1
|
Woxholt S, Ueland T, Aukrust P, Anstensrud AK, Broch K, Tøllefsen IM, Seljeflot I, Halvorsen B, Dahl TB, Huse C, Andersen GØ, Gullestad L, Wiseth R, Damås JK, Kleveland O. Effect of tocilizumab on endothelial and platelet-derived CXC-chemokines and their association with inflammation and myocardial injury in STEMI patients undergoing primary PCI. Int J Cardiol 2024; 418:132613. [PMID: 39374793 DOI: 10.1016/j.ijcard.2024.132613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/29/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Tocilizumab improves myocardial salvage in ST-elevation myocardial infarction (STEMI) patients when administered before percutaneous coronary intervention (PCI). The mechanisms underlying ischemia-reperfusion injury remain unclear. In this sub-study, we investigated whether endothelial and platelet-derived CXC chemokines are involved, as they represent inflammatory mediators from two cell types relevant to myocardial infarction. Associations between these chemokines and neutrophils, C-reactive protein (CRP), troponin T (TnT), myocardial salvage index (MSI), microvascular obstruction (MVO), and infarct size. METHODS This is a sub-study of the ASSAIL-MI trial, a double-blind clinical trial that randomized 199 STEMI patients to receive either 280 mg tocilizumab (n = 101) or placebo (n = 98) intravenously before PCI. Blood samples were collected prior to infusion, at day 1-2, 3-7, and at 3 and 6 months. Heparin was administered before baseline in 150 patients, while 49 received it after. We measured CXC-chemokines CXCL4, CXCL5, CXCL6, CXCL7, and CXCL12 using immunoassays. Cardiac MRI was performed in the first week and at 6 months. RESULTS Tocilizumab did not significantly affect CXC-chemokines levels. Although some correlations were observed between chemokine levels and neutrophil counts and CRP, none of the CXC chemokines were associated with infarct size, MSI, MVO, or TnT levels. Notably, CXCL 12 levels increased in patients who received heparin before baseline, while other CXC-chemokines decreased significantly. CONCLUSION This study suggests that the beneficial effects of tocilizumab in STEMI patients are not due to changes in circulating endothelial or platelet-derived CXC-chemokines, compared to placebo. However, heparin significantly influences the levels of these chemokines.
Collapse
Affiliation(s)
- Sindre Woxholt
- Clinic of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Thrombosis Research and Expertise Center (TREC), The Arctic University of Norway, Tromsø, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Section of Clinical Immunology and Infectious Disease, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | | | - Ingebjørg Seljeflot
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Oslo Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Camilla Huse
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Geir Øystein Andersen
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K. G. Jebsen Cardiac Research Centre and Centre for Heart Failure Research, University of Oslo, Oslo, Norway; Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Rune Wiseth
- Clinic of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Jan Kristian Damås
- Department of Infectious Disease, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway; Department of clinical and Molecular medicine, Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ola Kleveland
- Clinic of Cardiology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
2
|
Scott MA, Valeris-Chacin R, Thompson AC, Woolums AR, Karisch BB. Comprehensive time-course gene expression evaluation of high-risk beef cattle to establish immunological characteristics associated with undifferentiated bovine respiratory disease. Front Immunol 2024; 15:1412766. [PMID: 39346910 PMCID: PMC11427276 DOI: 10.3389/fimmu.2024.1412766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024] Open
Abstract
Bovine respiratory disease (BRD) remains the leading infectious disease in beef cattle production systems. Host gene expression upon facility arrival may indicate risk of BRD development and severity. However, a time-course approach would better define how BRD development influences immunological and inflammatory responses after disease occurrences. Here, we evaluated whole blood transcriptomes of high-risk beef cattle at three time points to elucidate BRD-associated host response. Sequenced jugular whole blood mRNA from 36 cattle (2015: n = 9; 2017: n = 27) across three time points (n = 100 samples; days [D]0, D28, and D63) were processed through ARS-UCD1.2 reference-guided assembly (HISAT2/Stringtie2). Samples were categorized into BRD-severity cohorts (Healthy, n = 14; Treated 1, n = 11; Treated 2+, n = 11) via frequency of antimicrobial clinical treatment. Assessment of gene expression patterns over time within each BRD cohort was modeled through an autoregressive hidden Markov model (EBSeq-HMM; posterior probability ≥ 0.5, FDR < 0.01). Mixed-effects negative binomial models (glmmSeq; FDR < 0.05) and edgeR (FDR < 0.10) identified differentially expressed genes between and across cohorts overtime. A total of 2,580, 2,216, and 2,381 genes were dynamically expressed across time in Healthy, Treated 1, and Treated 2+ cattle, respectively. Genes involved in the production of specialized resolving mediators (SPMs) decreased at D28 and then increased by D63 across all three cohorts. Accordingly, SPM production and alternative complement were differentially expressed between Healthy and Treated 2+ at D0, but not statistically different between the three groups by D63. Magnitude, but not directionality, of gene expression related to SPM production, alternative complement, and innate immune response signified Healthy and Treated 2+ cattle. Differences in gene expression at D63 across the three groups were related to oxygen binding and carrier activity, natural killer cell-mediated cytotoxicity, cathelicidin production, and neutrophil degranulation, possibly indicating prolonged airway pathology and inflammation weeks after clinical treatment for BRD. These findings indicate genomic mechanisms indicative of BRD development and severity over time.
Collapse
Affiliation(s)
- Matthew A Scott
- Veterinary Education, Research, and Outreach Program, Texas A&M University, Canyon, TX, United States
| | - Robert Valeris-Chacin
- Veterinary Education, Research, and Outreach Program, Texas A&M University, Canyon, TX, United States
| | - Alexis C Thompson
- Texas A&M Veterinary Medical Diagnostic Laboratory, Canyon, TX, United States
| | - Amelia R Woolums
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Brandi B Karisch
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, United States
| |
Collapse
|
3
|
Ito Y, Sun T, Tawada M, Kinashi H, Yamaguchi M, Katsuno T, Kim H, Mizuno M, Ishimoto T. Pathophysiological Mechanisms of Peritoneal Fibrosis and Peritoneal Membrane Dysfunction in Peritoneal Dialysis. Int J Mol Sci 2024; 25:8607. [PMID: 39201294 PMCID: PMC11354376 DOI: 10.3390/ijms25168607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
The characteristic feature of chronic peritoneal damage in peritoneal dialysis (PD) is a decline in ultrafiltration capacity associated with pathological fibrosis and angiogenesis. The pathogenesis of peritoneal fibrosis is attributed to bioincompatible factors of PD fluid and peritonitis. Uremia is associated with peritoneal membrane inflammation that affects fibrosis, neoangiogenesis, and baseline peritoneal membrane function. Net ultrafiltration volume is affected by capillary surface area, vasculopathy, peritoneal fibrosis, and lymphangiogenesis. Many inflammatory cytokines induce fibrogenic growth factors, with crosstalk between macrophages and fibroblasts. Transforming growth factor (TGF)-β and vascular endothelial growth factor (VEGF)-A are the key mediators of fibrosis and angiogenesis, respectively. Bioincompatible factors of PD fluid upregulate TGF-β expression by mesothelial cells that contributes to the development of fibrosis. Angiogenesis and lymphangiogenesis can progress during fibrosis via TGF-β-VEGF-A/C pathways. Complement activation occurs in fungal peritonitis and progresses insidiously during PD. Analyses of the human peritoneal membrane have clarified the mechanisms by which encapsulating peritoneal sclerosis develops. Different effects of dialysates on the peritoneal membrane were also recognized, particularly in terms of vascular damage. Understanding the pathophysiologies of the peritoneal membrane will lead to preservation of peritoneal membrane function and improvements in technical survival, mortality, and quality of life for PD patients.
Collapse
Affiliation(s)
- Yasuhiko Ito
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute 480-1195, Japan (H.K.); (M.Y.); (T.I.)
| | - Ting Sun
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute 480-1195, Japan (H.K.); (M.Y.); (T.I.)
| | - Mitsuhiro Tawada
- Department of Nephrology, Imaike Jin Clinic, Nagoya 464-0850, Japan
| | - Hiroshi Kinashi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute 480-1195, Japan (H.K.); (M.Y.); (T.I.)
| | - Makoto Yamaguchi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute 480-1195, Japan (H.K.); (M.Y.); (T.I.)
| | - Takayuki Katsuno
- Department of Nephrology and Rheumatology, Aichi Medical University Medical Center, Okazaki 444-2148, Japan;
| | - Hangsoo Kim
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (H.K.); (M.M.)
| | - Masashi Mizuno
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (H.K.); (M.M.)
| | - Takuji Ishimoto
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute 480-1195, Japan (H.K.); (M.Y.); (T.I.)
| |
Collapse
|
4
|
Huang L, Tan X, Xuan W, Luo Q, Xie L, Xi Y, Li R, Li L, Li F, Zhao M, Jiang Y, Wu X. Ficolin-A/2 Aggravates Severe Lung Injury through Neutrophil Extracellular Traps Mediated by Gasdermin D-Induced Pyroptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:989-1006. [PMID: 38442803 DOI: 10.1016/j.ajpath.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
Neutrophil extracellular traps (NETs) and pyroptosis are critical events in lung injury. This study investigated whether ficolin-A influenced NET formation through pyroptosis to exacerbate lipopolysaccharide (LPS)-induced lung injury. The expression of ficolin-A/2, NETs, and pyroptosis-related molecules was investigated in animal and cell models. Knockout and knockdown (recombinant protein) methods were used to elucidate regulatory mechanisms. The Pearson correlation coefficient was used to analyze the correlation between ficolins and pyroptosis- and NET-related markers in clinical samples. In this study, ficolin-2 (similar to ficolin-A) showed significant overexpression in patients with acute respiratory distress syndrome. In vivo, knockout of Fcna, but not Fcnb, attenuated lung inflammation and inhibited NET formation in the LPS-induced mouse model. DNase I further alleviated lung inflammation and NET formation in Fcna knockout mice. In vitro, neutrophils derived from Fcna-/- mice showed less pyroptosis and necroptosis than those from the control group after LPS stimulation. Additionally, GSDMD knockdown or Nod-like receptor protein 3 inhibitor reduced NET formation. Addition of recombinant ficolin-2 protein to human peripheral blood neutrophils promoted NET formation and pyroptosis after LPS stimulation, whereas Fcn2 knockdown had the opposite effect. Acute respiratory distress syndrome patients showed increased levels of pyroptosis- and NET-related markers, which were correlated positively with ficolin-2 levels. In conclusion, these results suggested that ficolin-A/2 exacerbated NET formation and LPS-induced lung injury via gasdermin D-mediated pyroptosis.
Collapse
Affiliation(s)
- Li Huang
- Department of Pediatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China; Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xiaowu Tan
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Weixia Xuan
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Qing Luo
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Xie
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yunzhu Xi
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Rong Li
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Li Li
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Feifan Li
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Meiyun Zhao
- Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yongliang Jiang
- Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China.
| | - Xu Wu
- Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China; Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
5
|
Guo J, Zhang QY, Xu L, Li M, Sun QY. Icariin ameliorates LPS-induced acute lung injury in mice via complement C5a-C5aR1 and TLR4 signaling pathways. Int Immunopharmacol 2024; 131:111802. [PMID: 38467082 DOI: 10.1016/j.intimp.2024.111802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/22/2024] [Accepted: 03/02/2024] [Indexed: 03/13/2024]
Abstract
Acute lung injury (ALI) is an acute respiratory-related progressive disorder, which lacks specific pharmacotherapy. Icariin (ICA) has been shown to be effective in treating ALI. However, the targets and pharmacological mechanisms underlying the effects of ICA in the treatment of ALI are relatively lacking. Based on network pharmacology and molecular docking analyses, the gene functions and potential target pathways of ICA in the treatment of ALI were determined. In addition, the underlying mechanisms of ICA were verified by immunohistochemistry, immunofluorescence, quantitative Real-time PCR, and Western blot in LPS-induced ALI mice. The biological processes targeted by ICA in the treatment of ALI included the pathological changes, inflammatory response, and cell signal transduction. Network pharmacology, molecular docking, and in vivo experimental results revealed that ICA inhibited the complement C5a-C5aR1 axis, TLR4 mediated NF-κB, MAPK, and JAK2-STAT3 signaling pathways related gene and protein expressions, and decreased inflammatory cytokine, chemokine, adhesion molecule expressions, and mitochondrial apoptosis in LPS-induced ALI.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China; School of Chinese Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, China
| | - Qi-Yun Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Lin Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Min Li
- General Ward, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Qian-Yun Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; Natural Products Research Center of Guizhou Province, Guiyang 550014, China.
| |
Collapse
|
6
|
Khadilkar P, Chougule D, Tipnis T, Khopkar U, Nadkar M, Rajadhyaksha A, Kini S, Kharkar V, Athvale A, Athvale T, Madkaikar M, Pradhan V. A comparative study of modulatory interaction between cytokines and apoptotic proteins among Scleroderma patients with and without pulmonary involvement. Cytokine 2023; 166:156183. [PMID: 37011542 DOI: 10.1016/j.cyto.2023.156183] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 04/04/2023]
Abstract
BACKGROUND Interstitial lung disease (ILD) and pulmonary arterial hypertension (PAH) are the most eminent forms of pulmonary involvement in Scleroderma. In this study we investigate the interaction between cytokines and apoptotic proteins in treatment naive Scleroderma (SSc) patients with and without pulmonary involvement. METHODS Newly diagnosed treatment naïve Scleroderma (SSc) patients (n = 100) and healthy controls (n = 100) were enrolled. Patients were classified as ILD-SSc, PAH-SSc and non-pulmonary SSc (np-SSc). Study variables like mRSS score, autoantibody profile, serum cytokines, serum TGF-β (1,2,3) and apoptotic proteins were assessed for these patients. RESULTS Scleroderma patients showed elevated levels of serum cytokines, but significantly lower IL-22 and TGF- β1 when compared to healthy controls (p < 0.05). Apoptotic proteins were significantly elevated among Scleroderma patients, but the patient groups also showed significant lower caspase 1/3/9 levels when compared to healthy controls (p < 0.05). ILD-SSc patients reported higher mRSS score (p = 0.0436) when compared with PAH-SSc and np-SSc. In ILD-SSc patients, finger tightening (p = 0.0481) and calcinosis/lesions (p = 0.0481) were significant clinical presentations whereas, digital ulcers were significantly prominent in np-SSc patients (p = 0.0132). Elevated TGF-β3 levels (p = 0.02) in SSC-ILD and reduced IL-4 levels (p = 0.02) in SSC-PAH were significant cytokines as compared to np-SSc. Significant correlations were obtained among serum cytokines and apoptotic proteins in Scleroderma patients with and without pulmonary involvement. (p < 0.05) CONCLUSION: Our study highlights the correlation between mRSS score, cytokines and apoptotic proteins in SSc patients with pulmonary involvement. A longitudinal follow up in these patients with assessment of these immunological parameters may be helpful in monitoring the disease.
Collapse
Affiliation(s)
- Prasad Khadilkar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Durga Chougule
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Tanaya Tipnis
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Uday Khopkar
- Department of Skin, STD and Leprosy, G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Milind Nadkar
- Department of Medicine, TNMC & BYLN Hospital, Mumbai, India
| | | | - Seema Kini
- Department of Medicine, G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Vidya Kharkar
- Department of Skin, STD and Leprosy, G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Amita Athvale
- Department of Pulmonary Medicine, G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Tanya Athvale
- Department of Pulmonary Medicine, G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Manisha Madkaikar
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Vandana Pradhan
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India.
| |
Collapse
|
7
|
Ali YM, Lynch NJ, Shaaban AA, Rizk DE, Abdel-Rahman SH, Khatri P, Yabuki M, Yaseen S, Dudler T, Demopulos G, Schwaeble WJ. Inhibition of the lectin pathway of complement activation reduces LPS-induced acute respiratory distress syndrome in mice. Front Immunol 2023; 14:1192767. [PMID: 37325666 PMCID: PMC10262210 DOI: 10.3389/fimmu.2023.1192767] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening disorder with a high rate of mortality. Complement activation in ARDS initiates a robust inflammatory reaction that can cause progressive endothelial injury in the lung. Here, we tested whether inhibition of the lectin pathway of complement could reduce the pathology and improve the outcomes in a murine model of LPS-induced lung injury that closely mimics ARDS in human. In vitro, LPS binds to murine and human collectin 11, human MBL and murine MBL-A, but not to C1q, the recognition subcomponent of the classical pathway. This binding initiates deposition of the complement activation products C3b, C4b and C5b-9 on LPS via the lectin pathway. HG-4, a monoclonal antibody that targets MASP-2, a key enzyme in the lectin pathway, inhibited lectin pathway functional activity in vitro, with an IC50 of circa 10nM. Administration of HG4 (5mg/kg) in mice led to almost complete inhibition of the lectin pathway activation for 48hrs, and 50% inhibition at 60hrs post administration. Inhibition of the lectin pathway in mice prior to LPS-induced lung injury improved all pathological markers tested. HG4 reduces the protein concentration in bronchoalveolar lavage fluid (p<0.0001) and levels of myeloid peroxide (p<0.0001), LDH (p<0.0001), TNFα and IL6 (both p<0.0001). Lung injury was significantly reduced (p<0.001) and the survival time of the mice increased (p<0.01). From the previous findings we concluded that inhibition of the lectin pathway has the potential to prevent ARDS pathology.
Collapse
Affiliation(s)
- Youssif M. Ali
- Department of Veterinary Medicine, School of Biological Sciences, University of Cambridge, Cambridge, United Kingdom
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Nicholas J. Lynch
- Department of Veterinary Medicine, School of Biological Sciences, University of Cambridge, Cambridge, United Kingdom
| | - Ahmed A. Shaaban
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Dina E. Rizk
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Shaymaa H. Abdel-Rahman
- Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Priyanka Khatri
- Department of Veterinary Medicine, School of Biological Sciences, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | - Wilhelm J. Schwaeble
- Department of Veterinary Medicine, School of Biological Sciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
8
|
Triggianese P, Conigliaro P, De Martino E, Monosi B, Chimenti MS. Overview on the Link Between the Complement System and Auto-Immune Articular and Pulmonary Disease. Open Access Rheumatol 2023; 15:65-79. [PMID: 37214353 PMCID: PMC10198272 DOI: 10.2147/oarrr.s318826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Complement system (CS) dysregulation is a key factor in the pathogenesis of different autoimmune diseases playing a central role in many immune innate and adaptive processes. Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by ta breach of self-tolerance leading to a synovitis and extra-articular manifestations. The CS is activated in RA and seems not only to mediate direct tissue damage but also play a role in the initiation of RA pathogenetic mechanisms through interactions with citrullinated proteins. Interstitial lung disease (ILD) represents the most common extra-articular manifestation that can lead to progressive fibrosis. In this review, we focused on the evidence of CS dysregulation in RA and in ILD, and highlighted the role of the CS in both the innate and adaptive immune responses in the development of diseases, by using idiopathic pulmonary fibrosis as a model of lung disease. As a proof of concept, we dissected the evidence that several treatments used to treat RA and ILD such as glucocorticoids, pirfenidone, disease modifying antirheumatic drugs, targeted biologics such as tumor necrosis factor (TNF)-inhibitors, rituximab, tocilizumab, and nintedanib may act indirectly on the CS, suggesting that the CS might represent a potential therapeutic target in these complex diseases.
Collapse
Affiliation(s)
- Paola Triggianese
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Erica De Martino
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Benedetta Monosi
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Maria Sole Chimenti
- Department of Systems Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
9
|
Dasgupta S, Das SS, Patidar S, Kajaria V, Chowdhury SR, Chaudhury K. Identification of Common Dysregulated Genes in COVID-19 and Hypersensitivity Pneumonitis: A Systems Biology and Machine Learning Approach. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:205-214. [PMID: 37062762 DOI: 10.1089/omi.2022.0171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
A comprehensive knowledge on systems biology of severe acute respiratory syndrome coronavirus 2 is crucial for differential diagnosis of COVID-19. Interestingly, the radiological and pathological features of COVID-19 mimic that of hypersensitivity pneumonitis (HP), another pulmonary fibrotic phenotype. This motivated us to explore the overlapping pathophysiology of COVID-19 and HP, if any, and using a systems biology approach. Two datasets were obtained from the Gene Expression Omnibus database (GSE147507 and GSE150910) and common differentially expressed genes (DEGs) for both diseases identified. Fourteen common DEGs, significantly altered in both diseases, were found to be implicated in complement activation and growth factor activity. A total of five microRNAs (hsa-miR-1-3p, hsa-miR-20a-5p, hsa-miR-107, hsa-miR-16-5p, and hsa-miR-34b-5p) and five transcription factors (KLF6, ZBTB7A, ELF1, NFIL3, and ZBT33) exhibited highest interaction with these common genes. Next, C3, CFB, MMP-9, and IL1A were identified as common hub genes for both COVID-19 and HP. Finally, these top-ranked genes (hub genes) were evaluated using random forest classifier to discriminate between the disease and control group (coronavirus disease 2019 [COVID-19] vs. controls, and HP vs. controls). This supervised machine learning approach demonstrated 100% and 87.6% accuracy in differentiating COVID-19 from controls, and HP from controls, respectively. These findings provide new molecular leads that inform COVID-19 and HP diagnostics and therapeutics research and innovation.
Collapse
Affiliation(s)
- Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Sankha Subhra Das
- Department of Human Genetics, University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Sankalp Patidar
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Vaibhav Kajaria
- Department of Pulmonology, Fortis Hospital Anandapur, Kolkata, India
| | | | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
10
|
Shute JK. Heparin, Low Molecular Weight Heparin, and Non-Anticoagulant Derivatives for the Treatment of Inflammatory Lung Disease. Pharmaceuticals (Basel) 2023; 16:ph16040584. [PMID: 37111341 PMCID: PMC10141002 DOI: 10.3390/ph16040584] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Unfractionated heparin has multiple pharmacological activities beyond anticoagulation. These anti-inflammatory, anti-microbial, and mucoactive activities are shared in part by low molecular weight and non-anticoagulant heparin derivatives. Anti-inflammatory activities include inhibition of chemokine activity and cytokine synthesis, inhibitory effects on the mechanisms of adhesion and diapedesis involved in neutrophil recruitment, inhibition of heparanase activity, inhibition of the proteases of the coagulation and complement cascades, inhibition of neutrophil elastase activity, neutralisation of toxic basic histones, and inhibition of HMGB1 activity. This review considers the potential for heparin and its derivatives to treat inflammatory lung disease, including COVID-19, ALI, ARDS, cystic fibrosis, asthma, and COPD via the inhaled route.
Collapse
Affiliation(s)
- Janis Kay Shute
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2UP, UK
| |
Collapse
|
11
|
Sveiven SN, Anesko K, Morgan J, Nair MG, Nordgren TM. Lipid-Sensing Receptor FFAR4 Modulates Pulmonary Epithelial Homeostasis following Immunogenic Exposures Independently of the FFAR4 Ligand Docosahexaenoic Acid (DHA). Int J Mol Sci 2023; 24:ijms24087072. [PMID: 37108233 PMCID: PMC10138935 DOI: 10.3390/ijms24087072] [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: 02/28/2023] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The role of pulmonary free fatty acid receptor 4 (FFAR4) is not fully elucidated and we aimed to clarify the impact of FFAR4 on the pulmonary immune response and return to homeostasis. We employed a known high-risk human pulmonary immunogenic exposure to extracts of dust from swine confinement facilities (DE). WT and Ffar4-null mice were repetitively exposed to DE via intranasal instillation and supplemented with docosahexaenoic acid (DHA) by oral gavage. We sought to understand if previous findings of DHA-mediated attenuation of the DE-induced inflammatory response are FFAR4-dependent. We identified that DHA mediates anti-inflammatory effects independent of FFAR4 expression, and that DE-exposed mice lacking FFAR4 had reduced immune cells in the airways, epithelial dysplasia, and impaired pulmonary barrier integrity. Analysis of transcripts using an immunology gene expression panel revealed a role for FFAR4 in lungs related to innate immune initiation of inflammation, cytoprotection, and immune cell migration. Ultimately, the presence of FFAR4 in the lung may regulate cell survival and repair following immune injury, suggestive of potential therapeutic directions for pulmonary disease.
Collapse
Affiliation(s)
- Stefanie N Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA 92521, USA
| | - Kyle Anesko
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA 92521, USA
| | - Joshua Morgan
- Department of Bioengineering, Bourns College of Engineering, University of California-Riverside, Riverside, CA 92521, USA
| | - Meera G Nair
- Division of Biomedical Sciences, School of Medicine, University of California-Riverside, Riverside, CA 92521, USA
| | - Tara M Nordgren
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
12
|
Yang Z, Nicholson SE, Cancio TS, Cancio LC, Li Y. Complement as a vital nexus of the pathobiological connectome for acute respiratory distress syndrome: An emerging therapeutic target. Front Immunol 2023; 14:1100461. [PMID: 37006238 PMCID: PMC10064147 DOI: 10.3389/fimmu.2023.1100461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
The hallmark of acute respiratory distress syndrome (ARDS) pathobiology is unchecked inflammation-driven diffuse alveolar damage and alveolar-capillary barrier dysfunction. Currently, therapeutic interventions for ARDS remain largely limited to pulmonary-supportive strategies, and there is an unmet demand for pharmacologic therapies targeting the underlying pathology of ARDS in patients suffering from the illness. The complement cascade (ComC) plays an integral role in the regulation of both innate and adaptive immune responses. ComC activation can prime an overzealous cytokine storm and tissue/organ damage. The ARDS and acute lung injury (ALI) have an established relationship with early maladaptive ComC activation. In this review, we have collected evidence from the current studies linking ALI/ARDS with ComC dysregulation, focusing on elucidating the new emerging roles of the extracellular (canonical) and intracellular (non-canonical or complosome), ComC (complementome) in ALI/ARDS pathobiology, and highlighting complementome as a vital nexus of the pathobiological connectome for ALI/ARDS via its crosstalking with other systems of the immunome, DAMPome, PAMPome, coagulome, metabolome, and microbiome. We have also discussed the diagnostic/therapeutic potential and future direction of ALI/ARDS care with the ultimate goal of better defining mechanistic subtypes (endotypes and theratypes) through new methodologies in order to facilitate a more precise and effective complement-targeted therapy for treating these comorbidities. This information leads to support for a therapeutic anti-inflammatory strategy by targeting the ComC, where the arsenal of clinical-stage complement-specific drugs is available, especially for patients with ALI/ARDS due to COVID-19.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Susannah E. Nicholson
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Tomas S. Cancio
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Leopoldo C. Cancio
- United States (US) Army Burn Center, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Yansong Li
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- The Geneva Foundation, Immunological Damage Control Resuscitation Program, Tacoma, WA, United States
- *Correspondence: Yansong Li,
| |
Collapse
|
13
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
14
|
Detsika MG, Theochari E, Palamaris K, Gakiopoulou H, Lianos EA. Effect of Heme Oxygenase-1 Depletion on Complement Regulatory Proteins Expression in the Rat. Antioxidants (Basel) 2022; 12:61. [PMID: 36670923 PMCID: PMC9854825 DOI: 10.3390/antiox12010061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Heme oxygenase has been implicated in the regulation of various immune responses including complement activation. Using a transgenic rat model of HO-1 depletion, the present study assessed the effect of HO-1 absence on the expression of complement regulatory proteins: decay accelerating factor (DAF), CR1-related gene/protein Y (Crry) and CD59, which act to attenuate complement activation. Protein expression was assessed by immunohistochemistry in kidney, liver, lung and spleen tissues. DAF protein was reduced in all tissues retrieved from rats lacking HO-1 (Hmox1-/-) apart from spleen tissue sections. Crry protein was also reduced, but only in Hmox1-/- kidney and liver tissue. C3b staining was augmented in the kidney and spleen from Hmox1-/- rats, suggesting that the decrease of DAF and Crry was sufficient to increase C3b deposition. The observations support an important role of HO-1 as a regulator of the complement system.
Collapse
Affiliation(s)
- Maria G. Detsika
- GP Livanos and M Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, National and Kapodistrian University of Athens, 10675 Athens, Greece
| | - Eirini Theochari
- Department of Pathology, School of Medicine, University of Athens, 11527 Athens, Greece
| | - Kostas Palamaris
- Department of Pathology, School of Medicine, University of Athens, 11527 Athens, Greece
| | - Harikleia Gakiopoulou
- Department of Pathology, School of Medicine, University of Athens, 11527 Athens, Greece
| | - Elias A. Lianos
- Veterans Affairs Medical Center and Virginia Tech, Carilion School of Medicine, Salem, VA 24153, USA
| |
Collapse
|
15
|
Sparreman Mikus M, Kolmert J, Andersson LI, Dahlén SE, James A. Reply: U-BIOPRED/BIOAIR proteins: inflammation or infection? Eur Respir J 2022; 60:2201795. [PMID: 36202410 PMCID: PMC9712852 DOI: 10.1183/13993003.01795-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 12/14/2022]
Abstract
We thank D.L. Hahn and W. Webley for their interesting interpretation of the data in our original article, in which we reported on plasma proteins associated with asthma severity in two independent cohorts, U-BIOPRED and BIOAIR [1]. Protein profiles in plasma remained associated with asthma severity in the European asthma cohort U-BIOPRED when controlling for previous history of respiratory infections https://bit.ly/3rdEL0O
Collapse
Affiliation(s)
- Maria Sparreman Mikus
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- The Clinical Laboratory for Lung and Allergy Research, Department of Respiratory Medicine and Allergy, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Johan Kolmert
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars I Andersson
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- The Clinical Laboratory for Lung and Allergy Research, Department of Respiratory Medicine and Allergy, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- The Clinical Laboratory for Lung and Allergy Research, Department of Respiratory Medicine and Allergy, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna James
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Blanc P, Liu Y, Reveneau N, Cavell B, Gorringe A, Renauld-Mongénie G. The role of bactericidal and opsonic activity in immunity against Bordetella pertussis. Expert Rev Vaccines 2022; 21:1727-1738. [PMID: 36369768 DOI: 10.1080/14760584.2022.2137145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
INTRODUCTION Pertussis vaccines have drastically reduced the disease burden in humans since their implementation. Despite their success, pertussis remains an important global public health challenge. Bordetella pertussis resurgence could be a result of greater surveillance combined with improved diagnosis methods, changes in Bordetella pertussis biology, vaccine schedules, and/or coverage. Additionally, mechanisms of protection conferred by acellular pertussis (aP) and whole-cell pertussis (wP) vaccines differ qualitatively. There are no clear immune correlates of protection for pertussis vaccines. Pertussis antigens can induce toxin neutralizing antibodies, block adherence or engage complement mediated phagocytic/bactericidal killing. AREAS COVERED We reviewed the existing evidence on antibody-mediated serum bactericidal and opsonophagocytic activity and discussed the relevance of these functional antibodies in the development of next-generation pertussis vaccines. EXPERT OPINION Current paradigm proposes that wP vaccines may confer greater herd protection than aP vaccines due to their enhanced clearance of bacteria from the nasopharynx in animal models. Functional antibodies may contribute to the reduction of nasal colonization, which differentiates aP and wP vaccines. Understanding the intrinsic differences in protective immune responses elicited by each class of vaccines will help to identify biomarkers that can be used as immunological end points in clinical trials.
Collapse
Affiliation(s)
- Pascal Blanc
- Research & Development, Sanofi, Marcy l'Etoile, France
| | - Yuanqing Liu
- Research & Development, Sanofi, Marcy l'Etoile, France
| | | | - Breeze Cavell
- Department of Research and Evaluation, United Kingdom (UK) Health Security Agency, Salisbury, UK
| | - Andrew Gorringe
- Department of Research and Evaluation, United Kingdom (UK) Health Security Agency, Salisbury, UK
| | | |
Collapse
|
17
|
Qin R, Kurz E, Chen S, Zeck B, Chiribogas L, Jackson D, Herchen A, Attia T, Carlock M, Rapkiewicz A, Bar-Sagi D, Ritchie B, Ross TM, Mahal LK. α2,6-Sialylation Is Upregulated in Severe COVID-19, Implicating the Complement Cascade. ACS Infect Dis 2022; 8:2348-2361. [PMID: 36219583 PMCID: PMC9578644 DOI: 10.1021/acsinfecdis.2c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Indexed: 01/29/2023]
Abstract
Better understanding of the molecular mechanisms underlying COVID-19 severity is desperately needed in current times. Although hyper-inflammation drives severe COVID-19, precise mechanisms triggering this cascade and what role glycosylation might play therein are unknown. Here we report the first high-throughput glycomic analysis of COVID-19 plasma samples and autopsy tissues. We find that α2,6-sialylation is upregulated in the plasma of patients with severe COVID-19 and in autopsied lung tissue. This glycan motif is enriched on members of the complement cascade (e.g., C5, C9), which show higher levels of sialylation in severe COVID-19. In the lung tissue, we observe increased complement deposition, associated with elevated α2,6-sialylation levels, corresponding to elevated markers of poor prognosis (IL-6) and fibrotic response. We also observe upregulation of the α2,6-sialylation enzyme ST6GAL1 in patients who succumbed to COVID-19. Our work identifies a heretofore undescribed relationship between sialylation and complement in severe COVID-19, potentially informing future therapeutic development.
Collapse
Affiliation(s)
- Rui Qin
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Emma Kurz
- Department
of Cell Biology, NYU Grossman School of
Medicine, 550 First Avenue, New York, New York 10016, United
States
| | - Shuhui Chen
- Department
of Chemistry, Biomedical Research Institute, New York University, New York, New York10003, United States
| | - Briana Zeck
- Center
for Biospecimen Research and Development, NYU Langone, New York, New York 10016, United
States
| | - Luis Chiribogas
- Center
for Biospecimen Research and Development, NYU Langone, New York, New York 10016, United
States
| | - Dana Jackson
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Alex Herchen
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Tyson Attia
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Michael Carlock
- Center for
Vaccines and Immunology, University of Georgia, Athens, Georgia 30605, United States
| | - Amy Rapkiewicz
- Department
of Pathology, NYU Long Island School of
Medicine, Mineola, New York 11501, United
States
| | - Dafna Bar-Sagi
- Department
of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Bruce Ritchie
- University
of Alberta Hospital, Edmonton, Alberta T6G 2B7, Canada
| | - Ted M. Ross
- Center for
Vaccines and Immunology, University of Georgia, Athens, Georgia 30605, United States
| | - Lara K. Mahal
- Department
of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
18
|
Guo J, Liu QZ, Zhu FJ, Li M, Li J, Guo L, Sun QY, Yang QX. Acteoside attenuates acute lung injury following administration of cobra venom factor to mice. Heliyon 2022; 8:e11622. [PMID: 36411899 PMCID: PMC9674544 DOI: 10.1016/j.heliyon.2022.e11622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/17/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Background Acteoside, a water-soluble active constituent of diverse valuable medicinal vegetation, has shown strong anti-inflammatory property. However, studies on the anti-inflammatory property of acteoside in complement-induced acute lung injury (ALI) are limited. Therefore, this study aims to evaluate the anti-inflammatory activity of acteoside in cobra venom factor (CVF)-stimulated human microvascular endothelial cells (HMEC) and in ALI mice model. Methods In this study, we investigated the effects of acteoside (20, 10, and 5 μg/mL) in vitro in CVF induced HMECs and the activity of acteoside (100, 50, and 20 mg/kg/day bodyweight) in vivo in CVF induced ALI mice. Each eight male mice were orally administered acteoside or the positive drug PDTC (100 mg/kg/day) for 7 days before CVF (35 μg/kg) injection. After injection for 1 h, the pharmacological effects of acteoside were investigated by spectrophotometry, pathological examination, enzyme-linked immunosorbent assay, and immunohistochemistry. Results In vitro, acteoside (20, 10, and 5 μg/mL) reduced the protein expression of adhesion molecules and pro-inflammatory cytokines and transcriptional activity of NF-κB (P < 0.01). In vivo studies showed that acteoside dose-dependently alleviated lung histopathologic lesion, inhibited the production of the protein content of BALF, leukocyte cell number, lung MPO activity, and expression levels of IL-6, TNF-α, and ICAM-1, and suppressed the C5b-9 deposition and NF-κB activation in CVF-induced acute lung inflammation in mice (P < 0.05, 0.01). Conclusion This study demonstrates that acteoside exerts strong anti-inflammatory activities in the CVF-induced acute lung inflammation model and suggests that acteoside is a potential therapeutic agent for complement-related inflammatory diseases.
Collapse
Affiliation(s)
- Jing Guo
- School of Karst Science, Guizhou Normal University/State Engineering Technology Institute for Karst Desertification Control, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products, Guizhou Province and Chinese Academy of Sciences, Guiyang, China
- Mordern Research Center for Traditional Chinese Medicine, Institute of Molecular Science, Shanxi University, Taiyuan 030006, China
| | - Qiao-Zhou Liu
- School of Karst Science, Guizhou Normal University/State Engineering Technology Institute for Karst Desertification Control, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products, Guizhou Province and Chinese Academy of Sciences, Guiyang, China
- Guyuan No. 8 Middle School, Guyuan, China
| | - Fang-Juan Zhu
- School of Karst Science, Guizhou Normal University/State Engineering Technology Institute for Karst Desertification Control, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products, Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| | - Min Li
- General Ward, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jiao Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products, Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| | - Li Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products, Guizhou Province and Chinese Academy of Sciences, Guiyang, China
| | - Qian-Yun Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products, Guizhou Province and Chinese Academy of Sciences, Guiyang, China
- Corresponding author.
| | - Qing-Xiong Yang
- School of Karst Science, Guizhou Normal University/State Engineering Technology Institute for Karst Desertification Control, Guiyang, China
- Corresponding author.
| |
Collapse
|
19
|
The Complement System, Aging, and Aging-Related Diseases. Int J Mol Sci 2022; 23:ijms23158689. [PMID: 35955822 PMCID: PMC9369321 DOI: 10.3390/ijms23158689] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 12/10/2022] Open
Abstract
The complement system is a part of the immune system and consists of multiple complement components with biological functions such as defense against pathogens and immunomodulation. The complement system has three activation pathways: the classical pathway, the lectin pathway, and the alternative pathway. Increasing evidence indicates that the complement system plays a role in aging. Complement plays a role in inflammatory processes, metabolism, apoptosis, mitochondrial function, and Wnt signaling pathways. In addition, the complement system plays a significant role in aging-related diseases, including Alzheimer’s disease, age-related macular degeneration, and osteoarthritis. However, the effect of complement on aging and aging-related diseases is still unclear. Thus, a better understanding of the potential relationship between complement, aging, and aging-related diseases will provide molecular targets for treating aging, while focusing on the balance of complement in during treatment. Inhibition of a single component does not result in a good outcome. In this review, we discussed the research progress and effects of complement in aging and aging-related diseases.
Collapse
|
20
|
Gianni P, Goldin M, Ngu S, Zafeiropoulos S, Geropoulos G, Giannis D. Complement-mediated microvascular injury and thrombosis in the pathogenesis of severe COVID-19: A review. World J Exp Med 2022; 12:53-67. [PMID: 36157337 PMCID: PMC9350720 DOI: 10.5493/wjem.v12.i4.53] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) causes acute microvascular thrombosis in both venous and arterial structures which is highly associated with increased mortality. The mechanisms leading to thromboembolism are still under investigation. Current evidence suggests that excessive complement activation with severe amplification of the inflammatory response (cytokine storm) hastens disease progression and initiates complement-dependent cytotoxic tissue damage with resultant prothrombotic complications. The concept of thromboinflammation, involving overt inflammation and activation of the coagulation cascade causing thrombotic microangiopathy and end-organ damage, has emerged as one of the core components of COVID-19 pathogenesis. The complement system is a major mediator of the innate immune response and inflammation and thus an appealing treatment target. In this review, we discuss the role of complement in the development of thrombotic microangiopathy and summarize the current data on complement inhibitors as COVID-19 therapeutics.
Collapse
Affiliation(s)
- Panagiota Gianni
- Department of Internal Medicine III, Hematology, Oncology, Palliative Medicine, Rheumatology and Infectious Diseases, University Hospital Ulm, Ulm 89070, Germany
| | - Mark Goldin
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New York, NY 11549, United States
- Feinstein Institutes for Medical Research at Northwell Health, Feinstein Institutes , New York, NY 11030, United States
| | - Sam Ngu
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New York, NY 11549, United States
| | - Stefanos Zafeiropoulos
- Elmezzi Graduate School of Molecular Medicine, Northwell Health, New York, NY 11030, United States
| | - Georgios Geropoulos
- Department of General Surgery, University College London Hospitals, London NW12BU, United Kingdom
| | - Dimitrios Giannis
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, New York, NY 11549, United States
- North Shore/Long Island Jewish General Surgery, Northwell Health, New York, NY 11021, United States
| |
Collapse
|
21
|
Chaudhary N, Jayaraman A, Reinhardt C, Campbell JD, Bosmann M. A single-cell lung atlas of complement genes identifies the mesothelium and epithelium as prominent sources of extrahepatic complement proteins. Mucosal Immunol 2022; 15:927-939. [PMID: 35672453 PMCID: PMC9173662 DOI: 10.1038/s41385-022-00534-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/05/2022] [Accepted: 05/22/2022] [Indexed: 02/04/2023]
Abstract
To understand functional duality of the complement system in host defense and lung injury, a more comprehensive view of its localized production in the lung, and the impact of age on complement production are essential. Here, we explored the expression of complement genes through computational analysis of preexisting single cell RNA sequencing data from lung transcriptomes of healthy young (3 months) and old C57BL/6 mice (24 months), and humans. We characterized the distribution of 48 complement genes. Across 28 distinct immune and non-immune cell types in mice, mesothelial cells expressed the greatest number of complement genes (e.g., C1ra, C2, C3), and regulators (e.g., Serping1, Cfh). C5 was abundant in type II alveolar epithelial cells and C1q in interstitial lung macrophages. There were only moderate differences in gene expression between young and old mice. Among 57 human lung cell types, mesothelial cells showed abundant complement expression. A few differences in gene expression (e.g., FCN1, CFI, C6, C7) were also evident between mice and human lung cells. Our findings present a novel perspective on the expression patterns of complement genes in normal lungs. These findings highlight the potential functions of complement in tissue-specific homeostasis and immunity and may foster a mechanistic understanding of its role in lung health and disease.
Collapse
Affiliation(s)
- Neha Chaudhary
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Archana Jayaraman
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Joshua D Campbell
- Division of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
22
|
Targeting vascular inflammation through emerging methods and drug carriers. Adv Drug Deliv Rev 2022; 184:114180. [PMID: 35271986 PMCID: PMC9035126 DOI: 10.1016/j.addr.2022.114180] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a common dangerous component of pathogenesis of many prevalent conditions with high morbidity and mortality including sepsis, thrombosis, acute respiratory distress syndrome (ARDS), COVID-19, myocardial and cerebral ischemia-reperfusion, infection, and trauma. Inflammatory changes of the vasculature and blood mediate the course and outcome of the pathology in the tissue site of insult, remote organs and systemically. Endothelial cells lining the luminal surface of the vasculature play the key regulatory functions in the body, distinct under normal vs. pathological conditions. In theory, pharmacological interventions in the endothelial cells might enable therapeutic correction of the overzealous damaging pro-inflammatory and pro-thrombotic changes in the vasculature. However, current agents and drug delivery systems (DDS) have inadequate pharmacokinetics and lack the spatiotemporal precision of vascular delivery in the context of acute inflammation. To attain this level of precision, many groups design DDS targeted to specific endothelial surface determinants. These DDS are able to provide specificity for desired tissues, organs, cells, and sub-cellular compartments needed for a particular intervention. We provide a brief overview of endothelial determinants, design of DDS targeted to these molecules, their performance in experimental models with focus on animal studies and appraisal of emerging new approaches. Particular attention is paid to challenges and perspectives of targeted therapeutics and nanomedicine for advanced management of acute inflammation.
Collapse
|
23
|
Functional, transcriptional, and microbial shifts associated with healthy pulmonary aging in rhesus macaques. Cell Rep 2022; 39:110725. [PMID: 35443183 PMCID: PMC9096119 DOI: 10.1016/j.celrep.2022.110725] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/09/2022] [Accepted: 03/30/2022] [Indexed: 01/04/2023] Open
Abstract
Older individuals are at increased risk of developing severe respiratory infections. However, our understanding of the impact of aging on the respiratory tract remains limited as samples from healthy humans are challenging to obtain and results can be confounded by variables such as smoking and diet. Here, we carry out a comprehensive cross-sectional study (n = 34 adult, n = 49 aged) to define the consequences of aging on the lung using the rhesus macaque model. Pulmonary function testing establishes similar age and sex differences as humans. Additionally, we report increased abundance of alveolar and infiltrating macrophages and a concomitant decrease in T cells were in aged animals. scRNAseq reveals shifts from GRZMB to IFN expressing CD8+ T cells in the lungs. These data provide insight into age-related changes in the lungs’ functional, microbial, and immunological landscape that explain increased prevalence and severity of respiratory diseases in the elderly. Rhoades et al. describe age-associated functional, microbial, and immunological changes in the lung using the rhesus macaque model. These data will support further studies aimed at designing and testing interventions to mitigate the impact of age-associated shifts in the lung environment to reduce age-related pulmonary disease in the elderly.
Collapse
|
24
|
Krishna NK, Cunnion KM, Parker GA. The EPICC Family of Anti-Inflammatory Peptides: Next Generation Peptides, Additional Mechanisms of Action, and In Vivo and Ex Vivo Efficacy. Front Immunol 2022; 13:752315. [PMID: 35222367 PMCID: PMC8863753 DOI: 10.3389/fimmu.2022.752315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/17/2022] [Indexed: 11/23/2022] Open
Abstract
The EPICC peptides are a family of peptides that have been developed from the sequence of the capsid protein of human astrovirus type 1 and previously shown to inhibit the classical and lectin pathways of complement. The EPICC peptides have been further optimized to increase aqueous solubility and identify additional mechanisms of action. Our laboratory has developed the lead EPICC molecule, PA-dPEG24 (also known as RLS-0071), which is composed of a 15 amino acid peptide with a C-terminal monodisperse 24-mer PEGylated moiety. RLS-0071 has been demonstrated to possess other mechanisms of action in addition to complement blockade that include the inhibition of neutrophil-driven myeloperoxidase (MPO) activity, inhibition of neutrophil extracellular trap (NET) formation as well as intrinsic antioxidant activity mediated by vicinal cysteine residues contained within the peptide sequence. RLS-0071 has been tested in various ex vivo and in vivo systems and has shown promise for the treatment of both immune-mediated hematological diseases where alterations in the classical complement pathway plays an important pathogenic role as well as in models of tissue-based diseases such as acute lung injury and hypoxic ischemic encephalopathy driven by both complement and neutrophil-mediated pathways (i.e., MPO activity and NET formation). Next generation EPICC peptides containing a sarcosine residue substitution in various positions within the peptide sequence possess aqueous solubility in the absence of PEGylation and demonstrate enhanced complement and neutrophil inhibitory activity compared to RLS-0071. This review details the development of the EPICC peptides, elucidation of their dual-acting complement and neutrophil inhibitory activities and efficacy in ex vivo systems using human clinical specimens and in vivo efficacy in animal disease models.
Collapse
Affiliation(s)
- Neel K Krishna
- Division of Research, ReAlta Life Sciences, Norfolk, VA, United States
| | - Kenji M Cunnion
- Division of Research, ReAlta Life Sciences, Norfolk, VA, United States.,Department of Pediatrics, Children's Hospital of The King's Daughters, Norfolk, VA, United States.,Children's Specialty Group, Norfolk, VA, United States.,Department of Pediatrics, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Grace A Parker
- Division of Research, ReAlta Life Sciences, Norfolk, VA, United States
| |
Collapse
|
25
|
Lebedenko AA, Afonin AA, Semernik OE, Loginova IG, Gunko VO, Larichkin AV, Alliluyev IA, Galkina GA, Panova IV. Proteomic analysis of blood serum - a new approach to the search for diagnostic markers of bronchial asthma in children. Klin Lab Diagn 2022; 67:81-84. [PMID: 35192752 DOI: 10.51620/0869-2084-2022-67-2-81-84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Currently, bronchial asthma (BA) is one of the most pressing medical and social problems, the molecular aspects of the formation and development of BA are insufficiently studied and the diagnosis is not perfect. Carrying out proteomic analysis of BA will not only reveal new biomarkers specific to this disease, but also bring us closer to understanding its pathogenetic mechanisms. The purpose of the study: to study the proteomic profile of blood serum of children with BA to identify proteins associated with this disease A comprehensive clinical and laboratory examination of children suffering from BA and control group patients was performed. Proteomic analysis of depleted blood serum included high-resolution two-dimensional electrophoresis (1 direction: immobiline strips 17cm, pH 3-10, 2 direction: denaturing electrophoresis in 12.5% polyacrylamide gel), protein staining on gels with fluorescent dye Flamingo, protein identification by MALDI-TOF mass spectrometry using the search algorithm Mascot and the Swiss-Prot database. Comparison of the proteomic profile of BA serum and the control group patients serum allowed us to establish that the production of a number of proteins is reduced in this pathology. Among them, proteins in the molecular weight range of 16-33 kDa (p<0.05) were identified: glutathione peroxidase 3, transtyretin, complement components C4b and C3. Research shows that changes in the children's serum proteome occur in BA, affecting proteins that play an important role in immune responses, ligand transport, and antioxidant protection. Special attention should be paid to the differences identified in the course of this work (glutathione peroxidase, transtyretin, C3 and C4 fragments of the complement system) or their combinations. Studying the features of their expression will expand our understanding of the molecular mechanisms underlying chronic inflammation of this disease.
Collapse
|
26
|
Liu Z, Chen L, Gao X, Zou R, Meng Q, Fu Q, Xie Y, Miao Q, Chen L, Tang X, Zhang S, Zhang H, Schroyen M. Quantitative proteomics reveals tissue-specific toxic mechanisms for acute hydrogen sulfide-induced injury of diverse organs in pig. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 806:150365. [PMID: 34555611 DOI: 10.1016/j.scitotenv.2021.150365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
Hydrogen sulfide (H2S) is a highly toxic gas in many environmental and occupational places. It can induce multiple organ injuries particularly in lung, trachea and liver, but the relevant mechanisms remain poorly understood. In this study, we used a TMT-based discovery proteomics to identify key proteins and correlated molecular pathways involved in the pathogenesis of acute H2S-induced toxicity in porcine lung, trachea and liver tissues. Pigs were subjected to acute inhalation exposure of up to 250 ppm of H2S for 5 h for the first time. Changes in hematology and biochemical indexes, serum inflammatory cytokines and histopathology demonstrated that acute H2S exposure induced organs inflammatory injury and dysfunction in the porcine lung, trachea and liver. The proteomic data showed 51, 99 and 84 proteins that were significantly altered in lung, trachea and liver, respectively. Gene ontology (GO) annotation, KEGG pathway and protein-protein interaction (PPI) network analysis revealed that acute H2S exposure affected the three organs via different mechanisms that were relatively similar between lung and trachea. Further analysis showed that acute H2S exposure caused inflammatory damages in the porcine lung and trachea through activating complement and coagulation cascades, and regulating the hyaluronan metabolic process. Whereas antigen presentation was found in the lung but oxidative stress and cell apoptosis was observed exclusively in the trachea. In the liver, an induced dysfunction was associated with protein processing in the endoplasmic reticulum and lipid metabolism. Further validation of some H2S responsive proteins using western blotting indicated that our proteomics data were highly reliable. Collectively, these findings provide insight into toxic molecular mechanisms that could potentially be targeted for therapeutic intervention for acute H2S intoxication.
Collapse
Affiliation(s)
- Zhen Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, University of Liège, Passage des Déportés 2, Gembloux 5030, Belgium
| | - Liang Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xin Gao
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China
| | - Ruixia Zou
- Graduate School, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Qingshi Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qin Fu
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Yanjiao Xie
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qixiang Miao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lei Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Sheng Zhang
- Proteomics and Metabolomics Facility, Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA Teaching and Research Centre, University of Liège, Passage des Déportés 2, Gembloux 5030, Belgium
| |
Collapse
|
27
|
Al-Allaff RGM, Badal Rasheed K. Evaluation of Haemolytic Activity in Smokers by Using Non-Linear Regression. Pak J Biol Sci 2022; 25:509-515. [PMID: 36098186 DOI: 10.3923/pjbs.2022.509.515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
<b>Background and Objective:</b> Smokers are poorly protected against pathogens. Many studies have focused on the concentrations of complement components in comparison to smokers while ignoring the practical sequencing of complement components. There are numerous methods for estimating haemolytic activity (CH50%), all of which need a large number of samples and dilution, in addition to a typically expensive test kit. This novel study attempts to use statistical analysis and use the non-linear regression 'power equation' to extract the CH50% by using 5 serum dilutions only. <b>Materials and Methods:</b> The power equation can multiply the five practical dilutions into hundreds of mathematical loops within the sample range. The (CH50%) value is highly accurate for both the study and comparison sample and was evaluated in11 smokers. <b>Results:</b> The results were contrasted with a control composed of 11 individuals, matched by age and sex. The power equation showed a 6.48% significant reduction in (CH50%) in smokers compared with non-smokers, where a 17.54% reduction was observed. <b>Conclusion:</b> The current study suggests a decrease in the function of the classical complement pathway (CH50%) in smokers. On the other hand, the study provided a new statistical pattern, linking the practical values with default values within the range of dilution and formulating an equation that could be used to extract the value of CH50% with high accuracy.
Collapse
|
28
|
Sager TM, Umbright CM, Mustafa GM, Roberts JR, Orandle MS, Cumpston JL, McKinney WG, Boots T, Kashon ML, Joseph P. Pulmonary toxicity and gene expression changes in response to whole-body inhalation exposure to multi-walled carbon nanotubes in rats. Inhal Toxicol 2022; 34:200-218. [PMID: 35648795 PMCID: PMC9885491 DOI: 10.1080/08958378.2022.2081386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Purpose: To investigate the molecular mechanisms underlying the pulmonary toxicity induced by exposure to one form of multi-walled carbon nanotubes (MWCNT-7).Materials and methods: Rats were exposed, by whole-body inhalation, to air or an aerosol containing MWCNT-7 particles at target cumulative doses (concentration x time) ranging from 22.5 to 180 (mg/m3)h over a three-day (6 hours/day) period and toxicity and global gene expression profiles were determined in the lungs.Results: MWCNT-7 particles, associated with alveolar macrophages (AMs), were detected in rat lungs following the exposure. Mild to moderate lung pathological changes consisting of increased cellularity, thickening of the alveolar wall, alveolitis, fibrosis, and granuloma formation were detected. Bronchoalveolar lavage (BAL) toxicity parameters such as lactate dehydrogenase activity, number of AMs and polymorphonuclear leukocytes (PMNs), intracellular oxidant generation by phagocytes, and levels of cytokines were significantly (p < 0.05) increased in response to exposure to MWCNT-7. Global gene expression profiling identified several significantly differentially expressed genes (fold change >1.5 and FDR p value <0.05) in all the MWCNT-7 exposed rats. Bioinformatic analysis of the gene expression data identified significant enrichment of several diseases/biological function categories (for example, cancer, leukocyte migration, inflammatory response, mitosis, and movement of phagocytes) and canonical pathways (for example, kinetochore metaphase signaling pathway, granulocyte and agranulocyte adhesion and diapedesis, acute phase response, and LXR/RXR activation). The alterations in the lung toxicity parameters and gene expression changes exhibited a dose-response to the MWCNT exposure.Conclusions: Taken together, the data provided insights into the molecular mechanisms underlying the pulmonary toxicity induced by inhalation exposure of rats to MWCNT-7.
Collapse
Affiliation(s)
- Tina M. Sager
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Christina M. Umbright
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Gul Mehnaz Mustafa
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Jenny R. Roberts
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Marlene S. Orandle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Jared L. Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Walter G. McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Theresa Boots
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Michael L. Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - Pius Joseph
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| |
Collapse
|
29
|
Multipopulational transcriptome analysis of post-weaned beef cattle at arrival further validates candidate biomarkers for predicting clinical bovine respiratory disease. Sci Rep 2021; 11:23877. [PMID: 34903778 PMCID: PMC8669006 DOI: 10.1038/s41598-021-03355-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Bovine respiratory disease (BRD) remains the leading infectious disease in post-weaned beef cattle. The objective of this investigation was to contrast the at-arrival blood transcriptomes from cattle derived from two distinct populations that developed BRD in the 28 days following arrival versus cattle that did not. Forty-eight blood samples from two populations were selected for mRNA sequencing based on even distribution of development (n = 24) or lack of (n = 24) clinical BRD within 28 days following arrival; cattle which developed BRD were further stratified into BRD severity cohorts based on frequency of antimicrobial treatment: treated once (treated_1) or treated twice or more and/or died (treated_2+). Sequenced reads (~ 50 M/sample, 150 bp paired-end) were aligned to the ARS-UCD1.2 bovine genome assembly. One hundred and thirty-two unique differentially expressed genes (DEGs) were identified between groups stratified by disease severity (healthy, n = 24; treated_1, n = 13; treated_2+, n = 11) with edgeR (FDR ≤ 0.05). Differentially expressed genes in treated_1 relative to both healthy and treated_2+ were predicted to increase neutrophil activation, cellular cornification/keratinization, and antimicrobial peptide production. Differentially expressed genes in treated_2+ relative to both healthy and treated_1 were predicted to increase alternative complement activation, decrease leukocyte activity, and increase nitric oxide production. Receiver operating characteristic (ROC) curves generated from expression data for six DEGs identified in our current and previous studies (MARCO, CFB, MCF2L, ALOX15, LOC100335828 (aka CD200R1), and SLC18A2) demonstrated good-to-excellent (AUC: 0.800–0.899; ≥ 0.900) predictability for classifying disease occurrence and severity. This investigation identifies candidate biomarkers and functional mechanisms in at arrival blood that predicted development and severity of BRD.
Collapse
|
30
|
Mikus MS, Kolmert J, Andersson LI, Östling J, Knowles RG, Gómez C, Ericsson M, Thörngren JO, Khoonsari PE, Dahlén B, Kupczyk M, De Meulder B, Auffray C, Bakke PS, Beghe B, Bel EH, Caruso M, Chanez P, Chawes B, Fowler SJ, Gaga M, Geiser T, Gjomarkaj M, Horváth I, Howarth PH, Johnston SL, Joos G, Krug N, Montuschi P, Musial J, Niżankowska-Mogilnicka E, Olsson HK, Papi A, Rabe KF, Sandström T, Shaw DE, Siafakas NM, Uhlen M, Riley JH, Bates S, Middelveld RJM, Wheelock CE, Chung KF, Adcock IM, Sterk PJ, Djukanovic R, Nilsson P, Dahlén SE, James A. Plasma proteins elevated in severe asthma despite oral steroid use and unrelated to Type-2 inflammation. Eur Respir J 2021; 59:13993003.00142-2021. [PMID: 34737220 PMCID: PMC8850689 DOI: 10.1183/13993003.00142-2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/24/2021] [Indexed: 12/02/2022]
Abstract
Rationale Asthma phenotyping requires novel biomarker discovery. Objectives To identify plasma biomarkers associated with asthma phenotypes by application of a new proteomic panel to samples from two well-characterised cohorts of severe (SA) and mild-to-moderate (MMA) asthmatics, COPD subjects and healthy controls (HCs). Methods An antibody-based array targeting 177 proteins predominantly involved in pathways relevant to inflammation, lipid metabolism, signal transduction and extracellular matrix was applied to plasma from 525 asthmatics and HCs in the U-BIOPRED cohort, and 142 subjects with asthma and COPD from the validation cohort BIOAIR. Effects of oral corticosteroids (OCS) were determined by a 2-week, placebo-controlled OCS trial in BIOAIR, and confirmed by relation to objective OCS measures in U-BIOPRED. Results In U-BIOPRED, 110 proteins were significantly different, mostly elevated, in SA compared to MMA and HCs. 10 proteins were elevated in SA versus MMA in both U-BIOPRED and BIOAIR (alpha-1-antichymotrypsin, apolipoprotein-E, complement component 9, complement factor I, macrophage inflammatory protein-3, interleukin-6, sphingomyelin phosphodiesterase 3, TNF receptor superfamily member 11a, transforming growth factor-β and glutathione S-transferase). OCS treatment decreased most proteins, yet differences between SA and MMA remained following correction for OCS use. Consensus clustering of U-BIOPRED protein data yielded six clusters associated with asthma control, quality of life, blood neutrophils, high-sensitivity C-reactive protein and body mass index, but not Type-2 inflammatory biomarkers. The mast cell specific enzyme carboxypeptidase A3 was one major contributor to cluster differentiation. Conclusions The plasma proteomic panel revealed previously unexplored yet potentially useful Type-2-independent biomarkers and validated several proteins with established involvement in the pathophysiology of SA. Application of new proteomic panel in two established European asthma cohorts identifies plasma proteins associated with disease severity independently of Type-2 inflammation, suggesting potentially useful novel biomarkers and therapeutic targets.https://bit.ly/3jtTq5m
Collapse
Affiliation(s)
- Maria Sparreman Mikus
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden .,Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Kolmert
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Lars I Andersson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Cristina Gómez
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Ericsson
- Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - John-Olof Thörngren
- Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Payam Emami Khoonsari
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Solna, Sweden
| | - Barbro Dahlén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Maciej Kupczyk
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, University of Lodz, Lodz, Poland
| | | | - Charles Auffray
- European Institute for Systems Biology and Medicine, Lyon, France
| | - Per S Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bianca Beghe
- Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabeth H Bel
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Pascal Chanez
- Assistance Publique des Hôpitaux de Marseille, Clinique des Bronches, Allergies et Sommeil, Aix Marseille Université, Marseille, France
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester; Manchester Academic Health Science Centre and NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Mina Gaga
- Respiratory Medicine Dept and Asthma Centre, Athens Chest Hospital "Sotiria", University of Athens, Athens, Greece
| | - Thomas Geiser
- Department for Pulmonary Medicine, University Hospital and University of Bern, Bern, Switzerland
| | - Mark Gjomarkaj
- Institute for Research and Biomedical Innovation, Italian National Research Council, Palermo, Italy
| | - Ildikó Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Peter H Howarth
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Guy Joos
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Paolo Montuschi
- Department of Pharmacology, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Jacek Musial
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland
| | | | - Henric K Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alberto Papi
- Division of lnternal and Cardiorespiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F Rabe
- Department of Internal Medicine, Christian Albrechts University Kiel, Kiel, Germany
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Dominick E Shaw
- Respiratory Research Unit, University of Nottingham, Nottingham, UK
| | - Nikolaos M Siafakas
- Department of Thoracic Medicine, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Mathias Uhlen
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - John H Riley
- Respiratory Therapeutic Unit, GlaxoSmithKline, London, UK
| | - Stewart Bates
- Respiratory Therapeutic Unit, GlaxoSmithKline, London, UK
| | - Roelinde J M Middelveld
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Craig E Wheelock
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Peter J Sterk
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ratko Djukanovic
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, and Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Peter Nilsson
- Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Sven-Erik Dahlén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Anna James
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
31
|
Peptide inhibition of acute lung injury in a novel two-hit rat model. PLoS One 2021; 16:e0259133. [PMID: 34710157 PMCID: PMC8553074 DOI: 10.1371/journal.pone.0259133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 10/14/2021] [Indexed: 12/19/2022] Open
Abstract
Acute lung injury (ALI) often causes severe trauma that may progress to significant morbidity and mortality. ALI results from a combination of the underlying clinical condition of the patient (e.g., inflammation) with a secondary insult such as viral pneumonia or a blood transfusion. While the secondary insult may be variable, the rapidly progressive disease process leading to pulmonary failure is typically mediated by an overwhelming innate immunological or inflammatory reaction driven by excessive complement and neutrophil-mediated inflammatory responses. We recently developed a ‘two-hit’ ALI rat model mediated by lipopolysaccharide followed by transfusion of incompatible human erythrocytes resulting in complement activation, neutrophil-mediated ALI and free DNA in the blood indicative of neutrophil extracellular trap formation. The objective of this study was to evaluate the role of peptide inhibitor of complement C1 (RLS-0071), a classical complement pathway inhibitor and neutrophil modulator in this animal model. Adolescent male Wistar rats were infused with lipopolysaccharide followed by transfusion of incompatible erythrocytes in the presence or absence of RLS-0071. Blood was collected at various time points to assess complement C5a levels, free DNA and cytokines in isolated plasma. Four hours following erythrocyte transfusion, lung tissue was recovered and assayed for ALI by histology. Compared to animals not receiving RLS-0071, lungs of animals treated with a single dose of RLS-0071 showed significant reduction in ALI as well as reduced levels of C5a, free DNA and inflammatory cytokines in the blood. These results demonstrate that RLS-0071 can modulate neutrophil-mediated ALI in this novel rat model.
Collapse
|
32
|
Kokelj S, Östling J, Georgi B, Fromell K, Ekdahl KN, Olsson HK, Olin AC. Smoking induces sex-specific changes in the small airway proteome. Respir Res 2021; 22:234. [PMID: 34429114 PMCID: PMC8385797 DOI: 10.1186/s12931-021-01825-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Abstract
Introduction Cigarette smoke triggers many cellular and signaling responses in the lung and the resulting inflammation plays a central role in smoke-related lung diseases, such as COPD. We explored the effects of smoking on the small airway proteome in samples obtained by collection of exhaled particles with the aim to identify specific proteins dysregulated by smoking. Methods Exhaled particles were obtained from 38 current smokers, 47 former smokers and 22 healthy controls with the PExA method. 120 ng of sample was collected from individual subjects and analyzed with the SOMAscan proteomics platform. General linear model-based statistics were performed. Results Two hundred and three proteins were detected in at least half of 107 total samples. Active smoking exerted a significant impact on the protein composition of respiratory tract lining fluid (RTLF), with 81 proteins altered in current smokers compared to never smokers (p < 0.05, q < 0.124). Among the proteins most clearly discriminating between current and never smokers were sRAGE, FSTL3, SPOCK2 and protein S, all of them being less abundant in current smokers. Analysis stratified for sex unveiled sex differences with more pronounced proteomic alterations due to active smoking in females than males. Proteins whose abundance was altered by active smoking in women were to a larger extent related to the complement system. The small airway protein profile of former smokers appeared to be more similar to that observed in never smokers. Conclusions The study shows that smoking has a strong impact on protein expression in the small airways, and that smoking affects men and women differently, suggesting PExA sampling combined with high sensitivity protein analysis offers a promising platform for early detection of COPD and identification of novel COPD drug targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01825-6.
Collapse
Affiliation(s)
- Spela Kokelj
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Inst. of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 414, 405 30, Gothenburg, Sweden.
| | - Jörgen Östling
- PExA AB, Gothenburg, Sweden.,Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Benjamin Georgi
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Karin Fromell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Kristina Nilsson Ekdahl
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.,Linnaeus Centre for Biomaterials Chemistry, Linnaeus University, Kalmar, Sweden
| | - Henric K Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, AstraZeneca, BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Anna-Carin Olin
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Inst. of Medicine, Sahlgrenska Academy, University of Gothenburg, Box 414, 405 30, Gothenburg, Sweden
| |
Collapse
|
33
|
Zewde NT, Hsu RV, Morikis D, Palermo G. Systems Biology Modeling of the Complement System Under Immune Susceptible Pathogens. FRONTIERS IN PHYSICS 2021; 9:603704. [PMID: 35145963 PMCID: PMC8827490 DOI: 10.3389/fphy.2021.603704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The complement system is assembled from a network of proteins that function to bring about the first line of defense of the body against invading pathogens. However, complement deficiencies or invasive pathogens can hijack complement to subsequently increase susceptibility of the body to infections. Moreover, invasive pathogens are increasingly becoming resistant to the currently available therapies. Hence, it is important to gain insights into the highly dynamic interaction between complement and invading microbes in the frontlines of immunity. Here, we developed a mathematical model of the complement system composed of 670 ordinary differential equations with 328 kinetic parameters, which describes all three complement pathways (alternative, classical, and lectin) and includes description of mannose-binding lectin, collectins, ficolins, factor H-related proteins, immunoglobulin M, and pentraxins. Additionally, we incorporate two pathogens: (type 1) complement susceptible pathogen and (type 2) Neisseria meningitidis located in either nasopharynx or bloodstream. In both cases, we generate time profiles of the pathogen surface occupied by complement components and the membrane attack complex (MAC). Our model shows both pathogen types in bloodstream are saturated by complement proteins, whereas MACs occupy <<1.0% of the pathogen surface. Conversely, the MAC production in nasopharynx occupies about 1.5-10% of the total N. meningitidis surface, thus making nasal MAC levels at least about eight orders of magnitude higher. Altogether, we predict complement-imbalance, favoring overactivation, is associated with nasopharynx homeostasis. Conversely, orientating toward complement-balance may cause disruption to the nasopharynx homeostasis. Thus, for sporadic meningococcal disease, our model predicts rising nasal levels of complement regulators as early infection biomarkers.
Collapse
Affiliation(s)
- Nehemiah T. Zewde
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| | - Rohaine V. Hsu
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| | - Dimitrios Morikis
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
- Correspondence: Giulia Palermo, , Dimitrios Morikis,
| | - Giulia Palermo
- Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
- Department of Chemistry, University of California, Riverside, Riverside, CA, United States
- Correspondence: Giulia Palermo, , Dimitrios Morikis,
| |
Collapse
|
34
|
Mayr CH, Simon LM, Leuschner G, Ansari M, Schniering J, Geyer PE, Angelidis I, Strunz M, Singh P, Kneidinger N, Reichenberger F, Silbernagel E, Böhm S, Adler H, Lindner M, Maurer B, Hilgendorff A, Prasse A, Behr J, Mann M, Eickelberg O, Theis FJ, Schiller HB. Integrative analysis of cell state changes in lung fibrosis with peripheral protein biomarkers. EMBO Mol Med 2021; 13:e12871. [PMID: 33650774 PMCID: PMC8033531 DOI: 10.15252/emmm.202012871] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/05/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
The correspondence of cell state changes in diseased organs to peripheral protein signatures is currently unknown. Here, we generated and integrated single-cell transcriptomic and proteomic data from multiple large pulmonary fibrosis patient cohorts. Integration of 233,638 single-cell transcriptomes (n = 61) across three independent cohorts enabled us to derive shifts in cell type proportions and a robust core set of genes altered in lung fibrosis for 45 cell types. Mass spectrometry analysis of lung lavage fluid (n = 124) and plasma (n = 141) proteomes identified distinct protein signatures correlated with diagnosis, lung function, and injury status. A novel SSTR2+ pericyte state correlated with disease severity and was reflected in lavage fluid by increased levels of the complement regulatory factor CFHR1. We further discovered CRTAC1 as a biomarker of alveolar type-2 epithelial cell health status in lavage fluid and plasma. Using cross-modal analysis and machine learning, we identified the cellular source of biomarkers and demonstrated that information transfer between modalities correctly predicts disease status, suggesting feasibility of clinical cell state monitoring through longitudinal sampling of body fluid proteomes.
Collapse
Affiliation(s)
- Christoph H Mayr
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Lukas M Simon
- Institute of Computational BiologyHelmholtz Zentrum MünchenMunichGermany
| | - Gabriela Leuschner
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
- Department of Internal Medicine VLudwig‐Maximilians University (LMU) MunichMember of the German Center for Lung Research (DZL), CPC‐M bioArchiveMunichGermany
| | - Meshal Ansari
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
- Institute of Computational BiologyHelmholtz Zentrum MünchenMunichGermany
| | - Janine Schniering
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
- Department of RheumatologyCenter of Experimental RheumatologyUniversity & University Hospital ZurichZurichSwitzerland
| | - Philipp E Geyer
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Ilias Angelidis
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Maximilian Strunz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Pawandeep Singh
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Nikolaus Kneidinger
- Department of Internal Medicine VLudwig‐Maximilians University (LMU) MunichMember of the German Center for Lung Research (DZL), CPC‐M bioArchiveMunichGermany
| | - Frank Reichenberger
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Edith Silbernagel
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Stephan Böhm
- Faculty of MedicineMax von Pettenkofer‐Institute, VirologyNational Reference Center for RetrovirusesLMU MünchenMunichGermany
| | - Heiko Adler
- Helmholtz Zentrum MünchenResearch Unit Lung Repair and Regeneration, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Michael Lindner
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
- University Department of Visceral and Thoracic Surgery SalzburgParacelsus Medical UniversitySalzburgAustria
| | - Britta Maurer
- Department of RheumatologyCenter of Experimental RheumatologyUniversity & University Hospital ZurichZurichSwitzerland
| | - Anne Hilgendorff
- Center for Comprehensive Developmental Care (CDeCLMU)Member of the German Center for Lung Research (DZL)Hospital of the Ludwig‐Maximilians University (LMU)CPC‐M bioArchiveMunichGermany
| | - Antje Prasse
- Department of PneumologyHannover Medical School, Member of the German Center for Lung Research (DZL)HannoverGermany
| | - Jürgen Behr
- Department of Internal Medicine VLudwig‐Maximilians University (LMU) MunichMember of the German Center for Lung Research (DZL), CPC‐M bioArchiveMunichGermany
- Asklepios Fachkliniken Munich‐GautingCPC‐M bioArchive, Member of the German Center for Lung Research (DZL)MunichGermany
| | - Matthias Mann
- Department of Proteomics and Signal TransductionMax Planck Institute of BiochemistryMartinsriedGermany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care MedicineDepartment of MedicineUniversity of PittsburghPittsburghPAUSA
| | - Fabian J Theis
- Institute of Computational BiologyHelmholtz Zentrum MünchenMunichGermany
| | - Herbert B Schiller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC–M bioArchiveHelmholtz Zentrum München, Member of the German Center for Lung Research (DZL)MunichGermany
| |
Collapse
|
35
|
Genomic biomarkers in chronic beryllium disease and sarcoidosis. Respir Med 2021; 187:106390. [PMID: 34399367 DOI: 10.1016/j.rmed.2021.106390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 11/20/2022]
Abstract
Background Previous gene expression studies have identified genes IFNγ, TNFα, RNase 3, CXCL9, and CD55 as potential biomarkers for sarcoidosis and/or chronic beryllium disease (CBD). We hypothesized that differential expression of these genes could function as diagnostic biomarkers for sarcoidosis and CBD, and prognostic biomarkers for sarcoidosis. Study Design/Methods We performed RT-qPCR on whole blood samples from CBD (n = 132), beryllium sensitized (BeS) (n = 109), and sarcoidosis (n = 99) cases and non-diseased controls (n = 97) to determine differential expression of target genes. We then performed logistic regression modeling and generated ROC curves to determine which genes could most accurately differentiate: 1) CBD versus sarcoidosis 2) CBD versus BeS 3) sarcoidosis versus controls 4) non-progressive versus progressive sarcoidosis. Results CD55 and TNFα were significantly upregulated, while CXCL9 was significantly downregulated in CBD compared to sarcoidosis (p < 0.05). The ROC curve from the logistic regression model demonstrated high discriminatory ability of the combination of CD55, TNFα, and CXCL9 to distinguish between CBD and sarcoidosis with an AUC of 0.98. CD55 and TNFα were significantly downregulated in sarcoidosis compared to controls (p < 0.05). The ROC curve from the model showed a reasonable discriminatory ability of CD55 and TNFα to distinguish between sarcoidosis and controls with an AUC of 0.86. There was no combination of genes that could accurately differentiate between CBD and BeS or sarcoidosis phenotypes. Interpretation CD55, TNFα and CXCL9 expression levels can accurately differentiate between CBD and sarcoidosis, while CD55 and TNFα expression levels can accurately differentiate sarcoidosis and controls.
Collapse
|
36
|
Joseph P, Umbright CM, Roberts JR, Cumpston JL, Orandle MS, McKinney WG, Sager TM. Lung toxicity and gene expression changes in response to whole-body inhalation exposure to cellulose nanocrystal in rats. Inhal Toxicol 2021; 33:66-80. [PMID: 33602020 PMCID: PMC10442725 DOI: 10.1080/08958378.2021.1884320] [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: 11/30/2020] [Accepted: 01/27/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Human exposure to cellulose nanocrystal (CNC) is possible during the production and/or use of products containing CNC. The objectives of the current study were to determine the lung toxicity of CNC and the underlying molecular mechanisms of the toxicity. METHODS Rats were exposed to air or CNC (20 mg/m3, six hours/day, 14 d) by whole-body inhalation and lung toxicity and global gene expression profile were determined. RESULTS Significant increases in lactate dehydrogenase activity, pro-inflammatory cytokine levels, phagocyte oxidant production, and macrophage and neutrophil counts were detected in the bronchoalveolar lavage cells or fluid from the CNC exposed rats. Mild lung histological changes, such as the accumulation of macrophages and neutrophils, were detected in the CNC exposed rats. Gene expression profiling by next generation sequencing identified 531 genes whose expressions were significantly different in the lungs of the CNC exposed rats, compared with the controls. Bioinformatic analysis of the lung gene expression data identified significant enrichment in several biological functions and canonical pathways including those related to inflammation (cellular movement, immune cell trafficking, inflammatory diseases and response, respiratory disease, complement system, acute phase response, leukocyte extravasation signaling, granulocyte and agranulocyte adhesion and diapedesis, IL-10 signaling, and phagosome formation and maturation) and oxidative stress (NRF2-mediated oxidative stress response, production of nitric oxide and reactive oxygen species in macrophages, and free radical scavenging). CONCLUSION Our data demonstrated that inhalation exposure of rats to CNC resulted in lung toxicity mediated mainly through the induction of inflammation and oxidative stress.
Collapse
Affiliation(s)
- Pius Joseph
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Christina M Umbright
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Jenny R Roberts
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Jared L Cumpston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Marlene S Orandle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Walter G McKinney
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Tina M Sager
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA
| |
Collapse
|
37
|
Zhang Z, Wang T, Liu F, Zhu A, Gu G, Luo J, Xu J, Zhao J, Li Y, Li Y, Liu X, Zhong N, Lu W. The proteomic characteristics of airway mucus from critical ill COVID-19 patients. Life Sci 2021; 269:119046. [PMID: 33453245 PMCID: PMC7806453 DOI: 10.1016/j.lfs.2021.119046] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/01/2021] [Accepted: 01/07/2021] [Indexed: 01/08/2023]
Abstract
Background The pandemic of the coronavirus disease 2019 (COVID-19) has brought a global public health crisis. However, the pathogenesis underlying COVID-19 are barely understood. Methods In this study, we performed proteomic analyses of airway mucus obtained by bronchoscopy from severe COVID-19 patients. In total, 2351 and 2073 proteins were identified and quantified in COVID-19 patients and healthy controls, respectively. Results Among them, 92 differentiated expressed proteins (DEPs) (46 up-regulated and 46 down-regulated) were found with a fold change >1.5 or <0.67 and a p-value <0.05, and 375 proteins were uniquely present in airway mucus from COVID-19 patients. Pathway and network enrichment analyses revealed that the 92 DEPs were mostly associated with metabolic, complement and coagulation cascades, lysosome, and cholesterol metabolism pathways, and the 375 COVID-19 only proteins were mainly enriched in amino acid degradation (Valine, Leucine and Isoleucine degradation), amino acid metabolism (beta-Alanine, Tryptophan, Cysteine and Methionine metabolism), oxidative phosphorylation, phagosome, and cholesterol metabolism pathways. Conclusions This study aims to provide fundamental data for elucidating proteomic changes of COVID-19, which may implicate further investigation of molecular targets directing at specific therapy.
Collapse
Affiliation(s)
- Zili Zhang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tao Wang
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fei Liu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guoping Gu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jieping Luo
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingyi Xu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiming Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuanyuan Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Annane D, Heming N, Grimaldi-Bensouda L, Frémeaux-Bacchi V, Vigan M, Roux AL, Marchal A, Michelon H, Rottman M, Moine P. Eculizumab as an emergency treatment for adult patients with severe COVID-19 in the intensive care unit: A proof-of-concept study. EClinicalMedicine 2020; 28:100590. [PMID: 33173853 PMCID: PMC7644240 DOI: 10.1016/j.eclinm.2020.100590] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Complement pathway inhibition may provide benefit for severe acute respiratory illnesses caused by viral infections such as COVID-19. We present results from a nonrandomized proof-of-concept study of complement C5 inhibitor eculizumab for treatment of severe COVID-19. METHODS All patients (N = 80) with confirmed SARS-CoV-2 infection and severe COVID-19 admitted to our intensive care unit between March 10 and May 5, 2020 were included. Forty-five patients were treated with standard care and 35 with standard care plus eculizumab through expanded-access emergency treatment. The prespecified primary outcome was day-15 survival. Clinical laboratory values and biomarkers, complement levels, and treatment-emergent serious adverse events (TESAEs) were also assessed. FINDINGS At day 15, estimated survival was 82.9% (95% CI: 70.4%‒95.3%) with eculizumab and 62.2% (48.1%‒76.4%) without eculizumab (log-rank test, P = 0.04). Patients treated with eculizumab experienced a significantly more rapid decrease in lactate, blood urea nitrogen, total and conjugated bilirubin levels and a significantly more rapid increase in platelet count, prothrombin time, and in the ratio of arterial oxygen tension over fraction of inspired oxygen versus patients treated without eculizumab. Eculizumab-associated changes in complement levels, laboratory values, and biomarkers were consistent with terminal complement inhibition, reduced hypoxia, and decreased inflammation. TESAEs of special interest occurring in >5% of patients treated with/without eculizumab were ventilator-associated pneumonia (51%/24%), bacteremia (11%/2%), gastroduodenal hemorrhage (14%/16%), and hemolysis (3%/18%). INTERPRETATION Findings from this proof-of-concept study suggest eculizumab may improve survival and reduce hypoxia in patients with severe COVID-19. Randomized studies evaluating the efficacy and safety of this treatment approach are needed. FUNDING Programme d'Investissements d'Avenir: ANR-18-RHUS60004.
Collapse
Affiliation(s)
- Djillali Annane
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, 104 Boulevard Raymond Poincaré, Garches 92380, France
| | - Nicholas Heming
- Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Lamiae Grimaldi-Bensouda
- Clinical Research Unit AP-HP, Paris-Saclay, Hôpital Raymond Poincaré, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, CESP Anti-Infective Evasion and Pharmacoepidemiology Team, F-78180 Montigny-Le-Bretonneux, France
| | - Véronique Frémeaux-Bacchi
- Laboratory of Immunology, Georges-Pompidou European Hospital (APHP), Invasive Bacterial Infection Unit and National Reference Center for Meningococci, Pasteur Institute, Centre de Recherche des Cordeliers, Paris, France
| | - Marie Vigan
- Clinical Research Unit, AP-HP, Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
| | - Anne-Laure Roux
- Department of Microbiology and Innovative Biomarkers Platform, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Armance Marchal
- Laboratory of Immunology, Georges-Pompidou European Hospital (APHP), Paris, France
| | - Hugues Michelon
- Pharmacy Department, Hôpital Raymond Poincaré (APHP), University Paris Saclay, Garches, France
| | - Martin Rottman
- Department of Microbiology and Innovative Biomarkers Platform, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Pierre Moine
- Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - for the Garches COVID 19 Collaborative Group
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, 104 Boulevard Raymond Poincaré, Garches 92380, France
- Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
- Clinical Research Unit AP-HP, Paris-Saclay, Hôpital Raymond Poincaré, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, CESP Anti-Infective Evasion and Pharmacoepidemiology Team, F-78180 Montigny-Le-Bretonneux, France
- Laboratory of Immunology, Georges-Pompidou European Hospital (APHP), Invasive Bacterial Infection Unit and National Reference Center for Meningococci, Pasteur Institute, Centre de Recherche des Cordeliers, Paris, France
- Clinical Research Unit, AP-HP, Paris-Saclay, Hôpital Raymond Poincaré, Garches, France
- Department of Microbiology and Innovative Biomarkers Platform, Hôpital Raymond Poincaré (APHP), Laboratory of Infection & Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
- Laboratory of Immunology, Georges-Pompidou European Hospital (APHP), Paris, France
- Pharmacy Department, Hôpital Raymond Poincaré (APHP), University Paris Saclay, Garches, France
| |
Collapse
|
39
|
Sriram K, Insel PA. Inflammation and thrombosis in COVID-19 pathophysiology: proteinase-activated and purinergic receptors as drivers and candidate therapeutic targets. Physiol Rev 2020; 101:545-567. [PMID: 33124941 PMCID: PMC8238137 DOI: 10.1152/physrev.00035.2020] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Paul A Insel
- Department of Pharmacology and Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
40
|
Świerzko AS, Cedzyński M. The Influence of the Lectin Pathway of Complement Activation on Infections of the Respiratory System. Front Immunol 2020; 11:585243. [PMID: 33193407 PMCID: PMC7609860 DOI: 10.3389/fimmu.2020.585243] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022] Open
Abstract
Lung diseases are among the leading causes of morbidity and mortality. Complement activation may prevent a variety of respiratory infections, but on the other hand, could exacerbate tissue damage or contribute to adverse side effects. In this review, the associations of factors specific for complement activation via the lectin pathway (LP) with infections of the respiratory system, from birth to adulthood, are discussed. The most extensive data concern mannose-binding lectin (MBL) which together with other collectins (collectin-10, collectin-11) and the ficolins (ficolin-1, ficolin-2, ficolin-3) belong to pattern-recognition molecules (PRM) specific for the LP. Those PRM form complexes with MBL-associated serine proteases (MASP-1, MASP-2, MASP-3) and related non-enzymatic factors (MAp19, MAp44). Beside diseases affecting humanity for centuries like tuberculosis or neonatal pneumonia, some recently published data concerning COVID-19 are summarized.
Collapse
Affiliation(s)
- Anna S Świerzko
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Łódź, Poland
| | - Maciej Cedzyński
- Laboratory of Immunobiology of Infections, Institute of Medical Biology, Polish Academy of Sciences, Łódź, Poland
| |
Collapse
|
41
|
Sananez I, Raiden S, Holgado MP, Seery V, De Lillo L, Davenport C, Ferrero F, Peeples ME, Geffner J, Arruvito L. Upregulation of CD32 in T Cells from Infants with Severe Respiratory Syncytial Virus Disease: A New Costimulatory Pathway? Am J Respir Cell Mol Biol 2020; 63:133-136. [PMID: 32609012 DOI: 10.1165/rcmb.2020-0025le] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Inés Sananez
- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida.,Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Silvina Raiden
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina.,Hospital General de Niños "Pedro de Elizalde"Buenos Aires, Argentinaand
| | - María P Holgado
- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida.,Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Vanesa Seery
- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida.,Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Leonardo De Lillo
- Hospital General de Niños "Pedro de Elizalde"Buenos Aires, Argentinaand
| | | | - Fernando Ferrero
- Hospital General de Niños "Pedro de Elizalde"Buenos Aires, Argentinaand
| | - Mark E Peeples
- Abigail Wexner Research Institute at Nationwide Children's HospitalColumbus, Ohio
| | - Jorge Geffner
- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida.,Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| | - Lourdes Arruvito
- Universidad de Buenos Aires, Instituto de Investigaciones Biomédicas en Retrovirus y Sida.,Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires, Argentina
| |
Collapse
|
42
|
Guo J, Li M, Yang Y, Zhang L, Zhang LW, Sun QY. Pretreatment with atorvastatin ameliorates cobra venom factor-induced acute lung inflammation in mice. BMC Pulm Med 2020; 20:263. [PMID: 33046059 PMCID: PMC7552367 DOI: 10.1186/s12890-020-01307-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 10/04/2020] [Indexed: 11/28/2022] Open
Abstract
Background The complement system plays a critical role as the pathogenic factor in the models of acute lung injury due to various causes. Cobra venom factor (CVF) is a commonly used complement research tool. The CVF can cause acute inflammation in the lung by producing complement activation components. Atorvastatin (ATR) is a 3-hydroxy-3-methylglutaryl coenzyme A inhibitor approved for control of plasma cholesterol levels. This inhibitor can reduce the acute pulmonary inflammatory response. However, the ability of ATR in treating acute lung inflammation caused by complement activation is still unknown. Therefore, we investigated the effect of ATR on lung inflammation in mice induced by activation of the complement alternative pathway in this study. Methods ATR (10 mg/kg/day via oral gavage) was administered for 7 days before tail vein injection of CVF (25 μg/kg). On the seventh day, all mice were sacrificed 1 h after injection. The lung lobe, bronchoalveolar lavage fluid (BALF), and blood samples were collected. The myeloperoxidase (MPO) activity of the lung homogenate, the leukocyte cell count, and the protein content of BALF were measured. The levels of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), P-selectin, and Intercellular cell adhesion molecule-1 (ICAM-1) in BALF and serum were determined by enzyme-linked immunosorbent assay. The pathological change of the lung tissue was observed by hematoxylin and eosin staining. The deposition of C5b-9 in the lung tissue was detected by immunohistochemistry. The phosphorylation of NF-κB p65 in the lung tissues was examined by immunohistochemistry and western blotting. Results The lung inflammation levels were determined by measuring the leukocyte cell numbers and protein content of BALF, the lung MPO activity, and expression and staining of the inflammatory mediators (IL-6 and TNF-α), and adhesion molecules (P-selectin and ICAM-1) for lung lesion. A significant reduction in the lung inflammation levels was observed after 7 days in ATR pre-treated mice with a CVF-induced lung disease. Deposition of C5b-9 was significantly alleviated by ATR pretreatment. Early intervention with ATR significantly reduced the development of acute lung inflammation on the basis of phosphorylation of NF-κB p65 in the lung. Conclusion These findings suggest the identification of ATR treatment for the lung inflammation induced by activating the complement system on the basis of its anti-inflammatory response. Together with the model replicating the complement activating characteristics of acute lung injury, the results may be translatable to the overactivated complement relevant diseases.
Collapse
Affiliation(s)
- Jing Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.,Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China.,Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, China
| | - Min Li
- General Ward, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Yi Yang
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Lin Zhang
- Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Li-Wei Zhang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, 030006, China.,Key Laboratory of Chemical Biology and Molecular Engineering of Education Ministry, Institute of Molecular Science, Shanxi University, Taiyuan, 030006, China
| | - Qian-Yun Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China. .,Center for Pharmacology and Bioactivity Research, The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China.
| |
Collapse
|
43
|
Hoevenaar M, Goossens D, Roorda J. Angiotensin-converting enzyme 2, the complement system, the kallikrein-kinin system, type-2 diabetes, interleukin-6, and their interactions regarding the complex COVID-19 pathophysiological crossroads. J Renin Angiotensin Aldosterone Syst 2020; 21:1470320320979097. [PMID: 33283602 PMCID: PMC7724427 DOI: 10.1177/1470320320979097] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Because of the current COVID-19-pandemic, the world is currently being held hostage in various lockdowns. ACE2 facilitates SARS-CoV-2 cell-entry, and is at the very center of several pathophysiological pathways regarding the RAAS, CS, KKS, T2DM, and IL-6. Their interactions with severe COVID-19 complications (e.g. ARDS and thrombosis), and potential therapeutic targets for pharmacological intervention, will be reviewed.
Collapse
Affiliation(s)
| | | | - Janne Roorda
- Medical Doctor, General Practice
van Dijk, Oisterwijk, The Netherlands
| |
Collapse
|
44
|
Neupane AS, Willson M, Chojnacki AK, Vargas E Silva Castanheira F, Morehouse C, Carestia A, Keller AE, Peiseler M, DiGiandomenico A, Kelly MM, Amrein M, Jenne C, Thanabalasuriar A, Kubes P. Patrolling Alveolar Macrophages Conceal Bacteria from the Immune System to Maintain Homeostasis. Cell 2020; 183:110-125.e11. [PMID: 32888431 DOI: 10.1016/j.cell.2020.08.020] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 07/14/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
During respiration, humans breathe in more than 10,000 liters of non-sterile air daily, allowing some pathogens access to alveoli. Interestingly, alveoli outnumber alveolar macrophages (AMs), which favors alveoli devoid of AMs. If AMs, like most tissue macrophages, are sessile, then this numerical advantage would be exploited by pathogens unless neutrophils from the blood stream intervened. However, this would translate to omnipresent persistent inflammation. Developing in vivo real-time intravital imaging of alveoli revealed AMs crawling in and between alveoli using the pores of Kohn. Importantly, these macrophages sensed, chemotaxed, and, with high efficiency, phagocytosed inhaled bacterial pathogens such as P. aeruginosa and S. aureus, cloaking the bacteria from neutrophils. Impairing AM chemotaxis toward bacteria induced superfluous neutrophil recruitment, leading to inappropriate inflammation and injury. In a disease context, influenza A virus infection impaired AM crawling via the type II interferon signaling pathway, and this greatly increased secondary bacterial co-infection.
Collapse
Affiliation(s)
- Arpan Sharma Neupane
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Michelle Willson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Fernanda Vargas E Silva Castanheira
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Christopher Morehouse
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Agostina Carestia
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Ashley Elaine Keller
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Moritz Peiseler
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Antonio DiGiandomenico
- Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA
| | - Margaret Mary Kelly
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Matthias Amrein
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Craig Jenne
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Ajitha Thanabalasuriar
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal Canada H3G1Y6; Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, 1 MedImmune Way, Gaithersburg, MD 20878, USA.
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Microbiology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada; Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
45
|
Frye CC, Bery AI, Kreisel D, Kulkarni HS. Sterile inflammation in thoracic transplantation. Cell Mol Life Sci 2020; 78:581-601. [PMID: 32803398 DOI: 10.1007/s00018-020-03615-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/20/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The life-saving benefits of organ transplantation can be thwarted by allograft dysfunction due to both infectious and sterile inflammation post-surgery. Sterile inflammation can occur after necrotic cell death due to the release of endogenous ligands [such as damage-associated molecular patterns (DAMPs) and alarmins], which perpetuate inflammation and ongoing cellular injury via various signaling cascades. Ischemia-reperfusion injury (IRI) is a significant contributor to sterile inflammation after organ transplantation and is associated with detrimental short- and long-term outcomes. While the vicious cycle of sterile inflammation and cellular injury is remarkably consistent amongst different organs and even species, we have begun understanding its mechanistic basis only over the last few decades. This understanding has resulted in the developments of novel, yet non-specific therapies for mitigating IRI-induced graft damage, albeit with moderate results. Thus, further understanding of the mechanisms underlying sterile inflammation after transplantation is critical for identifying personalized therapies to prevent or interrupt this vicious cycle and mitigating allograft dysfunction. In this review, we identify common and distinct pathways of post-transplant sterile inflammation across both heart and lung transplantation that can potentially be targeted.
Collapse
Affiliation(s)
- C Corbin Frye
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA.
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA
| |
Collapse
|
46
|
Kamminga T, Benis N, Martins Dos Santos V, Bijlsma JJE, Schaap PJ. Combined Transcriptome Sequencing of Mycoplasma hyopneumoniae and Infected Pig Lung Tissue Reveals Up-Regulation of Bacterial F1-Like ATPase and Down-Regulation of the P102 Cilium Adhesin in vivo. Front Microbiol 2020; 11:1679. [PMID: 32765473 PMCID: PMC7379848 DOI: 10.3389/fmicb.2020.01679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/26/2020] [Indexed: 12/21/2022] Open
Abstract
Mycoplasma hyopneumoniae (M. hyopneumoniae) causes enzootic pneumonia in pigs but it is still largely unknown which host-pathogen interactions enable persistent infection and cause disease. In this study, we analyzed the host and bacterial transcriptomes during infection using RNA sequencing. Comparison of the transcriptome of lung lesion tissue from infected pigs with lung tissue from non-infected animals, identified 424 differentially expressed genes (FDR < 0.01 and fold change > 1.5LOG2). These genes were part of the following major pathways of the immune system: interleukin signaling (type 4, 10, 13, and 18), regulation of Toll-like receptors by endogenous ligand and activation of C3 and C5 in the complement system. Besides analyzing the lung transcriptome, a sampling protocol was developed to obtain enough bacterial mRNA from infected lung tissue for RNA sequencing. This was done by flushing infected lobes in the lung, and subsequently enriching for bacterial RNA. On average, 2.2 million bacterial reads were obtained per biological replicate to analyze the bacterial in vivo transcriptome. We compared the in vivo bacterial transcriptome with the transcriptome of bacteria grown in vitro and identified 22 up-regulated and 30 down-regulated genes (FDR < 0.01 and fold change > 2LOG2). Six out of seven genes in the operon encoding the mycoplasma specific F1-like ATPase (MHP_RS02445-MHP_RS02475) and all genes in the operon MHP_RS01965-MHP_RS01990 with functions related to nucleotide metabolism, spermidine transport and glycerol-3-phoshate transport were up-regulated in vivo. Down-regulated in vivo were genes related to glycerol uptake, cilium adhesion (P102), cell division and myo-inositol metabolism. In addition to providing a novel method to isolate bacterial mRNA from infected lung, this study provided insights into changes in gene expression during infection, which could help development of novel treatment strategies against enzootic pneumonia caused by M. hyopneumoniae.
Collapse
Affiliation(s)
- Tjerko Kamminga
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands.,Bioprocess Technology and Support, MSD Animal Health, Boxmeer, Netherlands
| | - Nirupama Benis
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | - Vitor Martins Dos Santos
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| | | | - Peter J Schaap
- Laboratory of Systems and Synthetic Biology, Department of Agrotechnology and Food Sciences, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
47
|
Polycarpou A, Howard M, Farrar CA, Greenlaw R, Fanelli G, Wallis R, Klavinskis LS, Sacks S. Rationale for targeting complement in COVID-19. EMBO Mol Med 2020; 12:e12642. [PMID: 32559343 PMCID: PMC7323084 DOI: 10.15252/emmm.202012642] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/28/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022] Open
Abstract
A novel coronavirus, SARS-CoV-2, has recently emerged in China and spread internationally, posing a health emergency to the global community. COVID-19 caused by SARS-CoV-2 is associated with an acute respiratory illness that varies from mild to the life-threatening acute respiratory distress syndrome (ARDS). The complement system is part of the innate immune arsenal against pathogens, in which many viruses can evade or employ to mediate cell entry. The immunopathology and acute lung injury orchestrated through the influx of pro-inflammatory macrophages and neutrophils can be directly activated by complement components to prime an overzealous cytokine storm. The manifestations of severe COVID-19 such as the ARDS, sepsis and multiorgan failure have an established relationship with activation of the complement cascade. We have collected evidence from all the current studies we are aware of on SARS-CoV-2 immunopathogenesis and the preceding literature on SARS-CoV-1 and MERS-CoV infection linking severe COVID-19 disease directly with dysfunction of the complement pathways. This information lends support for a therapeutic anti-inflammatory strategy against complement, where a number of clinically ready potential therapeutic agents are available.
Collapse
MESH Headings
- Adult
- Alveolar Epithelial Cells/immunology
- Alveolar Epithelial Cells/metabolism
- Alveolar Epithelial Cells/virology
- Angiotensin-Converting Enzyme 2
- Animals
- Betacoronavirus/physiology
- COVID-19
- Child
- Complement Activation/drug effects
- Complement C3b/antagonists & inhibitors
- Complement C3b/physiology
- Complement Inactivating Agents/pharmacology
- Complement Inactivating Agents/therapeutic use
- Coronavirus Infections/drug therapy
- Coronavirus Infections/immunology
- Cytokine Release Syndrome/drug therapy
- Cytokine Release Syndrome/etiology
- Cytokine Release Syndrome/immunology
- Glycosylation
- Humans
- Immunity, Innate
- Ligands
- Mice
- Models, Animal
- Models, Molecular
- Pandemics
- Pattern Recognition, Automated
- Peptidyl-Dipeptidase A/metabolism
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/immunology
- Protein Conformation
- Protein Processing, Post-Translational
- Receptors, Virus/metabolism
- Respiratory Distress Syndrome/etiology
- Respiratory Distress Syndrome/immunology
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/metabolism
- COVID-19 Drug Treatment
Collapse
Affiliation(s)
- Anastasia Polycarpou
- MRC Centre of TransplantationPeter Gorer Department of ImmunobiologySchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| | - Mark Howard
- MRC Centre of TransplantationPeter Gorer Department of ImmunobiologySchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| | - Conrad A Farrar
- MRC Centre of TransplantationPeter Gorer Department of ImmunobiologySchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| | - Roseanna Greenlaw
- MRC Centre of TransplantationPeter Gorer Department of ImmunobiologySchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| | - Giorgia Fanelli
- MRC Centre of TransplantationPeter Gorer Department of ImmunobiologySchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| | - Russell Wallis
- Department of Respiratory Science and InfectionLeicester Institute of Chemical and Structural BiologyUniversity of LeicesterLeicesterUK
| | - Linda S Klavinskis
- Department of Infectious DiseasesSchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| | - Steven Sacks
- MRC Centre of TransplantationPeter Gorer Department of ImmunobiologySchool of Immunology and Microbial SciencesGuy's HospitalKing's College LondonLondonUK
| |
Collapse
|
48
|
Sadeghi S, Tapak M, Ghazanfari T, Mosaffa N. A review of Sulfur Mustard-induced pulmonary immunopathology: An Alveolar Macrophage Approach. Toxicol Lett 2020; 333:115-129. [PMID: 32758513 DOI: 10.1016/j.toxlet.2020.07.035] [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] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/27/2022]
Abstract
Despite many studies investigating the mechanism of Sulfur Mustard (SM) induced lung injury, the underlying mechanism is still unclear. Inflammatory and subsequent fibroproliferative stages of SM-toxicity are based upon several highly-related series of events controlled by the immune system. The inhalation of SM gas variably affects different cell populations within the lungs. Various studies have shown the critical role of macrophages in triggering a pulmonary inflammatory response as well as its maintenance, resolution, and repair. Importantly, macrophages can serve as either pro-inflammatory or anti-inflammatory populations depending on the present conditions at any pathological stage. Different characteristics of macrophages, including their differentiation, phenotypic, and functional properties, as well as interactions with other cell populations determine the outcomes of lung diseases and the extent of long- or short-term pulmonary damage induced by SM. In this paper, we summarize the current state of knowledge regarding the role of alveolar macrophages and their mediators in the pathogenesis of SM in pulmonary injury. Investigating the specific cells and mechanisms involved in SM-lung injury may be useful in finding new target opportunities for treatment of this injury.
Collapse
Affiliation(s)
- Somaye Sadeghi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahtab Tapak
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran; Department of Immunology, Shahed University, Tehran, Iran.
| | - Nariman Mosaffa
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Proteomic analysis of serum-derived extracellular vesicles in ankylosing spondylitis patients. Int Immunopharmacol 2020; 87:106773. [PMID: 32679547 DOI: 10.1016/j.intimp.2020.106773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic inflammatory disease, whose pathogenesis is still unclear. Many studies show the proteins in extracellular vesicle (EVs) would change regularly in many diseases. The study aims to explore the proteins contents of serum-derived EVs in AS patients. METHODS EVs were separated by ExoQuickTM kit. The protein profiles of AS patients and healthy subjects were analyzed by Label-free-liquid chromatography mass spectrometry (LC-MS/MS) technology. Enzyme-linked immunosorbent assay (ELISA) was used to verify the levels of the differently expressed proteins. Receiver operation characteristic (ROC) curves and bioinformatic analysis were conducted. RESULTS Six hundred and ten serum-derived EVs proteins from AS patients were detected. Seventy-three diferentially expressed proteins were found in AS group, compared with healthy subjects. Of these, 31 proteins were up-regulated in AS group, while 42 proteins were down-regulated. ELISA result showed that EVs-derived serum amyloid A-1 (SAA1) was higher in AS group, which was consistent with the Label-free-LC-MS/MS data. ROC curves result revealed that the area under curve (AUC) value of EVs-derived SAA1 for AS was 0.768 (0.652-0.885). Bioinformatic analysis revealed that the differently expressed proteins in AS group were significantly involved in "complement and coagulation cascades", "staphylococcus aureus infection", "systemic lupus erythematosus" and "PI3K-Akt signaling pathway". CONCLUSIONS The protein profiles of serum-derived EVs in AS patients and healthy subjects were different. EVs-derived SAA1 may be a potential biomarkes of AS. The function analysis indicated that the differentially expressed proteins may potentially participate in immune response.
Collapse
|
50
|
Domingo-Gonzalez R, Zanini F, Che X, Liu M, Jones RC, Swift MA, Quake SR, Cornfield DN, Alvira CM. Diverse homeostatic and immunomodulatory roles of immune cells in the developing mouse lung at single cell resolution. eLife 2020; 9:e56890. [PMID: 32484158 PMCID: PMC7358008 DOI: 10.7554/elife.56890] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
At birth, the lungs rapidly transition from a pathogen-free, hypoxic environment to a pathogen-rich, rhythmically distended air-liquid interface. Although many studies have focused on the adult lung, the perinatal lung remains unexplored. Here, we present an atlas of the murine lung immune compartment during early postnatal development. We show that the late embryonic lung is dominated by specialized proliferative macrophages with a surprising physical interaction with the developing vasculature. These macrophages disappear after birth and are replaced by a dynamic mixture of macrophage subtypes, dendritic cells, granulocytes, and lymphocytes. Detailed characterization of macrophage diversity revealed an orchestration of distinct subpopulations across postnatal development to fill context-specific functions in tissue remodeling, angiogenesis, and immunity. These data both broaden the putative roles for immune cells in the developing lung and provide a framework for understanding how external insults alter immune cell phenotype during a period of rapid lung growth and heightened vulnerability.
Collapse
Affiliation(s)
- Racquel Domingo-Gonzalez
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Fabio Zanini
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South WalesSydneyAustralia
| | - Xibing Che
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Michael A Swift
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - David N Cornfield
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|