101
|
Naumenko V, Turk M, Jenne CN, Kim SJ. Neutrophils in viral infection. Cell Tissue Res 2018; 371:505-516. [PMID: 29327081 DOI: 10.1007/s00441-017-2763-0] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022]
Abstract
Neutrophils are the first wave of recruited immune cells to sites of injury or infection and are crucial players in controlling bacterial and fungal infections. Although the role of neutrophils during bacterial or fungal infections is well understood, their impact on antiviral immunity is much less studied. Furthermore, neutrophil function in tumor pathogenesis and cancer treatment has recently received much attention, particularly within the context of oncolytic virus infection where neutrophils produce antitumor cytokines and enhance oncolysis. In this review, multiple functions of neutrophils in viral infections and immunity are discussed. Understanding the role of neutrophils during viral infection may provide insight into the pathogenesis of virus infections and the outcome of virus-based therapies.
Collapse
Affiliation(s)
- Victor Naumenko
- Department of Microbiology, Immunology and Infectious Diseases, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, HRIC 3330 Hospital Drive N.W, Calgary, Alberta, T2N 4N1, Canada.,National University of Science and Technology "MISIS", Leninskiy prospect 4, 119991, Moscow, Russia
| | - Madison Turk
- Department of Microbiology, Immunology and Infectious Diseases, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, HRIC 3330 Hospital Drive N.W, Calgary, Alberta, T2N 4N1, Canada
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, HRIC 3330 Hospital Drive N.W, Calgary, Alberta, T2N 4N1, Canada. .,Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, HRIC 2C26, 3280 Hospital Drive N.W., Calgary, Alberta, T2N 4N1, Canada.
| | - Seok-Joo Kim
- Department of Microbiology, Immunology and Infectious Diseases, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, HRIC 3330 Hospital Drive N.W, Calgary, Alberta, T2N 4N1, Canada. .,Calvin, Phoebe & Joan Snyder Institute for Chronic Diseases, Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, HRIC 4C49, 3280 Hospital Drive N.W., Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
102
|
Dai J, Gu L, Su Y, Wang Q, Zhao Y, Chen X, Deng H, Li W, Wang G, Li K. Inhibition of curcumin on influenza A virus infection and influenzal pneumonia via oxidative stress, TLR2/4, p38/JNK MAPK and NF-κB pathways. Int Immunopharmacol 2018; 54:177-187. [PMID: 29153953 DOI: 10.1016/j.intimp.2017.11.009] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Oxidative stress, Nrf2-HO-1 and TLR-MAPK/NF-κB signaling pathways have been proved to be involved in influenza A virus (IAV) replication and influenzal pneumonia. In the previous studies, we have performed several high-throughput drug screenings based on the TLR pathways. In the present study, through plaque inhibition test, luciferase reporter assay, TCID50, qRT-PCR, western blotting, ELISA and siRNA assays, we investigated the effect and mechanism of action of curcumin against IAV infection in vitro and in vivo. The results showed that curcumin could directly inactivate IAV, blocked IAV adsorption and inhibited IAV proliferation. As for the underlying mechanisms, we found that curcumin could significantly inhibit IAV-induced oxidative stress, increased Nrf2, HO-1, NQO1, GSTA3 and IFN-β production, and suppressed IAV-induced activation of TLR2/4/7, Akt, p38/JNK MAPK and NF-κB pathways. Suppression of Nrf2 via siRNA significantly abolished the stimulatory effect of curcumin on HO-1, NQO1, GSTA3 and IFN-β production and meanwhile blocked the inhibitory effect of curcumin on IAV M2 production. Oxidant H2O2 and TLR2/4, p38/JNK and NF-κB agonists could significantly antagonize the anti-IAV activity of curcumin in vitro. Additionally, curcumin significantly increased the survival rate of mice, reduced lung index, inflammatory cytokines and lung IAV titer, and finally improved pulmonary histopathological changes after IAV infection. In conclusion, curcumin can directly inactivate IAV, inhibits IAV adsorption and replication; and its inhibition on IAV replication may be via activating Nrf2 signal and inhibiting IAV-induced activation of TLR2/4, p38/JNK MAPK and NF-κB pathways.
Collapse
Affiliation(s)
- Jianping Dai
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China.
| | - Liming Gu
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Yun Su
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Qianwen Wang
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Ying Zhao
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Xiaoxua Chen
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Huixiong Deng
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Weizhong Li
- Department of Veterinary Medicine, University of Maryland, College Park, Virginia-Maryland Regional College of Veterinary Medicine, 159 College Park Rd, MD 20742, USA
| | - Gefei Wang
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Shantou University Medical College, 22 Xingling Rd, Shantou 515041, China
| |
Collapse
|
103
|
Dey R, Joshi AB, Oliveira F, Pereira L, Guimarães-Costa AB, Serafim TD, de Castro W, Coutinho-Abreu IV, Bhattacharya P, Townsend S, Aslan H, Perkins A, Karmakar S, Ismail N, Karetnick M, Meneses C, Duncan R, Nakhasi HL, Valenzuela JG, Kamhawi S. Gut Microbes Egested during Bites of Infected Sand Flies Augment Severity of Leishmaniasis via Inflammasome-Derived IL-1β. Cell Host Microbe 2017; 23:134-143.e6. [PMID: 29290574 DOI: 10.1016/j.chom.2017.12.002] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022]
Abstract
Leishmania donovani parasites are the cause of visceral leishmaniasis and are transmitted by bites from phlebotomine sand flies. A prominent feature of vector-transmitted Leishmania is the persistence of neutrophils at bite sites, where they protect captured parasites, leading to enhanced disease. Here, we demonstrate that gut microbes from the sand fly are egested into host skin alongside Leishmania parasites. The egested microbes trigger the inflammasome, leading to a rapid production of interleukin-1β (IL-1β), which sustains neutrophil infiltration. Reducing midgut microbiota by pretreatment of Leishmania-infected sand flies with antibiotics or neutralizing the effect of IL-1β in bitten mice abrogates neutrophil recruitment. These early events are associated with impairment of parasite visceralization, indicating that both gut microbiota and IL-1β are important for the establishment of Leishmania infections. Considering that arthropods harbor a rich microbiota, its potential egestion after bites may be a shared mechanism that contributes to severity of vector-borne disease.
Collapse
Affiliation(s)
- Ranadhir Dey
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Amritanshu B Joshi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Lais Pereira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Anderson B Guimarães-Costa
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Tiago D Serafim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Waldionê de Castro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Iliano V Coutinho-Abreu
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Shannon Townsend
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Hamide Aslan
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Alec Perkins
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Subir Karmakar
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Nevien Ismail
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Morgan Karetnick
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Robert Duncan
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
104
|
Royer PJ, Henrio K, Pain M, Loy J, Roux A, Tissot A, Lacoste P, Pison C, Brouard S, Magnan A. TLR3 promotes MMP-9 production in primary human airway epithelial cells through Wnt/β-catenin signaling. Respir Res 2017; 18:208. [PMID: 29237464 PMCID: PMC5729411 DOI: 10.1186/s12931-017-0690-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/28/2017] [Indexed: 12/27/2022] Open
Abstract
Background Airway epithelial cells (AEC) act as the first line of defence in case of lung infections. They constitute a physical barrier against pathogens and they participate in the initiation of the immune response. Yet, the modalities of pathogen recognition by AEC and the consequences on the epithelial barrier remain poorly documented. Method We investigated the response of primary human AEC to viral (polyinosinic-polycytidylic acid, poly(I:C)) and bacterial (lipopolysaccharide, LPS) stimulations in combination with the lung remodeling factor Transforming Growth Factor-β (TGF-β). Results We showed a strong production of pro-inflammatory cytokines (Interleukin (IL)-6, Tumor Necrosis Factor α, TNFα) or chemokines (CCL2, CCL3, CCL4, CXCL10, CXCL11) by AEC stimulated with poly(I:C). Cytokine and chemokine production, except CXCL10, was Toll Like Receptor (TLR)-3 dependent and although they express TLR4, we found no cytokine production after LPS stimulation. Poly(I:C), but not LPS, synergised with TGF-β for the production of matrix metalloproteinase-9 (MMP-9) and fibronectin. Mechanistic analyses suggest the secretion of Wnt ligands by AEC along with a degradation of the cellular junctions after poly(I:C) exposure, leading to the release of β-catenin from the cell membrane and stimulation of the Wnt/β-catenin pathway. Conclusion Our results highlight the cross talk between TGF-β and TLR signaling in bronchial epithelium and its impact on the remodeling process. Electronic supplementary material The online version of this article (10.1186/s12931-017-0690-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- P-J Royer
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France.
| | - K Henrio
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - M Pain
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - J Loy
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - A Roux
- Hopital Foch, Pneumology, Adult Cystic Fibrosis Center and Lung Transplantation Department,Suresnes, France, Université Versailles Saint-Quentin-en-Yvelines, UPRESS EA220, Montigny le Bretonneux, Grenoble, France
| | - A Tissot
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - P Lacoste
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | - C Pison
- Clinique Universitaire de Pneumologie, Pôle Thorax et Vaisseaux, CHU Grenoble Alpes, Grenoble, France.,Université Grenoble Alpes, Grenoble, France.,Laboratoire de Bioénergétique Fondamentale et Appliquée, Inserm, 1055, Grenoble, France
| | - S Brouard
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,Faculté de Médecine, Université de Nantes, Nantes, France.,CIC Biotherapy, CHU Nantes, Nantes, France
| | - A Magnan
- UMR_S 1087 CNRS UMR_6291, l'Institut du Thorax, Université de Nantes, CHU de Nantes, Centre National de Référence Mucoviscidose Nantes-Roscoff, Nantes, France
| | | |
Collapse
|
105
|
Lee YH, Huang JH, Chang TH, Yang HC, Wu-Hsieh BA. Mucosa-Associated Lymphoid Tissue Lymphoma Translocation Protein 1 Positively Modulates Matrix Metalloproteinase-9 Production in Alveolar Macrophages upon Toll-Like Receptor 7 Signaling and Influenza Virus Infection. Front Immunol 2017; 8:1177. [PMID: 29018444 PMCID: PMC5614920 DOI: 10.3389/fimmu.2017.01177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/06/2017] [Indexed: 01/14/2023] Open
Abstract
Influenza A virus (IAV) infection causes significant morbidity and mortality worldwide. Matrix metalloproteinase-9 (MMP-9) degrades extracellular matrix and is involved in the pathology of influenza. It has been reported that MMP-9 mediates neutrophil migration in IAV infection. Whether alveolar macrophages, the first immune cells that encounter IAV, produce MMP-9, and the mechanism of its regulation have never been investigated. As Toll-like receptor 7 (TLR7) is one of the receptors in innate immune cells that recognize IAV, we used TLR7 agonists and IAV to stimulate alveolar macrophage MH-S cells, primary macrophages, and bone marrow neutrophils. Results showed that MMP-9 expression in macrophages is inducible by TLR7 agonists and IAV, yet, MMP-9 production by neutrophils is not inducible by either one of them. We hypothesized that MMP-9 production in macrophages is mediated through TLR7-NF-κB pathway and used microarray to analyze TLR7 agonist-induced NF-κB-related genes. Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1), a positive regulator of NF-κB, is amongst the top highly induced genes. By use of MALT1 inhibitor (z-VRPR-fmk) and alveolar macrophages from MALT1-deficient mice, we found that MMP-9 production is MALT1-dependent. While MALT1 can act as a paracaspase in lymphocytes through degrading various signaling proteins, we discovered that MALT1 functions to reduce a negative regulator of NF-κB, cylindromatosis (CYLD), in alveolar macrophages. IAV-induced MMP-9, TNF, and IL-6 in lungs of MALT1-deficient mice are significantly lower than in wild-type mice after intratracheal infection. MALT1-deficient mice also have less body weight loss and longer survival after infection. Taken together, we demonstrated a novel role of MALT1 in regulating alveolar macrophage MMP-9 production whose presence exacerbates the severity of influenza.
Collapse
Affiliation(s)
- Yu-Hsiang Lee
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Juin-Hua Huang
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tzu-Hsuan Chang
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Chih Yang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Betty A Wu-Hsieh
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
106
|
Cannabinoid Receptor 2 Modulates Neutrophil Recruitment in a Murine Model of Endotoxemia. Mediators Inflamm 2017; 2017:4315412. [PMID: 28852269 PMCID: PMC5567445 DOI: 10.1155/2017/4315412] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 12/25/2022] Open
Abstract
The endocannabinoid system consists of endogenous lipid mediators and cannabinoid receptors (CB) 1 and 2. It has previously been demonstrated that activation of the leukocyte-expressed CB2 has anti-inflammatory effects in vivo. Here, we report its role under baseline conditions and in a model of low-dose endotoxemia by comparing CB2 knockout to littermate control mice. CB2-deficient mice displayed significantly more neutrophils and fewer monocytes in the bone marrow under steady state. In initial validation experiments, administration of 1 mg/kg LPS to male C57BL/6J mice was shown to transiently upregulate systemic proinflammatory mediators (peaked at 2 hours) and mobilise bone marrow neutrophils and monocytes into circulation. In CB2 knockout mice, the level of the metalloproteinase MMP-9 was significantly elevated by 2 hours and we also observed augmented recruitment of neutrophils to the spleen in addition to increased levels of Ccl2, Ccl3, Cxcl10, and Il6. Collectively, our data show that the absence of CB2 receptor increases the levels of innate immune cell populations in the bone marrow under steady state. Furthermore, during an acute systemic inflammatory insult, we observe a highly reproducible and site-specific increase in neutrophil recruitment and proinflammatory chemokine expression in the spleen of CB2 knockout mice.
Collapse
|
107
|
Abstract
Myeloid cells have diverse roles in regulating immunity, inflammation, and extracellular matrix turnover. To accomplish these tasks, myeloid cells carry an arsenal of metalloproteinases, which include the matrix metalloproteinases and the adamalysins. These enzymes have diverse substrate repertoires, and are thus involved in mediating proteolytic cascades, cell migration, and cell signaling. Dysregulation of metalloproteinases contributes to pathogenic processes, including inflammation, fibrosis, and cancer. Metalloproteinases also have important nonproteolytic functions in controlling cytoskeletal dynamics during macrophage fusion and enhancing transcription to promote antiviral immunity. This review highlights the diverse contributions of metalloproteinases to myeloid cell functions.
Collapse
|
108
|
Álvares PR, Arruda JAAD, Silva LPD, Nascimento GJFD, Silveira MFD, Sobral APV. Immunohistochemical expression of TGF-β1 and MMP-9 in periapical lesions. Braz Oral Res 2017; 31:e51. [PMID: 28678970 DOI: 10.1590/1807-3107bor-2017.vol31.0051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/21/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of this study was to evaluate the expression of matrix metalloproteinase 9 (MMP-9) and transforming growth factor beta (TGF-β1) in periapical lesion samples correlated with the intensity of the inflammatory infiltrate and thickness of the epithelial lining. Forty-five cases of periapical lesions (23 periapical granulomas and 22 radicular cysts) were subjected to morphological and immunohistochemical analyses using anti-MMP-9 and anti-TGF-β1 antibodies. The data were analyzed using the following tests: non-parametric Mann-Whitney, chi-square, Fisher's exact test and Spearman's correlation test (P<0.05). Analysis of inflammatory infiltrate revealed that 78% of periapical granulomas presented infiltrate grade III, in contrast with 32% of radicular cysts (P<0.001). Morphological evaluation of the epithelial thickness in radicular cysts revealed the presence of atrophic epithelium in 86% of the cysts. The immunostaining of MMP-9 was score 2 in 67% of the granulomas and 77% of the cysts. Both lesions were predominantly score 1 for TGF-β1. Significant differences were confirmed between the expression scores of TGF-β1 and MMP-9 in periapical granulomas (p = 0.004) and in radicular cysts (p < 0.001). Expression of TGF-β1 was different for periapical granulomas and radicular cysts. This immunoregulatory cytokine seems more representative in asymptomatic lesions. The extracellular matrix remodeling process dependent on MMP-9 seems to be similar for both periapical granulomas and radicular cysts. TGF-β1 and MMP-9 may play an important role in the maintenance of periapical lesions.
Collapse
Affiliation(s)
- Pâmella Recco Álvares
- Universidade de Pernambuco - UPE, School of Dentistry, Department of Oral Pathology, Camaragibe, PB, Brazil
| | | | - Leorik Pereira da Silva
- Universidade Federal do Rio Grande do Norte - UFRN, School of Dentistry, Department of Oral Pathology, Natal, RN, Brazil
| | | | - Maria Fonseca da Silveira
- Universidade de Pernambuco - UPE, School of Dentistry, Department of Stomatology, Camaragibe, PB, Brazil
| | - Ana Paula Veras Sobral
- Universidade de Pernambuco - UPE, School of Dentistry, Department of Oral Pathology, Camaragibe, PB, Brazil
| |
Collapse
|
109
|
Weiss G, Lai C, Fife ME, Grabiec AM, Tildy B, Snelgrove RJ, Xin G, Lloyd CM, Hussell T. Reversal of TREM-1 ectodomain shedding and improved bacterial clearance by intranasal metalloproteinase inhibitors. Mucosal Immunol 2017; 10:1021-1030. [PMID: 27966555 DOI: 10.1038/mi.2016.104] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 10/07/2016] [Indexed: 02/04/2023]
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is expressed on neutrophils and monocyte/macrophages and amplifies Toll-like receptor-mediated inflammation during infection. TREM-1 also exists in an antagonistic soluble form (sTREM-1) that has been used as a peripheral biomarker in sepsis, though the mechanisms of its release are not entirely clear. The requirement of TREM-1 in single microbial infections is controversial, with some studies showing a protective role and others a contribution to immunopathology. Furthermore, the role of membrane-bound and sTREM-1 in polygenic infections is currently unknown. In a mouse co-infection model where preceding viral infection greatly enhances bacteria co-infection, we now determine a mechanisms for the striking increase in sTREM-1 and the loss of TREM-1 on surface of neutrophils. We identified a matrix metalloproteinase (MMP)-9 cleavage site in TREM-1 and that the increase of MMP-9 in bronchoalveolar lavage fluid mirrors sTREM-1 release. In vitro studies with neutrophils and MMP-9 and the reduction of sTREM-1 in vivo after MMP-9 inhibition verifies that this enzyme cleaves TREM-1. Intriguingly, MMP-9 inhibition significantly reduces bacterial load and ensuing immunopathology in a co-infection model. This highlights MMP-9 inhibition as a potential therapeutic via blocking cleavage of TREM-1.
Collapse
Affiliation(s)
- G Weiss
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
| | - C Lai
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
| | - M E Fife
- Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester, UK
| | - A M Grabiec
- Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester, UK
| | - B Tildy
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
| | - R J Snelgrove
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
| | - G Xin
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
| | - C M Lloyd
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
| | - T Hussell
- National Heart and Lung Institute, Department of Inflammation, Development &Repair, Imperial College London, London, UK
- Manchester Collaborative Centre for Inflammation Research (MCCIR), Manchester, UK
| |
Collapse
|
110
|
Levin M, Udi Y, Solomonov I, Sagi I. Next generation matrix metalloproteinase inhibitors - Novel strategies bring new prospects. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017. [PMID: 28636874 DOI: 10.1016/j.bbamcr.2017.06.009] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Enzymatic proteolysis of cell surface proteins and extracellular matrix (ECM) is critical for tissue homeostasis and cell signaling. These proteolytic activities are mediated predominantly by a family of proteases termed matrix metalloproteinases (MMPs). The growing evidence in recent years that ECM and non-ECM bioactive molecules (e.g., growth factors, cytokines, chemokines, on top of matrikines and matricryptins) have versatile functions redefines our view on the roles matrix remodeling enzymes play in many physiological and pathological processes, and underscores the notion that ECM proteolytic reaction mechanisms represent master switches in the regulation of critical biological processes and govern cell behavior. Accordingly, MMPs are not only responsible for direct degradation of ECM molecules but are also key modulators of cardinal bioactive factors. Many attempts were made to manipulate ECM degradation by targeting MMPs using small peptidic and organic inhibitors. However, due to the high structural homology shared by these enzymes, the majority of the developed compounds are broad-spectrum inhibitors affecting the proteolytic activity of various MMPs and other zinc-related proteases. These inhibitors, in many cases, failed as therapeutic agents, mainly due to the bilateral role of MMPs in pathological conditions such as cancer, in which MMPs have both pro- and anti-tumorigenic effects. Despite the important role of MMPs in many human diseases, none of the broad-range synthetic MMP inhibitors that were designed have successfully passed clinical trials. It appears that, designing highly selective MMP inhibitors that are also effective in vivo, is not trivial. The challenges related to designing selective and effective metalloprotease inhibitors, are associated in part with the aforesaid high structural homology and the dynamic nature of their protein scaffolds. Great progress was achieved in the last decade in understanding the biochemistry and biology of MMPs activity. This knowledge, combined with lessons from the past has drawn new "boundaries" for the development of the next-generation MMP inhibitors. These novel agents are currently designed to be highly specific, capable to discriminate between the homologous MMPs and ideally administered as a short-term topical treatment. In this review we discuss the latest progress in the fields of MMP inhibitors in terms of structure, function and their specific activity. The development of novel highly specific inhibitors targeting MMPs paves the path to study complex biological processes associated with ECM proteolysis in health and disease. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.
Collapse
Affiliation(s)
- Maxim Levin
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yael Udi
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY 10065, USA
| | - Inna Solomonov
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
111
|
Sekelova Z, Stepanova H, Polansky O, Varmuzova K, Faldynova M, Fedr R, Rychlik I, Vlasatikova L. Differential protein expression in chicken macrophages and heterophils in vivo following infection with Salmonella Enteritidis. Vet Res 2017. [PMID: 28623956 PMCID: PMC5473982 DOI: 10.1186/s13567-017-0439-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In this study we compared the proteomes of macrophages and heterophils isolated from the spleen 4 days after intravenous infection of chickens with Salmonella Enteritidis. Heterophils were characterized by expression of MMP9, MRP126, LECT2, CATHL1, CATHL2, CATHL3, LYG2, LYZ and RSFR. Macrophages specifically expressed receptor proteins, e.g. MRC1L, LRP1, LGALS1, LRPAP1 and a DMBT1L. Following infection, heterophils decreased ALB and FN1, and released MMP9 to enable their translocation to the site of infection. In addition, the endoplasmic reticulum proteins increased in heterophils which resulted in the release of granular proteins. Since transcription of genes encoding granular proteins did not decrease, these genes remained continuously transcribed and translated even after initial degranulation. Macrophages increased amounts of fatty acid elongation pathway proteins, lysosomal and phagosomal proteins. Macrophages were less responsive to acute infection than heterophils and an increase in proteins like CATHL1, CATHL2, RSFR, LECT2 and GAL1 in the absence of any change in their expression at RNA level could even be explained by capturing these proteins from the external environment into which these could have been released by heterophils.
Collapse
Affiliation(s)
- Zuzana Sekelova
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | - Hana Stepanova
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | - Ondrej Polansky
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | | | - Marcela Faldynova
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| | - Radek Fedr
- Department of Cytokinetics, Institute of Biophysics of the CAS, Kralovopolska 135, 612 65, Brno, Czech Republic.,Center of Biomolecular and Cellular Engineering, International, Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, 656 91, Brno, Czech Republic
| | - Ivan Rychlik
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic.
| | - Lenka Vlasatikova
- Veterinary Research Institute, Hudcova 70, 621 00, Brno, Czech Republic
| |
Collapse
|
112
|
Camp JV, Jonsson CB. A Role for Neutrophils in Viral Respiratory Disease. Front Immunol 2017; 8:550. [PMID: 28553293 PMCID: PMC5427094 DOI: 10.3389/fimmu.2017.00550] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 04/24/2017] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are immune cells that are well known to be present during many types of lung diseases associated with acute respiratory distress syndrome (ARDS) and may contribute to acute lung injury. Neutrophils are poorly studied with respect to viral infection, and specifically to respiratory viral disease. Influenza A virus (IAV) infection is the cause of a respiratory disease that poses a significant global public health concern. Influenza disease presents as a relatively mild and self-limiting although highly pathogenic forms exist. Neutrophils increase in the respiratory tract during infection with mild seasonal IAV, moderate and severe epidemic IAV infection, and emerging highly pathogenic avian influenza (HPAI). During severe influenza pneumonia and HPAI infection, the number of neutrophils in the lower respiratory tract is correlated with disease severity. Thus, comparative analyses of the relationship between IAV infection and neutrophils provide insights into the relative contribution of host and viral factors that contribute to disease severity. Herein, we review the contribution of neutrophils to IAV disease pathogenesis and to other respiratory virus infections.
Collapse
Affiliation(s)
- Jeremy V Camp
- Institute of Virology, University of Veterinary Medicine at Vienna, Vienna, Austria
| | - Colleen B Jonsson
- Department of Microbiology, University of Tennessee-Knoxville, Knoxville, TN, USA
| |
Collapse
|
113
|
Abstract
Activated microglial cells are present in dengue virus (DENV)-infected brains; however, the possible effects of DENV on microglia remain unclear. Here, we demonstrated DENV caused infection, including viral entry, RNA replication, viral protein expression, and virus release, in the murine microglial cell line BV2. DENV infection caused an increase in the formation of the multipolar phenotype in vitro and in vivo without affecting cell growth and cytotoxicity. DENV infection considerably increased cell motility and disrupting either actin filaments or clathrin retarded such effect. Increase in cell migration was only occurred by DENV infection following a clathrin-regulated endocytosis of DENV entry. Ultraviolet-inactivated DENV did not affect cell migration, and pharmacologically blocking toll-like receptor (TLR) 3 and TLR3-related signaling pathways reduced the DENV-induced increase in cell migration. These results demonstrate an advanced effect of DENV infection on microglial migration via a mechanism involving viral entry, RNA release, and TLR3 signal activation.
Collapse
|
114
|
Agraz-Cibrian JM, Giraldo DM, Mary FM, Urcuqui-Inchima S. Understanding the molecular mechanisms of NETs and their role in antiviral innate immunity. Virus Res 2016; 228:124-133. [PMID: 27923601 DOI: 10.1016/j.virusres.2016.11.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 01/09/2023]
Abstract
Polymorphonuclear neutrophils (PMNs) are the most abundant cells in the context of innate immunity; they are one of the first cells to arrive at the site of viral infection constituting the first line of defense in response to invading pathogens. Indeed, neutrophils are provided with several defense mechanisms including release of cytokines, cytotoxic granules and the last recently described neutrophil extracellular traps (NETs). The main components of NETs are DNA, granular antimicrobial peptides, and nuclear and cytoplasmic proteins, that together play an important role in the innate immune response. While NETs were first described as a mechanism against bacteria and fungi, recently, several studies are beginning to elucidate how NETs are involved in the host antiviral response and the prominent characteristics of this new mechanism are discussed in the present review.
Collapse
Affiliation(s)
- Juan Manuel Agraz-Cibrian
- Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, Mexico.
| | - Diana M Giraldo
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Fafutis-Morris Mary
- Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
115
|
Cohen TS. Role of MicroRNA in the Lung's Innate Immune Response. J Innate Immun 2016; 9:243-249. [PMID: 27915347 DOI: 10.1159/000452669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 10/18/2016] [Indexed: 12/22/2022] Open
Abstract
The immune response to respiratory pathogens must be robust enough to defend the host yet properly constrained such that inflammation-induced tissue damage is avoided. MicroRNA (miRNA) are small noncoding RNA which posttranscriptionally influence gene expression. In this review, we discuss recent experimental evidence of the contribution of miRNA to the lung's response to bacterial and viral pathogens.
Collapse
Affiliation(s)
- Taylor S Cohen
- Department of Infectious Disease, Medimmune, Gaithersburg, MD, USA
| |
Collapse
|
116
|
Abstract
The respiratory immune response consists of multiple tiers of cellular responses that are engaged in a sequential manner in order to control infections. The stepwise engagement of effector functions with progressively increasing host fitness costs limits tissue damage. In addition, specific mechanisms are in place to promote disease tolerance in response to respiratory infections. Environmental factors, obesity and the ageing process can alter the efficiency and regulation of this tiered response, increasing pathology and mortality as a result. In this Review, we describe the cell types that coordinate pathogen clearance and tissue repair through the serial secretion of cytokines, and discuss how the environment and comorbidity influence this response.
Collapse
|
117
|
McMahon M, Ye S, Izzard L, Dlugolenski D, Tripp RA, Bean AGD, McCulloch DR, Stambas J. ADAMTS5 Is a Critical Regulator of Virus-Specific T Cell Immunity. PLoS Biol 2016; 14:e1002580. [PMID: 27855162 PMCID: PMC5113859 DOI: 10.1371/journal.pbio.1002580] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 10/25/2016] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix (ECM) provides physical scaffolding for cellular constituents and initiates biochemical and biomechanical cues that are required for physiological activity of living tissues. The ECM enzyme ADAMTS5, a member of the ADAMTS (A Disintegrin-like and Metalloproteinase with Thrombospondin-1 motifs) protein family, cleaves large proteoglycans such as aggrecan, leading to the destruction of cartilage and osteoarthritis. However, its contribution to viral pathogenesis and immunity is currently undefined. Here, we use a combination of in vitro and in vivo models to show that ADAMTS5 enzymatic activity plays a key role in the development of influenza-specific immunity. Influenza virus infection of Adamts5-/- mice resulted in delayed virus clearance, compromised T cell migration and immunity and accumulation of versican, an ADAMTS5 proteoglycan substrate. Our research emphasises the importance of ADAMTS5 expression in the control of influenza virus infection and highlights the potential for development of ADAMTS5-based therapeutic strategies to reduce morbidity and mortality. The extracellular matrix enzyme ADAMTS5 enhances the clearance of viruses by facilitating migration of T lymphocytes to the periphery following influenza virus infection. Movement of immune cells is critical for effective clearance of pathogens. The response to influenza virus infection requires immune cell trafficking between the lung, mediastinal lymph node and other peripheral lymphoid organs such as the spleen. We set out to assess the contribution of a specific extracellular matrix enzyme, ADAMTS5, to migration of lymphocytes and overall pathogenesis following infection. In our studies, we demonstrate that mice lacking Adamts5 have fewer influenza-specific lymphocytes in the lung and spleen following infection. These observations correlated with an accumulation of influenza-specific lymphocytes in the mediastinal lymph node and increased virus titres. This work suggests that ADAMTS5 is necessary for immune cell migration to the periphery, where lymphocyte function is required to fight infection.
Collapse
Affiliation(s)
- Meagan McMahon
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Siying Ye
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | - Leonard Izzard
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
| | | | - Ralph A. Tripp
- College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Andrew G. D. Bean
- Australian Animal Health Laboratory, CSIRO, East Geelong, Victoria, Australia
| | | | - John Stambas
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia
- * E-mail:
| |
Collapse
|
118
|
Vidaña B, Martínez J, Martorell J, Montoya M, Córdoba L, Pérez M, Majó N. Involvement of the different lung compartments in the pathogenesis of pH1N1 influenza virus infection in ferrets. Vet Res 2016; 47:113. [PMID: 27825367 PMCID: PMC5101722 DOI: 10.1186/s13567-016-0395-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/10/2016] [Indexed: 12/29/2022] Open
Abstract
Severe cases after pH1N1 infection are consequence of interstitial pneumonia triggered by alveolar viral replication and an exacerbated host immune response, characterized by the up-regulation of pro-inflammatory cytokines and the influx of inflammatory leukocytes to the lungs. Different lung cell populations have been suggested as culprits in the unregulated innate immune responses observed in these cases. This study aims to clarify this question by studying the different induction of innate immune molecules by the distinct lung anatomic compartments (vascular, alveolar and bronchiolar) of ferrets intratracheally infected with a human pH1N1 viral isolate, by means of laser microdissection techniques. The obtained results were then analysed in relation to viral quantification in the different anatomic areas and the histopathological lesions observed. More severe lung lesions were observed at 24 h post infection (hpi) correlating with viral antigen detection in bronchiolar and alveolar epithelial cells. However, high levels of viral RNA were detected in all anatomic compartments throughout infection. Bronchiolar areas were the first source of IFN-α and most pro-inflammatory cytokines, through the activation of RIG-I. In contrast, vascular areas contributed with the highest induction of CCL2 and other pro-inflammatory cytokines, through the activation of TLR3.
Collapse
Affiliation(s)
- Beatriz Vidaña
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Jorge Martínez
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain. .,UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.
| | - Jaime Martorell
- Departament de Medicina i Cirurgia Animals, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - María Montoya
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Lorena Córdoba
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mónica Pérez
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Natàlia Majó
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
119
|
Vidy A, Maisonnasse P, Da Costa B, Delmas B, Chevalier C, Le Goffic R. The Influenza Virus Protein PB1-F2 Increases Viral Pathogenesis through Neutrophil Recruitment and NK Cells Inhibition. PLoS One 2016; 11:e0165361. [PMID: 27798704 PMCID: PMC5087861 DOI: 10.1371/journal.pone.0165361] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022] Open
Abstract
The influenza A virus (IAV) PB1-F2 protein is a virulence factor contributing to the pathogenesis observed during IAV infections in mammals. In this study, using a mouse model, we compared the host response associated with PB1-F2 with an early transcriptomic signature that was previously associated with neutrophils and consecutively fatal IAV infections. This allowed us to show that PB1-F2 is partly involved in neutrophil-related mechanisms leading to death. Using neutropenic mice, we confirmed that the harmful effect of PB1-F2 is due to an excessive inflammation mediated by an increased neutrophil mobilization. We identified the downstream effects of this PB1-F2-exacerbated neutrophil recruitment. PB1-F2 had no impact on the lymphocyte recruitment in the airways at day 8 pi. However, functional genomics analysis and flow cytometry in broncho-alveolar lavages at 4 days pi revealed that PB1-F2 induced a NK cells deficiency. Thus, our results identify PB1-F2 as an important immune disruptive factor during the IAV infection.
Collapse
Affiliation(s)
- Aurore Vidy
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | | | - Bruno Da Costa
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Bernard Delmas
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | | | - Ronan Le Goffic
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- * E-mail:
| |
Collapse
|
120
|
Duan M, Hibbs ML, Chen W. The contributions of lung macrophage and monocyte heterogeneity to influenza pathogenesis. Immunol Cell Biol 2016; 95:225-235. [DOI: 10.1038/icb.2016.97] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/15/2016] [Accepted: 09/20/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Mubing Duan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne Victoria Australia
| | - Margaret L Hibbs
- Department of Immunology and Pathology, Monash University, Alfred Medical Research and Education Precinct, 89 Commercial Rd Melbourne Victoria Australia
| | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne Victoria Australia
| |
Collapse
|
121
|
Talmi-Frank D, Altboum Z, Solomonov I, Udi Y, Jaitin D, Klepfish M, David E, Zhuravlev A, Keren-Shaul H, Winter D, Gat-Viks I, Mandelboim M, Ziv T, Amit I, Sagi I. Extracellular Matrix Proteolysis by MT1-MMP Contributes to Influenza-Related Tissue Damage and Mortality. Cell Host Microbe 2016; 20:458-470. [DOI: 10.1016/j.chom.2016.09.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 06/19/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022]
|
122
|
Pleiotropic regulations of neutrophil receptors response to sepsis. Inflamm Res 2016; 66:197-207. [DOI: 10.1007/s00011-016-0993-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/24/2016] [Accepted: 09/20/2016] [Indexed: 12/21/2022] Open
|
123
|
Genital Chlamydia trachomatis: understanding the roles of innate and adaptive immunity in vaccine research. Clin Microbiol Rev 2016; 27:346-70. [PMID: 24696438 DOI: 10.1128/cmr.00105-13] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted disease worldwide, and despite significant advances in chlamydial research, a prophylactic vaccine has yet to be developed. This Gram-negative obligate intracellular bacterium, which often causes asymptomatic infection, may cause pelvic inflammatory disease (PID), ectopic pregnancies, scarring of the fallopian tubes, miscarriage, and infertility when left untreated. In the genital tract, Chlamydia trachomatis infects primarily epithelial cells and requires Th1 immunity for optimal clearance. This review first focuses on the immune cells important in a chlamydial infection. Second, we summarize the research and challenges associated with developing a chlamydial vaccine that elicits a protective Th1-mediated immune response without inducing adverse immunopathologies.
Collapse
|
124
|
The origin of Pasteurella multocida impacts pathology and inflammation when assessed in a mouse model. Res Vet Sci 2016; 105:139-42. [DOI: 10.1016/j.rvsc.2016.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
|
125
|
Oh DY, Dowling DJ, Ahmed S, Choi H, Brightman S, Bergelson I, Berger ST, Sauld JF, Pettengill M, Kho AT, Pollack HJ, Steen H, Levy O. Adjuvant-induced Human Monocyte Secretome Profiles Reveal Adjuvant- and Age-specific Protein Signatures. Mol Cell Proteomics 2016; 15:1877-94. [PMID: 26933193 PMCID: PMC5083103 DOI: 10.1074/mcp.m115.055541] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Indexed: 12/16/2022] Open
Abstract
Adjuvants boost vaccine responses, enhancing protective immunity against infections that are most common among the very young. Many adjuvants activate innate immunity, some via Toll-Like Receptors (TLRs), whose activities varies with age. Accordingly, characterization of age-specific adjuvant-induced immune responses may inform rational adjuvant design targeting vulnerable populations. In this study, we employed proteomics to characterize the adjuvant-induced changes of secretomes from human newborn and adult monocytes in response to Alum, the most commonly used adjuvant in licensed vaccines; Monophosphoryl Lipid A (MPLA), a TLR4-activating adjuvant component of a licensed Human Papilloma Virus vaccine; and R848 an imidazoquinoline TLR7/8 agonist that is a candidate adjuvant for early life vaccines. Monocytes were incubated in vitro for 24 h with vehicle, Alum, MPLA, or R848 and supernatants collected for proteomic analysis employing liquid chromatography-mass spectrometry (LC-MS) (data available via ProteomeXchange, ID PXD003534). 1894 non-redundant proteins were identified, of which ∼30 - 40% were common to all treatment conditions and ∼5% were treatment-specific. Adjuvant-stimulated secretome profiles, as identified by cluster analyses of over-represented proteins, varied with age and adjuvant type. Adjuvants, especially Alum, activated multiple innate immune pathways as assessed by functional enrichment analyses. Release of lactoferrin, pentraxin 3, and matrix metalloproteinase-9 was confirmed in newborn and adult whole blood and blood monocytes stimulated with adjuvants alone or adjuvanted licensed vaccines with distinct clinical reactogenicity profiles. MPLA-induced adult monocyte secretome profiles correlated in silico with transcriptome profiles induced in adults immunized with the MPLA-adjuvanted RTS,S malaria vaccine (Mosquirix™). Overall, adjuvants such as Alum, MPLA and R848 give rise to distinct and age-specific monocyte secretome profiles, paralleling responses to adjuvant-containing vaccines in vivo. Age-specific in vitro modeling coupled with proteomics may provide fresh insight into the ontogeny of adjuvant action thereby informing targeted adjuvanted vaccine development for distinct age groups.
Collapse
Affiliation(s)
- Djin-Ye Oh
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts; ¶Division of Pediatric Infectious Diseases, New York University Medical School, New York; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| | - David J Dowling
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts
| | - Saima Ahmed
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hyungwon Choi
- **Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Spencer Brightman
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and
| | - Ilana Bergelson
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and
| | - Sebastian T Berger
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - John F Sauld
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Matthew Pettengill
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| | - Alvin T Kho
- ‡‡Children's Hospital Informatics Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Henry J Pollack
- ¶Division of Pediatric Infectious Diseases, New York University Medical School, New York
| | - Hanno Steen
- ‖Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| | - Ofer Levy
- From the ‡Department of Medicine, Division of Infectious Diseases, Boston Children's Hospital and §Harvard Medical School, Boston, Massachusetts; Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
126
|
Supasorn O, Sringkarin N, Srimanote P, Angkasekwinai P. Matrix metalloproteinases contribute to the regulation of chemokine expression and pulmonary inflammation in Cryptococcus infection. Clin Exp Immunol 2015; 183:431-40. [PMID: 26445891 DOI: 10.1111/cei.12725] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2015] [Indexed: 12/28/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of extracellular proteases that play roles in regulating the immune response in inflammatory processes. Previous studies indicated that different MMPs were involved in the host defence and tissue damage in response to different pathogens. However, the contributions of MMPs during Cryptococcus infection have not been addressed clearly. Here, we examined the expression and activity of MMPs during Cryptococcus infection. Among MMP family members, we found significant increases of MMP-3 and MMP-12 mRNA levels and MMP12 zymographic activities in response to C. neoformans but not C. gattii infection. The expression of MMP12 was induced in RAW cells after C. neoformans treatment and in alveolar macrophages purified from C. neoformans-infected mice. Interestingly, administration of MMP inhibitor GM6001 into C. neoformans-infected mice resulted in a significantly increased pulmonary fungal burden with attenuated inflammatory cell infiltration. Corresponding to this finding, the expression of the macrophage- and neutrophil-attracting chemokines CCL2 and CXCL1 was inhibited in the GM6001-treated group and MMP12 levels were found to be correlated strongly with CCL2 mRNA expression. Thus, our data suggest that the induction of MMPs by C. neoformans infection potentiates inflammatory cell infiltration by modulating pulmonary chemokines, thereby promoting effective host immunity to pulmonary Cryptococcus infection.
Collapse
Affiliation(s)
- O Supasorn
- Department of Medical Technology, Faculty of Allied Health Sciences.,Graduate Program in Biomedical Science, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand
| | - N Sringkarin
- Graduate Program in Medical Technology, Faculty of Allied Health Sciences
| | - P Srimanote
- Graduate Program in Biomedical Science, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand
| | - P Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences.,Graduate Program in Medical Technology, Faculty of Allied Health Sciences
| |
Collapse
|
127
|
Lim K, Hyun YM, Lambert-Emo K, Capece T, Bae S, Miller R, Topham DJ, Kim M. Neutrophil trails guide influenza-specific CD8⁺ T cells in the airways. Science 2015; 349:aaa4352. [PMID: 26339033 DOI: 10.1126/science.aaa4352] [Citation(s) in RCA: 291] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
During viral infections, chemokines guide activated effector T cells to infection sites. However, the cells responsible for producing these chemokines and how such chemokines recruit T cells are unknown. Here, we show that the early recruitment of neutrophils into influenza-infected trachea is essential for CD8(+) T cell-mediated immune protection in mice. We observed that migrating neutrophils leave behind long-lasting trails that are enriched in the chemokine CXCL12. Experiments with granulocyte-specific CXCL12 conditionally depleted mice and a CXCR4 antagonist revealed that CXCL12 derived from neutrophil trails is critical for virus-specific CD8(+) T cell recruitment and effector functions. Collectively, these results suggest that neutrophils deposit long-lasting, chemokine-containing trails, which may provide both chemotactic and haptotactic cues for efficient CD8(+) T cell migration and localization in influenza-infected tissues.
Collapse
Affiliation(s)
- Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Young-Min Hyun
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Kris Lambert-Emo
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Tara Capece
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Seyeon Bae
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Richard Miller
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - David J Topham
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
128
|
Tang FSM, Van Ly D, Spann K, Reading PC, Burgess JK, Hartl D, Baines KJ, Oliver BG. Differential neutrophil activation in viral infections: Enhanced TLR-7/8-mediated CXCL8 release in asthma. Respirology 2015; 21:172-9. [PMID: 26477783 PMCID: PMC5324549 DOI: 10.1111/resp.12657] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/14/2015] [Accepted: 07/24/2015] [Indexed: 02/06/2023]
Abstract
Background and objective Respiratory viral infections are a major cause of asthma exacerbations. Neutrophils accumulate in the airways and the mechanisms that link neutrophilic inflammation, viral infections and exacerbations are unclear. This study aims to investigate anti‐viral responses in neutrophils from patients with and without asthma and to investigate if neutrophils can be directly activated by respiratory viruses. Methods Neutrophils from peripheral blood from asthmatic and non‐asthmatic individuals were isolated and stimulated with lipopolysaccharide (LPS) (1 μg/mL), f‐met‐leu‐phe (fMLP) (100 nM), imiquimod (3 μg/mL), R848 (1.5 μg/mL), poly I:C (10 μg/mL), RV16 (multiplicity of infection (MOI)1), respiratory syncytial virus (RSV) (MOI1) or influenza virus (MOI1). Cell‐free supernatants were collected after 1 h of neutrophil elastase (NE) and matrix metalloproteinase (MMP)‐9 release, or after 24 h for CXCL8 release. Results LPS, fMLP, imiquimod and R848 stimulated the release of CXCL8, NE and MMP‐9 whereas poly I:C selectively induced CXCL8 release only. R848‐induced CXCL8 release was enhanced in neutrophils from asthmatics compared with non‐asthmatic cells (P < 0.01). RSV triggered the release of CXCL8 and NE from neutrophils, whereas RV16 or influenza had no effect. Conclusion Neutrophils release CXCL8, NE and MMP‐9 in response to viral surrogates with R848‐induced CXCL8 release being specifically enhanced in asthmatic neutrophils. Toll‐like receptor (TLR7/8) dysregulation may play a role in neutrophilic inflammation in viral‐induced exacerbations. We aimed to investigate and compare neutrophil responses to bacterial compounds and viral mimetics as well as compare responses between people with and without asthma. We also investigated neutrophil responses to live respiratory viruses. Here we provide a novel comprehensive comparison showing differential and specific activation in innate immune cells. See Editorial, page 10
Collapse
Affiliation(s)
- Francesca S M Tang
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine, The University of Sydney, Sydney, New South Wales, Australia
| | - David Van Ly
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Genome Integrity Group, The Children's Medical Research Institute, Sydney, New South Wales, Australia
| | - Kirsten Spann
- School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Patrick C Reading
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Janette K Burgess
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Pharmacology, School of Medical Sciences, Faculty of Medicine, The University of Sydney, Sydney, New South Wales, Australia
| | - Dominik Hartl
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Katherine J Baines
- Priority Research Centre for Asthma and Respiratory Disease, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Brian G Oliver
- Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
129
|
Conejeros I, Gibson AJ, Werling D, Muñoz-Caro T, Hermosilla C, Taubert A, Burgos RA. Effect of the synthetic Toll-like receptor ligands LPS, Pam3CSK4, HKLM and FSL-1 in the function of bovine polymorphonuclear neutrophils. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 52:215-225. [PMID: 26026246 DOI: 10.1016/j.dci.2015.05.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 05/23/2015] [Accepted: 05/23/2015] [Indexed: 06/04/2023]
Abstract
Toll-like receptors (TLR) are a family of pattern recognition receptors that sense microbial associated molecular patterns (MAMP) such as microbial membrane components and nucleic acids of bacterial origin. Polymorphonuclear neutrophils (PMN) are the first cell of the innate immune system to arrive at the site of infection or injury and elicit oxidative and non-oxidative microbicidal mechanisms. Observations in human and mouse suggest that TLR ligands can induce direct responses in PMN. So far, there is no information of the effect of synthetic TLR ligands on the response of bovine PMN. The objective of this study was to evaluate the functional response of bovine PMN incubated with four synthetic TLR ligands: ultrapure LPS (TLR4), Pam(3)CSK(4) (TLR2/1), HKLM (TLR2) and FSL-1 (TLR2/6). The results show that all the ligands increment cells size as identified by changes in the FSC-SSC as part of the flow cytometric analysis. Interestingly, only Pam(3)CSK(4) consistently induced a calcium influx, increased ROS production and secretion of gelatinase granules, whereas no response was seen using other ligands. Furthermore, exposure of bovine PMN to ultrapure LPS, Pam(3)CSK(4), HKLM or FSL-1 for 24 hours did not impact on apoptosis of these cells. Our data provide evidence for a selective response of bovine PMNs to TLR ligands.
Collapse
Affiliation(s)
- Iván Conejeros
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de Chile, Valdivia, Chile.
| | - Amanda J Gibson
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, Hatfield AL9 7TA, UK
| | - Dirk Werling
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hawkshead Lane, Hatfield AL9 7TA, UK
| | - Tamara Muñoz-Caro
- Institute of Parasitology, Justus Liebig University Giessen, Schubertstr. 81, BFS, 35392 Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Justus Liebig University Giessen, Schubertstr. 81, BFS, 35392 Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Justus Liebig University Giessen, Schubertstr. 81, BFS, 35392 Giessen, Germany
| | - Rafael A Burgos
- Laboratory of Inflammation Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
130
|
Lämmermann T. In the eye of the neutrophil swarm-navigation signals that bring neutrophils together in inflamed and infected tissues. J Leukoc Biol 2015; 100:55-63. [PMID: 26416718 DOI: 10.1189/jlb.1mr0915-403] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/08/2015] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are sentinel cells that express in higher vertebrates >30 chemokine and chemoattractant receptors to sense and quickly react to tissue damage signals. Intravital microscopy studies in mouse models of wounding, inflammation, and infection have revealed that neutrophils form cell swarms at local sites of tissue injury and cell death. This swarming response is choreographed by chemokines, lipids, and other chemoattractants, controlling sequential phases of highly coordinated chemotaxis, intercellular signal relay, and cluster formation among neutrophils. This review will give a brief overview about the basic principles and key molecules that have led to the refined multistep model of how neutrophils come together to isolate sites of tissue injury and microbial invasion from healthy tissue. Whereas auto- and paracrine signaling among neutrophils during later phases of swarming can provide a level of self-organization for robust navigation in diverse inflammatory settings, guidance factors from primary tissue lesions, resident bystander cells, and dying cells regulate the initial phases of the swarming response. This review will discuss how the specific environmental context and mixture of attractants at the locally inflamed site can lead to variants of the multistep attraction model and influence the extent of neutrophil swarming, ranging from accumulations of only few individual cells to the aggregation of several hundreds of neutrophils, as found in abscesses. Given the critical roles of neutrophils in both host protection and tissue destruction, novel insights on neutrophil swarming might provide useful for the therapeutic modulation of neutrophil-dependent inflammatory processes.
Collapse
Affiliation(s)
- Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| |
Collapse
|
131
|
Abstract
The heart pumps blood to maintain circulation and ensure the delivery of oxygenated blood to all the organs of the body. Mechanics play a critical role in governing and regulating heart function under both normal and pathological conditions. Biological processes and mechanical stress are coupled together in regulating myocyte function and extracellular matrix structure thus controlling heart function. Here, we offer a brief introduction to the biomechanics of left ventricular function and then summarize recent progress in the study of the effects of mechanical stress on ventricular wall remodeling and cardiac function as well as the effects of wall mechanical properties on cardiac function in normal and dysfunctional hearts. Various mechanical models to determine wall stress and cardiac function in normal and diseased hearts with both systolic and diastolic dysfunction are discussed. The results of these studies have enhanced our understanding of the biomechanical mechanism in the development and remodeling of normal and dysfunctional hearts. Biomechanics provide a tool to understand the mechanism of left ventricular remodeling in diastolic and systolic dysfunction and guidance in designing and developing new treatments.
Collapse
Affiliation(s)
- Andrew P. Voorhees
- Department of Mechanical Engineering, The University of Texas at San Antonio, Biomedical Engineering Program, UTSA-UTHSCSA
| | - Hai-Chao Han
- Department of Mechanical Engineering, The University of Texas at San Antonio, Biomedical Engineering Program, UTSA-UTHSCSA
| |
Collapse
|
132
|
Dabo AJ, Cummins N, Eden E, Geraghty P. Matrix Metalloproteinase 9 Exerts Antiviral Activity against Respiratory Syncytial Virus. PLoS One 2015; 10:e0135970. [PMID: 26284919 PMCID: PMC4540458 DOI: 10.1371/journal.pone.0135970] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/28/2015] [Indexed: 01/01/2023] Open
Abstract
Increased lung levels of matrix metalloproteinase 9 (MMP9) are frequently observed during respiratory syncytial virus (RSV) infection and elevated MMP9 concentrations are associated with severe disease. However little is known of the functional role of MMP9 during lung infection with RSV. To determine whether MMP9 exerted direct antiviral potential, active MMP9 was incubated with RSV, which showed that MMP9 directly prevented RSV infectivity to airway epithelial cells. Using knockout mice the effect of the loss of Mmp9 expression was examined during RSV infection to demonstrate MMP9’s role in viral clearance and disease progression. Seven days following RSV infection, Mmp9-/- mice displayed substantial weight loss, increased RSV-induced airway hyperresponsiveness (AHR) and reduced clearance of RSV from the lungs compared to wild type mice. Although total bronchoalveolar lavage fluid (BALF) cell counts were similar in both groups, neutrophil recruitment to the lungs during RSV infection was significantly reduced in Mmp9-/- mice. Reduced neutrophil recruitment coincided with diminished RANTES, IL-1β, SCF, G-CSF expression and p38 phosphorylation. Induction of p38 signaling was required for RANTES and G-CSF expression during RSV infection in airway epithelial cells. Therefore, MMP9 in RSV lung infection significantly enhances neutrophil recruitment, cytokine production and viral clearance while reducing AHR.
Collapse
Affiliation(s)
- Abdoulaye J. Dabo
- Mount Sinai St. Luke’s Medical Center, Mount Sinai Health System, Division of Pulmonary, Critical Care and Sleep Medicine, New York, NY, United States of America
| | - Neville Cummins
- Mount Sinai St. Luke’s Medical Center, Mount Sinai Health System, Division of Pulmonary, Critical Care and Sleep Medicine, New York, NY, United States of America
| | - Edward Eden
- Mount Sinai St. Luke’s Medical Center, Mount Sinai Health System, Division of Pulmonary, Critical Care and Sleep Medicine, New York, NY, United States of America
| | - Patrick Geraghty
- Mount Sinai St. Luke’s Medical Center, Mount Sinai Health System, Division of Pulmonary, Critical Care and Sleep Medicine, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
133
|
Geerdink RJ, Pillay J, Meyaard L, Bont L. Neutrophils in respiratory syncytial virus infection: A target for asthma prevention. J Allergy Clin Immunol 2015; 136:838-47. [PMID: 26277597 PMCID: PMC7112351 DOI: 10.1016/j.jaci.2015.06.034] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/30/2015] [Accepted: 06/05/2015] [Indexed: 12/25/2022]
Abstract
Lower respiratory tract infections by respiratory syncytial virus (RSV) are the foremost cause of infant hospitalization and are implicated in lasting pulmonary impairment and the development of asthma. Neutrophils infiltrate the airways of pediatric patients with RSV-induced bronchiolitis in vast numbers: approximately 80% of infiltrated cells are neutrophils. However, why neutrophils are recruited to the site of viral respiratory tract infection is not clear. In this review we discuss the beneficial and pathologic contributions of neutrophils to the immune response against RSV infection. Neutrophils can limit viral replication and spread, as well as stimulate an effective antiviral adaptive immune response. However, low specificity of neutrophil antimicrobial armaments allows for collateral tissue damage. Neutrophil-induced injury to the airways during the delicate period of infant lung development has lasting adverse consequences for pulmonary architecture and might promote the onset of asthma in susceptible subjects. We suggest that pharmacologic modulation of neutrophils should be explored as a viable future therapy for severe RSV-induced bronchiolitis and thereby prevent the inception of subsequent asthma. The antiviral functions of neutrophils suggest that targeting of neutrophils in patients with RSV-induced bronchiolitis is best performed under the umbrella of antiviral treatment.
Collapse
Affiliation(s)
- Ruben J Geerdink
- Department of Immunology, Laboratory for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Janesh Pillay
- Department of Respiratory Medicine, Laboratory for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands; Department of Anaesthesiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Linde Meyaard
- Department of Immunology, Laboratory for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Louis Bont
- Department of Immunology, Laboratory for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands; Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
134
|
Voorhees AP, DeLeon-Pennell KY, Ma Y, Halade GV, Yabluchanskiy A, Iyer RP, Flynn E, Cates CA, Lindsey ML, Han HC. Building a better infarct: Modulation of collagen cross-linking to increase infarct stiffness and reduce left ventricular dilation post-myocardial infarction. J Mol Cell Cardiol 2015; 85:229-39. [PMID: 26080361 PMCID: PMC4530076 DOI: 10.1016/j.yjmcc.2015.06.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 06/08/2015] [Accepted: 06/09/2015] [Indexed: 11/29/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) deletion attenuates collagen accumulation and dilation of the left ventricle (LV) post-myocardial infarction (MI); however the biomechanical mechanisms underlying the improved outcome are poorly understood. The aim of this study was to determine the mechanisms whereby MMP-9 deletion alters collagen network composition and assembly in the LV post-MI to modulate the mechanical properties of myocardial scar tissue. Adult C57BL/6J wild-type (WT; n=88) and MMP-9 null (MMP-9(-/-); n=92) mice of both sexes underwent permanent coronary artery ligation and were compared to day 0 controls (n=42). At day 7 post-MI, WT LVs displayed a 3-fold increase in end-diastolic volume, while MMP-9(-/-) showed only a 2-fold increase (p<0.05). Biaxial mechanical testing revealed that MMP-9(-/-) infarcts were stiffer than WT infarcts, as indicated by a 1.3-fold reduction in predicted in vivo circumferential stretch (p<0.05). Paradoxically, MMP-9(-/-) infarcts had a 1.8-fold reduction in collagen deposition (p<0.05). This apparent contradiction was explained by a 3.1-fold increase in lysyl oxidase (p<0.05) in MMP-9(-/-) infarcts, indicating that MMP-9 deletion increased collagen cross-linking activity. Furthermore, MMP-9 deletion led to a 3.0-fold increase in bone morphogenetic protein-1, the metalloproteinase that cleaves pro-collagen and pro-lysyl oxidase (p<0.05) and reduced fibronectin fragmentation by 49% (p<0.05) to enhance lysyl oxidase activity. We conclude that MMP-9 deletion increases infarct stiffness and prevents LV dilation by reducing collagen degradation and facilitating collagen assembly and cross-linking through preservation of the fibronectin network and activation of lysyl oxidase.
Collapse
Affiliation(s)
- Andrew P Voorhees
- Department of Mechanical Engineering, The University of Texas at San Antonio, USA; Joint Biomedical Engineering Program, UTSA-UTHSCSA, USA; San Antonio Cardiovascular Proteomics Center, USA
| | - Kristine Y DeLeon-Pennell
- San Antonio Cardiovascular Proteomics Center, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA
| | - Yonggang Ma
- San Antonio Cardiovascular Proteomics Center, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA
| | - Ganesh V Halade
- San Antonio Cardiovascular Proteomics Center, USA; Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, USA
| | - Andriy Yabluchanskiy
- San Antonio Cardiovascular Proteomics Center, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, USA
| | - Rugmani Padmanabhan Iyer
- San Antonio Cardiovascular Proteomics Center, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA
| | - Elizabeth Flynn
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA
| | - Courtney A Cates
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA
| | - Merry L Lindsey
- Joint Biomedical Engineering Program, UTSA-UTHSCSA, USA; San Antonio Cardiovascular Proteomics Center, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, USA
| | - Hai-Chao Han
- Department of Mechanical Engineering, The University of Texas at San Antonio, USA; Joint Biomedical Engineering Program, UTSA-UTHSCSA, USA; San Antonio Cardiovascular Proteomics Center, USA.
| |
Collapse
|
135
|
Gui J, Greene CS, Sullivan C, Taylor W, Moore JH, Kim C. Testing multiple hypotheses through IMP weighted FDR based on a genetic functional network with application to a new zebrafish transcriptome study. BioData Min 2015; 8:17. [PMID: 26097506 PMCID: PMC4474579 DOI: 10.1186/s13040-015-0050-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 06/08/2015] [Indexed: 11/10/2022] Open
Abstract
In genome-wide studies, hundreds of thousands of hypothesis tests are performed simultaneously. Bonferroni correction and False Discovery Rate (FDR) can effectively control type I error but often yield a high false negative rate. We aim to develop a more powerful method to detect differentially expressed genes. We present a Weighted False Discovery Rate (WFDR) method that incorporate biological knowledge from genetic networks. We first identify weights using Integrative Multi-species Prediction (IMP) and then apply the weights in WFDR to identify differentially expressed genes through an IMP-WFDR algorithm. We performed a gene expression experiment to identify zebrafish genes that change expression in the presence of arsenic during a systemic Pseudomonas aeruginosa infection. Zebrafish were exposed to arsenic at 10 parts per billion and/or infected with P. aeruginosa. Appropriate controls were included. We then applied IMP-WFDR during the analysis of differentially expressed genes. We compared the mRNA expression for each group and found over 200 differentially expressed genes and several enriched pathways including defense response pathways, arsenic response pathways, and the Notch signaling pathway.
Collapse
Affiliation(s)
- Jiang Gui
- Department of Biomedical Data Science, Geisel school of medicine, Dartmouth College, Hanover, NH USA.,Dartmouth-Hitchcock Medical Center, 883 Rubin Bldg, HB7927, One Medical Center Dr., Lebanon, NH USA
| | - Casey S Greene
- Department of Genetics, Geisel school of medicine, Dartmouth College, Hanover, NH USA
| | - Con Sullivan
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME USA.,Graduate School of Biomedical Science and Engineeering, University of Maine, Orono, ME USA
| | - Walter Taylor
- Department of Genetics, Geisel school of medicine, Dartmouth College, Hanover, NH USA
| | - Jason H Moore
- Department of Biostatistics and Epidemiology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Carol Kim
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, ME USA.,Graduate School of Biomedical Science and Engineeering, University of Maine, Orono, ME USA
| |
Collapse
|
136
|
Pan J, Mor G, Ju W, Zhong J, Luo X, Aldo PB, Zhong M, Yu Y, Jenkins EC, Brown WT, Zhong N. Viral Infection-Induced Differential Expression of LncRNAs Associated with Collagen in Mouse Placentas and Amniotic Sacs. Am J Reprod Immunol 2015; 74:237-57. [PMID: 26073538 DOI: 10.1111/aji.12406] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 05/13/2015] [Indexed: 12/29/2022] Open
Abstract
PROBLEM We have previously determined that long non-coding RNAs (lncRNAs) are differentially expressed in preterm premature rupture of membranes (PPROM) and hypothesized that the collagenolysis ubiquitin-proteasome system may be activated by infection and inflammation. However, direct evidence of the involvement of lncRNAs in transcriptional and posttranscriptional regulation of the infection-triggered alteration of collagen is lacking. METHOD OF STUDY A previously developed mouse model with MHV68 viral infection was assessed to determine whether viral infection may induce differential expression of lncRNAs in mouse placentas and amniotic sacs. RESULTS Differential expression of lncRNAs that are associated with collagen was found in HMV68 viral-infected, compared to non-infected, mouse placentas and amniotic sacs. Differential expression of messenger RNAs (mRNAs) of collagen was also documented. CONCLUSIONS Our data demonstrate, for the first time, that viral infection may induce the differential expression of lncRNAs that are associated with collagen. Based on this finding, we propose that lncRNA may have involved in regulating of infection-induced collagen transcription.
Collapse
Affiliation(s)
- Jing Pan
- Center of Translational Medicine for Maternal and Children's Health, Lianyungang Maternal and Children's Hospital, Lianyungang, Jiangsu, China
| | - Gil Mor
- Department of Obstetrics Gynecology and Reproductive Sciences, Reproductive Immunology Unit, School of Medicine, Yale University, New Haven, CT, USA
| | - Weina Ju
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Julia Zhong
- Hunter College High School, New York, NY, USA
| | - Xiucui Luo
- Center of Translational Medicine for Maternal and Children's Health, Lianyungang Maternal and Children's Hospital, Lianyungang, Jiangsu, China
| | - Paulomi Bole Aldo
- Department of Obstetrics Gynecology and Reproductive Sciences, Reproductive Immunology Unit, School of Medicine, Yale University, New Haven, CT, USA
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanhong Yu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Edmund C Jenkins
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - William T Brown
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Nanbert Zhong
- Center of Translational Medicine for Maternal and Children's Health, Lianyungang Maternal and Children's Hospital, Lianyungang, Jiangsu, China.,Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
137
|
Mukund K, Subramaniam S. Dysregulated mechanisms underlying Duchenne muscular dystrophy from co-expression network preservation analysis. BMC Res Notes 2015; 8:182. [PMID: 25935398 PMCID: PMC4424514 DOI: 10.1186/s13104-015-1141-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/22/2015] [Indexed: 01/21/2023] Open
Abstract
Background Duchenne Muscular Dystrophy (DMD) is an X-linked recessive disorder with its primary insult on the skeletal muscle. Severe muscle wasting, chronic inflammation and fibrosis characterize dystrophic muscle. Here we identify dysregulated pathways in DMD utilizing a co-expression network approach as described in Weighted Gene Co-expression Network Analysis (WGCNA). Specifically, we utilize WGCNA’s “preservation” statistics to identify gene modules that exhibit a weak conservation of network topology within healthy and dystrophic networks. Preservation statistics rank modules based on their topological metrics such as node density, connectivity and separability between networks. Methods Raw data for DMD was downloaded from Gene Expression Omnibus (GSE6011) and suitably preprocessed. Co-expression networks for each condition (healthy and dystrophic) were generated using the WGCNA library in R. Preservation of healthy network edges was evaluated with respect to dystrophic muscle and vice versa using WGCNA. Highly exclusive gene pairs for each of the low preserved modules within both networks were also determined using a specificity measure. Results A total of 11 and 10 co-expressed modules were identified in the networks generated from 13 healthy and 23 dystrophic samples respectively. 5 out of the 11, and 4 out of the 10 modules were identified as exhibiting none-to-weak preservation. Functional enrichment analysis identified that these weakly preserved modules were highly relevant to the condition under study. For instance, weakly preserved dystrophic module D2 exhibited the highest fraction of genes exclusive to DMD. The highly specific gene pairs identified within these modules were enriched for genes activated in response to wounding and affect the extracellular matrix including several markers such as SPP1, MMP9 and ITGB2. Conclusion The proposed approach allowed us to identify clusters of genes that are non-randomly associated with the disease. Furthermore, highly specific gene pairs pointed to interactions between known markers of disease and identification of putative markers likely associated with disease. The analysis also helped identify putative novel interactions associated with the progression of DMD. Electronic supplementary material The online version of this article (doi:10.1186/s13104-015-1141-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kavitha Mukund
- Bioinformatics and System Biology Graduate Program, University of California San Diego, 9500 Gilman Drive, MC0412, La Jolla, CA, 92093, USA.
| | - Shankar Subramaniam
- Bioinformatics and System Biology Graduate Program, University of California San Diego, 9500 Gilman Drive, MC0412, La Jolla, CA, 92093, USA. .,Departments of Bioengineering, Computer Science & Engineering, Cellular & Molecular Medicine and Chemistry & Biochemistry University of California, San Diego, 9500 Gilman Drive, MC0412, La Jolla, CA, 92093, USA.
| |
Collapse
|
138
|
Chiba N, Shimada K, Chen S, Jones HD, Alsabeh R, Slepenkin AV, Peterson E, Crother TR, Arditi M. Mast cells play an important role in chlamydia pneumoniae lung infection by facilitating immune cell recruitment into the airway. THE JOURNAL OF IMMUNOLOGY 2015; 194:3840-51. [PMID: 25754739 DOI: 10.4049/jimmunol.1402685] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/02/2015] [Indexed: 01/17/2023]
Abstract
Mast cells are known as central players in allergy and anaphylaxis, and they play a pivotal role in host defense against certain pathogens. Chlamydia pneumoniae is an important human pathogen, but it is unclear what role mast cells play during C. pneumoniae infection. We infected C57BL/6 (wild-type [WT]) and mast cell-deficient mice (Kit(W-sh/W-sh) [Wsh]) with C. pneumoniae. Wsh mice showed improved survival compared with WT mice, with fewer cells in Wsh bronchoalveolar lavage fluid (BALF), despite similar levels of cytokines and chemokines. We also found a more rapid clearance of bacteria from the lungs of Wsh mice compared with WT mice. Cromolyn, a mast cell stabilizer, reduced BALF cells and bacterial burden similar to the levels seen in Wsh mice; conversely, Compound 48/80, a mast cell degranulator, increased the number of BALF cells and bacterial burden. Histology showed that WT lungs had diffuse inflammation, whereas Wsh mice had patchy accumulations of neutrophils and perivascular accumulations of lymphocytes. Infected Wsh mice had reduced amounts of matrix metalloprotease-9 in BALF and were resistant to epithelial integral membrane protein degradation, suggesting that barrier integrity remains intact in Wsh mice. Mast cell reconstitution in Wsh mice led to enhanced bacterial growth and normal epithelial integral membrane protein degradation, highlighting the specific role of mast cells in this model. These data suggest that mast cells play a detrimental role during C. pneumoniae infection by facilitating immune cell infiltration into the airspace and providing a more favorable replicative environment for C. pneumoniae.
Collapse
Affiliation(s)
- Norika Chiba
- Division of Pediatric Infectious Diseases and Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048
| | - Kenichi Shimada
- Division of Pediatric Infectious Diseases and Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048
| | - Shuang Chen
- Division of Pediatric Infectious Diseases and Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048
| | - Heather D Jones
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048
| | - Randa Alsabeh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048; and
| | | | - Ellena Peterson
- Department of Pathology, University of California Irvine, Irvine, CA 92697
| | - Timothy R Crother
- Division of Pediatric Infectious Diseases and Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048
| | - Moshe Arditi
- Division of Pediatric Infectious Diseases and Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center and David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90048;
| |
Collapse
|
139
|
Therapy with plasma purified alpha1-antitrypsin (Prolastin®) induces time-dependent changes in plasma levels of MMP-9 and MPO. PLoS One 2015; 10:e0117497. [PMID: 25635861 PMCID: PMC4311911 DOI: 10.1371/journal.pone.0117497] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/25/2014] [Indexed: 01/04/2023] Open
Abstract
The common Z mutation (Glu342Lys) of α1-antitrypsin (A1AT) results in the polymerization and intracellular retention of A1AT protein. The concomitant deficiency of functional A1AT predisposes PiZZ subjects to early onset emphysema. Clinical studies have implied that, among the biomarkers associated with emphysema, matrix metalloproteinase 9 (MMP-9) is of particular importance. Increased plasma MMP-9 levels are proposed to predict the decline of lung function as well as greater COPD exacerbations in A1AT deficiency-associated emphysema. The aim of the present study was to investigate the effect of A1AT therapy (Prolastin) on plasma MMP-9 and myeloperoxidase (MPO) levels. In total 34 PiZZ emphysema patients were recruited: 12 patients without and 22 with weekly intravenous (60 mg/kg body weight) A1AT therapy. The quantitative analysis of A1AT, MMP-9 and MPO was performed in serum and in supernatants of blood neutrophils isolated from patients before and after therapy. Patients with Prolastin therapy showed significantly lower serum MMP-9 and MPO levels than those without therapy. However, parallel analysis revealed that a rapid infusion of Prolastin is accompanied by a transient elevation of plasma MMP-9 and MPO levels. Experiments with freshly isolated blood neutrophils confirmed that therapy with Prolastin causes transient MMP-9 and MPO release. Prolastin induced the rapid release of MMP-9 and MPO when added directly to neutrophil cultures and this reaction was associated with the presence of IgA in A1AT preparation. Our data support the conclusion that changes in plasma levels of MMP-9 and MPO mirror the effect of Prolastin on blood neutrophils.
Collapse
|
140
|
Ramana CV, DeBerge MP, Kumar A, Alia CS, Durbin JE, Enelow RI. Inflammatory impact of IFN-γ in CD8+ T cell-mediated lung injury is mediated by both Stat1-dependent and -independent pathways. Am J Physiol Lung Cell Mol Physiol 2015; 308:L650-7. [PMID: 25617378 DOI: 10.1152/ajplung.00360.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/29/2014] [Indexed: 01/24/2023] Open
Abstract
Influenza infection results in considerable pulmonary pathology, a significant component of which is mediated by CD8(+) T cell effector functions. To isolate the specific contribution of CD8(+) T cells to lung immunopathology, we utilized a nonviral murine model in which alveolar epithelial cells express an influenza antigen and injury is initiated by adoptive transfer of influenza-specific CD8(+) T cells. We report that IFN-γ production by adoptively transferred influenza-specific CD8(+) T cells is a significant contributor to acute lung injury following influenza antigen recognition, in isolation from its impact on viral clearance. CD8(+) T cell production of IFN-γ enhanced lung epithelial cell expression of chemokines and the subsequent recruitment of inflammatory cells into the airways. Surprisingly, Stat1 deficiency in the adoptive-transfer recipients exacerbated the lung injury that was mediated by the transferred influenza-specific CD8(+) T cells but was still dependent on IFN-γ production by these cells. Loss of Stat1 resulted in sustained activation of Stat3 signaling, dysregulated chemokine expression, and increased infiltration of the airways by inflammatory cells. Taken together, these data identify important roles for IFN-γ signaling and Stat1-independent IFN-γ signaling in regulating CD8(+) T cell-mediated acute lung injury. This is the first study to demonstrate an anti-inflammatory effect of Stat1 on CD8(+) T cell-mediated lung immunopathology without the complication of differences in viral load.
Collapse
Affiliation(s)
- Chilakamarti V Ramana
- Department of Medicine, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire
| | - Matthew P DeBerge
- Department of Pathology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois
| | - Aseem Kumar
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Christopher S Alia
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Joan E Durbin
- Department of Pathology, Rutgers-New Jersey Medical School of Medicine, Newark, New Jersey; and
| | - Richard I Enelow
- Department of Medicine, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire; Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, New Hampshire
| |
Collapse
|
141
|
Type-I interferons induce lung protease responses following respiratory syncytial virus infection via RIG-I-like receptors. Mucosal Immunol 2015; 8:161-75. [PMID: 25005357 PMCID: PMC4268269 DOI: 10.1038/mi.2014.54] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 05/20/2014] [Accepted: 05/21/2014] [Indexed: 02/04/2023]
Abstract
The role of proteases in viral infection of the lung is poorly understood. Thus, we examined matrix metalloproteinases (MMPs) and cathepsin proteases in respiratory syncytial virus (RSV)-infected mouse lungs. RSV-induced gene expression for MMPs -2, -3, -7, -8, -9, -10, -12, -13, -14, -16, -17, -19, -20, -25, -27, and -28 and cathepsins B, C, E, G, H, K, L1, S, W, and Z in the airways of Friend leukemia virus B sensitive strain mice. Increased proteases were present in the bronchoalveolar lavage fluid (BALF) and lung tissue during infection. Mitochondrial antiviral-signaling protein (MAVS) and TIR-domain-containing adapter-inducing interferon-β-deficient mice were exposed to RSV. Mavs-deficient mice had significantly lower expression of airway MMP-2, -3, -7, -8, -9, -10, -12, -13, and -28 and cathepsins C, G, K, S, W, and Z. In lung epithelial cells, retinoic acid-inducible gene-1 (RIG-I) was identified as the major RIG-I-like receptor required for RSV-induced protease expression via MAVS. Overexpression of RIG-I or treatment with interferon-β in these cells induced MMP and cathepsin gene and protein expression. The significance of RIG-1 protease induction was demonstrated by the fact that inhibiting proteases with batimastat, E64 or ribavirin prevented airway hyperresponsiveness and enhanced viral clearance in RSV-infected mice.
Collapse
|
142
|
Sobinoff AP, Dando SJ, Redgrove KA, Sutherland JM, Stanger SJ, Armitage CW, Timms P, McLaughlin EA, Beagley KW. Chlamydia muridarum infection-induced destruction of male germ cells and sertoli cells is partially prevented by Chlamydia major outer membrane protein-specific immune CD4 cells. Biol Reprod 2014; 92:27. [PMID: 25472923 DOI: 10.1095/biolreprod.114.124180] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Chlamydia trachomatis infections are increasingly prevalent worldwide. Male chlamydial infections are associated with urethritis, epididymitis, and orchitis; however, the role of Chlamydia in prostatitis and male factor infertility remains controversial. Using a model of Chlamydia muridarum infection in male C57BL/6 mice, we investigated the effects of chlamydial infection on spermatogenesis and determined the potential of immune T cells to prevent infection-induced outcomes. Antigen-specific CD4 T cells significantly reduced the infectious burden in the penile urethra, epididymis, and vas deferens. Infection disrupted seminiferous tubules, causing loss of germ cells at 4 and 8 wk after infection, with the most severely affected tubules containing only Sertoli cells. Increased mitotic proliferation, DNA repair, and apoptosis in spermatogonial cells and damaged germ cells were evident in atrophic tubules. Activated caspase 3 (casp3) staining revealed increased (6-fold) numbers of Sertoli cells with abnormal morphology that were casp3 positive in tubules of infected mice, indicating increased levels of apoptosis. Sperm count and motility were both decreased in infected mice, and there was a significant decrease in morphologically normal spermatozoa. Assessment of the spermatogonial stem cell population revealed a decrease in promyelocytic leukemia zinc finger (PLZF)-positive cells in the seminiferous tubules. Interestingly, adoptive transfer of antigen-specific CD4 cells, particularly T-helper 2-like cells, prior to infection prevented these effects in spermatogenesis and Sertoli cells. These data suggest that chlamydial infection adversely affects spermatogenesis and male fertility, and that vaccination can potentially prevent the spread of infection and these adverse outcomes.
Collapse
Affiliation(s)
- Alexander P Sobinoff
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Samantha J Dando
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kate A Redgrove
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jessie M Sutherland
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Simone J Stanger
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Charles W Armitage
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Peter Timms
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Eileen A McLaughlin
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
143
|
Hoang LT, Tolfvenstam T, Ooi EE, Khor CC, Naim ANM, Ho EXP, Ong SH, Wertheim HF, Fox A, Van Vinh Nguyen C, Nghiem NM, Ha TM, Thi Ngoc Tran A, Tambayah P, Lin R, Sangsajja C, Manosuthi W, Chuchottaworn C, Sansayunh P, Chotpitayasunondh T, Suntarattiwong P, Chokephaibulkit K, Puthavathana P, de Jong MD, Farrar J, van Doorn HR, Hibberd ML. Patient-based transcriptome-wide analysis identify interferon and ubiquination pathways as potential predictors of influenza A disease severity. PLoS One 2014; 9:e111640. [PMID: 25365328 PMCID: PMC4218794 DOI: 10.1371/journal.pone.0111640] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/19/2014] [Indexed: 12/21/2022] Open
Abstract
Background The influenza A virus is an RNA virus that is responsible for seasonal epidemics worldwide with up to five million cases of severe illness and 500,000 deaths annually according to the World Health Organization estimates. The factors associated with severe diseases are not well defined, but more severe disease is more often seen among persons aged >65 years, infants, pregnant women, and individuals of any age with underlying health conditions. Methodology/Principal Findings Using gene expression microarrays, the transcriptomic profiles of influenza-infected patients with severe (N = 11), moderate (N = 40) and mild (N = 83) symptoms were compared with the febrile patients of unknown etiology (N = 73). We found that influenza-infected patients, regardless of their clinical outcomes, had a stronger induction of antiviral and cytokine responses and a stronger attenuation of NK and T cell responses in comparison with those with unknown etiology. More importantly, we found that both interferon and ubiquitination signaling were strongly attenuated in patients with the most severe outcomes in comparison with those with moderate and mild outcomes, suggesting the protective roles of these pathways in disease pathogenesis. Conclusion/Significances The attenuation of interferon and ubiquitination pathways may associate with the clinical outcomes of influenza patients.
Collapse
Affiliation(s)
| | - Thomas Tolfvenstam
- Infection Immunology, Respiratory Infections, Karolinska Institutet, Solna, Sweden
| | - Eng Eong Ooi
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Swee Hoe Ong
- Genome Institute of Singapore, Singapore, Singapore
| | - Heiman F. Wertheim
- National Hospital of Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Hanoi, Vietnam, and Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Annette Fox
- National Hospital of Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Hanoi, Vietnam, and Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Ngoc My Nghiem
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | | | | | | | - Raymond Lin
- National University Hospital, Singapore, Singapore
| | | | | | | | | | | | | | | | | | - Menno D. de Jong
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam, and Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Medical Microbiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeremy Farrar
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam, and Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - H. Rogier van Doorn
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Programme, Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam, and Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
144
|
Redgrove KA, McLaughlin EA. The Role of the Immune Response in Chlamydia trachomatis Infection of the Male Genital Tract: A Double-Edged Sword. Front Immunol 2014; 5:534. [PMID: 25386180 PMCID: PMC4209867 DOI: 10.3389/fimmu.2014.00534] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/09/2014] [Indexed: 01/16/2023] Open
Abstract
Chlamydia trachomatis (CT) is the most prevalent bacterial sexually transmitted infection in the world, with more than 100 million cases reported annually. While there have been extensive studies into the adverse effects that CT infection has on the female genital tract, and on the subsequent ability of these women to conceive, studies into the consequences on male fertility have been limited and controversial. This is in part due to the asymptomatic nature of the infection, where it is estimated that 50% of men with Chlamydia fail to show any symptoms. It is accepted, however, that acute and/or persistent CT infection is the causative agent for conditions such as urethritis, epididymitis, epididymo-orchitis, and potentially prostatitis. As with most infections, the immune system plays a fundamental role in the body’s attempts to eradicate the infection. The first and most important immune response to Chlamydia infection is a local one, whereby immune cells such as leukocytes are recruited to the site of infections, and subsequently secrete pro-inflammatory cytokines and chemokines such as interferon gamma. Immune cells also work to initiate and potentiate chronic inflammation through the production of reactive oxygen species (ROS), and the release of molecules with degradative properties including defensins, elastase, collagenase, cathespins, and lysozyme. This long-term inflammation can lead to cell proliferation (a possible precursor to cancer), tissue remodeling, and scarring, as well as being linked to the onset of autoimmune responses in genetically disposed individuals. This review will focus on the ability of the immune system to recognize and clear acute and persistent chlamydial infections in the male genital tract, and on the paradoxical damage that chronic inflammation resulting from the infection can cause on the reproductive health of the individual.
Collapse
Affiliation(s)
- Kate A Redgrove
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| | - Eileen A McLaughlin
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| |
Collapse
|
145
|
Hematopoietic but not endothelial cell MyD88 contributes to host defense during gram-negative pneumonia derived sepsis. PLoS Pathog 2014; 10:e1004368. [PMID: 25254554 PMCID: PMC4177915 DOI: 10.1371/journal.ppat.1004368] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 07/31/2014] [Indexed: 12/12/2022] Open
Abstract
Klebsiella pneumoniae is an important cause of sepsis. The common Toll-like receptor adapter myeloid differentiation primary response gene (MyD)88 is crucial for host defense against Klebsiella. Here we investigated the role of MyD88 in myeloid and endothelial cells during Klebsiella pneumosepsis. Mice deficient for MyD88 in myeloid (LysM-Myd88(-/-)) and myeloid plus endothelial (Tie2-Myd88(-/-)) cells showed enhanced lethality and bacterial growth. Tie2-Myd88(-/-) mice reconstituted with control bone marrow, representing mice with a selective MyD88 deficiency in endothelial cells, showed an unremarkable antibacterial defense. Myeloid or endothelial cell MyD88 deficiency did not impact on lung pathology or distant organ injury during late stage sepsis, while LysM-Myd88(-/-) mice demonstrated a strongly attenuated inflammatory response in the airways early after infection. These data suggest that myeloid but not endothelial MyD88 is important for host defense during gram-negative pneumonia derived sepsis.
Collapse
|
146
|
DeLeon-Pennell KY, de Castro Brás LE, Iyer RP, Bratton DR, Jin YF, Ripplinger CM, Lindsey ML. P. gingivalis lipopolysaccharide intensifies inflammation post-myocardial infarction through matrix metalloproteinase-9. J Mol Cell Cardiol 2014; 76:218-26. [PMID: 25240641 DOI: 10.1016/j.yjmcc.2014.09.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 09/02/2014] [Accepted: 09/07/2014] [Indexed: 01/21/2023]
Abstract
Periodontal disease (PD) strongly correlates with increased mortality post-myocardial infarction (MI); however, the underlying mechanisms are unknown. Matrix metalloproteinase (MMP)-9 levels directly correlate with dysfunction and remodeling of the left ventricle (LV) post-MI. Post-MI, MMP-9 is produced by leukocytes and modulates inflammation. We have shown that exposure to Porphyromonas gingivalis lipopolysaccharide (PgLPS), an immunomodulatory molecule identified in PD patients, increases LV MMP-9 levels in mice and leads to cardiac inflammation and dysfunction. The aim of the study was to determine if circulating PgLPS exacerbates the LV inflammatory response post-MI through MMP-9 dependent mechanisms. We exposed wild type C57BL/6J and MMP-9(-/-) mice to PgLPS (ATCC 33277) for a period of 28 days before performing MI, and continued to deliver PgLPS for up to 7 days post-MI. We found systemic levels of PgLPS 1) increased MMP-9 levels in both plasma and infarcted LV resulting in reduced wall thickness and increased incidence of LV rupture post-MI and 2) increased systemic and local macrophage chemotaxis leading to accelerated M1 macrophage infiltration post-MI and decreased LV function. MMP-9 deletion played a protective role by attenuating the inflammation induced by systemic delivery of PgLPS. In conclusion, MMP-9 deletion has a cardioprotective role against PgLPS exposure, by attenuating macrophage mediated inflammation.
Collapse
Affiliation(s)
- Kristine Y DeLeon-Pennell
- San Antonio Cardiovascular Proteomics Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Lisandra E de Castro Brás
- San Antonio Cardiovascular Proteomics Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Rugmani Padmanabhan Iyer
- San Antonio Cardiovascular Proteomics Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Dustin R Bratton
- San Antonio Cardiovascular Proteomics Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Yu-Fang Jin
- San Antonio Cardiovascular Proteomics Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Electrical and Computer Engineering, University of Texas San Antonio, San Antonio, TX 78249, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Merry L Lindsey
- San Antonio Cardiovascular Proteomics Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
147
|
Vidaña B, Martínez J, Martínez-Orellana P, García Migura L, Montoya M, Martorell J, Majó N. Heterogeneous pathological outcomes after experimental pH1N1 influenza infection in ferrets correlate with viral replication and host immune responses in the lung. Vet Res 2014; 45:85. [PMID: 25163545 PMCID: PMC4161856 DOI: 10.1186/s13567-014-0085-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 07/31/2014] [Indexed: 01/13/2023] Open
Abstract
The swine-origin pandemic (p) H1N1 influenza A virus causes mild upper-respiratory tract disease in most human patients. However, some patients developed severe lower-respiratory tract infections with fatal consequences, and the cause of these infections remain unknown. Recently, it has been suggested that different populations have different degrees of susceptibility to pH1N1 strains due to host genetic variations that are associated with inappropriate immune responses against viral genetic characteristics. Here, we tested whether the pathologic patterns of influenza strains that produce different disease outcomes in humans could be reproduced in a ferret model. Our results revealed that the severities of infection did not correspond to particular viral isolate and were not associated with the clinical phenotypes of the corresponding patients. Severe pathological outcomes were associated with higher viral replication, especially in alveolar areas, and with an exacerbated innate cellular immune response that was characterised by substantial phagocytic and cytotoxic cell migration into the lungs. Moreover, detrimental innate cellular responses were linked to the up-regulation of several proinflammatory cytokines and chemokines and the down-regulation of IFNα in the lungs. Additionally, severe lung lesions were associated with greater up-regulations of pro-apoptotic markers and higher levels of apoptotic neutrophils and macrophages. In conclusion, this study confirmed that the clinicopathological outcomes of pH1N1 infection in ferrets were not only due to viral replication abilities but also depended on the hosts’ capacities to mount efficient immune responses to control viral infection of the lung.
Collapse
Affiliation(s)
- Beatriz Vidaña
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain ; Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain
| | - Jorge Martínez
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain ; Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain
| | - Pamela Martínez-Orellana
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain
| | - Lourdes García Migura
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain
| | - María Montoya
- Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain ; Institut de Recerca i Tecnologia Agroalimentaria (IRTA), Barcelona, Spain
| | - Jaime Martorell
- Departament de Medicina i Cirurgia Animals, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain
| | - Natàlia Majó
- Departament de Sanitat i Anatomia Animals, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain ; Centre de Recerca en Sanitat Animal (CReSA), UAB-IRTA, Campus de la Universitat Autònoma de Barcelona, Cerdanyola del Vallés, 08193 Bellaterra Spain
| |
Collapse
|
148
|
Lindner D, Zietsch C, Tank J, Sossalla S, Fluschnik N, Hinrichs S, Maier L, Poller W, Blankenberg S, Schultheiss HP, Tschöpe C, Westermann D. Cardiac fibroblasts support cardiac inflammation in heart failure. Basic Res Cardiol 2014; 109:428. [PMID: 25086637 DOI: 10.1007/s00395-014-0428-7] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 07/02/2014] [Accepted: 07/18/2014] [Indexed: 12/23/2022]
Abstract
Cardiac remodeling and inflammation are hallmarks of cardiac failure and correlate with outcome in patients. However, the basis for the development of both remains unclear. We have previously reported that cardiac inflammation triggers transdifferentiation of fibroblasts to myofibroblasts and therefore increase accumulation of cardiac collagen, one key pathology in cardiac remodeling. Hence, identifying key pathways for inflammation would be beneficial for patients suffering from heart failure also. Besides their well-characterized function in matrix regulation, we here investigate the role of fibroblasts in the inflammatory process. We address for the first time the role of fibroblasts as inflammatory supporter cells in heart failure. Using endomyocardial biopsies from patients with heart failure and dilated cardiomyopathy, we created a primary human cardiac fibroblast cell culture system. We found that mechanical stretch mimicking cardiac dilation in heart failure induces activation of fibroblasts and not only stimulates production of extracellular matrix but more interestingly up-regulates chemokine production and triggers typical inflammatory pathways in vitro. Moreover, the cell culture supernatant of stretched fibroblasts activates inflammatory cells and induces further recruitment of monocytes by allowing transendothelial migration into the cardiac tissue. Our findings reveal that cardiac fibroblasts provide pro-inflammatory mediators and may act as sentinel cells activated by mechanical stress. Those cells are able to recruit inflammatory cells into the cardiac tissue, a process known to aggravate outcome of patients. This might be important in different forms of heart failure and therefore may be one general mechanism specific for fibroblasts.
Collapse
Affiliation(s)
- Diana Lindner
- Clinic for General and Interventional Cardiology, University Heart Center Hamburg, Martinistr. 52, 20246, Hamburg, Germany,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Hu F, Ku MC, Markovic D, Dzaye ODA, Lehnardt S, Synowitz M, Wolf SA, Kettenmann H. Glioma-associated microglial MMP9 expression is upregulated by TLR2 signaling and sensitive to minocycline. Int J Cancer 2014; 135:2569-78. [PMID: 24752463 DOI: 10.1002/ijc.28908] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/03/2014] [Indexed: 11/10/2022]
Abstract
The invasiveness of malignant gliomas is one of the major obstacles in glioma therapy and the reason for the poor survival of patients. Glioma cells infiltrate into the brain parenchyma and thereby escape surgical resection. Glioma associated microglia/macrophages support glioma infiltration into the brain parenchyma by increased expression and activation of extracellular matrix degrading proteases such as matrix metalloprotease (MMP) 2, MMP9 and membrane-type 1 MMP. In this work we demonstrate that, MMP9 is predominantly expressed by glioma associated microglia/macrophages in mouse and human glioma tissue but not by the glioma cells. Supernatant from glioma cells induced the expression of MMP9 in cultured microglial cells. Using mice deficient for different Toll-like receptors we identified Toll-like receptor 2/6 as the signaling pathway for the glioma induced upregulation of microglial MMP9. Also in an experimental mouse glioma model, Toll-like receptor 2 deficiency attenuated the upregulation of microglial MMP9. Moreover, glioma supernatant triggered an upregulation of Toll-like receptor 2 expression in microglia. Both, the upregulation of MMP9 and Toll-like receptor 2 were attenuated by the antibiotic minocycline and a p38 mitogen-activated protein kinase antagonist in vitro. Minocycline also extended the survival rate of glioma bearing mice when given to the drinking water. Thus glioma cells change the phenotype of glioma associated microglia/macrophages in a complex fashion using Toll-like receptor 2 as an important signaling pathway and minocycline further proved to be a potential candidate for adjuvant glioma therapy.
Collapse
Affiliation(s)
- Feng Hu
- Cellular Neurosciences, Max Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
150
|
MMP9 Deficiency Increased the Size of Experimentally Induced Apical Periodontitis. J Endod 2014; 40:658-64. [DOI: 10.1016/j.joen.2014.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 10/01/2013] [Accepted: 01/02/2014] [Indexed: 12/12/2022]
|