affiliation(s)
- Geyang Luo
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity and Shanghai Public Health Clinical Center, Fudan University, 130 Dongan Rd., Xuhui District, 200032 Shanghai, People's Republic of China
| | - Dong Zeng
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Rd., Jinshan District, 201508 Shanghai, People's Republic of China
| | - Jianxin Liu
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Rd., Jinshan District, 201508 Shanghai, People's Republic of China
- School of Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai JiaoTong University, 639 Manufacturing Bureau Rd., Huangpu District, 200011 Shanghai, People's Republic of China
| | - Duoduo Li
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Rd., Jinshan District, 201508 Shanghai, People's Republic of China
| | - Howard E Takiff
- Instituto Venezolano de Investigaciones Científicas, Centro de Microbiología y Biología Celular, Caracas, 1020A, Venezuela
| | - Shu Song
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Rd., Jinshan District, 201508 Shanghai, People's Republic of China
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Shanghai Institute of Infectious Disease and Biosecurity and Shanghai Public Health Clinical Center, Fudan University, 130 Dongan Rd., Xuhui District, 200032 Shanghai, People's Republic of China
| | - Bo Yan
- Center for Tuberculosis Research, Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Rd., Jinshan District, 201508 Shanghai, People's Republic of China
| |
Collapse
|
7
|
Craig EW, Black EC, Goo CE, Swearer AA, Yee NG, Rasmussen JP. Dendritic atoh1a+ cells serve as transient intermediates during zebrafish Merkel cell development and regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557830. [PMID: 37745341 PMCID: PMC10515958 DOI: 10.1101/2023.09.14.557830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Sensory cells often adopt specific morphologies that aid in the detection of external stimuli. Merkel cells encode gentle touch stimuli in vertebrate skin and adopt a reproducible shape characterized by spiky, actin-rich microvilli that emanate from the cell surface. The mechanism by which Merkel cells acquire this stereotyped morphology from basal keratinocyte progenitors is unknown. Here, we establish that dendritic Merkel cells (dMCs) express atonal homolog 1a (atoh1a), extend dynamic filopodial processes, and arise in transient waves during zebrafish skin development and regeneration. We find that dMCs share molecular similarities with both basal keratinocytes and Merkel cells, yet display mesenchymal-like behaviors, including local cell motility and proliferation within the epidermis. Furthermore, dMCs can directly adopt the mature, microvilliated Merkel cell morphology through substantial remodeling of the actin cytoskeleton. Loss of Ectodysplasin A signaling alters the morphology of dMCs and Merkel cells within specific skin regions. Our results show that dMCs represent an intermediate state in the Merkel cell maturation program and identify Ectodysplasin A signaling as a key regulator of Merkel cell morphology.
Collapse
Affiliation(s)
- Evan W. Craig
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Erik C. Black
- Department of Biology, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Camille E.A. Goo
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Avery Angell Swearer
- Department of Biology, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, USA
| | - Nathaniel G. Yee
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Jeffrey P. Rasmussen
- Department of Biology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
8
|
Wu S, Liang T, Jiang J, Zhu J, Chen T, Zhou C, Huang S, Yao Y, Guo H, Ye Z, Chen L, Chen W, Fan B, Qin J, Liu L, Wu S, Ma F, Zhan X, Liu C. Proteomic analysis to identification of hypoxia related markers in spinal tuberculosis: a study based on weighted gene co-expression network analysis and machine learning. BMC Med Genomics 2023; 16:142. [PMID: 37340462 DOI: 10.1186/s12920-023-01566-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
OBJECTIVE This article aims at exploring the role of hypoxia-related genes and immune cells in spinal tuberculosis and tuberculosis involving other organs. METHODS In this study, label-free quantitative proteomics analysis was performed on the intervertebral discs (fibrous cartilaginous tissues) obtained from five spinal tuberculosis (TB) patients. Key proteins associated with hypoxia were identified using molecular complex detection (MCODE), weighted gene co-expression network analysis(WGCNA), least absolute shrinkage and selection operator (LASSO), and support vector machine recursive feature Elimination (SVM-REF) methods, and their diagnostic and predictive values were assessed. Immune cell correlation analysis was then performed using the Single Sample Gene Set Enrichment Analysis (ssGSEA) method. In addition, a pharmaco-transcriptomic analysis was also performed to identify targets for treatment. RESULTS The three genes, namely proteasome 20 S subunit beta 9 (PSMB9), signal transducer and activator of transcription 1 (STAT1), and transporter 1 (TAP1), were identified in the present study. The expression of these genes was found to be particularly high in patients with spinal TB and other extrapulmonary TB, as well as in TB and multidrug-resistant TB (p-value < 0.05). They revealed high diagnostic and predictive values and were closely related to the expression of multiple immune cells (p-value < 0.05). It was inferred that the expression of PSMB9, STAT 1, and TAP1 could be regulated by different medicinal chemicals. CONCLUSION PSMB9, STAT1, and TAP1, might play a key role in the pathogenesis of TB, including spinal TB, and the protein product of the genes can be served as diagnostic markers and potential therapeutic target for TB.
Collapse
Affiliation(s)
- Shaofeng Wu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tuo Liang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jie Jiang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jichong Zhu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tianyou Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chenxing Zhou
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shengsheng Huang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yuanlin Yao
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hao Guo
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhen Ye
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liyi Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wuhua Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Binguang Fan
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiahui Qin
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lu Liu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Siling Wu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fengzhi Ma
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinli Zhan
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| | - Chong Liu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China.
| |
Collapse
|
9
|
Kennard AS, Sathe M, Labuz EC, Prinz CK, Theriot JA. Post-injury hydraulic fracturing drives fissure formation in the zebrafish basal epidermal cell layer. Curr Biol 2023:S0960-9822(23)00616-4. [PMID: 37290442 DOI: 10.1016/j.cub.2023.05.021] [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: 07/19/2022] [Revised: 03/05/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023]
Abstract
The skin epithelium acts as the barrier between an organism's internal and external environments. In zebrafish and other freshwater organisms, this barrier function requires withstanding a large osmotic gradient across the epidermis. Wounds breach this epithelium, causing a large disruption to the tissue microenvironment due to the mixing of isotonic interstitial fluid with the external hypotonic fresh water. Here, we show that, following acute injury, the larval zebrafish epidermis undergoes a dramatic fissuring process that resembles hydraulic fracturing, driven by the influx of external fluid. After the wound has sealed-preventing efflux of this external fluid-fissuring starts in the basal epidermal layer at the location nearest to the wound and then propagates at a constant rate through the tissue, spanning over 100 μm. During this process, the outermost superficial epidermal layer remains intact. Fissuring is completely inhibited when larvae are wounded in isotonic external media, suggesting that osmotic gradients are required for fissure formation. Additionally, fissuring partially depends on myosin II activity, as myosin II inhibition reduces the distance of fissure propagation away from the wound. During and after fissuring, the basal layer forms large macropinosomes (with cross-sectional areas ranging from 1 to 10 μm2). We conclude that excess external fluid entry through the wound and subsequent closure of the wound through actomyosin purse-string contraction in the superficial cell layer causes fluid pressure buildup in the extracellular space of the zebrafish epidermis. This excess fluid pressure causes tissue to fissure, and eventually the fluid is cleared through macropinocytosis.
Collapse
Affiliation(s)
- Andrew S Kennard
- Biophysics Program, Stanford University, Stanford, CA 94305, USA; Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Mugdha Sathe
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Ellen C Labuz
- Biophysics Program, Stanford University, Stanford, CA 94305, USA; Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Christopher K Prinz
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
10
|
Rosa JB, Nassman KY, Sagasti A. Sensory axons induce epithelial lipid microdomain remodeling and determine the distribution of junctions in the epidermis. Mol Biol Cell 2023; 34:ar5. [PMID: 36322392 PMCID: PMC9816649 DOI: 10.1091/mbc.e22-09-0396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022] Open
Abstract
Epithelial cell properties are determined by the polarized distribution of membrane lipids, the cytoskeleton, and adhesive junctions. Epithelia are often profusely innervated, but little work has addressed how neurites affect epithelial organization. We previously found that basal keratinocytes in the zebrafish epidermis enclose axons in ensheathment channels sealed by autotypic junctions. Here we characterized how axons remodel cell membranes, the cytoskeleton, and junctions in basal keratinocytes. At the apical surface of basal keratinocytes, axons organized lipid microdomains quantitatively enriched in reporters for PI(4,5)P2 and liquid-ordered (Lo) membranes. Lipid microdomains supported the formation of cadherin-enriched, F-actin protrusions, which wrapped around axons, likely initiating ensheathment. In the absence of axons, cadherin-enriched microdomains formed on basal cells but did not organize into contiguous domains. Instead, these isolated domains formed heterotypic junctions with periderm cells, a distinct epithelial cell type. Thus, axon endings dramatically remodel polarized epithelial components and regulate epidermal adhesion.
Collapse
Affiliation(s)
- Jeffrey B. Rosa
- Department of Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Khaled Y. Nassman
- Department of Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095
| | - Alvaro Sagasti
- Department of Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
11
|
Tobin DM. Modelling infectious disease to support human health. Dis Model Mech 2022; 15:276457. [PMID: 36037003 PMCID: PMC9459390 DOI: 10.1242/dmm.049824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
During the current COVID-19 pandemic, there has been renewed scientific and public focus on understanding the pathogenesis of infectious diseases and investigating vaccines and therapies to combat them. In addition to the tragic toll of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), we also recognize increased threats from antibiotic-resistant bacterial strains, the effects of climate change on the prevalence and spread of human pathogens, and the recalcitrance of other infectious diseases – including tuberculosis, malaria, human immunodeficiency virus (HIV) and fungal infections – that continue to cause millions of deaths annually. Large amounts of funding have rightly been redirected toward vaccine development and clinical trials for COVID-19, but we must continue to pursue fundamental and translational research on other pathogens and host immunity. Now more than ever, we need to support the next generation of researchers to develop and utilize models of infectious disease that serve as engines of discovery, innovation and therapy. Summary: This Editorial considers how knowledge from animal and other models of infectious disease can impact our understanding of human biology and potential therapies, focusing largely on zebrafish. It also highlights ways in which DMM is supporting these areas.
Collapse
Affiliation(s)
- David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
12
|
Perera DJ, Hassan AS, Liu SS, Elahi SM, Gadoury C, Weeratna RD, Gilbert R, Ndao M. A low dose adenovirus vectored vaccine expressing Schistosoma mansoni Cathepsin B protects from intestinal schistosomiasis in mice. EBioMedicine 2022; 80:104036. [PMID: 35500538 PMCID: PMC9065910 DOI: 10.1016/j.ebiom.2022.104036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/30/2022] [Accepted: 04/16/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Schistosomiasis is an underestimated neglected tropical disease which affects over 236.6 million people worldwide. According to the CDC, the impact of this disease is second to only malaria as the most devastating parasitic infection. Affected individuals manifest chronic pathology due to egg granuloma formation, destroying the liver over time. The only FDA approved drug, praziquantel, does not protect individuals from reinfection, highlighting the need for a prophylactic vaccine. Schistosoma mansoni Cathepsin B (SmCB) is a parasitic gut peptidase necessary for helminth growth and maturation and confers protection as a vaccine target for intestinal schistosomiasis. METHODS An SmCB expressing human adenovirus serotype 5 (AdSmCB) was constructed and delivered intramuscularly to female C57BL/6 mice in a heterologous prime and boost vaccine with recombinant protein. Vaccine induced immunity was described and subsequent protection from parasite infection was assessed by analysing parasite burden and liver pathology. FINDINGS Substantially higher humoral and cell-mediated immune responses, consisting of IgG2c, Th1 effectors, and polyfunctional CD4+ T cells, were induced by the heterologous administration of AdSmCB when compared to the other regimens. Though immune responses favoured Th1 immunity, Th2 responses provided by SmCB protein boosts were maintained. This mixed Th1/Th2 immune response resulted in significant protection from S. mansoni infection comparable to other vaccine formulations which are in clinical trials. Schistosomiasis associated liver pathology was also prevented in a murine model. INTERPRETATION Our study provides missing preclinical data supporting the use of adenoviral vectoring in vaccines for S. mansoni infection. Our vaccination method significantly reduces parasite burden and its associated liver pathology - both of which are critical considerations for this helminth vaccine. FUNDING This work was supported by the Canadian Institutes of Health Research, R. Howard Webster Foundation, and the Foundation of the McGill University Health Centre.
Collapse
Affiliation(s)
- Dilhan J Perera
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Room: EM3.3244, 1001 Decarie Blvd, Montréal, Québec H4A 3J1, Canada
| | - Adam S Hassan
- Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Room: EM3.3244, 1001 Decarie Blvd, Montréal, Québec H4A 3J1, Canada; Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Sunny S Liu
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | | | | | | | - Rénald Gilbert
- National Research Council Canada, Montréal, Québec, Canada
| | - Momar Ndao
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, Québec, Canada; Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Room: EM3.3244, 1001 Decarie Blvd, Montréal, Québec H4A 3J1, Canada; Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada; National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada.
| |
Collapse
|
13
|
Cronan MR. In the Thick of It: Formation of the Tuberculous Granuloma and Its Effects on Host and Therapeutic Responses. Front Immunol 2022; 13:820134. [PMID: 35320930 PMCID: PMC8934850 DOI: 10.3389/fimmu.2022.820134] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The defining pathology of tuberculosis is the granuloma, an organized structure derived from host immune cells that surrounds infecting Mycobacterium tuberculosis. As the location of much of the bacteria in the infected host, the granuloma is a central point of interaction between the host and the infecting bacterium. This review describes the signals and cellular reprogramming that drive granuloma formation. Further, as a central point of host-bacterial interactions, the granuloma shapes disease outcome by altering host immune responses and bacterial susceptibility to antibiotic treatment, as discussed herein. This new understanding of granuloma biology and the signaling behind it highlights the potential for host-directed therapies targeting the granuloma to enhance antibiotic access and tuberculosis-specific immune responses.
Collapse
Affiliation(s)
- Mark R. Cronan
-
In Vivo Cell Biology of Infection Group, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
14
|
Abstract
Pulmonary granulomas are widely considered the epicenters of the immune response to Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). Recent animal studies have revealed factors that either promote or restrict TB immunity within granulomas. These models, however, typically ignore the impact of preexisting immunity on cellular organization and function, an important consideration because most TB probably occurs through reinfection of previously exposed individuals. Human postmortem research from the pre-antibiotic era showed that infections in Mtb-naïve individuals (primary TB) versus those with prior Mtb exposure (postprimary TB) have distinct pathologic features. We review recent animal findings in TB granuloma biology, which largely reflect primary TB. We also discuss our current understanding of postprimary TB lesions, about which much less is known. Many knowledge gaps remain, particularly regarding how preexisting immunity shapes granuloma structure and local immune responses at Mtb infection sites. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sara B. Cohen
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Benjamin H. Gern
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Kevin B. Urdahl
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
15
|
Perveen S, Sharma R. Screening approaches and therapeutic targets: The two driving wheels of tuberculosis drug discovery. Biochem Pharmacol 2022; 197:114906. [PMID: 34990594 DOI: 10.1016/j.bcp.2021.114906] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) is an infectious disease, infecting a quarter of world's population. Drug resistant TB further exacerbates the grim scenario of the drying TB drug discovery pipeline. The limited arsenal to fight TB presses the need for thorough efforts for identifying promising hits to combat the disease. The review highlights the efforts in the field of tuberculosis drug discovery, with an emphasis on massive drug screening campaigns for identifying novel hits against Mtb in both industry and academia. As an intracellular pathogen, mycobacteria reside in a complicated intracellular environment with multiple factors at play. Here, we outline various strategies employed in an effort to mimic the intracellular milieu for bringing the screening models closer to the actual settings. The review also focuses on the novel targets and pathways that could aid in target-based drug discovery in TB. The recent high throughput screening efforts resulting in the identification of potent hits against Mtb has been summarized in this article. There is a pressing need for effective screening strategies and approaches employing innovative tools and recent technologies; including nanotechnology, gene-editing tools such as CRISPR-cas system, host-directed bacterial killing and high content screening to augment the TB drug discovery pipeline with safer and shorter drug regimens.
Collapse
Affiliation(s)
- Summaya Perveen
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rashmi Sharma
- Infectious Diseases Division, CSIR- Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
16
|
Mouse innate-like B-1 lymphocytes promote inhaled particle-induced in vitro granuloma formation and inflammation in conjunction with macrophages. Arch Toxicol 2021; 96:585-599. [PMID: 34935064 PMCID: PMC8837577 DOI: 10.1007/s00204-021-03200-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
The current paradigm for explaining lung granulomatous diseases induced by inhaled particles is mainly based on macrophages. This mechanism is now challenging because B lymphocytes also infiltrate injured tissue, and the deficiency in B lymphocytes is associated with limited lung granulomas in silica-treated mice. Here, we investigated how B lymphocytes respond to micro- and nanoparticles by combining in vivo and in vitro mouse models. We first demonstrated that innate-like B-1 lymphocytes (not conventional B-2 lymphocytes or plasma cells) specifically accumulated during granuloma formation in mice instilled with crystalline silica (DQ12, 2.5 mg/mouse) and carbon nanotubes (CNT Mitsui, 0.2 mg/mouse). In comparison to macrophages, peritoneal B-1 lymphocytes purified from naïve mice were resistant to the pyroptotic activity of reactive particles (up to 1 mg/mL) but clustered to establish in vitro cell/particle aggregates. Mouse B-1 lymphocytes (not B-2 lymphocytes) in coculture with macrophages and CNT (0.1 µg/mL) organized three-dimensional spheroid structures in Matrigel and stimulated the release of TIMP-1. Furthermore, purified B-1 lymphocytes are sensitive to nanosilica toxicity through radical generation in culture. Nanosilica-exposed B-1 lymphocytes released proinflammatory cytokines and alarmins. In conclusion, our data indicate that in addition to macrophages, B-1 lymphocytes participate in micrometric particle-induced granuloma formation and display inflammatory functions in response to nanoparticles.
Collapse
|
17
|
Blanc L, Ferraro GB, Tuck M, Prideaux B, Dartois V, Jain RK, Desbenoit N. Kendrick Mass Defect Variation to Decipher Isotopic Labeling in Brain Metastases Studied by Mass Spectrometry Imaging. Anal Chem 2021; 93:16314-16319. [PMID: 34860501 PMCID: PMC9841243 DOI: 10.1021/acs.analchem.1c03916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Besides many other applications, isotopic labeling is commonly used to decipher the metabolism of living biological systems. By giving a stable isotopically labeled compound as a substrate, the biological system will use this labeled nutrient as it would with a regular substrate and incorporate stable heavy atoms into new metabolites. Utilizing mass spectrometry, by comparing heavy atom enriched isotopic profiles and naturally occurring ones, it is possible to identify these metabolites and deduce valuable information about metabolism and biochemical pathways. The coupling of this approach with mass spectrometry imaging (MSI) allows one then to obtain 2D maps of metabolisms used by living specimens. As metabolic networks are convoluted, a global overview of the isotopically labeled data set to detect unexpected metabolites is crucial. Unfortunately, due to the complexity of MSI spectra, such untargeted processing approaches are difficult to decipher. In this technical note, we demonstrate the potential of a variation around the Kendrick analysis concept to detect the incorporation of stable heavy atoms into metabolites. The Kendrick analysis uses as a base unit the difference between the mass of the most abundant isotope and the mass of the corresponding stable isotopic tracer (namely, 12C and 13C). The resulting Kendrick plot offers an alternative method to process the MSI data set with a new perspective allowing for the rapid detection of the 13C-enriched metabolites and separating unrelated compounds. This processing method of MS data could therefore be a useful tool to decipher isotopic labeling and study metabolic networks, especially as it does not require advanced computational capabilities.
Collapse
Affiliation(s)
- Landry Blanc
- Univ. Bordeaux, CNRS, CBMN, UMR 5248, F-33600 Pessac, France
| | - Gino B. Ferraro
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Michael Tuck
- Univ. Bordeaux, CNRS, CBMN, UMR 5248, F-33600 Pessac, France
| | - Brendan Prideaux
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Department of Medical Sciences, Hackensack Meridian Health, Nutley, New Jersey 07601, United States
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | | |
Collapse
|
18
|
Ge G, Jiang H, Xiong J, Zhang W, Shi Y, Tao C, Wang H. Progress of the Art of Macrophage Polarization and Different Subtypes in Mycobacterial Infection. Front Immunol 2021; 12:752657. [PMID: 34899703 PMCID: PMC8660122 DOI: 10.3389/fimmu.2021.752657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Mycobacteriosis, mostly resulting from Mycobacterium tuberculosis (MTb), nontuberculous mycobacteria (NTM), and Mycobacterium leprae (M. leprae), is the long-standing granulomatous disease that ravages several organs including skin, lung, and peripheral nerves, and it has a spectrum of clinical-pathologic features based on the interaction of bacilli and host immune response. Histiocytes in infectious granulomas mainly consist of infected and uninfected macrophages (Mφs), multinucleated giant cells (MGCs), epithelioid cells (ECs), and foam cells (FCs), which are commonly discovered in lesions in patients with mycobacteriosis. Granuloma Mφ polarization or reprogramming is the crucial appearance of the host immune response to pathogen aggression, which gets a command of endocellular microbe persistence. Herein, we recapitulate the current gaps and challenges during Mφ polarization and the different subpopulations of mycobacteriosis.
Collapse
Affiliation(s)
- Gai Ge
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Haiqin Jiang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jingshu Xiong
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wenyue Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ying Shi
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Chenyue Tao
- Imperial College London, London, United Kingdom
| | - Hongsheng Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,National Center for Sexually Transmitted Disease and Leprosy Control, China Centers for Disease Control and Prevention, Nanjing, China.,Centre for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
19
|
Varela M, Meijer AH. A fresh look at mycobacterial pathogenicity with the zebrafish host model. Mol Microbiol 2021; 117:661-669. [PMID: 34714579 PMCID: PMC9297993 DOI: 10.1111/mmi.14838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022]
Abstract
The zebrafish has earned its place among animal models to study tuberculosis and other infections caused by pathogenic mycobacteria. This model host is especially useful to study the role of granulomas, the inflammatory lesions characteristic of mycobacterial disease. The optically transparent zebrafish larvae provide a window on the initial stages of granuloma development in the context of innate immunity. Application of fluorescent dyes and transgenic markers enabled real-time visualization of how innate immune mechanisms, such as autophagy and inflammasomes, are activated in infected macrophages and how propagating calcium signals drive communication between macrophages during granuloma formation. A combination of imaging, genetic, and chemical approaches has revealed that the interplay between macrophages and mycobacteria is the main driver of tissue dissemination and granuloma development, while neutrophils have a protective function in early granulomas. Different chemokine signaling axes, conserved between humans and zebrafish, have been shown to recruit macrophages permissive to mycobacterial growth, control their microbicidal capacity, drive their spreading and aggregation, and mediate granuloma vascularization. Finally, zebrafish larvae are now exploited to explore cell death processes, emerging as crucial factors in granuloma expansion. In this review, we discuss recent advances in the understanding of mycobacterial pathogenesis contributed by zebrafish models.
Collapse
Affiliation(s)
- Monica Varela
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Sarcoidosis remains a mysterious disease that presents many challenges both in pathogenetic understanding and in the management of patients. This review presents experimental models for sarcoidosis developed since 2016 and discusses their strengths and weaknesses and how they have contributed to the understanding and therapeutic approaches in this disease. In addition, future directions are proposed to overcome the limitations of current models. RECENT FINDINGS New cellular models using infectious antigen as trigger, and transgenic models in mice have recently been developed to study signaling pathways potentially implicated in the pathogenesis of sarcoidosis. SUMMARY No model fully reproduces sarcoidosis, but most of them generate data supporting key concepts and some open up therapeutic perspectives, like mTOR inhibition or IL-1β blocking. However, there are still many limitations to these models, largely related to the complexity of sarcoidosis, which might be overcome with new technologies, such as mathematical modeling.
Collapse
|
21
|
Cronan MR, Hughes EJ, Brewer WJ, Viswanathan G, Hunt EG, Singh B, Mehra S, Oehlers SH, Gregory SG, Kaushal D, Tobin DM. A non-canonical type 2 immune response coordinates tuberculous granuloma formation and epithelialization. Cell 2021; 184:1757-1774.e14. [PMID: 33761328 PMCID: PMC8055144 DOI: 10.1016/j.cell.2021.02.046] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/19/2022]
Abstract
The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified/genetics
- Animals, Genetically Modified/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Disease Models, Animal
- Epithelioid Cells/cytology
- Epithelioid Cells/immunology
- Epithelioid Cells/metabolism
- Granuloma/immunology
- Granuloma/metabolism
- Granuloma/pathology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Immunity/physiology
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Mycobacterium Infections, Nontuberculous/immunology
- Mycobacterium Infections, Nontuberculous/pathology
- Mycobacterium marinum/isolation & purification
- Mycobacterium marinum/physiology
- Necrosis
- Receptors, Interleukin-4/antagonists & inhibitors
- Receptors, Interleukin-4/genetics
- Receptors, Interleukin-4/metabolism
- STAT6 Transcription Factor/antagonists & inhibitors
- STAT6 Transcription Factor/genetics
- STAT6 Transcription Factor/metabolism
- Signal Transduction
- Zebrafish/growth & development
- Zebrafish/metabolism
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emily G Hunt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Ordonez AA, Tucker EW, Anderson CJ, Carter CL, Ganatra S, Kaushal D, Kramnik I, Lin PL, Madigan CA, Mendez S, Rao J, Savic RM, Tobin DM, Walzl G, Wilkinson RJ, Lacourciere KA, Via LE, Jain SK. Visualizing the dynamics of tuberculosis pathology using molecular imaging. J Clin Invest 2021; 131:145107. [PMID: 33645551 PMCID: PMC7919721 DOI: 10.1172/jci145107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Nearly 140 years after Robert Koch discovered Mycobacterium tuberculosis, tuberculosis (TB) remains a global threat and a deadly human pathogen. M. tuberculosis is notable for complex host-pathogen interactions that lead to poorly understood disease states ranging from latent infection to active disease. Additionally, multiple pathologies with a distinct local milieu (bacterial burden, antibiotic exposure, and host response) can coexist simultaneously within the same subject and change independently over time. Current tools cannot optimally measure these distinct pathologies or the spatiotemporal changes. Next-generation molecular imaging affords unparalleled opportunities to visualize infection by providing holistic, 3D spatial characterization and noninvasive, temporal monitoring within the same subject. This rapidly evolving technology could powerfully augment TB research by advancing fundamental knowledge and accelerating the development of novel diagnostics, biomarkers, and therapeutics.
Collapse
Affiliation(s)
- Alvaro A. Ordonez
- Center for Infection and Inflammation Imaging Research
- Center for Tuberculosis Research
- Department of Pediatrics, and
| | - Elizabeth W. Tucker
- Center for Infection and Inflammation Imaging Research
- Center for Tuberculosis Research
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Claire L. Carter
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, New Jersey, USA
| | - Shashank Ganatra
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusets, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Philana L. Lin
- Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Cressida A. Madigan
- Department of Biological Sciences, UCSD, San Diego, La Jolla, California, USA
| | - Susana Mendez
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Rockville, Maryland, USA
| | - Jianghong Rao
- Molecular Imaging Program at Stanford, Department of Radiology and Chemistry, Stanford University, Stanford, California, USA
| | - Rada M. Savic
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy and Medicine, UCSF, San Francisco, California, USA
| | - David M. Tobin
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Gerhard Walzl
- SAMRC Centre for Tuberculosis Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Robert J. Wilkinson
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
- Wellcome Centre for Infectious Diseases Research in Africa and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- The Francis Crick Institute, London, United Kingdom
| | - Karen A. Lacourciere
- National Institute of Allergy and Infectious Diseases (NIAID), NIH, Rockville, Maryland, USA
| | - Laura E. Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, and Tuberculosis Imaging Program, Division of Intramural Research, NIAID, NIH, Bethesda, Maryland, USA
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research
- Center for Tuberculosis Research
- Department of Pediatrics, and
| |
Collapse
|
23
|
Asai M, Sheehan G, Li Y, Robertson BD, Kavanagh K, Langford PR, Newton SM. Innate Immune Responses of Galleria mellonella to Mycobacterium bovis BCG Challenge Identified Using Proteomic and Molecular Approaches. Front Cell Infect Microbiol 2021; 11:619981. [PMID: 33634038 PMCID: PMC7900627 DOI: 10.3389/fcimb.2021.619981] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/04/2021] [Indexed: 01/10/2023] Open
Abstract
The larvae of the insect Galleria mellonella, have recently been established as a non-mammalian infection model for the Mycobacterium tuberculosis complex (MTBC). To gain further insight into the potential of this model, we applied proteomic (label-free quantification) and transcriptomic (gene expression) approaches to characterise the innate immune response of G. mellonella to infection with Mycobacterium bovis BCG lux over a 168 h time course. Proteomic analysis of the haemolymph from infected larvae revealed distinct changes in the proteome at all time points (4, 48, 168 h). Reverse transcriptase quantitative PCR confirmed induction of five genes (gloverin, cecropin, IMPI, hemolin, and Hdd11), which encoded proteins found to be differentially abundant from the proteomic analysis. However, the trend between gene expression and protein abundance were largely inconsistent (20%). Overall, the data are in agreement with previous phenotypic observations such as haemocyte internalization of mycobacterial bacilli (hemolin/β-actin), formation of granuloma-like structures (Hdd11), and melanization (phenoloxidase activating enzyme 3 and serpins). Furthermore, similarities in immune expression in G. mellonella, mouse, zebrafish and in vitro cell-line models of tuberculosis infection were also identified for the mechanism of phagocytosis (β-actin). Cecropins (antimicrobial peptides), which share the same α-helical motif as a highly potent peptide expressed in humans (h-CAP-18), were induced in G. mellonella in response to infection, giving insight into a potential starting point for novel antimycobacterial agents. We believe that these novel insights into the innate immune response further contribute to the validation of this cost-effective and ethically acceptable insect model to study members of the MTBC.
Collapse
Affiliation(s)
- Masanori Asai
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Gerard Sheehan
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, Maynooth, Ireland.,Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Yanwen Li
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Brian D Robertson
- MRC Centre for Molecular Bacteriology and Infection, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Kevin Kavanagh
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, Maynooth, Ireland
| | - Paul R Langford
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Sandra M Newton
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
24
|
Jagatia H, Tsolaki AG. The Role of Complement System and the Immune Response to Tuberculosis Infection. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:84. [PMID: 33498555 PMCID: PMC7909539 DOI: 10.3390/medicina57020084] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 11/30/2022]
Abstract
The complement system orchestrates a multi-faceted immune response to the invading pathogen, Mycobacterium tuberculosis. Macrophages engulf the mycobacterial bacilli through bacterial cell surface proteins or secrete proteins, which activate the complement pathway. The classical pathway is activated by C1q, which binds to antibody antigen complexes. While the alternative pathway is constitutively active and regulated by properdin, the direct interaction of properdin is capable of complement activation. The lectin-binding pathway is activated in response to bacterial cell surface carbohydrates such as mannose, fucose, and N-acetyl-d-glucosamine. All three pathways contribute to mounting an immune response for the clearance of mycobacteria. However, the bacilli can reside, persist, and evade clearance by the immune system once inside the macrophages using a number of mechanisms. The immune system can compartmentalise the infection into a granulomatous structure, which contains heterogenous sub-populations of M. tuberculosis. The granuloma consists of many types of immune cells, which aim to clear and contain the infection whilst sacrificing the affected host tissue. The full extent of the involvement of the complement system during infection with M. tuberculosis is not fully understood. Therefore, we reviewed the available literature on M. tuberculosis and other mycobacterial literature to understand the contribution of the complement system during infection.
Collapse
Affiliation(s)
- Heena Jagatia
- Department for Respiratory Sciences, University of Leicester, Leicester LE1 9HN, UK
| | - Anthony G. Tsolaki
- Department of Life Sciences, College of Health and Life Sciences, Brunel University of London, Uxbridge UB8 3PN, UK;
| |
Collapse
|
25
|
Levic DS, Ryan S, Marjoram L, Honeycutt J, Bagwell J, Bagnat M. Distinct roles for luminal acidification in apical protein sorting and trafficking in zebrafish. J Cell Biol 2020; 219:133852. [PMID: 32328632 PMCID: PMC7147097 DOI: 10.1083/jcb.201908225] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial cell physiology critically depends on the asymmetric distribution of channels and transporters. However, the mechanisms targeting membrane proteins to the apical surface are still poorly understood. Here, we performed a visual forward genetic screen in the zebrafish intestine and identified mutants with defective apical targeting of membrane proteins. One of these mutants, affecting the vacuolar H+-ATPase gene atp6ap1b, revealed specific requirements for luminal acidification in apical, but not basolateral, membrane protein sorting and transport. Using a low temperature block assay combined with genetic and pharmacologic perturbation of luminal pH, we monitored transport of newly synthesized membrane proteins from the TGN to apical membrane in live zebrafish. We show that vacuolar H+-ATPase activity regulates sorting of O-glycosylated proteins at the TGN, as well as Rab8-dependent post-Golgi trafficking of different classes of apical membrane proteins. Thus, luminal acidification plays distinct and specific roles in apical membrane biogenesis.
Collapse
Affiliation(s)
| | - Sean Ryan
- Department of Cell Biology, Duke University, Durham, NC
| | | | | | | | - Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC
| |
Collapse
|
26
|
Perera DJ, Hassan AS, Jia Y, Ricciardi A, McCluskie MJ, Weeratna RD, Ndao M. Adjuvanted Schistosoma mansoni-Cathepsin B With Sulfated Lactosyl Archaeol Archaeosomes or AddaVax™ Provides Protection in a Pre-Clinical Schistosomiasis Model. Front Immunol 2020; 11:605288. [PMID: 33304354 PMCID: PMC7701121 DOI: 10.3389/fimmu.2020.605288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/20/2020] [Indexed: 12/15/2022] Open
Abstract
Schistosomiasis threatens 800 million people worldwide. Chronic pathology manifests as hepatosplenomegaly, and intestinal schistosomiasis caused by Schistosoma mansoni can lead to liver fibrosis, cirrhosis, and blood in the stool. To assist the only FDA-approved drug, praziquantel, in parasite elimination, the development of a vaccine would be of high value. S. mansoni Cathepsin B (SmCB) is a well-documented vaccine target for intestinal schistosomiasis. Herein, we test the increased efficacy and immunogenicity of SmCB when combined with sulfated lactosyl archaeol (SLA) archaeosomes or AddaVax™ (a squalene based oil-in-water emulsion). Both vaccine formulations resulted in robust humoral and cell mediated immune responses. Impressively, both formulations were able to reduce parasite burden greater than 40% (WHO standard), with AddaVax™ reaching 86.8%. Additionally, SmCB with both adjuvants were able to reduce granuloma size and the amount of larval parasite hatched from feces, which would reduce transmission. Our data support SmCB as a target for S. mansoni vaccination; especially when used in an adjuvanted formulation.
Collapse
Affiliation(s)
- Dilhan J Perera
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.,Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Adam S Hassan
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Yimei Jia
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Alessandra Ricciardi
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Michael J McCluskie
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Risini D Weeratna
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Momar Ndao
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.,Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.,National Reference Center for Parasitology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
27
|
Do chance encounters between heterogeneous cells shape the outcome of tuberculosis infections? Curr Opin Microbiol 2020; 59:72-78. [PMID: 33049596 DOI: 10.1016/j.mib.2020.08.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
Abstract
The sum of all of the interactions between single bacteria and host cells determines if an infection is cleared, controlled, or progresses at the whole host-organism level. These individual interactions have independent trajectories defined by diverse and dynamic host-cell and bacterial responses. Focusing on Mycobacterium tuberculosis infection, we discuss how advances in single-cell technologies allow investigation of heterogeneity in host-pathogen interactions and how different layers of heterogeneity in the host affect disease outcome. At late stages of infection, many single interactions co-exist and different outcomes depend on inter-granuloma and intra-granuloma heterogeneity. However, during bottleneck events involving small numbers of bacteria, random events, such as chance interactions with more or less permissive host cells, play a decisive role and may explain why some exposed individuals never develop the disease.
Collapse
|
28
|
Berry SB, Gower MS, Su X, Seshadri C, Theberge AB. A Modular Microscale Granuloma Model for Immune-Microenvironment Signaling Studies in vitro. Front Bioeng Biotechnol 2020; 8:931. [PMID: 32974300 PMCID: PMC7461927 DOI: 10.3389/fbioe.2020.00931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB) is one of the most potent infectious diseases in the world, causing more deaths than any other single infectious agent. TB infection is caused by inhalation of Mycobacterium tuberculosis (Mtb) and subsequent phagocytosis and migration into the lung tissue by innate immune cells (e.g., alveolar macrophages, neutrophils, and dendritic cells), resulting in the formation of a fused mass of immune cells known as the granuloma. Considered the pathological hallmark of TB, the granuloma is a complex microenvironment that is crucial for pathogen containment as well as pathogen survival. Disruption of the delicate granuloma microenvironment via numerous stimuli, such as variations in cytokine secretions, nutrient availability, and the makeup of immune cell population, can lead to an active infection. Herein, we present a novel in vitro model to examine the soluble factor signaling between a mycobacterial infection and its surrounding environment. Adapting a newly developed suspended microfluidic platform, known as Stacks, we established a modular microscale infection model containing human immune cells and a model mycobacterial strain that can easily integrate with different microenvironmental cues through simple spatial and temporal "stacking" of each module of the platform. We validate the establishment of suspended microscale (4 μL) infection cultures that secrete increased levels of proinflammatory factors IL-6, VEGF, and TNFα upon infection and form 3D aggregates (granuloma model) encapsulating the mycobacteria. As a proof of concept to demonstrate the capability of our platform to examine soluble factor signaling, we cocultured an in vitro angiogenesis model with the granuloma model and quantified morphology changes in endothelial structures as a result of culture conditions (P < 0.05 when comparing infected vs. uninfected coculture systems). We envision our modular in vitro granuloma model can be further expanded and adapted for studies focusing on the complex interplay between granulomatous structures and their surrounding microenvironment, as well as a complementary tool to augment in vivo signaling and mechanistic studies.
Collapse
Affiliation(s)
- Samuel B. Berry
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Maia S. Gower
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Xiaojing Su
- Department of Chemistry, University of Washington, Seattle, WA, United States
| | - Chetan Seshadri
- Department of Medicine, University of Washington, Seattle, WA, United States
| | - Ashleigh B. Theberge
- Department of Chemistry, University of Washington, Seattle, WA, United States
- Department of Urology, University of Washington, Seattle, WA, United States
| |
Collapse
|
29
|
Parish T. In vitro drug discovery models for Mycobacterium tuberculosis relevant for host infection. Expert Opin Drug Discov 2020; 15:349-358. [PMID: 31899974 DOI: 10.1080/17460441.2020.1707801] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Tuberculosis is the leading cause of death from infectious disease. Current drug therapy requires a combination of antibiotics taken over >6 months. An urgent need for new agents that can shorten therapy is required. In order to develop new drugs, simple in vitro assays are required that can identify efficacious compounds rapidly and predict in vivo activity in the human.Areas covered: This review focusses on the most relevant in vitro assays that can be utilized in a drug discovery program and which mimic different aspects of infection or disease. The focus is largely on assays used to test >1000s of compounds reliably and robustly. However, some assays used for 10s to 100s of compounds are included where the utility outweighs the low capacity. Literature searches for high throughput screening, models and in vitro assays were undertaken.Expert opinion: Drug discovery and development in tuberculosis is extremely challenging due to the requirement for predicting drug efficacy in a disease with complex pathology in which bacteria exist in heterogeneous states in inaccesible locations. A combination of assays can be used to determine profiles against replicating, non-replicating, intracellular and tolerant bacteria.
Collapse
Affiliation(s)
- Tanya Parish
- Seattle Children's Research Institute, Center for Global Infectious Disease Research, Seattle, WA, USA
| |
Collapse
|
30
|
Vaccination against the digestive enzyme Cathepsin B using a YS1646 Salmonella enterica Typhimurium vector provides almost complete protection against Schistosoma mansoni challenge in a mouse model. PLoS Negl Trop Dis 2019; 13:e0007490. [PMID: 31790394 PMCID: PMC6907844 DOI: 10.1371/journal.pntd.0007490] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 12/12/2019] [Accepted: 10/01/2019] [Indexed: 12/03/2022] Open
Abstract
Schistosoma mansoni threatens hundreds of millions of people in >50 countries. Schistosomulae migrate through the lung and adult worms reside in blood vessels adjacent to the intestinal mucosa. Current candidate vaccines aren’t designed to elicit a mucosal response. We have repurposed an attenuated Salmonella enterica Typhimurium strain (YS1646) to produce such a vaccine targeting Cathepsin B (CatB), a digestive enzyme important for parasite survival. Promoter-Type 3 secretory signal pairs were screened for protein expression in vitro and transfected into YS1646 to generate candidate vaccine strains. Two strains were selected for in vivo evaluation (nirB_SspH1 and SspH1_SspH1). Female C57BL/6 mice were immunized twice, 3 weeks apart, using six strategies: i) saline gavage (control), ii) the ‘empty’ YS1646 vector orally (PO) followed by intramuscular (IM) recombinant CatB (20μg IM rCatB), iii) two doses of IM rCatB, iv) two PO doses of YS1646-CatB, v) IM rCatB then PO YS1646-CatB and vi) PO YS1646-CatB then IM rCatB. Serum IgG responses to CatB were monitored by ELISA. Three weeks after the second dose, mice were challenged with 150 cercariae and sacrificed 7 weeks later to assess adult worm and egg burden (liver and intestine), granuloma size and egg morphology. CatB-specific IgG antibodies were low/absent in the control and PO only groups but rose substantially in other groups (5898-6766ng/mL). The highest response was in animals that received nirB_SspH1 YS1646 PO then IM rCatB. In this group, reductions in worm and intestine/liver egg burden (vs. control) were 93.1% and 79.5%/90.3% respectively (all P < .0001). Granuloma size was reduced in all vaccinated groups (range 32.9–52.8 x103μm2) and most significantly in the nirB_SspH1 + CatB IM group (34.7±3.4 x103μm2vs. 62.2±6.1 x103μm2: vs. control P < .01). Many eggs in the vaccinated animals had abnormal morphology. Targeting CatB using a multi-modality approach can provide almost complete protection against S. mansoni challenge. Schistosomiasis is a parasitic disease that affects over 250 million people worldwide and over 800 million are at risk of infection. Of the three main species, Schistosoma mansoni is the most widely distributed and is endemic in the Caribbean, South America, and Africa. It causes a chronic disease with severe negative effects on quality of life. Mass drug administration of praziquantel is the only available course of action due to a current lack of vaccines. However, praziquantel does not protect from reinfection. Therefore, a vaccine would be beneficial as a long-term solution to reduce morbidity and transmission of the disease. Our group has repurposed the attenuated YS1646 strain of Salmonella Typhimurium as an oral vaccine vector for the digestive enzyme Cathepsin B of S. mansoni. Oral vaccination followed by an intramuscular dose of recombinant Cathepsin B lead to significant reductions in parasite burden in mice. These animals had the highest titers in serum IgG and intestinal IgA antibodies. This multimodal vaccination approach also elicited both Th1 and Th2 cytokines as seen by the increases in IFNγ and IL-5. Finally, vaccinated mice had reductions in granuloma size along with a higher proportion of morphologically-abnormal eggs. This work demonstrates that a YS1646-based, multimodality, prime-boost immunization schedule can provide nearly complete protection against S. mansoni in a well-established murine model.
Collapse
|
31
|
Cheng T, Kam JY, Johansen MD, Oehlers SH. High content analysis of granuloma histology and neutrophilic inflammation in adult zebrafish infected with Mycobacterium marinum. Micron 2019; 129:102782. [PMID: 31775097 DOI: 10.1016/j.micron.2019.102782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022]
Abstract
Infection of zebrafish with natural pathogen Mycobacterium marinum is a useful surrogate for studying the human granulomatous inflammatory response to infection by Mycobacterium tuberculosis. The adaptive immune system of the adult stage zebrafish offers an advance on the commonly used embryo infection model as adult zebrafish form granulomas with striking similarities to human-M. tuberculosis granulomas. Here, we present workflows to perform high content analyses of granulomas in adult zebrafish infected with M. marinum by cryosectioning to take advantage of strong endogenous transgenic fluorescence adapted from common zebrafish embryo infection tools. Specific guides to classifying granuloma necrosis and organisation, quantifying bacterial burden and leukocyte infiltration of granulomas, visualizing foam cell formation, analysing extracellular matrix remodelling and granuloma fibrosis are also provided. We use these methods to characterize neutrophil recruitment to M. marinum granulomas across time and find an inverse relation to granuloma necrosis suggesting granuloma necrosis is not a marker of immunopathology in the natural infection system of the adult zebrafish-M. marinum pairing. The methods can be easily translated to studying the zebrafish adaptive immune response to other chronic and granuloma-forming pathogens.
Collapse
Affiliation(s)
- Tina Cheng
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Julia Y Kam
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Matt D Johansen
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, 2050, Australia; The University of Sydney, Discipline of Infectious Diseases & Immunology and Marie Bashir Institute, Camperdown, NSW, 2050, Australia.
| |
Collapse
|
32
|
Cronan MR, Tobin DM. Endogenous Tagging at the cdh1 Locus for Live Visualization of E-Cadherin Dynamics. Zebrafish 2019; 16:324-325. [PMID: 30969163 DOI: 10.1089/zeb.2019.1746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mark R Cronan
- 1 Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina
| | - David M Tobin
- 1 Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina.,2 Department of Immunology, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
33
|
Jiang N, Rasmussen JP, Clanton JA, Rosenberg MF, Luedke KP, Cronan MR, Parker ED, Kim HJ, Vaughan JC, Sagasti A, Parrish JZ. A conserved morphogenetic mechanism for epidermal ensheathment of nociceptive sensory neurites. eLife 2019; 8:42455. [PMID: 30855229 PMCID: PMC6450671 DOI: 10.7554/elife.42455] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
Interactions between epithelial cells and neurons influence a range of sensory modalities including taste, touch, and smell. Vertebrate and invertebrate epidermal cells ensheath peripheral arbors of somatosensory neurons, including nociceptors, yet the developmental origins and functional roles of this ensheathment are largely unknown. Here, we describe an evolutionarily conserved morphogenetic mechanism for epidermal ensheathment of somatosensory neurites. We found that somatosensory neurons in Drosophila and zebrafish induce formation of epidermal sheaths, which wrap neurites of different types of neurons to different extents. Neurites induce formation of plasma membrane phosphatidylinositol 4,5-bisphosphate microdomains at nascent sheaths, followed by a filamentous actin network, and recruitment of junctional proteins that likely form autotypic junctions to seal sheaths. Finally, blocking epidermal sheath formation destabilized dendrite branches and reduced nociceptive sensitivity in Drosophila. Epidermal somatosensory neurite ensheathment is thus a deeply conserved cellular process that contributes to the morphogenesis and function of nociceptive sensory neurons. Humans and other animals perceive and interact with the outside world through their sensory nervous system. Nerve cells, acting as the body’s ‘telegraph wires’, convey signals from sensory organs – like the eyes – to the brain, which then processes this information and tells the body how to respond. There are different kinds of sensory nerve cells that carry different types of information, but they all associate closely with the tissues and organs they connect to the brain. Human skin contains sensory nerve cells, which underpin our senses of touch and pain. There is a highly specialized, complex connection between some of these nerve cells and cells in the skin: the skin cells wrap tightly around the nerve cells’ free ends, forming sheath-like structures. This ‘ensheathment’ process happens in a wide range of animals, including those with a backbone, like fish and humans, and those without, like insects. Ensheathment is thought to be important for the skin’s nerve cells to work properly. Yet it remains unclear how or when these connections first appear. Jiang et al. therefore wanted to determine the developmental origins of ensheathment and to find out if these were also similar in animals with and without backbones. Experiments using fruit fly and zebrafish embryos revealed that nerve cells, not skin cells, were responsible for forming and maintaining the sheaths. In embryos where groups of sensory nerve cells were selectively killed – either using a laser or by making the cells produce a toxin – ensheathment did not occur. Further studies, using a variety of microscopy techniques, revealed that the molecular machinery required to stabilize the sheaths was similar in both fish and flies, and therefore likely to be conserved across different groups of animals. Removing sheaths in fly embryos led to nerve cells becoming unstable; the animals were also less sensitive to touch. This confirmed that ensheathment was indeed necessary for sensory nerve cells to work properly. By revealing how ensheathment first emerges, these findings shed new light on how the sensory nervous system develops and how its activity is controlled. In humans, skin cells ensheath the nerve cells responsible for sensing pain. A better understanding of how ensheathments first arise could therefore lead to new avenues for treating chronic pain and related conditions.
Collapse
Affiliation(s)
- Nan Jiang
- Department of Biology, University of Washington, Seattle, United States
| | - Jeffrey P Rasmussen
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Joshua A Clanton
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Marci F Rosenberg
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Kory P Luedke
- Department of Biology, University of Washington, Seattle, United States
| | - Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University, Durham, United States
| | - Edward D Parker
- Department of Opthalmology, University of Washington, Seattle, United States
| | - Hyeon-Jin Kim
- Department of Chemistry, University of Washington, Seattle, United States.,Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, United States.,Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Alvaro Sagasti
- Department of Molecular Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, United States
| | - Jay Z Parrish
- Department of Biology, University of Washington, Seattle, United States
| |
Collapse
|
34
|
Matty MA, Knudsen DR, Walton EM, Beerman RW, Cronan MR, Pyle CJ, Hernandez RE, Tobin DM. Potentiation of P2RX7 as a host-directed strategy for control of mycobacterial infection. eLife 2019; 8:39123. [PMID: 30693866 PMCID: PMC6351102 DOI: 10.7554/elife.39123] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022] Open
Abstract
Mycobacterium tuberculosis is the leading worldwide cause of death due to a single infectious agent. Existing anti-tuberculous therapies require long treatments and are complicated by multi-drug-resistant strains. Host-directed therapies have been proposed as an orthogonal approach, but few have moved into clinical trials. Here, we use the zebrafish-Mycobacterium marinum infection model as a whole-animal screening platform to identify FDA-approved, host-directed compounds. We identify multiple compounds that modulate host immunity to limit mycobacterial disease, including the inexpensive, safe, and widely used drug clemastine. We find that clemastine alters macrophage calcium transients through potentiation of the purinergic receptor P2RX7. Host-directed drug activity in zebrafish larvae depends on both P2RX7 and inflammasome signaling. Thus, targeted activation of a P2RX7 axis provides a novel strategy for enhanced control of mycobacterial infections. Using a novel explant model, we find that clemastine is also effective within the complex granulomas that are the hallmark of mycobacterial infection.
Collapse
Affiliation(s)
- Molly A Matty
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States.,University Program in Genetics and Genomics, Duke University, Durham, United States
| | - Daphne R Knudsen
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Eric M Walton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Rebecca W Beerman
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Charlie J Pyle
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States
| | - Rafael E Hernandez
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, United States.,Department of Pediatrics, University of Washington, Seattle, United States
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, United States.,Department of Immunology, Duke University School of Medicine, Durham, United States
| |
Collapse
|