1
|
Gómez-Morón Á, Alegre-Gómez S, Ramirez-Muñoz R, Hernaiz-Esteban A, Carrasco-Padilla C, Scagnetti C, Aguilar-Sopeña Ó, García-Gil M, Borroto A, Torres-Ruiz R, Rodriguez-Perales S, Sánchez-Madrid F, Martín-Cófreces NB, Roda-Navarro P. Human T-cell receptor triggering requires inactivation of Lim kinase-1 by Slingshot-1 phosphatase. Commun Biol 2024; 7:918. [PMID: 39080357 PMCID: PMC11289303 DOI: 10.1038/s42003-024-06605-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024] Open
Abstract
Actin dynamics control early T-cell receptor (TCR) signalling during T-cell activation. However, the precise regulation of initial actin rearrangements is not completely understood. Here, we have investigated the regulatory role of the phosphatase Slingshot-1 (SSH1) in this process. Our data show that SSH1 rapidly polarises to nascent cognate synaptic contacts and later relocalises to peripheral F-actin networks organised at the mature immunological synapse. Knockdown of SSH1 expression by CRISPR/Cas9-mediated genome editing or small interfering RNA reveal a regulatory role for SSH1 in CD3ε conformational change, allowing Nck binding and proper downstream signalling and immunological synapse organisation. TCR triggering induces SSH1-mediated activation of actin dynamics through a mechanism mediated by Limk-1 inactivation. These data suggest that during early TCR activation, SSH1 is required for rapid F-actin rearrangements that mediate initial conformational changes of the TCR, integrin organisation and proximal signalling events for proper synapse organisation. Therefore, the SSH1 and Limk-1 axis is a key regulatory element for full T cell activation.
Collapse
Affiliation(s)
- Álvaro Gómez-Morón
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, UAM, 28006, Madrid, Spain
| | - Sergio Alegre-Gómez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
| | - Rocio Ramirez-Muñoz
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
| | - Alicia Hernaiz-Esteban
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
| | - Carlos Carrasco-Padilla
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
| | - Camila Scagnetti
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, UAM, 28006, Madrid, Spain
- Videomicroscopy Unit, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, UAM, 28006, Madrid, Spain
| | - Óscar Aguilar-Sopeña
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
| | - Marta García-Gil
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain
| | - Aldo Borroto
- Centro de Biología Molecular Severo Ochoa, Campus de Cantoblanco, 28049, Madrid, Spain
| | - Raul Torres-Ruiz
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
- Division of Hematopoietic Innovative Therapies, Biomedical Innovation Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnologicas (CIEMAT); Advanced Therapies Unit, Instituto de Investigacion Sanitaria Fundacion Jiménez Díaz; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM), 28040, Madrid, Spain
| | - Sandra Rodriguez-Perales
- Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics Program, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, UAM, 28006, Madrid, Spain
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Noa Beatriz Martín-Cófreces
- Immunology Service, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, UAM, 28006, Madrid, Spain.
- Videomicroscopy Unit, Instituto de Investigación Sanitaria del Hospital Universitario La Princesa, IIS-Princesa, UAM, 28006, Madrid, Spain.
- Area of Vascular Pathophysiology, Laboratory of Intercellular Communication, Fundación Centro Nacional de Investigaciones Cardiovasculares-Carlos III, 28029, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Pedro Roda-Navarro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
- 12 de Octubre Health Research Institute (imas12), 28040, Madrid, Spain.
| |
Collapse
|
2
|
Goodswen SJ, Kennedy PJ, Ellis JT. A state-of-the-art methodology for high-throughput in silico vaccine discovery against protozoan parasites and exemplified with discovered candidates for Toxoplasma gondii. Sci Rep 2023; 13:8243. [PMID: 37217589 DOI: 10.1038/s41598-023-34863-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/09/2023] [Indexed: 05/24/2023] Open
Abstract
Vaccine discovery against eukaryotic parasites is not trivial as highlighted by the limited number of known vaccines compared to the number of protozoal diseases that need one. Only three of 17 priority diseases have commercial vaccines. Live and attenuated vaccines have proved to be more effective than subunit vaccines but adversely pose more unacceptable risks. One promising approach for subunit vaccines is in silico vaccine discovery, which predicts protein vaccine candidates given thousands of target organism protein sequences. This approach, nonetheless, is an overarching concept with no standardised guidebook on implementation. No known subunit vaccines against protozoan parasites exist as a result of this approach, and consequently none to emulate. The study goal was to combine current in silico discovery knowledge specific to protozoan parasites and develop a workflow representing a state-of-the-art approach. This approach reflectively integrates a parasite's biology, a host's immune system defences, and importantly, bioinformatics programs needed to predict vaccine candidates. To demonstrate the workflow effectiveness, every Toxoplasma gondii protein was ranked in its capacity to provide long-term protective immunity. Although testing in animal models is required to validate these predictions, most of the top ranked candidates are supported by publications reinforcing our confidence in the approach.
Collapse
Affiliation(s)
- Stephen J Goodswen
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - Paul J Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology and the Australian Artificial Intelligence Institute, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - John T Ellis
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
3
|
Greco A, Straasheijm KR, Mul K, van den Heuvel A, van der Maarel SM, Joosten LA, van Engelen BG, Pruijn GJ. Profiling Serum Antibodies Against Muscle Antigens in Facioscapulohumeral Muscular Dystrophy Finds No Disease-Specific Autoantibodies. J Neuromuscul Dis 2021; 8:801-814. [PMID: 34024774 PMCID: PMC9789485 DOI: 10.3233/jnd-210653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND FSHD is caused by specific genetic mutations resulting in activation of the Double Homeobox 4 gene (DUX4). DUX4 targets hundreds of downstream genes eventually leading to muscle atrophy, oxidative stress, abnormal myogenesis, and muscle inflammation. We hypothesized that DUX4-induced aberrant expression of genes triggers a sustained autoimmune response against skeletal muscle cells. OBJECTIVE This study aimed at the identification of autoantibodies directed against muscle antigens in FSHD. Moreover, a possible relationship between serum antibody reactivity and DUX4 expression was also investigated. METHODS FSHD sera (N = 138, 48±16 years, 48% male) and healthy control sera (N = 20, 47±14 years, 50% male) were analyzed by immunoblotting for antibodies against several skeletal muscle protein extracts: healthy muscle, FSHD muscle, healthy and FSHD myotubes, and inducible DUX4 expressing myoblasts. In addition, DUX4 expressing myoblasts were analyzed by immunofluorescence with FSHD and healthy control sera. RESULTS The results showed that the reactivity of FSHD sera did not significantly differ from that of healthy controls, with all the tested muscle antigen extracts. Besides, the immunofluorescent staining of DUX4-expressing myoblasts was not different when incubated with either FSHD or healthy control sera. CONCLUSION Since the methodology used did not lead to the identification of disease-specific autoantibodies in the FSHD cohort, we suggest that autoantibody-mediated pathology may not be an important disease mechanism in FSHD. Nevertheless, it is crucial to further unravel if and which role the immune system plays in FSHD pathogenesis. Other innate as well as adaptive immune players could be involved in the complex DUX4 cascade of events and could become appealing druggable targets.
Collapse
Affiliation(s)
- Anna Greco
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands,Correspondence to: Anna Greco, MD, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Reinier Postlaan 4, 6525 GC, Nijmegen, The Netherlands. P.O. Box 9101, 6500 HB Nijmegen, The Netherlands. Tel.: +31 68 71 17 452; Fax: +31 24 354 1122; E-mail:
| | - Kirsten R. Straasheijm
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Karlien Mul
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anita van den Heuvel
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Baziel G.M. van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ger J.M. Pruijn
- Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
4
|
The Preventive Effect of the Phenotype of Tumour-Associated Macrophages, Regulated by CD39, on Colon Cancer in Mice. Int J Mol Sci 2021; 22:ijms22147478. [PMID: 34299098 PMCID: PMC8308112 DOI: 10.3390/ijms22147478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022] Open
Abstract
Background: This study was designed to investigate the effect of cluster differentiation (CD)39 and CD73 inhibitors on the expresion of tumour-associated macrophages (TAMs), M1- versus M2-tumour phenotypes in mice with colon cancer. Methods: An in vitro study of co-culture with colon cancer cells and immune cells from the bone marrow (BM) of mice was performed. After the confirmation of the effect of polyoxotungstate (POM-1) as an inhibitor of CD39 on TAMs, the mice were randomly divided into a control group without POM-1 and a study group with POM-1, respectively, after subcutaneous injection of CT26 cells. On day 14 after the injection, the mice were sacrificed, and TAMs were evaluated using fluorescence-activated cell sorting. Results: In the in vitro study, the co-culture with POM-1 significantly increased the apoptosis of CT26 cells. The cell population from the co-culture with POM-1 showed significant increases in the expression of CD11b+ for myeloid cells, lymphocyte antigen 6 complex, locus C (Ly6C+) for monocytes, M1-tumour phenotypes from TAMs, and F4/80+ for macrophages. In the in vivo study, tumour growth in the study group with POM-1 was significantly limited, compared with the control group without POM-1. The expressions of Ly6C+ and major histocompatibility complex class II+ for M1-tumour phenotypes from TAMs on F4/80+ from the tumour tissue in the study group had significantly higher values compared with the control group. Conclusion: The inhibition of CD39 with POM-1 prevented the growth of colon cancer in mice, and it was associated with the increased expression of M1-tumour phenotypes from TAMs in the cancer tissue.
Collapse
|
5
|
Ebelt ND, Zuniga E, Johnson BL, Diamond DJ, Manuel ER. 5-Azacytidine Potentiates Anti-tumor Immunity in a Model of Pancreatic Ductal Adenocarcinoma. Front Immunol 2020; 11:538. [PMID: 32296439 PMCID: PMC7136411 DOI: 10.3389/fimmu.2020.00538] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/09/2020] [Indexed: 12/26/2022] Open
Abstract
Tumors evolve a variety of mechanisms to escape immune detection while expressing tumor-promoting molecules that can be immunogenic. Here, we show that transposable elements (TE) and gene encoded, tumor-associated antigens (TAA), which can be both highly immunogenic and tumor-promoting, are significantly upregulated during the transition from pre-malignancy to malignancy in an inducible model of pancreatic ductal adenocarcinoma (PDAC). Coincident with the increased presence of TEs and TAAs was the downregulation of gene transcripts associated with antigen presentation, T cell recruitment and intrinsic anti-viral responses, suggesting a unique strategy employed by PDAC to possibly augment tumorigenesis while escaping detection by the immune system. In vitro treatment of mouse and human PDAC cell lines with the DNA methyltransferase inhibitor 5-azacytidine (Aza) resulted in augmented expression of transcripts for antigen presentation machinery and T cell chemokines. When immunocompetent mice implanted with PDAC were therapeutically treated with Aza, we observed significant tumor regression that was not observed in immunocompromised mice, implicating anti-tumor immunity as the principal mechanism of tumor growth control. Analysis of PDAC tumors, immediately following Aza treatment in immunocompetent mice, revealed a significantly greater infiltration of T cells and various innate immune subsets compared to control treatment, suggesting that Aza treatment enhances tumor immunogenicity. Thus, augmenting antigen presentation and T cell chemokine expression using DNA methyltransferase inhibitors could be leveraged to potentiate adaptive anti-tumor immune responses against PDAC.
Collapse
Affiliation(s)
- Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Edith Zuniga
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| | - Benjamin L. Johnson
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Don J. Diamond
- Department of Hematology and Hematopoietic Stem Cell Transplantation, City of Hope, Duarte, CA, United States
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, United States
| |
Collapse
|
6
|
Investigating Macrophages Plasticity Following Tumour-Immune Interactions During Oncolytic Therapies. Acta Biotheor 2019; 67:321-359. [PMID: 31410657 PMCID: PMC6825040 DOI: 10.1007/s10441-019-09357-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 08/02/2019] [Indexed: 12/22/2022]
Abstract
Over the last few years, oncolytic virus therapy has been recognised as a promising approach in cancer treatment, due to the potential of these viruses to induce systemic anti-tumour immunity and selectively killing tumour cells. However, the effectiveness of these viruses depends significantly on their interactions with the host immune responses, both innate (e.g., macrophages, which accumulate in high numbers inside solid tumours) and adaptive (e.g., \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {CD8}^{+}$$\end{document}CD8+ T cells). In this article, we consider a mathematical approach to investigate the possible outcomes of the complex interactions between two extreme types of macrophages (M1 and M2 cells), effector \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {CD8}^{+}$$\end{document}CD8+ T cells and an oncolytic Vesicular Stomatitis Virus (VSV), on the growth/elimination of B16F10 melanoma. We discuss, in terms of VSV, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {CD8}^{+}$$\end{document}CD8+ and macrophages levels, two different types of immune responses which could ensure tumour control and eventual elimination. We show that both innate and adaptive anti-tumour immune responses, as well as the oncolytic virus, could be very important in delaying tumour relapse and eventually eliminating the tumour. Overall this study supports the use mathematical modelling to increase our understanding of the complex immune interaction following oncolytic virotherapies. However, the complexity of the model combined with a lack of sufficient data for model parametrisation has an impact on the possibility of making quantitative predictions.
Collapse
|
7
|
Friedrich J, Heim L, Trufa DI, Sirbu H, Rieker RJ, Chiriac MT, Finotto S. STAT1 deficiency supports PD-1/PD-L1 signaling resulting in dysfunctional TNFα mediated immune responses in a model of NSCLC. Oncotarget 2018; 9:37157-37172. [PMID: 30647851 PMCID: PMC6324686 DOI: 10.18632/oncotarget.26441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 11/26/2018] [Indexed: 12/27/2022] Open
Abstract
In this study we described that Signal Transducer and Activator of Transcription 1 (STAT1) is a key point regulator of PD-1 in tumour infiltrating lymphocytes and PD-L1 in Tumour associated macrophages (TAM) in NSCLC. In our murine model of adenocarcinoma targeted deletion of Stat1 was found associated with enhanced tumour growth, impaired differentiation into M1-like macrophages from the bone marrow, the accumulation of tumor associated macrophages overexpressing PD-L1 and impaired T cell responses in the tumor microenvironment by affecting TNFα responses. In our human NSCLC patient cohort we found that loss of isoforms STAT1 α and STAT1β mRNA in the tumoural region of the lung correlates with increased tumor size in NSCLC patients. Therefore, STAT1 isoform regulation could be considered for future therapeutical strategies associated to current immune-checkpoint blockade therapy in NSCLC.
Collapse
Affiliation(s)
- Juliane Friedrich
- Department of Molecular Pneumology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Lisanne Heim
- Department of Molecular Pneumology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Denis I Trufa
- Department of Thoracic Surgery, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf J Rieker
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Mircea T Chiriac
- Department of Medicine 1-Gastroenterology, Pneumology and Endocrinology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Crouser ED, White P, Caceres EG, Julian MW, Papp AC, Locke LW, Sadee W, Schlesinger LS. A Novel In Vitro Human Granuloma Model of Sarcoidosis and Latent Tuberculosis Infection. Am J Respir Cell Mol Biol 2017; 57:487-498. [PMID: 28598206 DOI: 10.1165/rcmb.2016-0321oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Many aspects of pathogenic granuloma formation are poorly understood, requiring new relevant laboratory models that represent the complexity (genetics and diversity) of human disease. To address this need, we developed an in vitro model of granuloma formation using human peripheral blood mononuclear cells (PBMCs) derived from patients with active sarcoidosis, latent tuberculosis (TB) infection (LTBI), or normal healthy control subjects. PBMCs were incubated for 7 days with uncoated polystyrene beads or beads coated with purified protein derivative (PPD) or human serum albumin. In response to PPD-coated beads, PBMCs from donors with sarcoidosis and LTBI formed robust multicellular aggregates resembling granulomas, displaying a typical T-helper cell type 1 immune response, as assessed by cytokine analyses. In contrast, minimal PBMC aggregation occurred when control PBMCs were incubated with PPD-coated beads, whereas the response to uncoated beads was negligible in all groups. Sarcoidosis PBMCs responded to human serum albumin-coated beads with modest cellular aggregation and inflammatory cytokine release. Whereas the granuloma-like aggregates formed in response to PPD-coated beads were similar for sarcoidosis and LTBI, molecular profiles differed significantly. mRNA expression patterns revealed distinct pathways engaged in early granuloma formation in sarcoidosis and LTBI, and they resemble molecular patterns reported in diseased human tissues. This novel in vitro human granuloma model is proposed as a tool to investigate mechanisms of early granuloma formation and for preclinical drug discovery research of human granulomatous disorders. Clinical trial registered with www.clinicaltrials.gov (NCT01857401).
Collapse
Affiliation(s)
- Elliott D Crouser
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, the Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Peter White
- 2 Center for Microbial Pathogenesis, the Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and
| | - Evelyn Guirado Caceres
- 3 Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, and
| | - Mark W Julian
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, the Dorothy M. Davis Heart and Lung Research Institute, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Audrey C Papp
- 4 Department of Cancer Biology and Genetics, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Landon W Locke
- 3 Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, and
| | - Wolfgang Sadee
- 4 Department of Cancer Biology and Genetics, the Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Larry S Schlesinger
- 3 Department of Microbial Infection and Immunity, Center for Microbial Interface Biology, and
| |
Collapse
|
9
|
Sha LK, Sha W, Kuchler L, Daiber A, Giegerich AK, Weigert A, Knape T, Snodgrass R, Schröder K, Brandes RP, Brüne B, von Knethen A. Loss of Nrf2 in bone marrow-derived macrophages impairs antigen-driven CD8(+) T cell function by limiting GSH and Cys availability. Free Radic Biol Med 2015; 83:77-88. [PMID: 25687825 DOI: 10.1016/j.freeradbiomed.2015.02.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 01/01/2023]
Abstract
NF-E2-related factor 2 (Nrf2), known to protect against reactive oxygen species, has recently been reported to resolve acute inflammatory responses in activated macrophages. Consequently, disruption of Nrf2 promotes a proinflammatory macrophage phenotype. In the current study, we addressed the impact of this macrophage phenotype on CD8(+) T cell activation by using an antigen-driven coculture model consisting of Nrf2(-/-) and Nrf2(+/+) bone marrow-derived macrophages (BMDMΦ) and transgenic OT-1 CD8(+) T cells. OT-1 CD8(+) T cells encode a T cell receptor that specifically recognizes MHC class I-presented ovalbumin OVA(257-264) peptide, thereby causing a downstream T cell activation. Interestingly, coculture of OVA(257-264)-pulsed Nrf2(-/-) BMDMΦ with transgenic OT-1 CD8(+) T cells attenuated CD8(+) T cell activation, proliferation, and cytotoxic function. Since the provision of low-molecular-weight thiols such as glutathione (GSH) or cysteine (Cys) by macrophages limits antigen-driven CD8(+) T cell activation, we quantified the amounts of intracellular and extracellular GSH and Cys in both cocultures. Indeed, GSH levels were strongly decreased in Nrf2(-/-) cocultures compared to wild-type counterparts. Supplementation of thiols in Nrf2(-/-) cocultures via addition of glutathione ester, N-acetylcysteine, β-mercaptoethanol, or cysteine itself restored T cell proliferation as well as cytotoxicity by increasing intracellular GSH. Mechanistically, we identified two potential Nrf2-regulated genes involved in thiol synthesis in BMDMΦ: the cystine transporter subunit xCT and the modulatory subunit of the GSH-synthesizing enzyme γ-GCS (GCLM). Pharmacological inhibition of γ-GCS-dependent GSH synthesis as well as knockdown of the cystine antiporter xCT in Nrf2(+/+) BMDMΦ mimicked the effect of Nrf2(-/-) BMDMΦ on CD8(+) T cell function. Our findings demonstrate that reduced levels of GCLM as well as xCT in Nrf2(-/-) BMDMΦ limit GSH availability, thereby inhibiting antigen-induced CD8(+) T cell function.
Collapse
MESH Headings
- Animals
- Antioxidants/metabolism
- Apoptosis
- Blotting, Western
- Bone Marrow/immunology
- Bone Marrow/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cells, Cultured
- Cystine/metabolism
- Flow Cytometry
- Glutathione/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/metabolism
- Immunoenzyme Techniques
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- NF-E2-Related Factor 2/physiology
- Ovalbumin/immunology
- Ovalbumin/metabolism
- Oxidative Stress
- RNA, Messenger/genetics
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
Collapse
Affiliation(s)
- Lisa K Sha
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Weixiao Sha
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Laura Kuchler
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas Daiber
- Department of Medicine II, University Medical Center, Johannes Gutenberg-University Mainz, 55116 Mainz, Germany
| | - Annika K Giegerich
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Tilo Knape
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Project Group Translational Medicine & Pharmacology TMP, and Goethe-University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Ryan Snodgrass
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Katrin Schröder
- Institute of Cardiovascular Physiology-Physiology I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Ralf P Brandes
- Institute of Cardiovascular Physiology-Physiology I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Andreas von Knethen
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
10
|
Ammari MG, Harris AN, Stokes JV, Bailey RH, Pinchuk LM. NEGATIVE REGULATORY EFFECTS OF PHOSPHATIDYLINOSITOL3-KINASE PATHWAY ON PHAGOCYTOSIS AND MACROPINOCYTOSIS IN BOVINE MONOCYTES. JOURNAL OF VETERINARY MEDICINE AND RESEARCH 2014; 1:1008. [PMID: 25893219 PMCID: PMC4399709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Recent studies have shown that monocytes and macrophages not only present antigens to effector T cells and stimulate and shape T cell-mediated immune responses, but they also prime naïve T cells, thus initiating adaptive immune responses. Phosphatidylinositol 3-kinase functions at an early phase of toll-like receptor signaling pathways, modulates the magnitude of the primary immune responses, and is involved in the reorganization of the actin cytoskeleton during macropinocytic and phagocytic antigen uptakes, important early steps in triggering adaptive immune responses. We assessed by flow cytometry the endocytic capacities of bovine monocytes by using endocytic tracers and Salmonella transformed with a green fluorescence plasmid GFP to evaluate macropinocytosis, mannose receptor-mediated endocytosis, and phagocytosis in bovine professional antigen presenting cells, respectively. Our data reveal that wortmannin, an inhibitor of phosphatidylinositol 3-kinase signaling pathway, significantly increased macropinocytosis and phagocytosis but did not affect the mannose receptor-mediated antigen uptake in bovine monocytes. Protein expression data support these findings by showing decreased levels of phosphoinositide 3-kinase in the presence of wortmannin during macropinocytosis. We expanded further the key role of phosphatidylinositol 3-kinase as an endogenous suppressor of primary immune responses, suggesting a novel mechanism of phosphatidylinositol 3-kinase antigen uptake modulation that may provide a unique therapeutic target for controlling excessive inflammation.
Collapse
Affiliation(s)
- Mais G. Ammari
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Autumn N. Harris
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
- Department of Small Animal Internal Medicine, University of Florida, Gainesville, FL 32611, USA
| | - John V. Stokes
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Richard H. Bailey
- Department of Pathobiology and Population Medicine, Mississippi State, MS 39762, USA
| | - Lesya M. Pinchuk
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| |
Collapse
|
11
|
Weaver KF, Stokes JV, Gunnoe SA, Follows JS, Shafer L, Ammari MG, Archer TM, Thomason JM, Mackin AJ, Pinchuk LM. EFFECT OF LIPOSOMAL CLODRONATE-DEPENDENT DEPLETION OF PROFESSIONAL ANTIGEN PRESENTING CELLS ON NUMBERS AND PHENOTYPE OF CANINE CD4+CD25+FOXP3+ REGULATORY T CELLS. JOURNAL OF VETERINARY MEDICINE AND RESEARCH 2014; 1:1003. [PMID: 25950023 PMCID: PMC4418640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Regulatory T cells (Tregs) are known to control autoreactivity during and subsequent to the development of the peripheral immune system. Professional antigen presenting cells (APCs), dendritic cells (DCs) and monocytes, have an important role in inducing Tregs. For the first time, this study evaluated proportions and phenotypes of Tregs in canine peripheral blood depleted of professional APCs, utilizing liposomal clodronate (LC) and multicolor flow cytometry analysis. Our results demonstrate that LC exposure promoted short term decreases followed by significant increases in the proportions or absolute numbers of CD4+CD25+FOXP3+ Tregs in dogs. In general, the LC-dependent Treg fluctuations were similar to the changes in the levels of CD14+ monocytes in Walker hounds. However, the proportions of monocytes showed more dramatic changes compared to the proportions of Tregs that were visually unchanged after LC treatment over the study period. At the same time, absolute Treg numbers showed, similarly to the levels of CD14+ monocytes, significant compensatory gains as well as the recovery during the normalization period. We confirm the previous data that CD4+ T cells with the highest CD25 expression were highly enriched for FOXP3. Furthermore, for the first time, we report that CD4+CD25lowFOXP3+ is the major regulatory T cell subset affected by LC exposure. The increases within the lowest CD25 expressers of CD4+FOXP3+ cells together with compensatory gains in the proportion of CD14+ monocytes during compensatory and normalization periods suggest the possible direct or indirect roles of monocytes in active recruitment and generation of Tregs from naïve CD4+ T cells.
Collapse
Affiliation(s)
- Kriston F. Weaver
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - John V. Stokes
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Sagen A. Gunnoe
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Joyce S. Follows
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Lydia Shafer
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Mais G. Ammari
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Todd M. Archer
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - John M. Thomason
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Andrew J. Mackin
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| | - Lesya M. Pinchuk
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, 240 Wise Center Drive, Mississippi State, MS 39762, USA
| |
Collapse
|
12
|
Mulder R, Banete A, Basta S. Spleen-derived macrophages are readily polarized into classically activated (M1) or alternatively activated (M2) states. Immunobiology 2014; 219:737-45. [PMID: 24954891 DOI: 10.1016/j.imbio.2014.05.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/25/2014] [Accepted: 05/29/2014] [Indexed: 10/25/2022]
Abstract
Bone marrow derived macrophages (BM-MΦ) that differentiate from precursor cells can be polarized into classically activated pro-inflammatory (M1) or alternatively activated (M2) states depending upon the cytokine microenvironment. We questioned whether tissue MΦ, such as spleen-derived MΦ (Sp-MΦ), have the ability to differentiate into M1 or M2 cells. We show in response to activation with IFN-gamma (IFN-γ) and lipopolysaccharide (LPS), that the Sp-MΦ readily acquired an M1 status indicated by up-regulation of iNOS mRNA, nitric oxide (NO) production, and the co-stimulatory molecule CD86. Conversely, Sp-MΦ exposed to IL-4 exhibited increased levels of mannose receptor (CD 206), arginase-1 (Arg)-1 mRNA expression, and significant urea production typical of M2 cells. At this stage of differentiation, the M2 Sp-MΦ were more efficient at phagocytosis of cell-associated antigens than their M1 counterparts. This polarization was not indefinite as the cells could revert back to their original state upon the removal of stimuli and exhibited flexibility to convert from M2 to M1. Remarkably, both M1 and M2 Sp-MΦ induced more CD4 expression on their cells surface after stimulation. We also demonstrate that adherent macrophages cultured for a short term in IL-4 enhances ARG-1 and YM-1 mRNA along with increasing urea producing capacity: traits indicative of an M2 phenotype. Moreover, in response to in vivo virus infection, the adherent macrophages obtained from spleens rapidly express iNOS. These results provide new evidence for the polarization capabilities of Sp-MΦ when exposed to pro-inflammatory or anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Rylend Mulder
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Andra Banete
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
13
|
Alvarez B, Poderoso T, Alonso F, Ezquerra A, Domínguez J, Revilla C. Antigen targeting to APC: from mice to veterinary species. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:153-163. [PMID: 23648645 DOI: 10.1016/j.dci.2013.04.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 04/24/2013] [Accepted: 04/25/2013] [Indexed: 06/02/2023]
Abstract
Antigen delivery to receptors expressed on antigen presenting cells (APC) has shown to improve immunogenicity of vaccines in mice. An enhancement of cytotoxic T lymphocyte (CTL), helper T cell or humoral responses was obtained depending on the type of APC and the surface molecule targeted. Although this strategy is being also evaluated in livestock animals with promising results, some discrepancies have been found between species and pathogens. The genetic diversity of livestock animals, the different pattern of expression of some receptors among species, the use of different markers to characterize APC in large animals and sometimes the lack of reagents make difficult to compare results obtained in different species. In this review, we summarize the data available regarding antigen targeting to APC receptors in cattle, sheep and pig and discuss the results found in these animals in the context of what has been obtained in mice.
Collapse
Affiliation(s)
- B Alvarez
- Dpto. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
14
|
Barrio MM, Abes R, Colombo M, Pizzurro G, Boix C, Roberti MP, Gélizé E, Rodriguez-Zubieta M, Mordoh J, Teillaud JL. Human macrophages and dendritic cells can equally present MART-1 antigen to CD8(+) T cells after phagocytosis of gamma-irradiated melanoma cells. PLoS One 2012; 7:e40311. [PMID: 22768350 PMCID: PMC3388056 DOI: 10.1371/journal.pone.0040311] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 06/04/2012] [Indexed: 11/19/2022] Open
Abstract
Dendritic cells (DC) can achieve cross-presentation of naturally-occurring tumor-associated antigens after phagocytosis and processing of dying tumor cells. They have been used in different clinical settings to vaccinate cancer patients. We have previously used gamma-irradiated MART-1 expressing melanoma cells as a source of antigens to vaccinate melanoma patients by injecting irradiated cells with BCG and GM-CSF or to load immature DC and use them as a vaccine. Other clinical trials have used IFN-gamma activated macrophage killer cells (MAK) to treat cancer patients. However, the clinical use of MAK has been based on their direct tumoricidal activity rather than on their ability to act as antigen-presenting cells to stimulate an adaptive antitumor response. Thus, in the present work, we compared the fate of MART-1 after phagocytosis of gamma-irradiated cells by clinical grade DC or MAK as well as the ability of these cells to cross present MART-1 to CD8(+) T cells. Using a high affinity antibody against MART-1, 2A9, which specifically stains melanoma tumors, melanoma cell lines and normal melanocytes, the expression level of MART-1 in melanoma cell lines could be related to their ability to stimulate IFN-gamma production by a MART-1 specific HLA-A*0201-restricted CD8(+) T cell clone. Confocal microscopy with Alexa Fluor®(647)-labelled 2A9 also showed that MART-1 could be detected in tumor cells attached and/or fused to phagocytes and even inside these cells as early as 1 h and up to 24 h or 48 h after initiation of co-cultures between gamma-irradiated melanoma cells and MAK or DC, respectively. Interestingly, MART-1 was cross-presented to MART-1 specific T cells by both MAK and DC co-cultured with melanoma gamma-irradiated cells for different time-points. Thus, naturally occurring MART-1 melanoma antigen can be taken-up from dying melanoma cells into DC or MAK and both cell types can induce specific CD8(+) T cell cross-presentation thereafter.
Collapse
Affiliation(s)
- María Marcela Barrio
- Centro de Investigaciones Oncológicas,
Fundación Cáncer FUCA, Buenos Aires, Argentina
| | - Riad Abes
- INSERM UMR S 872, Paris, France
- Centre de Recherche des Cordeliers,
Université Pierre et Marie Curie – Paris6, UMR S 872, Paris,
France
- Université Paris Descartes,
UMR S 872, Paris, France
| | - Marina Colombo
- Instituto de Investigaciones Bioquímicas
de Buenos Aires, CONICET, Fundación Instituto Leloir, Buenos Aires,
Argentina
| | - Gabriela Pizzurro
- Centro de Investigaciones Oncológicas,
Fundación Cáncer FUCA, Buenos Aires, Argentina
| | - Charlotte Boix
- INSERM UMR S 872, Paris, France
- Centre de Recherche des Cordeliers,
Université Pierre et Marie Curie – Paris6, UMR S 872, Paris,
France
- Université Paris Descartes,
UMR S 872, Paris, France
| | - María Paula Roberti
- Centro de Investigaciones Oncológicas,
Fundación Cáncer FUCA, Buenos Aires, Argentina
| | - Emmanuelle Gélizé
- INSERM UMR S 872, Paris, France
- Centre de Recherche des Cordeliers,
Université Pierre et Marie Curie – Paris6, UMR S 872, Paris,
France
- Université Paris Descartes,
UMR S 872, Paris, France
| | - Mariana Rodriguez-Zubieta
- Instituto de Investigaciones Bioquímicas
de Buenos Aires, CONICET, Fundación Instituto Leloir, Buenos Aires,
Argentina
| | - José Mordoh
- Centro de Investigaciones Oncológicas,
Fundación Cáncer FUCA, Buenos Aires, Argentina
- Instituto de Investigaciones Bioquímicas
de Buenos Aires, CONICET, Fundación Instituto Leloir, Buenos Aires,
Argentina
| | - Jean-Luc Teillaud
- INSERM UMR S 872, Paris, France
- Centre de Recherche des Cordeliers,
Université Pierre et Marie Curie – Paris6, UMR S 872, Paris,
France
- Université Paris Descartes,
UMR S 872, Paris, France
| |
Collapse
|
15
|
Mukhopadhyay S, Nair S, Ghosh S. Pathogenesis in tuberculosis: transcriptomic approaches to unraveling virulence mechanisms and finding new drug targets. FEMS Microbiol Rev 2011; 36:463-85. [PMID: 22092372 DOI: 10.1111/j.1574-6976.2011.00302.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 07/31/2011] [Accepted: 08/05/2011] [Indexed: 01/12/2023] Open
Abstract
Tuberculosis (TB) remains a major health problem worldwide. Attempts to control this disease have proved difficult owing to our poor understanding of the pathobiology of Mycobacterium tuberculosis and the emergence of strains that are resistant to multiple drugs currently available for treatment. Genome-wide expression profiling has provided new insight into the transcriptome signatures of the bacterium during infection, notably of macrophages and dendritic cells. These data indicate that M. tuberculosis expresses numerous genes to evade the host immune responses, to suit its intracellular life style, and to respond to various antibiotic drugs. Among the intracellularly induced genes, several have functions in lipid metabolism, cell wall synthesis, iron uptake, oxidative stress resistance, protein secretion, or inhibition of apoptosis. Herein we review these findings and discuss possible ways to exploit the data to understand the complex etiology of TB and to find new effective drug targets.
Collapse
Affiliation(s)
- Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting and Diagnostics, Nampally, Hyderabad, India.
| | | | | |
Collapse
|
16
|
Bibliography. Current world literature. Curr Opin Endocrinol Diabetes Obes 2009; 16:328-37. [PMID: 19564733 DOI: 10.1097/med.0b013e32832eb365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Acosta-Iborra B, Elorza A, Olazabal IM, Martín-Cofreces NB, Martin-Puig S, Miró M, Calzada MJ, Aragonés J, Sánchez-Madrid F, Landázuri MO. Macrophage oxygen sensing modulates antigen presentation and phagocytic functions involving IFN-gamma production through the HIF-1 alpha transcription factor. THE JOURNAL OF IMMUNOLOGY 2009; 182:3155-64. [PMID: 19234213 DOI: 10.4049/jimmunol.0801710] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Low oxygen tension areas are found in inflamed or diseased tissues where hypoxic cells induce survival pathways by regulating the hypoxia-inducible transcription factor (HIF). Macrophages are essential regulators of inflammation and, therefore, we have analyzed their response to hypoxia. Murine peritoneal elicited macrophages cultured under hypoxia produced higher levels of IFN-gamma and IL-12 mRNA and protein than those cultured under normoxia. A similar IFN-gamma increment was obtained with in vivo models using macrophages from mice exposed to atmospheric hypoxia. Our studies showed that IFN-gamma induction was mediated through HIF-1alpha binding to its promoter on a new functional hypoxia response element. The requirement of HIF-alpha in the IFN-gamma induction was confirmed in RAW264.7 cells, where HIF-1alpha was knocked down, as well as in resident HIF-1alpha null macrophages. Moreover, Ag presentation capacity was enhanced in hypoxia through the up-regulation of costimulatory and Ag-presenting receptor expression. Hypoxic macrophages generated productive immune synapses with CD8 T cells that were more efficient for activation of TCR/CD3epsilon, CD3zeta and linker for activation of T cell phosphorylation, and T cell cytokine production. In addition, hypoxic macrophages bound opsonized particles with a higher efficiency, increasing their phagocytic uptake, through the up-regulated expression of phagocytic receptors. These hypoxia-increased immune responses were markedly reduced in HIF-1alpha- and in IFN-gamma-silenced macrophages, indicating a link between HIF-1alpha and IFN-gamma in the functional responses of macrophages to hypoxia. Our data underscore an important role of hypoxia in the activation of macrophage cytokine production, Ag-presenting activity, and phagocytic activity due to an HIF-1alpha-mediated increase in IFN-gamma levels.
Collapse
Affiliation(s)
- Bárbara Acosta-Iborra
- Servicio de Inmunología, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|