1
|
Kailankangas V, Katayama S, Gröndahl-Yli-Hannuksela K, Vilhonen J, Tervaniemi MH, Rantakokko-Jalava K, Seiskari T, Lönnqvist E, Kere J, Oksi J, Syrjänen J, Vuopio J. Low expression of the CCL5 gene and low serum concentrations of CCL5 in severe invasive group a streptococcal disease. Infection 2025; 53:51-59. [PMID: 38865072 PMCID: PMC11825563 DOI: 10.1007/s15010-024-02318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE Our objective was to elucidate host dependent factors of disease severity in invasive group A Streptococcal disease (iGAS) using transcriptome profiling of iGAS cases of varying degrees of severity at different timepoints. To our knowledge there are no previous transcriptome studies in iGAS patients. METHODS We recruited iGAS cases from June 2018 to July 2020. Whole blood samples for transcriptome analysis and serum for biomarker analysis were collected at three timepoints representing the acute (A), the convalescent (B) and the post-infection phase (C). Gene expression was compared against clinical traits and disease course. Serum chemokine ligand 5 (CCL5, an inflammatory cytokine) concentration was also measured. RESULTS Forty-five patients were enrolled. After disqualifying degraded or impure RNAs we had 34, 31 and 21 subjects at timepoints A, B, and C, respectively. Low expression of the CCL5 gene correlated strongly with severity (death or need for intensive care) at timepoint A (AUC = 0.92), supported by low concentrations of CCL5 in sera. CONCLUSIONS Low gene expression levels and low serum concentration of CCL5 in the early stages of an iGAS infection were associated with a more severe disease course. CCL5 might have potential as a predictor of disease severity. Low expression of genes of cytotoxic immunity, especially CCL5, and corresponding low serum concentrations of CCL5 associated with a severe disease course, i.e. death, or need for intensive care, in early phase of invasive group A Streptococcal disease.
Collapse
Affiliation(s)
- V Kailankangas
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland.
| | - S Katayama
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden
| | | | - J Vilhonen
- Department of Infectious Diseases, Turku University Hospital, Turku, Finland
| | - M H Tervaniemi
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - K Rantakokko-Jalava
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Clinical Microbiology, Laboratory Division, Turku University Hospital, Turku, Finland
| | - T Seiskari
- Department of Clinical Microbiology, Fimlab Laboratories, Tampere, Finland
| | - E Lönnqvist
- Department of Clinical Microbiology, Fimlab Laboratories, Tampere, Finland
| | - J Kere
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden
| | - J Oksi
- Department of Infectious Diseases, Turku University Hospital, Turku, Finland
- Faculty of Medicine, University of Turku, Turku, Finland
| | - J Syrjänen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - J Vuopio
- Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Clinical Microbiology, Laboratory Division, Turku University Hospital, Turku, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| |
Collapse
|
2
|
Filippov I, Schauser L, Peterson P. An integrated single-cell atlas of blood immune cells in aging. NPJ AGING 2024; 10:59. [PMID: 39613786 DOI: 10.1038/s41514-024-00185-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
Recent advances in single-cell technologies have facilitated studies on age-related alterations in the immune system. However, previous studies have often employed different marker genes to annotate immune cell populations, making it challenging to compare results. In this study, we combined seven single-cell transcriptomic datasets, comprising more than a million cells from one hundred and three donors, to create a unified atlas of human peripheral blood mononuclear cells (PBMC) from both young and old individuals. Using a consistent set of marker genes for immune cell annotation, we standardized the classification of immune cells and assessed their prevalence in both age groups. The integrated dataset revealed several consistent trends related to aging, including a decline in CD8+ naive T cells and MAIT cells and an expansion of non-classical monocyte compartments. However, we observed significant variability in other cell types. Our analysis of the long non-coding RNA MALAT1hi T cell population, previously implicated in age-related T cell exhaustion, showed that this population is highly heterogeneous with a mixture of naïve-like and memory-like cells. Despite substantial variation among the datasets when comparing gene expression between age groups, we identified a high-confidence signature of CD8+ naive T cell aging marked by an increased expression of pro-inflammatory genes. In conclusion, our study emphasizes the importance of standardizing existing single-cell datasets to enable the comprehensive examination of age-related cellular changes across multiple datasets.
Collapse
Affiliation(s)
- Igor Filippov
- QIAGEN Aarhus A/S, Aarhus, Denmark.
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
| | | | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
3
|
Wang S, Li H, Liu Y, Pang S, Qiao S, Su J, Wang S, Zhang Y. Connectivity Network Feature Sharing in Single-Cell RNA Sequencing Data Identifies Rare Cells. J Chem Inf Model 2024; 64:6596-6609. [PMID: 39096508 DOI: 10.1021/acs.jcim.4c00796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
Single-cell RNA sequencing is a valuable technique for identifying diverse cell subtypes. A key challenge in this process is that the detection of rare cells is often missed by conventional methods due to low abundance and subtle features of these cells. To overcome this, we developed SCLCNF (Local Connectivity Network Feature Sharing in Single-Cell RNA sequencing), a novel approach that identifies rare cells by analyzing features uniquely expressed in these cells. SCLCNF creates a cellular connectivity network, considering how each cell relates to its neighbors. This network helps to pinpoint coexpression patterns unique to rare cells, utilizing a rarity score to confirm their presence. Our method performs better in detecting rare cells than existing techniques, offering enhanced robustness. It has proven to be effective in human gastrula data sets for accurately pinpointing rare cells, and in sepsis data sets where it uncovers previously unidentified rare cell populations.
Collapse
Affiliation(s)
- Shudong Wang
- Qingdao Institute of Software, College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Hengxiao Li
- Qingdao Institute of Software, College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Yahui Liu
- College of Science, China University of Petroleum (East China), Qingdao 266580, China
| | - Shanchen Pang
- Qingdao Institute of Software, College of Computer Science and Technology, China University of Petroleum (East China), Qingdao 266580, China
| | - Sibo Qiao
- The College of Software, Tiangong University, Tianjin 300387, China
| | - Jionglong Su
- School of AI and Advanced Computing, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, Suzhou 215123, Jiangsu, China
| | - Shaoqiang Wang
- School of Information and Control Engineering, Qingdao University of Technology, Qingdao 266525, China
| | - Yulin Zhang
- College of Mathematics and Systems Science, Shandong University of Science and Technology, Qingdao 266590, China
| |
Collapse
|
4
|
Aubert A, Jung K, Hiroyasu S, Pardo J, Granville DJ. Granzyme serine proteases in inflammation and rheumatic diseases. Nat Rev Rheumatol 2024; 20:361-376. [PMID: 38689140 DOI: 10.1038/s41584-024-01109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 05/02/2024]
Abstract
Granzymes (granule-secreted enzymes) are a family of serine proteases that have been viewed as redundant cytotoxic enzymes since their discovery more than 30 years ago. Predominantly produced by cytotoxic lymphocytes and natural killer cells, granzymes are delivered into the cytoplasm of target cells through immunological synapses in cooperation with the pore-forming protein perforin. After internalization, granzymes can initiate cell death through the cleavage of intracellular substrates. However, evidence now also demonstrates the existence of non-cytotoxic, pro-inflammatory, intracellular and extracellular functions that are granzyme specific. Under pathological conditions, granzymes can be produced and secreted extracellularly by immune cells as well as by non-immune cells. Depending on the granzyme, accumulation in the extracellular milieu might contribute to inflammation, tissue injury, impaired wound healing, barrier dysfunction, osteoclastogenesis and/or autoantigen generation.
Collapse
Affiliation(s)
- Alexandre Aubert
- International Collaboration on Repair Discoveries (ICORD) Centre; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD) Centre; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sho Hiroyasu
- Department of Dermatology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA); Department of Microbiology, Radiology, Paediatrics and Public Health, University of Zaragoza, Zaragoza, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD) Centre; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- Centre for Heart Lung Innovation, Providence Research, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
5
|
Rasi V, Phelps KR, Paulson KR, Eickhoff CS, Chinnaraj M, Pozzi N, Di Gioia M, Zanoni I, Shakya S, Carlson HL, Ford DA, Kolar GR, Hoft DF. Homodimeric Granzyme A Opsonizes Mycobacterium tuberculosis and Inhibits Its Intracellular Growth in Human Monocytes via Toll-Like Receptor 4 and CD14. J Infect Dis 2024; 229:876-887. [PMID: 37671668 PMCID: PMC10938207 DOI: 10.1093/infdis/jiad378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb)-specific γ9δ2 T cells secrete granzyme A (GzmA) protective against intracellular Mtb growth. However, GzmA-enzymatic activity is unnecessary for pathogen inhibition, and the mechanisms of GzmA-mediated protection remain unknown. We show that GzmA homodimerization is essential for opsonization of mycobacteria, altered uptake into human monocytes, and subsequent pathogen clearance within the phagolysosome. Although monomeric and homodimeric GzmA bind mycobacteria, only homodimers also bind cluster of differentiation 14 (CD14) and Toll-like receptor 4 (TLR4). Without access to surface-expressed CD14 and TLR4, GzmA fails to inhibit intracellular Mtb. Upregulation of Rab11FIP1 was associated with inhibitory activity. Furthermore, GzmA colocalized with and was regulated by protein disulfide isomerase AI (PDIA1), which cleaves GzmA homodimers into monomers and prevents Mtb inhibitory activity. These studies identify a previously unrecognized role for homodimeric GzmA structure in opsonization, phagocytosis, and elimination of Mtb in human monocytes, and they highlight PDIA1 as a potential host-directed therapy for prevention and treatment of tuberculosis, a major human disease.
Collapse
Affiliation(s)
- Valerio Rasi
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Kathleen R Phelps
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Keegan R Paulson
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Christopher S Eickhoff
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Mathivanan Chinnaraj
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Nicola Pozzi
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Marco Di Gioia
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Shubha Shakya
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Haley L Carlson
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Grant R Kolar
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Daniel F Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Scimeca RC, Reichard MV. Differential gene expression response to acute and chronic Cytauzxoon felis infection in domestic cats (Felis catus). Ticks Tick Borne Dis 2023; 14:102242. [PMID: 37651848 DOI: 10.1016/j.ttbdis.2023.102242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/25/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Cytauxzoonosis is a severe tick transmitted protozoan disease of domestic cats, caused by Cytauxzoon felis. The disease is characterized by acute onset of high fever, depression, lethargy, inappentence, anorexia, icterus, dehydration, hemolytic anemia, and alteration of immune response. The aim of our study was to further detail the immune response of domestic cats to C. felis infection by comparing the differential expression of feline immune transcriptional elements during acute and chronic cytauxzoonosis. True single molecule sequencing (tSMS) was used to analyze the whole genome of acutely and chronically infected C. felis cats, focusing on the analysis of genes involved on the immune response. Two C. felis donor cats were infested with Amblyomma americanum nymphs, which after repletion were collected and kept in humidity chambers until they molted. The resulting A. americanum were randomly selected to infest three C. felis naïve principal cats. Infection of these cats was confirmed by nested PCR of the 18S rRNA C. felis gene and clinical signs. RNA was extracted from whole blood at different time points and used for tSMS analyses, the results revealed overexpression in transcripts involved in type I interferon signaling, cellular and cytokine responses during the acute stage of infection, while cell cycle, and metabolic processes were downregulated. Genes involved in cell adhesion increased their expression in the chronic infected cats, whereas inflammatory and apoptotic related genes were downregulated. This study provided information on the host immune response to C. felis in domestic cats, demonstrating that inflammatory, apoptotic, and cell adhesion are some of the pathways altered during acute and chronic infection.
Collapse
Affiliation(s)
- Ruth C Scimeca
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078.
| | - Mason V Reichard
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078
| |
Collapse
|
7
|
Clain JA, Boutrais S, Dewatines J, Racine G, Rabezanahary H, Droit A, Zghidi-Abouzid O, Estaquier J. Lipid metabolic reprogramming of hepatic CD4 + T cells during SIV infection. Microbiol Spectr 2023; 11:e0168723. [PMID: 37656815 PMCID: PMC10581067 DOI: 10.1128/spectrum.01687-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/24/2023] [Indexed: 09/03/2023] Open
Abstract
While liver inflammation is associated with AIDS, little is known so far about hepatic CD4+ T cells. By using the simian immunodeficiency virus (SIV)-infected rhesus macaque (RM) model, we aimed to characterize CD4+ T cells. The phenotype of CD4+ T cells was assessed by flow cytometry from uninfected (n = 3) and infected RMs, with either SIVmac251 (n = 6) or SHIVSF162p3 (n = 6). After cell sorting of hepatic CD4+ T cells, viral DNA quantification and RNA sequencing were performed.Thus, we demonstrated that liver CD4+ T cells strongly expressed the SIV coreceptor, CCR5. We showed that viremia was negatively correlated with the percentage of hepatic effector memory CD4+ T cells. Consistent with viral sensing, inflammatory and interferon gene transcripts were increased. We also highlighted the presence of harmful CD4+ T cells expressing GZMA and members of TGFB that could contribute to fuel inflammation and fibrosis. Whereas RNA sequencing demonstrated activated CD4+ T cells displaying higher levels of mitoribosome and membrane lipid synthesis transcripts, few genes were related to glycolysis and oxidative phosphorylation, which are essential to sustain activated T cells. Furthermore, we observed lower levels of mitochondrial DNA and higher levels of genes associated with damaged organelles (reticulophagy and mitophagy). Altogether, our data revealed that activated hepatic CD4+ T cells are reprogrammed to lipid metabolism. Thus, strategies aiming to reprogram T cell metabolism with effector function could be of interest for controlling viral infection and preventing liver disorders.IMPORTANCEHuman immunodeficiency virus (HIV) infection may cause liver diseases, associated with inflammation and tissue injury, contributing to comorbidity in people living with HIV. Paradoxically, the contribution of hepatic CD4+ T cells remains largely underestimated. Herein, we used the model of simian immunodeficiency virus (SIV)-infected rhesus macaques to access liver tissue. Our work demonstrates that hepatic CD4+ T cells express CCR5, the main viral coreceptor, and are infected. Viral infection is associated with the presence of inflamed and activated hepatic CD4+ T cells expressing cytotoxic molecules. Furthermore, hepatic CD4+ T cells are reprogrammed toward lipid metabolism after SIV infection. Altogether, our findings shed new light on hepatic CD4+ T cell profile that could contribute to liver injury following viral infection.
Collapse
Affiliation(s)
- Julien A. Clain
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada
| | - Steven Boutrais
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada
| | - Juliette Dewatines
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada
| | - Gina Racine
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada
| | | | - Arnaud Droit
- Proteomics Platform, CHU de Québec - Université Laval Research Center, Québec City, Québec, Canada
- Computational Biology Laboratory, CHU de Québec - Université Laval Research Center, Québec City, Québec, Canada
| | - Ouafa Zghidi-Abouzid
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada
| | - Jérôme Estaquier
- Centre de Recherche du CHU de Québec, Université Laval, Québec City, Québec, Canada
- INSERM U1124, Université Paris, Paris, France
| |
Collapse
|
8
|
Jia T, Yuan F, Tao J, Wang G, Zhang X, Zhang B, Li H. CRISPR/Cas13d targeting GZMA in PARs pathway regulates the function of osteoclasts in chronic apical periodontitis. Cell Mol Biol Lett 2023; 28:70. [PMID: 37626297 PMCID: PMC10464397 DOI: 10.1186/s11658-023-00477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic apical periodontitis is a prevalent oral disease characterized by bone loss, and its underlying mechanisms remain unclear. This study aimed to investigate the role and mechanism of the serine protease GZMA in osteoclasts during chronic apical periodontitis. To address this, we employed crRNA/Cas13d to inhibit GZMA expression and examined its impact on osteoclast behavior. Our findings revealed that GZMA plays a significant role in promoting osteoclast cell proliferation while inhibiting cell apoptosis. Additionally, the inhibition of GZMA led to a notable increase in miR-25-3p expression, which, in turn, downregulated the expression of TGF-β. Consequently, the reduction in TGF-β expression led to a decrease in PAR1 expression within the PARs pathway. These results suggest that GZMA might serve as a promising therapeutic target for the treatment of chronic apical periodontitis. Furthermore, our study highlights the potential of targeting GZMA using crRNA/Cas13d as a valuable approach for future therapeutic interventions.
Collapse
Affiliation(s)
- Tingting Jia
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Fang Yuan
- Department of Oncology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jingqiao Tao
- Department of Stomatology, Southern Medical Branch of PLA General Hospital, Beijing, China
| | - Gang Wang
- Medical School of Chinese PLA, Beijing, China
| | - Xianhua Zhang
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Bin Zhang
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China.
| | - Hongbo Li
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
9
|
Lin T, Qin T, Jiang S, Zhang C, Wang L. Anti-inflammatory and anti-biotic drug metronidazole loaded ZIF-90 nanoparticles as a pH responsive drug delivery system for improved pediatric sepsis management. Microb Pathog 2023; 176:105941. [PMID: 36509311 DOI: 10.1016/j.micpath.2022.105941] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Sepsis is a life-threatening disease caused by the dis-functioning of the immune response to pathogenic infections. Despite, the discovery of modern therapeutics and treatments of sepsis are lacking due to the resistance of pathogens. Metronidazole is an antibiotic commonly used to treat bacterial infections, but usage is limited and challenging by a short half-life period. In this research work, fabricate a pH-responsive drug delivery system for controlled release of metronidazole targeted molecules. We exemplified that, the encapsulation of hydrophilic metronidazole drug within a hydrophobic ZIF-90 framework can be enhanced the pH-responsive drug release under acidic conditions. The ZIF-90 frameworks only decompose in under acidic solutions, they are highly stable in physiological conditions. The pH-responsive protonation mechanism of ZIF-90 frameworks promotes the quick release of metronidazole within cells. The antimicrobial proficiency of zinc and metronidazole will expose a synergistic effect in ROS-mediated bacterial inhibition and auto-immunity boosting of normal cells. In vitro, antibacterial activity results revealed that the MI@ZIF-90 nano drug delivery system effectively eradicated human infectious pathogens at the lowest concentrations. In anti-fungal activity, studies show excellent growth inhibition against human pathogenic fungi Aspergillus fumigatus and Candida albicans. Finally, the PBMC cytocompatibility study concludes, that the fabricated MI@ZIF-90 drug delivery system is non-toxic to biomedical applications. The overall research findings highlight the design of a smart drug delivery system for sepsis treatment. In future it will be an efficient, low-cost, and biocompatible pharmaceutics for pediatric sepsis management processes.
Collapse
Affiliation(s)
- Tingting Lin
- Department of Neonatology, Wenling First People's Hospital, Wenling, Zhejiang Province, 317500, China.
| | - Tao Qin
- Department of Neonatology, Wenling First People's Hospital, Wenling, Zhejiang Province, 317500, China
| | - Shanshan Jiang
- Department of Neonatology, Wenling First People's Hospital, Wenling, Zhejiang Province, 317500, China
| | - Chunfeng Zhang
- Department of Neonatology, Wenling First People's Hospital, Wenling, Zhejiang Province, 317500, China
| | - Ling Wang
- Department of Neonatology, Wenling First People's Hospital, Wenling, Zhejiang Province, 317500, China.
| |
Collapse
|
10
|
Moreno-Martinez L, Santiago L, de la Torre M, Calvo AC, Pardo J, Osta R. Hemizygous Granzyme A Mice Expressing the hSOD1G93A Transgene Show Slightly Extended Lifespan. Int J Mol Sci 2022; 23:13554. [PMID: 36362341 PMCID: PMC9655466 DOI: 10.3390/ijms232113554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 08/30/2023] Open
Abstract
Granzyme A (gzmA), a serine protease involved in the modulation of the inflammatory immune response, is found at an elevated level in the serum from ALS patients. However, the influence of gzmA on the progression of ALS remains unclear. The aim of our work was to assess whether the absence of gzmA in an ALS murine model could help slow down the progression of the disease. Homozygous and hemizygous gzmA-deficient mice expressing the hSOD1G93A transgene were generated, and survival of these mice was monitored. Subsequently, gene and protein expression of inflammatory and oxidative stress markers was measured in the spinal cord and quadriceps of these mice. We observed the longest lifespan in gzmA+/- mice. GzmA gene and protein expression was downregulated in the spinal cord and serum from gmzA+/- mice, confirming that the increased survival of hemizygous mice is correlated with lower levels of gzmA. In addition, mRNA and protein levels of glutathione reductase (GSR), involved in oxidative stress, were found downregulated in the spinal cord and quadriceps of gmzA+/- mice, together with lower IL-1β and IL-6 mRNA levels in hemyzigous mice. In summary, our findings indicate for the first time that reduced levels, but not the absence, of gzmA could slightly ameliorate the disease progression in this animal model.
Collapse
Affiliation(s)
- Laura Moreno-Martinez
- LAGENBIO, Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain
- Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Biomedical Research Centre of Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- AgriFood Institute of Aragon-IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Llipsy Santiago
- Biomedical Research Centre of Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miriam de la Torre
- LAGENBIO, Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain
- Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Biomedical Research Centre of Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- AgriFood Institute of Aragon-IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Ana Cristina Calvo
- LAGENBIO, Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain
- Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Biomedical Research Centre of Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- AgriFood Institute of Aragon-IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Julián Pardo
- Biomedical Research Centre of Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009 Zaragoza, Spain
| | - Rosario Osta
- LAGENBIO, Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain
- Centre for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Biomedical Research Centre of Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- AgriFood Institute of Aragon-IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
11
|
Richardson KC, Jung K, Pardo J, Turner CT, Granville DJ. Noncytotoxic Roles of Granzymes in Health and Disease. Physiology (Bethesda) 2022; 37:323-348. [PMID: 35820180 DOI: 10.1152/physiol.00011.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Granzymes are serine proteases previously believed to play exclusive and somewhat redundant roles in lymphocyte-mediated target cell death. However, recent studies have challenged this paradigm. Distinct substrate profiles and functions have since emerged for each granzyme while their dysregulated proteolytic activities have been linked to diverse pathologies.
Collapse
Affiliation(s)
- Katlyn C Richardson
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Jung
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), Zaragoza, Spain.,Department of Microbiology, Radiology, Pediatrics and Public Health, University of Zaragoza, Zaragoza, Spain.,CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Zaragoza, Spain
| | - Christopher T Turner
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria, Australia.,Future Industries Institute, University of South Australia, Adelaide, South Australia, Australia
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), British Columbia Professional Firefighters' Wound Healing Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Chen J, Wang H, Guo R, Li H, Cui N. Early Expression of Functional Markers on CD4+ T Cells Predicts Outcomes in ICU Patients With Sepsis. Front Immunol 2022; 13:938538. [PMID: 35898496 PMCID: PMC9309518 DOI: 10.3389/fimmu.2022.938538] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/16/2022] [Indexed: 11/23/2022] Open
Abstract
Objective There is evidence that metabolic disorder, dysfunction and abnormal apoptosis of immune cells are closely related to immunosuppression in sepsis. Single monitoring of exhaustion receptors does not reflect well the immune status of septic patients; therefore, we monitored immune status in relation to metabolism, function and apoptosis of immune cells to find good prognostic indicators for sepsis. Design A single-center prospective observational study. Setting Teaching hospital including an academic tertiary care center. Patients 81 patients with sepsis and 22 without sepsis admitted to the ICU. Interventions Patients were divided according to Sequential Organ Failure Assessment (SOFA) score: mild sepsis 2–5 points and severe sepsis ≥6 points. SOFA score was recalculated daily. If it changed by ≥2 points within 2 days, T-cell metabolism, function and apoptotic makers [mammalian target of rapamycin (mTOR), T-bet, interferon (IFN)-γ, granzyme B, and programmed cell death (PD)-1] were continuously monitored on days 1, 3 and 5 after admission. Measurements and Main Results The overall status of immune cells was compared among patients with different severity of sepsis. Patients with severe sepsis, compared with mild and no sepsis, had lower lymphocyte counts, higher expression of receptors associated with cell metabolism, activation and apoptosis, and lower expression of functional receptors. Multivariate regression analysis revealed that frequency of CD4+ T cells expressing mTOR, IFN-γ and PD-1 at admission was an independent predictor of 28-day mortality. Receiver operating characteristic curve analysis indicated that frequency of CD4+ T cells expressing mTOR, IFN-γ and PD-1 predicted 28-day mortality, with cutoffs of 30.57%, 12.81% and 22.46%, respectively. The expression of related receptors on CD8+ T cells showed similar trend to that on CD4+ T cells, but no significant difference was found. Conclusions Abnormally increased expression of metabolic and apoptotic receptors on CD4+ T cells and decreased expression of functional factors are associated with poor prognosis in ICU patients with sepsis. Poor prognosis can be identified by early detection of expression of mammalian target of rapamycin (mTOR), IFN-γ and PD-1 on CD4+ T cells.
Collapse
Affiliation(s)
- Jianwei Chen
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Beijing Jishuitan Hospital, Beijing, China
| | - Ran Guo
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Na Cui,
| |
Collapse
|
13
|
Uranga-Murillo I, Morte E, Hidalgo S, Pesini C, García-Mulero S, Sierra JL, Santiago L, Arias M, De Miguel D, Encabo-Berzosa MDM, Gracia-Tello B, Sanz-Pamplona R, Martinez-Lostao L, Galvez EM, Paño-Pardo JR, Ramirez-Labrada A, Pardo J. Integrated analysis of circulating immune cellular and soluble mediators reveals specific COVID19 signatures at hospital admission with utility for prediction of clinical outcomes. Theranostics 2022; 12:290-306. [PMID: 34987646 PMCID: PMC8690910 DOI: 10.7150/thno.63463] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/28/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID19), caused by SARS-CoV-2, is a complex disease, with a variety of clinical manifestations ranging from asymptomatic infection or mild cold-like symptoms to more severe cases requiring hospitalization and critical care. The most severe presentations seem to be related with a delayed, deregulated immune response leading to exacerbated inflammation and organ damage with close similarities to sepsis. Methods: In order to improve the understanding on the relation between host immune response and disease course, we have studied the differences in the cellular (monocytes, CD8+ T and NK cells) and soluble (cytokines, chemokines and immunoregulatory ligands) immune response in blood between Healthy Donors (HD), COVID19 and a group of patients with non-COVID19 respiratory tract infections (NON-COV-RTI). In addition, the immune response profile has been analyzed in COVID19 patients according to disease severity. Results: In comparison to HDs and patients with NON-COV-RTI, COVID19 patients show a heterogeneous immune response with the presence of both activated and exhausted CD8+ T and NK cells characterised by the expression of the immune checkpoint LAG3 and the presence of the adaptive NK cell subset. An increased frequency of adaptive NK cells and a reduction of NK cells expressing the activating receptors NKp30 and NKp46 correlated with disease severity. Although both activated and exhausted NK cells expressing LAG3 were increased in moderate/severe cases, unsupervised cell clustering analyses revealed a more complex scenario with single NK cells expressing more than one immune checkpoint (PD1, TIM3 and/or LAG3). A general increased level of inflammatory cytokines and chemokines was found in COVID19 patients, some of which like IL18, IL1RA, IL36B and IL31, IL2, IFNα and TNFα, CXCL10, CCL2 and CCL8 were able to differentiate between COVID19 and NON-COV-RTI and correlated with bad prognosis (IL2, TNFα, IL1RA, CCL2, CXCL10 and CXCL9). Notably, we found that soluble NKG2D ligands from the MIC and ULBPs families were increased in COVID19 compared to NON-COV-RTI and correlated with disease severity. Conclusions: Our results provide a detailed comprehensive analysis of the presence of activated and exhausted CD8+T, NK and monocyte cell subsets as well as extracellular inflammatory factors beyond cytokines/chemokines, specifically associated to COVID19. Importantly, multivariate analysis including clinical, demographical and immunological experimental variables have allowed us to reveal specific immune signatures to i) differentiate COVID19 from other infections and ii) predict disease severity and the risk of death.
Collapse
|
14
|
Gardner MM, Kirschen MP, Wong HR, McKeone DJ, Halstead ES, Thompson J, Himebauch AS, Topjian AA, Yehya N. Biomarkers associated with mortality in pediatric patients with cardiac arrest and acute respiratory distress syndrome. Resuscitation 2022; 170:184-193. [PMID: 34871756 PMCID: PMC8799511 DOI: 10.1016/j.resuscitation.2021.11.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/16/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023]
Abstract
AIMS To identify plasma biomarkers associated with cardiac arrest in a cohort of children with acute respiratory distress syndrome (ARDS), and to assess the association of these biomarkers with mortality in children with cardiac arrest and ARDS (ARDS + CA). METHODS This was a secondary analysis of a single-center prospective cohort study of children with ARDS from 2014-2019 with 17 biomarkers measured. Clinical characteristics and biomarkers were compared between subjects with ARDS + CA and ARDS with univariate analysis. In a sub-cohort of ARDS + CA subjects, the association between biomarker levels and mortality was tested using univariate and bivariate logistic regression. RESULTS Biomarkers were measured in 333 subjects: 301 with ARDS (median age 5.3 years, 55.5% male) and 32 ARDS + CA (median age 8 years, 53.1% male). More arrests (69%) occurred out-of-hospital with a median CPR duration of 11 (IQR 5.5, 25) minutes. ARDS severity, PRISM III score, vasoactive-ionotropic score and extrapulmonary organ failures were worse in the ARDS + CA versus ARDS group. Eight biomarkers were elevated in the ARDS + CA versus ARDS cohort: sRAGE, nucleosomes, SP-D, CCL22, IL-6, HSP70, IL-8, and MIP-1b. sRAGE, SP-D, and CCL22 remained elevated when the cohorts were matched for illness severity. When controlling for severity of ARDS and cardiac arrest characteristics, sRAGE, IL-6 and granzyme B were associated with mortality in the ARDS + CA group. CONCLUSION sRAGE, IL-6 and granzyme B were associated with cardiac arrest mortality when controlling for illness severity. sRAGE was consistently higher in the ARDS + CA cohort compared to ARDS and retained independent association with mortality.
Collapse
Affiliation(s)
- Monique M. Gardner
- Division of Cardiac Critical Care Medicine, Department of Anesthesiology & Critical Care Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia Pennsylvania
| | - Matthew P. Kirschen
- Division of Critical Care Medicine, Department of Anesthesiology & Critical Care Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia Pennsylvania
| | - Hector R. Wong
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Daniel J. McKeone
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - E. Scott Halstead
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jill Thompson
- Division of Critical Care Medicine, Department of Anesthesiology & Critical Care Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia Pennsylvania
| | - Adam S. Himebauch
- Division of Critical Care Medicine, Department of Anesthesiology & Critical Care Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia Pennsylvania
| | - Alexis A. Topjian
- Division of Critical Care Medicine, Department of Anesthesiology & Critical Care Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia Pennsylvania
| | - Nadir Yehya
- Division of Critical Care Medicine, Department of Anesthesiology & Critical Care Medicine, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia Pennsylvania,Leonard Davis Institute, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
15
|
Uranga-Murillo I, Tapia E, Garzón-Tituaña M, Ramirez-Labrada A, Santiago L, Pesini C, Esteban P, Roig FJ, Galvez EM, Bird PI, Pardo J, Arias M. Biological relevance of Granzymes A and K during E. coli sepsis. Am J Cancer Res 2021; 11:9873-9883. [PMID: 34815792 PMCID: PMC8581435 DOI: 10.7150/thno.59418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/08/2021] [Indexed: 11/07/2022] Open
Abstract
Aims: Recent in vitro findings suggest that the serine protease Granzyme K (GzmK) may act as a proinflammatory mediator. However, its role in sepsis is unknown. Here we aim to understand the role of GzmK in a mouse model of bacterial sepsis and compare it to the biological relevance of Granzyme A (GzmA). Methods: Sepsis was induced in WT, GzmA-/- and GzmK-/- mice by an intraperitoneal injection of 2x108 CFU from E. coli. Mouse survival was monitored during 5 days. Levels of IL-1α, IL-1β, TNFα and IL-6 in plasma were measured and bacterial load in blood, liver and spleen was analyzed. Finally, profile of cellular expression of GzmA and GzmK was analyzed by FACS. Results: GzmA and GzmK are not involved in the control of bacterial infection. However, GzmA and GzmK deficient mice showed a lower sepsis score in comparison with WT mice, although only GzmA deficient mice exhibited increased survival. GzmA deficient mice also showed reduced expression of some proinflammatory cytokines like IL1-α, IL-β and IL-6. A similar result was found when extracellular GzmA was therapeutically inhibited in WT mice using serpinb6b, which improved survival and reduced IL-6 expression. Mechanistically, active extracellular GzmA induces the production of IL-6 in macrophages by a mechanism dependent on TLR4 and MyD88. Conclusions: These results suggest that although both proteases contribute to the clinical signs of E. coli-induced sepsis, inhibition of GzmA is sufficient to reduce inflammation and improve survival irrespectively of the presence of other inflammatory granzymes, like GzmK.
Collapse
|
16
|
Zhou S, Lu H, Xiong M. Identifying Immune Cell Infiltration and Effective Diagnostic Biomarkers in Rheumatoid Arthritis by Bioinformatics Analysis. Front Immunol 2021; 12:726747. [PMID: 34484236 PMCID: PMC8411707 DOI: 10.3389/fimmu.2021.726747] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/30/2021] [Indexed: 01/16/2023] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder characterized by inflammatory cell infiltration, leading to persistent synovitis and joint destruction. The pathogenesis of RA remains unclear. This study aims to explore the immune molecular mechanism of RA through bioinformatics analysis. Methods Five microarray datasets and a high throughput sequencing dataset were downloaded. CIBERSORT algorithm was performed to evaluate immune cell infiltration in synovial tissues between RA and healthy control (HC). Wilcoxon test and Least Absolute Shrinkage and Selection Operator (LASSO) regression were conducted to identify the significantly different infiltrates of immune cells. Differentially expressed genes (DEGs) were screened by "Batch correction" and "RobustRankAggreg" methods. Functional correlation of DEGs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Candidate biomarkers were identified by cytoHubba of Cytoscape, and their diagnostic effectiveness was predicted by Receiver Operator Characteristic Curve (ROC) analysis. The association of the identified biomarkers with infiltrating immune cells was explored using Spearman's rank correlation analysis in R software. Results Ten significantly different types of immune cells between RA and HC were identified. A total of 202 DEGs were obtained by intersection of DEGs screened by two methods. The function of DEGs were significantly associated with immune cells. Five hub genes (CXCR4, CCL5, CD8A, CD247, and GZMA) were screened by R package "UpSet". CCL5+CXCR4 and GZMA+CD8A were verified to have the capability to diagnose RA and early RA with the most excellent specificity and sensitivity, respectively. The correlation between immune cells and biomarkers showed that CCL5 was positively correlated with M1 macrophages, CXCR4 was positively correlated with memory activated CD4+ T cells and follicular helper T (Tfh) cells, and GZMA was positively correlated with Tfh cells. Conclusions CCL5, CXCR4, GZMA, and CD8A can be used as diagnostic biomarker for RA. GZMA-Tfh cells, CCL5-M1 macrophages, and CXCR4- memory activated CD4+ T cells/Tfh cells may participate in the occurrence and development of RA, especially GZMA-Tfh cells for the early pathogenesis of RA.
Collapse
Affiliation(s)
- Sheng Zhou
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Hongcheng Lu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Xiong
- Department of Orthopedics, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
17
|
Rasi V, Wood DC, Eickhoff CS, Xia M, Pozzi N, Edwards RL, Walch M, Bovenschen N, Hoft DF. Granzyme A Produced by γ 9δ 2 T Cells Activates ER Stress Responses and ATP Production, and Protects Against Intracellular Mycobacterial Replication Independent of Enzymatic Activity. Front Immunol 2021; 12:712678. [PMID: 34413857 PMCID: PMC8368726 DOI: 10.3389/fimmu.2021.712678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/12/2021] [Indexed: 01/14/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the pathological agent that causes tuberculosis (TB) is the number one infectious killer worldwide with one fourth of the world's population currently infected. Data indicate that γ9δ2 T cells secrete Granzyme A (GzmA) in the extracellular space triggering the infected monocyte to inhibit growth of intracellular mycobacteria. Accordingly, deletion of GZMA from γ9δ2 T cells reverses their inhibitory capacity. Through mechanistic studies, GzmA's action was investigated in monocytes from human PBMCs. The use of recombinant human GzmA expressed in a mammalian system induced inhibition of intracellular mycobacteria to the same degree as previous human native protein findings. Our data indicate that: 1) GzmA is internalized within mycobacteria-infected cells, suggesting that GzmA uptake could prevent infection and 2) that the active site is not required to inhibit intracellular replication. Global proteomic analysis demonstrated that the ER stress response and ATP producing proteins were upregulated after GzmA treatment, and these proteins abundancies were confirmed by examining their expression in an independent set of patient samples. Our data suggest that immunotherapeutic host interventions of these pathways may contribute to better control of the current TB epidemic.
Collapse
Affiliation(s)
- Valerio Rasi
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States,Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - David C. Wood
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Christopher S. Eickhoff
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Mei Xia
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Nicola Pozzi
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Rachel L. Edwards
- Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Michael Walch
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands,Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Daniel F. Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States,Department of Internal Medicine, Saint Louis University School of Medicine, Saint Louis, MO, United States,*Correspondence: Daniel F. Hoft,
| |
Collapse
|
18
|
Fung ITH, Zhang Y, Shin DS, Sankar P, Sun X, D'Souza SS, Song R, Kuentzel ML, Chittur SV, Zuloaga KL, Yang Q. Group 2 innate lymphoid cells are numerically and functionally deficient in the triple transgenic mouse model of Alzheimer's disease. J Neuroinflammation 2021; 18:152. [PMID: 34229727 PMCID: PMC8261980 DOI: 10.1186/s12974-021-02202-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 06/23/2021] [Indexed: 01/02/2023] Open
Abstract
Background The immune pathways in Alzheimer’s disease (AD) remain incompletely understood. Our recent study indicates that tissue-resident group 2 innate lymphoid cells (ILC2) accumulate in the brain barriers of aged mice and that their activation alleviates aging-associated cognitive decline. The regulation and function of ILC2 in AD, however, remain unknown. Methods In this study, we examined the numbers and functional capability of ILC2 from the triple transgenic AD mice (3xTg-AD) and control wild-type mice. We investigated the effects of treatment with IL-5, a cytokine produced by ILC2, on the cognitive function of 3xTg-AD mice. Results We demonstrate that brain-associated ILC2 are numerically and functionally defective in the triple transgenic AD mouse model (3xTg-AD). The numbers of brain-associated ILC2 were greatly reduced in 7-month-old 3xTg-AD mice of both sexes, compared to those in age- and sex-matched control wild-type mice. The remaining ILC2 in 3xTg-AD mice failed to efficiently produce the type 2 cytokine IL-5 but gained the capability to express a number of proinflammatory genes. Administration of IL-5, a cytokine produced by ILC2, transiently improved spatial recognition and learning in 3xTg-AD mice. Conclusion Our results collectively indicate that numerical and functional deficiency of ILC2 might contribute to the cognitive impairment of 3xTg-AD mice. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02202-2.
Collapse
Affiliation(s)
- Ivan Ting Hin Fung
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Yuanyue Zhang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Damian S Shin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Poornima Sankar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Xiangwan Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Shanti S D'Souza
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Renjie Song
- Biochemistry & Immunology Core Facility at Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Marcy L Kuentzel
- Center for Functional Genomics, University at Albany-SUNY, Rensselaer, NY, 12144, USA
| | - Sridar V Chittur
- Center for Functional Genomics, University at Albany-SUNY, Rensselaer, NY, 12144, USA
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Qi Yang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA.
| |
Collapse
|
19
|
Santiago L, Castro M, Sanz-Pamplona R, Garzón M, Ramirez-Labrada A, Tapia E, Moreno V, Layunta E, Gil-Gómez G, Garrido M, Peña R, Lanuza PM, Comas L, Jaime-Sanchez P, Uranga-Murillo I, Del Campo R, Pelegrín P, Camerer E, Martínez-Lostao L, Muñoz G, Uranga JA, Alcalde A, Galvez EM, Ferrandez A, Bird PI, Metkar S, Arias MA, Pardo J. Extracellular Granzyme A Promotes Colorectal Cancer Development by Enhancing Gut Inflammation. Cell Rep 2021; 32:107847. [PMID: 32640217 DOI: 10.1016/j.celrep.2020.107847] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/11/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
If not properly regulated, the inflammatory immune response can promote carcinogenesis, as evident in colorectal cancer (CRC). Aiming to gain mechanistic insight into the link between inflammation and CRC, we perform transcriptomics analysis of human CRC, identifying a strong correlation between expression of the serine protease granzyme A (GzmA) and inflammation. In a dextran sodium sulfate and azoxymethane (DSS/AOM) mouse model, deficiency and pharmacological inhibition of extracellular GzmA both attenuate gut inflammation and prevent CRC development, including the initial steps of cell transformation and epithelial-to-mesenchymal transition. Mechanistically, extracellular GzmA induces NF-κB-dependent IL-6 production in macrophages, which in turn promotes STAT3 activation in cultured CRC cells. Accordingly, colon tissues from DSS/AOM-treated, GzmA-deficient animals present reduced levels of pSTAT3. By identifying GzmA as a proinflammatory protease that promotes CRC development, these findings provide information on mechanisms that link immune cell infiltration to cancer progression and present GzmA as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Llipsy Santiago
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Marta Castro
- Department of Pharmacology and Physiology, Faculty of Health and Sports Sciences, University of Zaragoza, 22002 Huesca, Spain
| | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Marcela Garzón
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Ariel Ramirez-Labrada
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Elena Tapia
- Animal Unit, University of Zaragoza, 50009 Zaragoza, Spain
| | - Víctor Moreno
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESP, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Elena Layunta
- Department of Pharmacology and Physiology, Faculty of Veterinary, University of Zaragoza, 50013 Zaragoza, Spain
| | - Gabriel Gil-Gómez
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona
| | - Marta Garrido
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona
| | - Raúl Peña
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), 08003 Barcelona
| | - Pilar M Lanuza
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Laura Comas
- Instituto de Carboquímica ICB-CSIC, 50018 Zaragoza, Spain
| | - Paula Jaime-Sanchez
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Iratxe Uranga-Murillo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Rosa Del Campo
- Department of Microbiology, University Hospital Ramón y Cajal & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Pablo Pelegrín
- Unidad de Inflamación Molecular y Cirugía Experimental, Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Eric Camerer
- Université de Paris, Paris Cardiovascular Research Center, INSERM U970, 75015 Paris, France
| | - Luis Martínez-Lostao
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Department of Immunology, University Clinic Hospital Lozano Blesa, 50009, Zaragoza, Spain and Department of Pathology, University Clinic Hospital Lozano Blesa, University of Zaragoza, IIS Aragón, CIBEREHD, 50009 Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; Department Biochemistry and Molecular and Cell Biology and Department Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009 Zaragoza, Spain
| | - Guillermo Muñoz
- Department of Immunology, University Clinic Hospital Lozano Blesa, 50009, Zaragoza, Spain and Department of Pathology, University Clinic Hospital Lozano Blesa, University of Zaragoza, IIS Aragón, CIBEREHD, 50009 Zaragoza, Spain
| | - José A Uranga
- Department of Basis Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, 28922 Madrid, Spain
| | - Anabel Alcalde
- Department of Pharmacology and Physiology, Faculty of Veterinary, University of Zaragoza, 50013 Zaragoza, Spain
| | - Eva M Galvez
- Instituto de Carboquímica ICB-CSIC, 50018 Zaragoza, Spain
| | - Angel Ferrandez
- Service of Digestive Diseases, University Clinic Hospital Lozano Blesa, University of Zaragoza, IIS Aragón, CIBEREHD, Zaragoza, Spain
| | - Phillip I Bird
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University 3800 Melbourne, Australia
| | | | - Maykel A Arias
- Instituto de Carboquímica ICB-CSIC, 50018 Zaragoza, Spain.
| | - Julian Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Aragon I+D Foundation (ARAID), Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; Department Biochemistry and Molecular and Cell Biology and Department Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009 Zaragoza, Spain; CIBER-BBN, Madrid, Spain.
| |
Collapse
|
20
|
Garzón-Tituaña M, Sierra-Monzón JL, Comas L, Santiago L, Khaliulina-Ushakova T, Uranga-Murillo I, Ramirez-Labrada A, Tapia E, Morte-Romea E, Algarate S, Couty L, Camerer E, Bird PI, Seral C, Luque P, Paño-Pardo JR, Galvez EM, Pardo J, Arias M. Granzyme A inhibition reduces inflammation and increases survival during abdominal sepsis. Theranostics 2021; 11:3781-3795. [PMID: 33664861 PMCID: PMC7914344 DOI: 10.7150/thno.49288] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
Aims: Peritonitis is one of the most common causes of sepsis, a serious syndrome characterized by a dysregulated systemic inflammatory response. Recent evidence suggests that Granzyme A (GzmA), a serine protease mainly expressed by NK and T cells, could act as a proinflammatory mediator and could play an important role in the pathogenesis of sepsis. This work aims to analyze the role and the therapeutic potential of GzmA in the pathogenesis of peritoneal sepsis. Methods: The level of extracellular GzmA as well as GzmA activity were analyzed in serum from healthy volunteers and patients with confirmed peritonitis and were correlated with the Sequential Organ Failure Assessment (SOFA) score. Peritonitis was induced in C57Bl/6 (WT) and GzmA-/- mice by cecal ligation and puncture (CLP). Mice were treated intraperitoneally with antibiotics alone or in combination serpinb6b, a specific GzmA inhibitor, for 5 days. Mouse survival was monitored during 14 days, levels of some proinflammatory cytokines were measured in serum and bacterial load and diversity was analyzed in blood and spleen at different times. Results: Clinically, elevated GzmA was observed in serum from patients with abdominal sepsis suggesting that GzmA plays an important role in this pathology. In the CLP model GzmA deficient mice, or WT mice treated with an extracellular GzmA inhibitor, showed increased survival, which correlated with a reduction in proinflammatory markers in both serum and peritoneal lavage fluid. GzmA deficiency did not influence bacterial load in blood and spleen and GzmA did not affect bacterial replication in macrophages in vitro, indicating that GzmA has no role in bacterial control. Analysis of GzmA in lymphoid cells following CLP showed that it was mainly expressed by NK cells. Mechanistically, we found that extracellular active GzmA acts as a proinflammatory mediator in macrophages by inducing the TLR4-dependent expression of IL-6 and TNFα. Conclusions: Our findings implicate GzmA as a key regulator of the inflammatory response during abdominal sepsis and provide solid evidences about its therapeutic potential for the treatment of this severe pathology.
Collapse
Affiliation(s)
- Marcela Garzón-Tituaña
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - José L Sierra-Monzón
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - Laura Comas
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
| | - Llipsy Santiago
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Tatiana Khaliulina-Ushakova
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - Iratxe Uranga-Murillo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Ariel Ramirez-Labrada
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
| | - Elena Tapia
- Animal Unit, University of Zaragoza, 50009, Zaragoza, Spain
| | - Elena Morte-Romea
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - Sonia Algarate
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology and Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - Ludovic Couty
- INSERM U970, Paris Cardiovascular Research Centre, Université de Paris, 75015, Paris, France
| | - Eric Camerer
- INSERM U970, Paris Cardiovascular Research Centre, Université de Paris, 75015, Paris, France
| | - Phillip I Bird
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, 3800, Clayton VIC, Australia
| | - Cristina Seral
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology and Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - Pilar Luque
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - José R Paño-Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, 50009, Zaragoza, Spain
| | - Eva M Galvez
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
| | - Julián Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), 50009, Zaragoza, Spain
- Aragon I+D Foundation (ARAID), 50018, Zaragoza, Spain
- Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018, Zaragoza, Spain
- Department of Biochemistry and Molecular and Cell Biology and Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
| | - Maykel Arias
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
| |
Collapse
|
21
|
Ma L, Li Q, Cai S, Peng H, Huyan T, Yang H. The role of NK cells in fighting the virus infection and sepsis. Int J Med Sci 2021; 18:3236-3248. [PMID: 34400893 PMCID: PMC8364442 DOI: 10.7150/ijms.59898] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/12/2021] [Indexed: 12/22/2022] Open
Abstract
Natural killer cells, one of the important types of innate immune cells, play a pivotal role in the antiviral process in vivo. It has been shown that increasing NK cell activity may promote the alleviation of viral infections, even severe infection-induced sepsis. Given the current state of the novel coronavirus (SARS-CoV-2) global pandemic, clarifying the anti-viral function of NK cells would be helpful for revealing the mechanism of host immune responses and decipher the progression of COVID-19 and providing important clues for combating this pandemic. In this review, we summarize the roles of NK cells in viral infection and sepsis as well as the potential possibilities of NK cell-based immunotherapy for treating COVID-19.
Collapse
Affiliation(s)
- Lu Ma
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qi Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Suna Cai
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hourong Peng
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ting Huyan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
22
|
Granzymes in cardiovascular injury and disease. Cell Signal 2020; 76:109804. [PMID: 33035645 DOI: 10.1016/j.cellsig.2020.109804] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 12/17/2022]
Abstract
Chronic inflammation and impaired wound healing play important roles in the pathophysiology of cardiovascular diseases. Moreover, the aberrant secretion of proteases plays a critical role in pathological tissue remodeling in chronic inflammatory conditions. Human Granzymes (Granule secreted enzymes - Gzms) comprise a family of five (GzmA, B, H, K, M) cell-secreted serine proteases. Although each unique in function and substrate specificities, Gzms were originally thought to share redundant, intracellular roles in cytotoxic lymphocyte-induced cell death. However, an abundance of evidence has challenged this dogma. It is now recognized, that individual Gzms exhibit unique substrate repertoires and functions both intracellularly and extracellularly. In the extracellular milieu, Gzms contribute to inflammation, vascular dysfunction and permeability, reduced cell adhesion, release of matrix-sequestered growth factors, receptor activation, and extracellular matrix cleavage. Despite these recent findings, the non-cytotoxic functions of Gzms in the context of cardiovascular disease pathogenesis remain poorly understood. Minimally detected in tissues and bodily fluids of normal individuals, GzmB is elevated in patients with acute coronary syndromes, coronary artery disease, and myocardial infarction. Pre-clinical animal models have exemplified the importance of GzmB in atherosclerosis, aortic aneurysm, and cardiac fibrosis as animals deficient in GzmB exhibit reduced tissue remodeling, improved disease phenotypes and increased survival. Although a role for GzmB in cardiovascular disease is described, further work to elucidate the mechanisms that underpin the remaining human Gzms activity in cardiovascular disease is necessary. The present review provides a summary of the pre-clinical and clinical evidence, as well as emerging areas of research pertaining to Gzms in tissue remodeling and cardiovascular disease.
Collapse
|
23
|
Park S, Griesenauer B, Jiang H, Adom D, Mehrpouya-Bahrami P, Chakravorty S, Kazemian M, Imam T, Srivastava R, Hayes TA, Pardo J, Janga SC, Paczesny S, Kaplan MH, Olson MR. Granzyme A-producing T helper cells are critical for acute graft-versus-host disease. JCI Insight 2020; 5:124465. [PMID: 32809971 PMCID: PMC7526544 DOI: 10.1172/jci.insight.124465] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 08/05/2020] [Indexed: 12/20/2022] Open
Abstract
Acute graft-versus-host disease (aGVHD) can occur after hematopoietic cell transplant in patients undergoing treatment for hematological malignancies or inborn errors. Although CD4+ T helper (Th) cells play a major role in aGVHD, the mechanisms by which they contribute, particularly within the intestines, have remained elusive. We have identified a potentially novel subset of Th cells that accumulated in the intestines and produced the serine protease granzyme A (GrA). GrA+ Th cells were distinct from other Th lineages and exhibited a noncytolytic phenotype. In vitro, GrA+ Th cells differentiated in the presence of IL-4, IL-6, and IL-21 and were transcriptionally unique from cells cultured with either IL-4 or the IL-6/IL-21 combination alone. In vivo, both STAT3 and STAT6 were required for GrA+ Th cell differentiation and played roles in maintenance of the lineage identity. Importantly, GrA+ Th cells promoted aGVHD-associated morbidity and mortality and contributed to crypt destruction within intestines but were not required for the beneficial graft-versus-leukemia effect. Our data indicate that GrA+ Th cells represent a distinct Th subset and are critical mediators of aGVHD.
Collapse
Affiliation(s)
- Sungtae Park
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Brad Griesenauer
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hua Jiang
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Djamilatou Adom
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Srishti Chakravorty
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, Indiana, USA
| | - Majid Kazemian
- Departments of Biochemistry and Computer Science, Purdue University, West Lafayette, Indiana, USA
| | - Tanbeena Imam
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and
| | - Rajneesh Srivastava
- Department of Biohealth Informatics, School of Informatics and Computing, Indiana University-Purdue University, Indianapolis, Indianapolis, Indiana, USA
| | - Tristan A Hayes
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and
| | - Julian Pardo
- Biomedical Research Centre of Aragon (CIBA), Department of Microbiology, Preventative Medicine and Public Health, Nanoscience Institute of Aragon (INA), Aragon I+D Foundation, IIS Aragon/University of Zaragoza, Zaragoza, Spain
| | - Sarath Chandra Janga
- Department of Biohealth Informatics, School of Informatics and Computing, Indiana University-Purdue University, Indianapolis, Indianapolis, Indiana, USA
| | - Sophie Paczesny
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mark H Kaplan
- Department of Pediatrics and Herman B Wells Center for Pediatric Research and.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matthew R Olson
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
24
|
Saldanha MG, Pagliari C, Queiroz A, Machado PRL, Carvalho L, Scott P, Carvalho EM, Arruda S. Tissue Damage in Human Cutaneous Leishmaniasis: Correlations Between Inflammatory Cells and Molecule Expression. Front Cell Infect Microbiol 2020; 10:355. [PMID: 32766167 PMCID: PMC7381142 DOI: 10.3389/fcimb.2020.00355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/09/2020] [Indexed: 11/28/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is caused by the bite of the infected sand fly, which inoculates parasites of Leishmania spp and triggers an immune response. An exacerbated cutaneous inflammatory response is crucial for controlling parasite burden but can also promote tissue damage. This study aimed to characterize the populations of natural killer (NK), CD57+, CD4+, and CD8+ T cells, CD20+ B cells, as well as CD68+ macrophages, in biopsies of ulcerated CL lesions, and quantify the production of perforin+, grazyme B+, interleukin 1 beta (IL-1β+) and Tumor Necrosis Factor (TNF-α+ cells). We then correlated these parameters with necrosis, inflammation and the number of amastigotes. CD4+ T cells were positively correlated to the extent of inflammation, B cells and IL-1β+ were associated with the extent of necrosis, CD68+ macrophages and perforin were correlated with the number of amastigotes, and CD57+ NK cells was correlated to CD68+ macrophages and amastigotes. In sum, the finding suggests that the production of cytotoxic granules and cytokines by inflammatory cells contributes to tissue damage in CL lesions.
Collapse
Affiliation(s)
| | - Carla Pagliari
- Departamento de Patologia, Faculdade de Medicina, Universidade São Paulo, São Paulo, Brazil
| | - Adriano Queiroz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
| | - Paulo Roberto Lima Machado
- Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Lucas Carvalho
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Edgar M Carvalho
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Serviço de Imunologia, Complexo Hospitalar Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Brazil
| | - Sérgio Arruda
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil.,Departamento de Ciências de Vida, Universidade Estadual da Bahia, Salvador, Brazil
| |
Collapse
|
25
|
Garzón-Tituaña M, Arias MA, Sierra-Monzón JL, Morte-Romea E, Santiago L, Ramirez-Labrada A, Martinez-Lostao L, Paño-Pardo JR, Galvez EM, Pardo J. The Multifaceted Function of Granzymes in Sepsis: Some Facts and a Lot to Discover. Front Immunol 2020; 11:1054. [PMID: 32655547 PMCID: PMC7325996 DOI: 10.3389/fimmu.2020.01054] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/30/2020] [Indexed: 12/21/2022] Open
Abstract
Sepsis is a serious global health problem. In addition to a high incidence, this syndrome has a high mortality and is responsible for huge health expenditure. The pathophysiology of sepsis is very complex and it is not well-understood yet. However, it is widely accepted that the initial phase of sepsis is characterized by a hyperinflammatory response while the late phase is characterized by immunosuppression and immune anergy, increasing the risk of secondary infections. Granzymes (Gzms) are a family of serine proteases classified according to their cleavage specificity. Traditionally, it was assumed that all Gzms acted as cytotoxic proteases. However, recent evidence suggests that GzmB is the one with the greatest cytotoxic capacity, while the cytotoxicity of others such as GzmA and GzmK is not clear. Recent studies have found that GzmA, GzmB, GzmK, and GzmM act as pro-inflammatory mediators. Specially, solid evidences show that GzmA and GzmK function as extracellular proteases that regulate the inflammatory response irrespectively of its ability to induce cell death. Indeed, studies in animal models indicate that GzmA is involved in the cytokine release syndrome characteristic of sepsis. Moreover, the GZM family also could regulate other biological processes involved in sepsis pathophysiology like the coagulation cascade, platelet function, endothelial barrier permeability, and, in addition, could be involved in the immunosuppressive stage of sepsis. In this review, we provide a comprehensive overview on the contribution of these novel functions of Gzms to sepsis and the new therapeutic opportunities emerging from targeting these proteases for the treatment of this serious health problem.
Collapse
Affiliation(s)
- Marcela Garzón-Tituaña
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | | | - José L Sierra-Monzón
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Elena Morte-Romea
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Llipsy Santiago
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Ariel Ramirez-Labrada
- Nanotoxicology and Immunotoxicology Unit (UNATI), Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain
| | - Luis Martinez-Lostao
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain.,Nanoscience Institute of Aragon (INA), University of Zaragoza, Zaragoza, Spain
| | - José R Paño-Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Eva M Galvez
- Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain
| | - Julián Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Nanoscience Institute of Aragon (INA), University of Zaragoza, Zaragoza, Spain.,Aragon I + D Foundation (ARAID), Zaragoza, Spain.,Department of Biochemistry and Molecular and Cell Biology and Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
26
|
Zhang Y, Fung ITH, Sankar P, Chen X, Robison LS, Ye L, D'Souza SS, Salinero AE, Kuentzel ML, Chittur SV, Zhang W, Zuloaga KL, Yang Q. Depletion of NK Cells Improves Cognitive Function in the Alzheimer Disease Mouse Model. THE JOURNAL OF IMMUNOLOGY 2020; 205:502-510. [PMID: 32503894 DOI: 10.4049/jimmunol.2000037] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022]
Abstract
Despite mounting evidence suggesting the involvement of the immune system in regulating brain function, the specific role of immune and inflammatory cells in neurodegenerative diseases remain poorly understood. In this study, we report that depletion of NK cells, a type of innate lymphocytes, alleviates neuroinflammation, stimulates neurogenesis, and improves cognitive function in a triple-transgenic Alzheimer disease (AD) mouse model. NK cells in the brains of triple-transgenic AD mouse model (3xTg-AD) mice exhibited an enhanced proinflammatory profile. Depletion of NK cells by anti-NK1.1 Abs drastically improved cognitive function of 3xTg-AD mice. NK cell depletion did not affect amyloid β concentrations but enhanced neurogenesis and reduced neuroinflammation. Notably, in 3xTg-AD mice depleted of NK cells, microglia demonstrated a homeostatic-like morphology, decreased proliferative response and reduced expression of neurodestructive proinflammatory cytokines. Together, our results suggest a proinflammatory role for NK cells in 3xTg-AD mice and indicate that targeting NK cells might unlock novel strategies to combat AD.
Collapse
Affiliation(s)
- Yuanyue Zhang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Ivan Ting Hin Fung
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Poornima Sankar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Xiangyu Chen
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208
| | - Longyun Ye
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Shanti S D'Souza
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208
| | - Marcy L Kuentzel
- Center for Functional Genomics, University at Albany-SUNY, Rensselaer, NY 12144; and
| | - Sridar V Chittur
- Center for Functional Genomics, University at Albany-SUNY, Rensselaer, NY 12144; and
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY 12208
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY 12208;
| | - Qi Yang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208;
| |
Collapse
|
27
|
van Daalen KR, Reijneveld JF, Bovenschen N. Modulation of Inflammation by Extracellular Granzyme A. Front Immunol 2020; 11:931. [PMID: 32508827 PMCID: PMC7248576 DOI: 10.3389/fimmu.2020.00931] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022] Open
Abstract
Granzyme A (GrA) has long been recognized as one of the key players in the induction of cell death of neoplastic, foreign or infected cells after granule delivery by cytotoxic cells. While the cytotoxic potential of GrA is controversial in current literature, accumulating evidence now indicates roles for extracellular GrA in modulating inflammation and inflammatory diseases. This paper aims to explore the literature presenting current knowledge on GrA as an extracellular modulator of inflammation by summarizing (i) the presence and role of extracellular GrA in several inflammatory diseases, and (ii) the potential molecular mechanisms of extracellular GrA in augmenting inflammation.
Collapse
Affiliation(s)
- Kim R. van Daalen
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | | | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
28
|
Schanoski AS, Le TT, Kaiserman D, Rowe C, Prow NA, Barboza DD, Santos CA, Zanotto PMA, Magalhães KG, Aurelio L, Muller D, Young P, Zhao P, Bird PI, Suhrbier A. Granzyme A in Chikungunya and Other Arboviral Infections. Front Immunol 2020; 10:3083. [PMID: 31993061 PMCID: PMC6971054 DOI: 10.3389/fimmu.2019.03083] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/17/2019] [Indexed: 12/23/2022] Open
Abstract
Granzyme A (GzmA) is secreted by cytotoxic lymphocytes and has traditionally been viewed as a mediator of cell death. However, a growing body of data suggests the physiological role of GzmA is promotion of inflammation. Here, we show that GzmA is significantly elevated in the sera of chikungunya virus (CHIKV) patients and that GzmA levels correlated with viral loads and disease scores in these patients. Serum GzmA levels were also elevated in CHIKV mouse models, with NK cells the likely source. Infection of mice deficient in type I interferon responses with CHIKV, Zika virus, or dengue virus resulted in high levels of circulating GzmA. We also show that subcutaneous injection of enzymically active recombinant mouse GzmA was able to mediate inflammation, both locally at the injection site as well as at a distant site. Protease activated receptors (PARs) may represent targets for GzmA, and we show that treatment with PAR antagonist ameliorated GzmA- and CHIKV-mediated inflammation.
Collapse
Affiliation(s)
| | - Thuy T Le
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Dion Kaiserman
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Caitlin Rowe
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Natalie A Prow
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Australian Infectious Disease Research Centre, University of Queensland, Brisbane, QLD, Australia
| | - Diego D Barboza
- Bacteriology Laboratory, Butantan Institute, São Paulo, Brazil
| | - Cliomar A Santos
- Health Foundation Parreiras Horta, Central Laboratory of Public Health, State Secretary for Health, Aracajú, Brazil
| | - Paolo M A Zanotto
- Laboratory of Molecular Evolution and Bioinformatics, Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Kelly G Magalhães
- Laboratory of Immunology and Inflammation, University of Brasilia, Brasilia, Brazil
| | - Luigi Aurelio
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - David Muller
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Paul Young
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| | - Peishen Zhao
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Phillip I Bird
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Australian Infectious Disease Research Centre, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
29
|
Vigneron C, Mirouse A, Merdji H, Rousseau C, Cousin C, Alby-Laurent F, Mira JP, Chiche JD, Llitjos JF, Pène F. Sepsis inhibits tumor growth in mice with cancer through Toll-like receptor 4-associated enhanced Natural Killer cell activity. Oncoimmunology 2019; 8:e1641391. [PMID: 31646090 DOI: 10.1080/2162402x.2019.1641391] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/26/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis-induced immune dysfunctions are likely to impact on malignant tumor growth. Sequential sepsis-then-cancer models of tumor transplantation in mice recovering from sepsis have shown that the post-septic immunosuppressive environment was able to promote tumor growth. We herein addressed the impact of sepsis on pre-established malignancy in a reverse cancer-then sepsis experimental model. Mice previously inoculated with MCA205 fibrosarcoma cells were subjected to septic challenges by polymicrobial peritonitis induced by cecal ligation and puncture or endotoxinic shock. The anti-tumoral immune response was assessed through the distribution of tumor-infiltrating immune cells, as well as the functions of cytotoxic cells. As compared to sham surgery, polymicrobial sepsis dampened malignant tumor growth in wild-type (WT) mice, but neither in Toll-like receptor 4 (Tlr4)-/- nor in Myd88-/- mice. Similar tumor growth inhibition was observed following a LPS challenge in WT mice, suggesting a regulatory role of Tlr4 in this setting. The low expression of MHC class 1 onto MCA205 cells suggested the involvement of Natural Killer (NK) cells in sepsis-induced tumor inhibition. Septic insults applied to mice with cancer promoted the main anti-tumoral NK functions of IFNγ production and degranulation. The anti-tumoral properties of NK cells obtained from septic mice were exacerbated when cultured with MHC1low MCA205 or YAC-1 cells. These results suggest that sepsis may harbor dual effects on tumor growth depending on the sequential experimental model. When applied in mice with cancer, sepsis prevents tumor growth in a Tlr4-dependent manner by enhancing the anti-tumoral functions of NK cells.
Collapse
Affiliation(s)
- Clara Vigneron
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Adrien Mirouse
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Hamid Merdji
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Christophe Rousseau
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Clément Cousin
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Fanny Alby-Laurent
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Jean-Paul Mira
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-Daniel Chiche
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Jean-François Llitjos
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Frédéric Pène
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris, France.,Université Paris Descartes, Paris, France.,Médecine Intensive & Réanimation, hôpital Cochin, Hôpitaux Universitaires Paris Centre, Assistance Publique - Hôpitaux de Paris, Paris, France
| |
Collapse
|
30
|
Kumar V. Natural killer cells in sepsis: Underprivileged innate immune cells. Eur J Cell Biol 2019; 98:81-93. [DOI: 10.1016/j.ejcb.2018.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/15/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
|
31
|
Egui A, Ledesma D, Pérez-Antón E, Montoya A, Gómez I, Robledo SM, Infante JJ, Vélez ID, López MC, Thomas MC. Phenotypic and Functional Profiles of Antigen-Specific CD4 + and CD8 + T Cells Associated With Infection Control in Patients With Cutaneous Leishmaniasis. Front Cell Infect Microbiol 2018; 8:393. [PMID: 30510917 PMCID: PMC6252334 DOI: 10.3389/fcimb.2018.00393] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/19/2018] [Indexed: 01/03/2023] Open
Abstract
The host immunological response is a key factor determining the pathogenesis of cutaneous leishmaniasis. It is known that a Th1 cellular response is associated with infection control and that antigen-specific memory T cells are necessary for the development of a rapid and strong protective cellular response. The present manuscript reports the analysis of the functional and phenotypic profiles of antigen-specific CD4+ and CD8+ T cells from patients cured of cutaneous leishmaniasis (CL), patients with an active process of cutaneous leishmaniasis, asymptomatic individuals with a positive Montenegro test and healthy donors (HD). Peripheral blood mononuclear cells (PBMCs) from the patients exhibited a lymphoproliferative capacity after stimulation with total soluble protein from either Leishmania panamensis (SLpA) or Leishmania infantum (SLiA) or with a recombinant paraflagellar rod protein-1 (rPFR1). Higher frequencies of antigen-specific TNAIVE cells, mainly following stimulation with rPFR1, were observed in asymptomatic and cured patients than in patients with active cutaneous leishmaniasis, while T cells from patients with active cutaneous leishmaniasis showed a higher percentage of effector memory T cells (TEM for CD4+ T cells and TEMRA for CD8+ T cells). The amount of antigen-specific CD57+/CD8+ TEMRA cells in patients with active cutaneous leishmaniasis was higher than that in cured patients and asymptomatic subjects. Regarding functionality, a more robust multifunctional CD8+ T cell response was detected in cured patients than in those with active cutaneous leishmaniasis. Moreover, cured patients showed a significant increase in the frequency of cells expressing a Th1-type cytotoxic production profile (IFN-γ+/granzyme-B/+perforin+). Patients with an active leishmaniosis process had a significantly higher frequency of CD8+ T cells expressing the inhibitory CD160 and 2B4 receptors than did cured patients. The expression profile observed in cured patients could be indicative of an imbalance toward a CD8+ Th1 response, which could be associated with infection control; consequently, the determination of this profile could be a useful tool for facilitating the clinical follow-up of patients with cutaneous leishmaniasis. The results also suggest a possible exhaustion process of CD8+ T cells associated with the evolution of Leishmania infection.
Collapse
Affiliation(s)
- Adriana Egui
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Darién Ledesma
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Elena Pérez-Antón
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Andrés Montoya
- Programa de Estudio y Control de Enfermedades Tropicales, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Inmaculada Gómez
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Sara María Robledo
- Programa de Estudio y Control de Enfermedades Tropicales, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Juan José Infante
- Bionaturis Group, Bioorganic Research and Services, S.A., Jerez de la Frontera, Spain
| | - Ivan Darío Vélez
- Programa de Estudio y Control de Enfermedades Tropicales, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Manuel C López
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - M Carmen Thomas
- Molecular Biology Department, Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
32
|
Jensen IJ, Winborn CS, Fosdick MG, Shao P, Tremblay MM, Shan Q, Tripathy SK, Snyder CM, Xue HH, Griffith TS, Houtman JC, Badovinac VP. Polymicrobial sepsis influences NK-cell-mediated immunity by diminishing NK-cell-intrinsic receptor-mediated effector responses to viral ligands or infections. PLoS Pathog 2018; 14:e1007405. [PMID: 30379932 PMCID: PMC6231673 DOI: 10.1371/journal.ppat.1007405] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 11/12/2018] [Accepted: 10/13/2018] [Indexed: 12/14/2022] Open
Abstract
The sepsis-induced cytokine storm leads to severe lymphopenia and reduced effector capacity of remaining/surviving cells. This results in a prolonged state of immunoparalysis, that contributes to enhanced morbidity/mortality of sepsis survivors upon secondary infection. The impact of sepsis on several lymphoid subsets has been characterized, yet its impact on NK-cells remains underappreciated-despite their critical role in controlling infection(s). Here, we observed numerical loss of NK-cells in multiple tissues after cecal-ligation-and-puncture (CLP)-induced sepsis. To elucidate the sepsis-induced lesions in surviving NK-cells, transcriptional profiles were evaluated and indicated changes consistent with impaired effector functionality. A corresponding deficit in NK-cell capacity to produce effector molecules following secondary infection and/or cytokine stimulation (IL-12,IL-18) further suggested a sepsis-induced NK-cell intrinsic impairment. To specifically probe NK-cell receptor-mediated function, the activating Ly49H receptor, that recognizes the murine cytomegalovirus (MCMV) m157 protein, served as a model receptor. Although relative expression of Ly49H receptor did not change, the number of Ly49H+ NK-cells in CLP hosts was reduced leading to impaired in vivo cytotoxicity and the capacity of NK-cells (on per-cell basis) to perform Ly49H-mediated degranulation, killing, and effector molecule production in vitro was also severely reduced. Mechanistically, Ly49H adaptor protein (DAP12) activation and clustering, assessed by TIRF microscopy, was compromised. This was further associated with diminished AKT phosphorylation and capacity to flux calcium following receptor stimulation. Importantly, DAP12 overexpression in NK-cells restored Ly49H/D receptors-mediated effector functions in CLP hosts. Finally, as a consequence of sepsis-dependent numerical and functional lesions in Ly49H+ NK-cells, host capacity to control MCMV infection was significantly impaired. Importantly, IL-2 complex (IL-2c) therapy after CLP improved numbers but not a function of NK-cells leading to enhanced immunity to MCMV challenge. Thus, the sepsis-induced immunoparalysis state includes numerical and NK-cell-intrinsic functional impairments, an instructive notion for future studies aimed in restoring NK-cell immunity in sepsis survivors.
Collapse
Affiliation(s)
- Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Christina S. Winborn
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Micaela G. Fosdick
- Interdisciplinary Graduate Program in Molecular Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Peng Shao
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Mikaela M. Tremblay
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Qiang Shan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Sandeep Kumar Tripathy
- Gastroenterology Division, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Christopher M. Snyder
- Department of Immunology and Microbiology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Hai-Hui Xue
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Thomas S. Griffith
- Microbiology, Immunology, and Cancer Biology Ph.D. Program, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Urology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Minneapolis VA Health Care, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jon C. Houtman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Interdisciplinary Graduate Program in Molecular Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
33
|
Jaime-Sánchez P, Catalán E, Uranga-Murillo I, Aguiló N, Santiago L, M Lanuza P, de Miguel D, A Arias M, Pardo J. Antigen-specific primed cytotoxic T cells eliminate tumour cells in vivo and prevent tumour development, regardless of the presence of anti-apoptotic mutations conferring drug resistance. Cell Death Differ 2018; 25:1536-1548. [PMID: 29743559 DOI: 10.1038/s41418-018-0112-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 02/06/2023] Open
Abstract
Cytotoxic CD8+ T (Tc) cells are the main executors of transformed and cancer cells during cancer immunotherapy. The latest clinical results evidence a high efficacy of novel immunotherapy agents that modulate Tc cell activity against bad prognosis cancers. However, it has not been determined yet whether the efficacy of these treatments can be affected by selection of tumoural cells with mutations in the cell death machinery, known to promote drug resistance and cancer recurrence. Here, using a model of prophylactic tumour vaccination based on the LCMV-gp33 antigen and the mouse EL4 T lymphoma, we analysed the molecular mechanism employed by Tc cells to eliminate cancer cells in vivo and the impact of mutations in the apoptotic machinery on tumour development. First of all, we found that Tc cells, and perf and gzmB are required to efficiently eliminate EL4.gp33 cells after LCMV immunisation during short-term assays (1-4 h), and to prevent tumour development in the long term. Furthermore, we show that antigen-pulsed chemoresistant EL4 cells overexpressing Bcl-XL or a dominant negative form of caspase-3 are specifically eliminated from the peritoneum of infected animals, as fast as parental EL4 cells. Notably, antigen-specific Tc cells control the tumour growth of the mutated cells, as efficiently as in the case of parental cells. Altogether, expression of the anti-apoptotic mutations does not confer any advantage for tumour cells neither in the short-term survival nor in long-term tumour formation. Although the mechanism involved in the elimination of the apoptosis-resistant tumour cells is not completely elucidated, neither necroptosis nor pyroptosis seem to be involved. Our results provide the first experimental proof that chemoresistant cancer cells with mutations in the main cell death pathways are efficiently eliminated by Ag-specific Tc cells in vivo during immunotherapy and, thus, provide the molecular basis to treat chemoresistant cancer cells with CD8 Tc-based immunotherapy.
Collapse
Affiliation(s)
- Paula Jaime-Sánchez
- Biomedical Research Centre of Aragon (CIBA), IIS Aragon/University of Zaragoza, Zaragoza, Spain
| | - Elena Catalán
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| | - Iratxe Uranga-Murillo
- Biomedical Research Centre of Aragon (CIBA), IIS Aragon/University of Zaragoza, Zaragoza, Spain
| | - Nacho Aguiló
- Dept. Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain.,CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Llipsy Santiago
- Biomedical Research Centre of Aragon (CIBA), IIS Aragon/University of Zaragoza, Zaragoza, Spain
| | - Pilar M Lanuza
- Biomedical Research Centre of Aragon (CIBA), IIS Aragon/University of Zaragoza, Zaragoza, Spain
| | - Diego de Miguel
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Maykel A Arias
- Biomedical Research Centre of Aragon (CIBA), IIS Aragon/University of Zaragoza, Zaragoza, Spain.
| | - Julián Pardo
- Biomedical Research Centre of Aragon (CIBA), IIS Aragon/University of Zaragoza, Zaragoza, Spain. .,Dept. Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain. .,Nanoscience Institute of Aragon (INA), University of Zaragoza, Zaragoza, Spain. .,Aragon I+D Foundation, Zaragoza, Spain.
| |
Collapse
|
34
|
Multiplex Cytokine Profiling Identifies Interleukin-27 as a Novel Biomarker For Neonatal Early Onset Sepsis. Shock 2018; 47:140-147. [PMID: 27648693 DOI: 10.1097/shk.0000000000000753] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Early onset sepsis (EOS) remains a major cause of mortality and morbidity in neonates, and traditional clinical markers effective for adults are less effective in these patients. This study aimed to assess the value of individual plasma biomarkers as well as biomarker combinations for predicting EOS in neonates. METHODS This prospective study included 151 neonates with suspected EOS. Plasma levels of interleukin (IL)-27, IL-6, IL-8, tumor necrosis factor (TNF)-α, heat shock protein (HSP) 70, macrophage inflammatory protein (MIP)-1α, MIP-1β, granzyme B, and matrix metalloproteinase (MMP)-8 were measured through multiplex cytokine profiling and assessed along with C-reactive protein (CRP) and procalcitonin (PCT). Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive ability of biomarkers individually and in combination. Logistic regression model was constructed to identify independent predictors of EOS. RESULTS The proven sepsis and probable sepsis groups were combined to form the infected group (n = 68), and the possible sepsis and low-risk sepsis groups were combined to form the uninfected group (n = 83). The ROC area under the curve was 0.747 for IL-27 (P <0.01). In addition, IL-6, TNF-α, HSP 70, MMP-8, PCT, and CRP were significantly predictive of EOS, whereas IL-8, granzyme B, MIP-1α, and MIP-1β were not. Both IL-27 and PCT were identified as independent predictors of EOS in the multivariate model, and the combined use of these markers showed significantly increased predictive ability for EOS. CONCLUSION Our results indicate that elevated IL-27 strongly correlates with EOS and may provide additional diagnostic value along with PCT.
Collapse
|
35
|
Immunoproteasome Subunits Are Required for CD8 + T Cell Function and Host Resistance to Brucella abortus Infection in Mice. Infect Immun 2018; 86:IAI.00615-17. [PMID: 29263103 DOI: 10.1128/iai.00615-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 12/12/2017] [Indexed: 01/18/2023] Open
Abstract
The immunoproteasome is a specific proteasome isoform composed of three subunits, termed β1i, β2i, and β5i. Its proteolytic activity enhances the quantity and quality of peptides to be presented by major histocompatibility complex class I (MHC-I) molecules to CD8+ T cells. However, the role of the combined deficiency of the three immunoproteasome subunits in protective immunity against bacterial pathogens has not been investigated. In this study, we addressed the role of the immunoproteasome during infection by Brucella abortus, an intracellular bacterium that requires CD8+ T cell responses for the control of infection. Here, we demonstrate that immunoproteasome triple-knockout (TKO) mice were more susceptible to Brucella infection. This observed susceptibility was accompanied by reduced interferon gamma (IFN-γ) production by mouse CD4+ and CD8+ T lymphocytes. Moreover, the absence of the immunoproteasome had an impact on MHC-I surface expression and antigen presentation by dendritic cells. CD8+ T cell function, which plays a pivotal role in B. abortus immunity, also presented a partial impairment of granzyme B expression and, consequently, reduced cytotoxic activity. In conclusion, these results strongly suggest that immunoproteasome subunits are important components in host resistance to B. abortus infection by impacting both the magnitude and quality of CD8+ T cell responses.
Collapse
|
36
|
Núñez D, Comas L, Lanuza PM, Sánchez-Martinez D, Pérez-Hernández M, Catalán E, Domingo MP, Velázquez-Campoy A, Pardo J, Gálvez EM. A Functional Analysis on the Interspecies Interaction between Mouse LFA-1 and Human Intercellular Adhesion Molecule-1 at the Cell Level. Front Immunol 2017; 8:1817. [PMID: 29312326 PMCID: PMC5742583 DOI: 10.3389/fimmu.2017.01817] [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: 04/06/2017] [Accepted: 12/04/2017] [Indexed: 01/31/2023] Open
Abstract
The interaction between intercellular adhesion molecules (ICAM) and the integrin leukocyte function-associated antigen-1 (LFA-1) is crucial for the regulation of several physiological and pathophysiological processes like cell-mediated elimination of tumor or virus infected cells, cancer metastasis, or inflammatory and autoimmune processes. Using purified proteins it was reported a species restriction for the interaction of ICAM-1 and LFA-1, being mouse ICAM-1 able to interact with human LFA-1 but not human ICAM-1 with mouse LFA-1. However, in vivo results employing tumor cells transfected with human ICAM-1 suggest that functionally mouse LFA-1 can recognize human ICAM-1. In order to clarify the interspecies cross-reactivity of the ICAM-1/LFA-1 interaction, we have performed functional studies analyzing the ability of human soluble ICAM-1 and human/mouse LFA-1 derived peptides to inhibit cell aggregation and adhesion as well as cell-mediated cytotoxicity in both mouse and human systems. In parallel, the affinity of the interaction between mouse LFA-1-derived peptides and human ICAM-1 was determined by calorimetry assays. According to the results obtained, it seems that human ICAM-1 is able to interact with mouse LFA-1 on intact cells, which should be taking into account when using humanized mice and xenograft models for the study of immune-related processes.
Collapse
Affiliation(s)
- David Núñez
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain.,Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain
| | - Laura Comas
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain.,Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain
| | - Pilar M Lanuza
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain
| | - Diego Sánchez-Martinez
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain
| | - Marta Pérez-Hernández
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain
| | - Elena Catalán
- Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain
| | | | - Adrián Velázquez-Campoy
- Department of Biochemistry and Molecular and Cell Biology, Fac. Ciencias, University of Zaragoza, Zaragoza, Spain.,Institute of Biocomputation and Physics of Complex Systems (BIFI), Unidad Asociada IQFR-CSIC-BIFI, Universidad de Zaragoza, Zaragoza, Spain.,Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain.,Nanoscience Institute of Aragon (INA), University of Zaragoza, Zaragoza, Spain.,Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Eva M Gálvez
- Immune Effector Cells Group, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Instituto de Carboquímica ICB-CSIC, Zaragoza, Spain
| |
Collapse
|
37
|
Campos TM, Costa R, Passos S, Carvalho LP. Cytotoxic activity in cutaneous leishmaniasis. Mem Inst Oswaldo Cruz 2017; 112:733-740. [PMID: 29091132 PMCID: PMC5661895 DOI: 10.1590/0074-02760170109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/10/2017] [Indexed: 01/27/2023] Open
Abstract
Cutaneous leishmaniasis (CL) is a chronic disease caused by species of the protozoan Leishmania and characterised by the presence of ulcerated skin lesions. Both parasite and host factors affect the clinical presentation of the disease. The development of skin ulcers in CL is associated with an inflammatory response mediated by cells that control parasite growth but also contribute to pathogenesis. CD8+ T cells contribute to deleterious inflammatory responses in patients with CL through cytotoxic mechanisms. In addition, natural killer cells also limit Leishmania infections by production of interferon-γ and cytotoxicity. In this review, we focus on studies of cytotoxicity in CL and its contribution to the pathogenesis of this disease.
Collapse
Affiliation(s)
- Taís M Campos
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA, Brasil.,Universidade Federal da Bahia, Faculdade de Medicina da Bahia, Programa de Pós-Graduação em Ciências da Saúde, Salvador, BA, Brasil
| | - Rúbia Costa
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA, Brasil.,Universidade Federal da Bahia, Faculdade de Medicina da Bahia, Programa de Pós-Graduação em Ciências da Saúde, Salvador, BA, Brasil
| | - Sara Passos
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA, Brasil.,Houston Methodist Research Institute, Department of Nanomedicine, Houston, TX, United States
| | - Lucas P Carvalho
- Universidade Federal da Bahia, Serviço de Imunologia, Salvador, BA, Brasil.,Universidade Federal da Bahia, Faculdade de Medicina da Bahia, Programa de Pós-Graduação em Ciências da Saúde, Salvador, BA, Brasil.,Instituto Nacional de Ciências e Tecnologia-Doenças Tropicais, Salvador, BA, Brasil.,Fundação Oswaldo Cruz-Fiocruz, Instituto Gonçalo Moniz, Laboratório Avançado de Saúde Pública, Salvador, BA, Brasil
| |
Collapse
|
38
|
Pérez-Hernández M, Moros M, Stepien G, Del Pino P, Menao S, de Las Heras M, Arias M, Mitchell SG, Pelaz B, Gálvez EM, de la Fuente JM, Pardo J. Multiparametric analysis of anti-proliferative and apoptotic effects of gold nanoprisms on mouse and human primary and transformed cells, biodistribution and toxicity in vivo. Part Fibre Toxicol 2017; 14:41. [PMID: 29073907 PMCID: PMC5658988 DOI: 10.1186/s12989-017-0222-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 10/17/2017] [Indexed: 12/02/2022] Open
Abstract
Background The special physicochemical properties of gold nanoprisms make them very useful for biomedical applications including biosensing and cancer therapy. However, it is not clear how gold nanoprisms may affect cellular physiology including viability and other critical functions. We report a multiparametric investigation on the impact of gold-nanoprisms on mice and human, transformed and primary cells as well as tissue distribution and toxicity in vivo after parental injection. Methods Cellular uptake of the gold-nanoprisms (NPRs) and the most crucial parameters of cell fitness such as generation of reactive oxygen species (ROS), mitochondria membrane potential, cell morphology and apoptosis were systematically assayed in cells. Organ distribution and toxicity including inflammatory response were analysed in vivo in mice at 3 days or 4 months after parental administration. Results Internalized gold-nanoprisms have a significant impact in cell morphology, mitochondrial function and ROS production, which however do not affect the potential of cells to proliferate and form colonies. In vivo NPRs were only detected in spleen and liver at 3 days and 4 months after administration, which correlated with some changes in tissue architecture. However, the main serum biochemical markers of organ damage and inflammation (TNFα and IFNγ) remained unaltered even after 4 months. In addition, animals did not show any macroscopic sign of toxicity and remained healthy during all the study period. Conclusion Our data indicate that these gold-nanoprisms are neither cytotoxic nor cytostatic in transformed and primary cells, and suggest that extensive parameters should be analysed in different cell types to draw useful conclusions on nanomaterials safety. Moreover, although there is a tendency for the NPRs to accumulate in liver and spleen, there is no observable negative impact on animal health. Electronic supplementary material The online version of this article (10.1186/s12989-017-0222-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marta Pérez-Hernández
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, 50009, Zaragoza, Spain. .,Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Centro de Investigación Biomédica de Aragón (CIBA), Universidad de Zaragoza, 50009, Zaragoza, Spain.
| | - María Moros
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, 50018, Zaragoza, Spain.,Institute of Applied Sciences and Intelligent Systems-CNR, Via Campi Flegrei, 34, 80078, Pozzuoli, Italy
| | - Grazyna Stepien
- Fundación Instituto Universitario de Nanociencia de Aragón (FINA), Universidad de Zaragoza, 50018, Zaragoza, Spain.,CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - Pablo Del Pino
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, 50018, Zaragoza, Spain.,Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CiQUS) y Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Sebastián Menao
- Departamento de Bioquímica clínica. H.C.U. Lozano Blesa, 50009, Zaragoza, Spain
| | - Marcelo de Las Heras
- Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, 50009, Zaragoza, Spain
| | - Maykel Arias
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Centro de Investigación Biomédica de Aragón (CIBA), Universidad de Zaragoza, 50009, Zaragoza, Spain
| | - Scott G Mitchell
- Instituto de Ciencia de Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza, Spain
| | - Beatriz Pelaz
- Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, 50018, Zaragoza, Spain.,Centro Singular de Investigación en Química Biológica y Materiales Moleculares (CiQUS) y Departamento de Física de Partículas, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Eva M Gálvez
- Instituto de Carboquímica ICB-CSIC, 50018, Zaragoza, Spain
| | - Jesús M de la Fuente
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain.,Instituto de Ciencia de Materiales de Aragón, CSIC-Universidad de Zaragoza, Zaragoza, Spain
| | - Julián Pardo
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Centro de Investigación Biomédica de Aragón (CIBA), Universidad de Zaragoza, 50009, Zaragoza, Spain.,Instituto Universitario de Nanociencia de Aragón (INA), Universidad de Zaragoza, 50018, Zaragoza, Spain.,Departamento de Microbiología, Medicina Preventiva y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, 50009, Zaragoza, Spain.,Aragón I+D Foundation (ARAID), Gobierno de Aragón, Zaragoza, Spain
| |
Collapse
|
39
|
Jaime P, García-Guerrero N, Estella R, Pardo J, García-Álvarez F, Martinez-Lostao L. CD56 +/CD16 - Natural Killer cells expressing the inflammatory protease granzyme A are enriched in synovial fluid from patients with osteoarthritis. Osteoarthritis Cartilage 2017; 25:1708-1718. [PMID: 28668542 DOI: 10.1016/j.joca.2017.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Natural killer (NK) cells have been involved in the pathology of different inflammatory and autoimmune disorders. Inflammation is an important regulator of osteoarthritis (OA), but the molecular and cellular mechanisms regulating this process are not well defined. DESIGN To understand the role of NK cells in OA, we have compared the phenotype (CD56 subsets and perforin and granzyme expression) and cytotoxic function of NK cells in peripheral blood and synovial fluid from patients with OA undergoing total knee arthroplasty. RESULTS In contrast to peripheral blood lymphocytes (PBLs), the majority of NK cells from the synovial fluid were CD56brightCD16(-) cells. As expected the expression of the cytolytic mediators perforin and granzyme B in CD56brightCD16(-) cells was low and correlated with a poor cytotoxic potential against K562 sensitive target cells. Surprisingly, this low cytotoxic NK cell subset expressed high levels of granzyme A (a protease recently characterized as a key modulator of inflammation in mouse models) in synovial fluid but not in peripheral blood. The presence of the CD56(+)brightCD16(-) cells expressing granzyme A correlated with increased levels of pro-inflammatory cytokines in synovial fluid from OA patients. CONCLUSION Our results indicate that NK cells from the synovium of patients with OA, which present an immunoregulatory non-cytotoxic phenotype, show different phenotype comparing with NK cells from peripheral blood, especially expressing granzyme A, a pro-inflammatory molecule which may contribute to the establishment of chronic articular inflammation in this type of patients.
Collapse
Affiliation(s)
- P Jaime
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (ISS Aragón), Zaragoza, Spain; Centro de Investigación Biomédica de Aragón (CIBA), Instituto de Investigaciones Sanitarias de Aragón (ISS Aragon), Zaragoza, Spain
| | - N García-Guerrero
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Investigación Sanitaria de Aragón (ISS Aragón), Zaragoza, Spain
| | - R Estella
- Centro de Investigación Biomédica de Aragón (CIBA), Instituto de Investigaciones Sanitarias de Aragón (ISS Aragon), Zaragoza, Spain; Servicio de Cirugía Ortopédica y Traumatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - J Pardo
- Centro de Investigación Biomédica de Aragón (CIBA), Instituto de Investigaciones Sanitarias de Aragón (ISS Aragon), Zaragoza, Spain; Departamento de Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Nanociencia de Aragón, IIS Aragón/Universidad de Zaragoza, Spain; Fundación Aragón I+D (ARAID), Gobierno de Aragón, Zaragoza, Spain.
| | - F García-Álvarez
- Centro de Investigación Biomédica de Aragón (CIBA), Instituto de Investigaciones Sanitarias de Aragón (ISS Aragon), Zaragoza, Spain; Servicio de Cirugía Ortopédica y Traumatología, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - L Martinez-Lostao
- Instituto de Investigación Sanitaria de Aragón (ISS Aragón), Zaragoza, Spain; Departamento de Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, Zaragoza, Spain; Instituto de Nanociencia de Aragón, IIS Aragón/Universidad de Zaragoza, Spain; Servicio de Inmunología Hospital Clínico Universitario Lorenzo Blesa, Zaragoza, Spain.
| |
Collapse
|
40
|
Santiago L, Menaa C, Arias M, Martin P, Jaime-Sánchez P, Metkar S, Comas L, Erill N, Gonzalez-Rumayor V, Esser E, Galvez EM, Raja S, Simon MM, Sprague SM, Gabay C, Martinez-Lostao L, Pardo J, Froelich CJ. Granzyme A Contributes to Inflammatory Arthritis in Mice Through Stimulation of Osteoclastogenesis. Arthritis Rheumatol 2017; 69:320-334. [PMID: 27598995 DOI: 10.1002/art.39857] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/25/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Granzyme A (GzmA) levels are elevated in the plasma and synovium of patients with rheumatoid arthritis (RA), suggesting involvement of this protease in the pathogenesis of the disease. GzmA contributes to sepsis by regulating the production of proinflammatory cytokines. The purpose of this study was to evaluate the contribution of GzmA to the pathogenesis of RA in vivo and to examine the possibility that GzmA acting via tumor necrosis factor (TNF) stimulates osteoclastogenesis. METHODS Inflammatory arthritis induced by type II collagen was evaluated in wild-type, GzmA-deficient, and perforin-deficient mice. The osteoclastogenic potential of GzmA was examined in vitro using bone marrow cells and colony-forming unit-granulocyte-macrophage (CFU-GM) cells and in vivo using GzmA-deficient mice. RESULTS Gene deletion of GzmA attenuated collagen-induced arthritis, including serum levels of proinflammatory cytokines, joint damage, and bone erosion in affected mice, suggesting that osteoclast activity is reduced in the absence of GzmA. Accordingly, GzmA-treated bone marrow cells produced multinucleated cells that fulfilled the criteria for mature osteoclasts: tartrate-resistant acid phosphatase (TRAP) activity, β integrin expression, calcitonin receptor expression, and resorptive activity on dentin slices. GzmA appeared to act without accessory cells, and its activity was not affected by osteoprotegerin, suggesting a minor contribution of RANKL. It also induced the expression and secretion of TNF. Neutralization of TNF or stimulation of CFU-GM cells from TNF-/- mice prevented GzmA-induced osteoclastogenesis. GzmA-deficient mice had reduced osteoclastogenesis in vivo (fewer calcitonin receptor-positive multinucleated cells and fewer transcripts for cathepsin K, matrix metalloproteinase 9, and TRAP in joints) and reduced serum levels of C-terminal telopeptide of type I collagen. CONCLUSION GzmA contributes to the joint destruction of RA partly by promoting osteoclast differentiation.
Collapse
Affiliation(s)
| | - Cheikh Menaa
- NorthShore University Healthcare System, Evanston, Illinois
| | - Maykel Arias
- Biomedical Research Centre of Aragon, Zaragoza, Spain
| | - Praxedis Martin
- University of Geneva and University Hospital, Geneva, Switzerland
| | | | - Sunil Metkar
- NorthShore University Healthcare System, Evanston, Illinois, and TheraTest Laboratories, Lombard, Illinois
| | - Laura Comas
- Biomedical Research Centre of Aragon and Instituto de Carboquímica, Zaragoza, Spain
| | | | | | - Erica Esser
- NorthShore University Healthcare System, Evanston, Illinois
| | | | - Sri Raja
- NorthShore University Healthcare System, Evanston, Illinois
| | - Markus M Simon
- Max Planck Institute for Immunology and Epigenetics, Freiburg, Germany
| | | | - Cem Gabay
- University of Geneva and University Hospital, Geneva, Switzerland
| | - Luis Martinez-Lostao
- Biomedical Research Centre of Aragon, University of Zaragoza, and Nanoscience Institute of Aragon, Zaragoza, Spain
| | - Julian Pardo
- Biomedical Research Centre of Aragon, University of Zaragoza, Fundación Aragon I+D, and Nanoscience Institute of Aragon, Zaragoza, Spain
| | | |
Collapse
|
41
|
Expression and Function of Granzymes A and B in Escherichia coli Peritonitis and Sepsis. Mediators Inflamm 2017; 2017:4137563. [PMID: 28694562 PMCID: PMC5485334 DOI: 10.1155/2017/4137563] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/05/2017] [Accepted: 04/20/2017] [Indexed: 12/14/2022] Open
Abstract
Escherichia (E.) coli is the most common causative pathogen in peritonitis, the second most common cause of sepsis. Granzymes (gzms) are serine proteases traditionally implicated in cytotoxicity and, more recently, in the inflammatory response. We here sought to investigate the role of gzms in the host response to E. coli-induced peritonitis and sepsis in vivo. For this purpose, we used a murine model of E. coli intraperitoneal infection, resembling the clinical condition commonly associated with septic peritonitis by this bacterium, in wild-type and gzmA-deficient (gzmA−/−), gzmB−/−, and gzmAxB−/−mice. GzmA and gzmB were predominantly expressed by natural killer cells, and during abdominal sepsis, the percentage of these cells expressing gzms in peritoneal lavage fluid decreased, while the amount of expression in the gzm+ cells increased. Deficiency of gzmA and/or gzmB was associated with increased bacterial loads, especially in the case of gzmB at the primary site of infection at late stage sepsis. While gzm deficiency did not impact neutrophil recruitment into the abdominal cavity, it was accompanied by enhanced nucleosome release at the primary site of infection, earlier hepatic necrosis, and more renal dysfunction. These results suggest that gzms influence bacterial growth and the host inflammatory response during abdominal sepsis caused by E. coli.
Collapse
|
42
|
Arias M, Martínez-Lostao L, Santiago L, Ferrandez A, Granville DJ, Pardo J. The Untold Story of Granzymes in Oncoimmunology: Novel Opportunities with Old Acquaintances. Trends Cancer 2017; 3:407-422. [PMID: 28718416 DOI: 10.1016/j.trecan.2017.04.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 12/16/2022]
Abstract
For more than 20 years perforin and granzymes (GZMs) have been recognized as key cell death executors of cytotoxic T (Tc) and natural killer (NK) cells during cancer immunosurveillance. In immune surveillance, perforin and GZMB, the most potent cytotoxic molecules, act mainly as antitumoral and anti-infectious factors. However, when expressed by immune regulatory cells they may contribute to immune evasion of specific cancer types. By contrast, the other major granzyme, GZMA, seems not to play a major role in Tc/NK cell-mediated cytotoxicity, but acts as a proinflammatory cytokine that might contribute to cancer development. Members of the GZM family also regulate other biological processes unrelated to cell death, such as angiogenesis, vascular integrity, extracellular matrix remodeling, and barrier function, all of which contribute to cancer initiation and progression. Thus, a new paradigm is emerging in the field of oncoimmunology. Can GZMs act as protumoral factors under some circumstances? We review the diverse roles of GZMs in cancer progression, and new therapeutic opportunities emerging from targeting these protumoral roles.
Collapse
Affiliation(s)
- Maykel Arias
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; These authors contributed equally to this work
| | - Luis Martínez-Lostao
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Department of Biochemistry and Molecular and Cell Biology, and Department of Microbiology, Preventive Medicine, and Public Health, University of Zaragoza, 50009 Zaragoza, Spain; Servicio de Inmunología Hospital Clínico Universitario Lorenzo Blesa, Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; These authors contributed equally to this work
| | - Llipsy Santiago
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain
| | - Angel Ferrandez
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Servicio de Aparato Digestivo, Hospital Clínico Universitario Lorenzo Blesa, Zaragoza, Spain
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Julián Pardo
- Fundación Instituto de Investigación Sanitaria Aragón (IIS Aragón), Biomedical Research Centre of Aragon (CIBA), 50009 Zaragoza, Spain; Department of Biochemistry and Molecular and Cell Biology, and Department of Microbiology, Preventive Medicine, and Public Health, University of Zaragoza, 50009 Zaragoza, Spain; Nanoscience Institute of Aragon (INA), University of Zaragoza, 50018 Zaragoza, Spain; Aragon I+D Foundation (ARAID), Zaragoza, Spain.
| |
Collapse
|
43
|
Cheuk S, Martini E, Bergh K, Chang D, Rethi B, Ståhle M, Eidsmo L. Granzyme A potentiates chemokine production in IL-17-stimulated keratinocytes. Exp Dermatol 2017; 26:824-827. [DOI: 10.1111/exd.13284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Stanley Cheuk
- Dermatology and Venereology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| | - Elisa Martini
- Dermatology and Venereology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| | - Kerstin Bergh
- Dermatology and Venereology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| | - David Chang
- Dermatology and Venereology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| | - Bence Rethi
- Rheumatology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| | - Mona Ståhle
- Dermatology and Venereology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| | - Liv Eidsmo
- Dermatology and Venereology Unit; Department of Medicine; Karolinska Institutet, Karolinska University Hospital; Solna Stockholm Sweden
| |
Collapse
|
44
|
van Eck JA, Shan L, Meeldijk J, Hack CE, Bovenschen N. A novel proinflammatory role for granzyme A. Cell Death Dis 2017; 8:e2630. [PMID: 28230859 PMCID: PMC5386495 DOI: 10.1038/cddis.2017.56] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Jacqueline A van Eck
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Liling Shan
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Jan Meeldijk
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Niels Bovenschen
- Department of Pathology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht 3584 CX, The Netherlands
| |
Collapse
|
45
|
RNA-Seq analysis of chikungunya virus infection and identification of granzyme A as a major promoter of arthritic inflammation. PLoS Pathog 2017; 13:e1006155. [PMID: 28207896 PMCID: PMC5312928 DOI: 10.1371/journal.ppat.1006155] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023] Open
Abstract
Chikungunya virus (CHIKV) is an arthritogenic alphavirus causing epidemics of acute and chronic arthritic disease. Herein we describe a comprehensive RNA-Seq analysis of feet and lymph nodes at peak viraemia (day 2 post infection), acute arthritis (day 7) and chronic disease (day 30) in the CHIKV adult wild-type mouse model. Genes previously shown to be up-regulated in CHIKV patients were also up-regulated in the mouse model. CHIKV sequence information was also obtained with up to ≈8% of the reads mapping to the viral genome; however, no adaptive viral genome changes were apparent. Although day 2, 7 and 30 represent distinct stages of infection and disease, there was a pronounced overlap in up-regulated host genes and pathways. Type I interferon response genes (IRGs) represented up to ≈50% of up-regulated genes, even after loss of type I interferon induction on days 7 and 30. Bioinformatic analyses suggested a number of interferon response factors were primarily responsible for maintaining type I IRG induction. A group of genes prominent in the RNA-Seq analysis and hitherto unexplored in viral arthropathies were granzymes A, B and K. Granzyme A-/- and to a lesser extent granzyme K-/-, but not granzyme B-/-, mice showed a pronounced reduction in foot swelling and arthritis, with analysis of granzyme A-/- mice showing no reductions in viral loads but reduced NK and T cell infiltrates post CHIKV infection. Treatment with Serpinb6b, a granzyme A inhibitor, also reduced arthritic inflammation in wild-type mice. In non-human primates circulating granzyme A levels were elevated after CHIKV infection, with the increase correlating with viral load. Elevated granzyme A levels were also seen in a small cohort of human CHIKV patients. Taken together these results suggest granzyme A is an important driver of arthritic inflammation and a potential target for therapy. Trial Registration: ClinicalTrials.gov NCT00281294 The largest chikungunya virus (CHIKV) epidemic ever recorded began in 2004 in Africa and spread across Asia reaching Europe and recently the Americas, with millions of cases reported. We undertook a detailed analysis of the mRNA expression profile during acute and chronic arthritis in an adult wild-type mouse model of CHIKV infection and disease. Gene induction profiles showed a high concordance with published human data, providing some validation of the mouse model. The host response was overwhelmingly dominated by type I interferon response genes, even after type I interferon induction was lost. The analysis also provided information on CHIKV RNA, with no adaptive viral genome changes identified. An important goal of the analysis was to identify new players in arthritic inflammation. Granzyme A was prominent in the RNA-Seq data and granzyme A deficient mice showed reduced arthritis, with no effects on viral loads. Arthritic disease could also be ameliorated in wild-type mice with a granzyme A inhibitor. Elevated circulating granzyme A levels were seen in non-human primates infected with CHIKV and in human CHIKV patients. Granzyme A thus emerges to be a major driver of CHIKV-mediated arthritic inflammation and a potential target for anti-inflammatory interventions.
Collapse
|
46
|
Arias MA, Santiago L, Costas-Ramon S, Jaime-Sánchez P, Freudenberg M, Jiménez De Bagüés MP, Pardo J. Toll-Like Receptors 2 and 4 Cooperate in the Control of the Emerging Pathogen Brucella microti. Front Cell Infect Microbiol 2017; 6:205. [PMID: 28119856 PMCID: PMC5220065 DOI: 10.3389/fcimb.2016.00205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/22/2016] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptors (TLRs) recognize pathogen-derived molecules and play a critical role during the host innate and adaptive immune response. Brucella spp. are intracellular gram-negative bacteria including several virulent species, which cause a chronic zoonotic infection in a wide range of mammalian hosts known as brucellosis. A new Brucella species, Brucella microti, was recently isolated from wild rodents and found to be highly pathogenic in mice. Using this species-specific model, it was previously found that CD8+ T cells are required to control this infection. In order to find out the role of TLR-mediated responses in the control of this pathogen, the course of infection of B. microti was analyzed over 3 weeks in wild-type (WT) and TLR knock out (KO) mice including TLR2-/-, TLR4-/-, TLR9-/-, TLR2×4-/- and TLR2×4×9-/-. WT and single TLR2, TLR4 and TLR9 KO mice similarly control infection in liver and spleen. In contrast, bacterial clearance was delayed in TLR2×4-/- and TLR2×4×9-/- mice at 7 and 14 days post-infection. This defect correlated with impaired maturation and pro-inflammatory cytokine production in B. microti-infected dendritic cells from TLR2×4-/- and TLR2×4×9-/- mice. Finally, it was found that Tc cells from TLR2×4-/- and TLR2×4×9-/- mice showed reduced ability to inhibit growth of B. microti in macrophages, suggesting the involvement of TLR2 and 4 in the generation of specific Tc cells. Our findings indicate that TLR2 and TLR4 are required to control B. microti infection in mice and that this effect could be related to its participation in the maturation of dendritic cells and the generation of specific CD8+ Tc cells.
Collapse
Affiliation(s)
- Maykel A Arias
- Cell Immunity in Cancer, Inflammation and Infection Group, Department of Biochemistry and Molecular and Cell Biology, Biomedical Research Centre of Aragon (CIBA), IIS Aragon, University of Zaragoza Zaragoza, Spain
| | - Llipsy Santiago
- Cell Immunity in Cancer, Inflammation and Infection Group, Department of Biochemistry and Molecular and Cell Biology, Biomedical Research Centre of Aragon (CIBA), IIS Aragon, University of Zaragoza Zaragoza, Spain
| | - Santiago Costas-Ramon
- Cell Immunity in Cancer, Inflammation and Infection Group, Department of Biochemistry and Molecular and Cell Biology, Biomedical Research Centre of Aragon (CIBA), IIS Aragon, University of Zaragoza Zaragoza, Spain
| | - Paula Jaime-Sánchez
- Cell Immunity in Cancer, Inflammation and Infection Group, Department of Biochemistry and Molecular and Cell Biology, Biomedical Research Centre of Aragon (CIBA), IIS Aragon, University of Zaragoza Zaragoza, Spain
| | - Marina Freudenberg
- Max-Planck Institute for Immunobiology and Epigenetics Freiburg, Germany
| | - Maria P Jiménez De Bagüés
- Unidad de Producción y Sanidad Animal, Centro de Investigación y Tecnología Agroalimentaria, Instituto Agroalimentario de Aragón - IA2, CITA-Universidad de Zaragoza Zaragoza, Spain
| | - Julián Pardo
- Cell Immunity in Cancer, Inflammation and Infection Group, Department of Biochemistry and Molecular and Cell Biology, Biomedical Research Centre of Aragon (CIBA), IIS Aragon, University of ZaragozaZaragoza, Spain; Nanoscience Institute of Aragon, University of ZaragozaZaragoza, Spain; Aragon I+D FoundationZaragoza, Spain
| |
Collapse
|
47
|
Granzymes A and K differentially potentiate LPS-induced cytokine response. Cell Death Discov 2016; 2:16084. [PMID: 28028441 PMCID: PMC5149580 DOI: 10.1038/cddiscovery.2016.84] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/01/2016] [Accepted: 09/30/2016] [Indexed: 01/09/2023] Open
Abstract
Granzymes are serine proteases that, upon release from cytotoxic cells, induce apoptosis in tumor cells and virally infected cells. In addition, a role of granzymes in inflammation is emerging. Recently, we have demonstrated that extracellular granzyme K (GrK) potentiates lipopolysaccharide (LPS)-induced cytokine response from monocytes. GrK interacts with LPS, disaggregates LPS micelles, and stimulates LPS-CD14 binding and Toll-like receptor signaling. Here we show that human GrA also potentiates cytokine responses in human monocytes initiated by LPS or Gram-negative bacteria. Similar to GrK, this effect is independent of GrA catalytic activity. Unlike GrK, however, GrA does not bind to LPS, has little influence on LPS micelle disaggregation, and does not augment LPS-CD14 complex formation. We conclude that GrA and GrK differentially modulate LPS-Toll-like receptor signaling in monocytes, suggesting functional redundancy among cytotoxic lymphocyte proteases in the anti-bacterial innate immune response.
Collapse
|
48
|
He S, Li X, Li R, Fang L, Sun L, Wang Y, Wu M. Annexin A2 Modulates ROS and Impacts Inflammatory Response via IL-17 Signaling in Polymicrobial Sepsis Mice. PLoS Pathog 2016; 12:e1005743. [PMID: 27389701 PMCID: PMC4936746 DOI: 10.1371/journal.ppat.1005743] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/14/2016] [Indexed: 02/05/2023] Open
Abstract
Sepsis is a progressive disease manifesting excessive inflammatory responses, severe tissue injury, organ dysfunction, and, ultimately, mortality. Since currently, there are limited therapeutic options for this disease, further understanding the molecular pathogenesis of sepsis may help develop effective treatments. Here we identify a novel role for Annexin A2 (AnxA2), a multi-compartmental protein, in inhibiting pro-inflammatory response by regulating reactive oxygen species (ROS) and IL-17 signaling during sepsis. In cecal ligation and puncture (CLP) sepsis models, anxa2-/- mice manifested increased pro-inflammatory cytokines and neutrophil infiltration, but decreased bacterial clearance and animal survival. In addition, AnxA2 deficiency led to intensified ROS and IL-17A. Using site directed mutagenesis, we uncovered that cysteine 9 of AnxA2 was the most important aa (site) for regulation of ROS levels. Furthermore, ROS appears to be responsible for elevated IL-17A levels and subsequently exaggerated inflammatory response. Depletion of IL-17 via CRISPR/Cas9 KO strategy down-regulated inflammation and conferred protection against sepsis in anxa2-/- mice. Our findings reveal a previously undemonstrated function for AnxA2 in inflammatory response in polymicrobial sepsis models via an AnxA2-ROS-IL-17 axis, providing insight into the regulation of pathophysiology of sepsis.
Collapse
Affiliation(s)
- Sisi He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, P. R. China
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Xuefeng Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, P. R. China
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Rongpeng Li
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Lizhu Fang
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, Jiangsu, P. R. China
| | - Yongsheng Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, P. R. China
| | - Min Wu
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| |
Collapse
|
49
|
Uranga S, Marinova D, Martin C, Pardo J, Aguilo N. Granzyme A Is Expressed in Mouse Lungs during Mycobacterium tuberculosis Infection but Does Not Contribute to Protection In Vivo. PLoS One 2016; 11:e0153028. [PMID: 27055232 PMCID: PMC4824395 DOI: 10.1371/journal.pone.0153028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/21/2016] [Indexed: 11/19/2022] Open
Abstract
Granzyme A, a serine protease expressed in the granules of cytotoxic T and Natural Killer cells, is involved in the generation of pro-inflammatory cytokines by macrophages. Granzyme A has been described to induce in macrophages in vitro the activation of pro-inflammatory pathways that impair intracellular mycobacterial replication. In the present study, we explored the physiological relevance of Granzyme A in the control of pulmonary Mycobacterium tuberculosis infection in vivo. Our results show that, even though Granzyme A is expressed by cytotoxic cells from mouse lungs during pulmonary infection, its deficiency in knockout mice does not have an effect in the control of M. tuberculosis infection. In addition our findings indicate that absence of Granzyme A does not affect the protection conferred by the live-attenuated M. tuberculosis vaccine MTBVAC. Altogether, our findings are in apparent contradiction with previously published in vitro results and suggest that Granzyme A does not have a crucial role in vivo in the protective response to tuberculosis.
Collapse
Affiliation(s)
- Santiago Uranga
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Dessislava Marinova
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martin
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Microbiología, Hospital Universitario Miguel Servet, ISS Aragón, Paseo Isabel la Católica 1–3, 50009, Zaragoza, Spain
| | - Julián Pardo
- Immune Effector Cells Group (ICE), 3 Aragón Health Research Institute (IIS Aragón), Edificio CIBA, Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Nanoscience Institute of Aragon (INA), University of Zaragoza, Zaragoza, Spain
- Fundación Aragón I+D (ARAID), Gobierno de Aragón, Zaragoza, Spain
| | - Nacho Aguilo
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, C/ Domingo Miral s/n, 50009, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
50
|
Sharma M, Merkulova Y, Raithatha S, Parkinson LG, Shen Y, Cooper D, Granville DJ. Extracellular granzyme K mediates endothelial activation through the cleavage of protease-activated receptor-1. FEBS J 2016; 283:1734-47. [PMID: 26936634 DOI: 10.1111/febs.13699] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 01/25/2016] [Accepted: 02/29/2016] [Indexed: 01/26/2023]
Abstract
Granzymes are a family of serine proteases that were once thought to function exclusively as mediators of cytotoxic lymphocyte-induced target cell death. However, non-apoptotic roles for granzymes, including granzyme K (GzK), have been proposed. As recent studies have observed elevated levels of GzK in the plasma of patients diagnosed with clinical sepsis, we hypothesized that extracellular GzK induces a proinflammatory response in endothelial cells. In the present study, extracellular GzK proteolytically activated protease-activated receptor-1 leading to increased interleukin 6 and monocyte chemotactic protein 1 production in endothelial cells. Enhanced expression of intercellular adhesion molecule 1 along with an increased capacity for adherence of THP-1 cells was also observed. Characterization of downstream pathways implicated the mitogen-activated protein kinase p38 pathway for intercellular adhesion molecule 1 expression, and both the p38 and the extracellular signal-regulated protein kinases 1 and 2 pathways in cytokine production. GzK also increased tumour necrosis factor α-induced inflammatory adhesion molecule expression. Furthermore, the physiological inhibitor of GzK, inter-α-inhibitor protein, significantly inhibited GzK activity in vitro. In summary, extracellular GzK promotes a proinflammatory response in endothelial cells.
Collapse
Affiliation(s)
- Mehul Sharma
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yulia Merkulova
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Sheetal Raithatha
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Leigh G Parkinson
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yue Shen
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dawn Cooper
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - David J Granville
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|