1
|
Sadler M, Cannata A, Mackie S, Mondi Anandhakrishna R, Argunhan F, Ferone E, Mohammad AA, Salim J, Tantichirasakul N, Lam MT, Ambon J, Shamsi A, Piper S, Napolitani G, Shah AM, McDonagh T, Scott PA, Quek L, Bromage DI. Ethnic variations in neutrophil count as predictors of prognosis following acute myocardial infarction. Atherosclerosis 2025; 403:119169. [PMID: 40157178 DOI: 10.1016/j.atherosclerosis.2025.119169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/03/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
AIMS Elevated neutrophils are associated with a poor prognosis after acute myocardial infarction (AMI) but it is not known if ethnicity influences the association between neutrophil count and outcome. We aimed to describe the temporal dynamics of neutrophils after AMI, and assess the interaction between ethnicity, neutrophil count, and outcomes after AMI. METHODS Consecutive patients presenting with AMI between 2016 and 2023 were divided into two groups according to their median neutrophil count. Ethnicity was dichotomised as white and other ethnic groups combined (referred to as 'ethnic minorities'). The primary outcome was in-hospital mortality, with a secondary outcome of 60-day mortality. RESULTS In our study of 3062 AMI patients (76 % white, 24 % from ethnic minority groups), we found that neutrophil counts rose early post AMI, which coincided with a nadir of the other cell groups. We identified a relative baseline neutropenia in ethnic minority individuals, compared to white individuals (6.85 vs 8.42 × 109/L). We observed a significant, independent association between elevated neutrophils at baseline and the primary outcome of in-hospital mortality (OR 2.06, p < 0.001) and secondary outcome of 60-day all-cause mortality (HR 1.08, p = 0.002). Sub-group analysis revealed a significant interaction between ethnicity and elevated neutrophils (p = 0.004), indicating that a comparable neutrophil count conferred an increased risk for ethnic minority patients for both outcomes. CONCLUSIONS We report ethnicity-specific leucocyte dynamics after AMI. Furthermore, neutrophil count is associated with a disproportionate risk in ethnic minority compared with white individuals. Understanding post-AMI inflammation and its interaction with ethnicity is essential in providing personalised prognostication and patient management.
Collapse
Affiliation(s)
- Matthew Sadler
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Antonio Cannata
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Sarah Mackie
- Myeloid Leukaemia Genomics and Biology Group, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE5 8AF, UK
| | - Rupavidhya Mondi Anandhakrishna
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Fulye Argunhan
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Emma Ferone
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Al-Agil Mohammad
- Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Jamila Salim
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Narun Tantichirasakul
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Mei Tung Lam
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Josel Ambon
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK
| | - Aamir Shamsi
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Susan Piper
- Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Giorgio Napolitani
- Myeloid Leukaemia Genomics and Biology Group, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE5 8AF, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Theresa McDonagh
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Paul A Scott
- Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK
| | - Lynn Quek
- Myeloid Leukaemia Genomics and Biology Group, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE5 8AF, UK; Department of Haematological Medicine, King's College Hospital NHS Foundation Trust, London, UK
| | - Daniel I Bromage
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, James Black Centre, 125 Coldharbour Lane, London, SE5 9NU, UK; Department of Cardiology, King's College Hospital, Denmark Hill, Brixton, London, SE5 9RS, UK.
| |
Collapse
|
2
|
Blackmon TJ, MacMahon JA, Bernardino PN, Hogans RE, Cheng MY, Vu J, Lee RD, Saito NH, Grodzki AC, Bruun DA, Wulff H, Woolard KD, Brooks-Kayal A, Harvey DJ, Gorin FA, Lein PJ. Spatiotemporal perturbations of the plasminogen activation system in a rat model of acute organophosphate intoxication. Acta Neuropathol Commun 2025; 13:62. [PMID: 40102979 PMCID: PMC11917081 DOI: 10.1186/s40478-025-01979-0] [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: 10/30/2024] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
Neuroinflammation is widely posited to be a key pathogenic mechanism linking acute organophosphate (OP)-induced status epilepticus (SE) to persistent brain injury and abnormal electrical activity that contribute to epilepsy and cognitive impairment. The plasminogen activation system (PAS) promotes neuroinflammation in diverse neurological diseases but whether it is activated following acute OP intoxication has yet to be evaluated. To address this data gap, we characterized the spatiotemporal expression patterns of multiple components of the PAS in a rat model of acute intoxication with the OP, diisopropylfluorophosphate (DFP). Adult male Sprague Dawley rats administered DFP (4 mg/kg, sc), atropine sulfate (2 mg/kg, im) and 2-pralidoxime (25 mg/kg, im) went into SE that persisted for hours. One day after acute DFP-induced SE, plasmin activity and protein concentrations of plasminogen activator inhibitor-1 (PAI-1) in the plasma were increased, though not significantly. In contrast, acute DFP intoxication significantly increased brain levels of PAI-1, tissue-type plasminogen activator (tPA), urokinase plasminogen activator (uPA), and transcripts of TGF-β in a time- and region-dependent manner. In the cortex and hippocampus, quantification of PAI-1, tPA, and uPA by ELISA indicated significantly increased levels at 1 day post-exposure (DPE). PAI-1 and uPA returned to control values by 7 DPE while tPA protein remained elevated at 28 DPE. Immunohistochemistry detected elevated PAI-1 expression in the DFP brain up to 28 DPE. Co-localization of PAI-1 with biomarkers of neurons, microglia, and astrocytes demonstrated that PAI-1 localized predominantly to a subpopulation of astrocytes. Cytologically, PAI-1 localized to astrocytic end feet, but not adjacent neurovascular endothelium. Electron microscopy revealed neuronal metabolic stress and neurodegeneration with disruption of adjacent neurovascular units in the hippocampus post-DFP exposure. These data indicate that acute DFP intoxication altered PAS expression in the brain, with aberrant PAI-1 expression in a subset of reactive astrocyte populations.
Collapse
Affiliation(s)
- Thomas J Blackmon
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Jeremy A MacMahon
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Pedro N Bernardino
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Ryan E Hogans
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Mei-Yun Cheng
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Joan Vu
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Ruth Diana Lee
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Naomi H Saito
- Department of Public Health Sciences, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Ana Cristina Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Donald A Bruun
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Kevin D Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Amy Brooks-Kayal
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Danielle J Harvey
- Department of Public Health Sciences, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Fredric A Gorin
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
- Department of Neurology, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
- Molecular Biosciences, UC Davis School of Veterinary Medicine, 1089 Veterinary Research Drive, Davis, CA, 95616, USA.
| |
Collapse
|
3
|
Zhang X, Jiang M, Zhang X, Zuo Y, Zhang H, Zhang T, Yang L, Lin J, Zhang Y, Dai X, Ge W, Sun C, Yang F, Zhang J, Liu Y, Wang Y, Qiang H, Yang X, Sun D. Adipsin improves diabetic hindlimb ischemia through SERPINE1 dependent angiogenesis. Cardiovasc Diabetol 2024; 23:429. [PMID: 39623437 PMCID: PMC11613494 DOI: 10.1186/s12933-024-02526-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Adipsin (complement factor D, CFD), as the first described adipokine, is well-known for its regulatory effects in diabetic cardiovascular complications. However, its role in diabetic hind-limb ischemia was not clarified. This study aimed to evaluate the possible therapeutic effect of Adipsin in hind-limb ischemia in type 2 diabetic mice and to elucidate the molecular mechanisms involved. METHODS A high-fat diet and streptozotocin (HFD/STZ)-induced diabetic mouse model, and a transgenic mouse model with adipose tissue-specific overexpression of Adipsin (Adipsin-Tg) were employed. Hindlimb ischemia was established by femoral artery ligation, and blood flow recovery was monitored using Laser Doppler perfusion imaging. Molecular mechanisms underlying Adipsin-potentiated angiogenesis were examined using RNA sequencing and co-immunoprecipitation/mass spectrometry (Co-IP/MS) analyses. RESULTS Adipsin expression was upregulated in non-diabetic mice following HLI, while suppressed in diabetic mice, indicating its potential role in ischemic recovery which is impaired in diabetes. Adipsin-Tg mice exhibited significantly improved blood flow recovery, increased capillary density, and enhanced muscle regeneration in comparison with non-transgenic (NTg) diabetic mice. Adipsin facilitated proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) under hyperglycemic and hypoxic conditions. Additionally, it enhanced phosphorylation of AKT, ERK, and eNOS pathways both in vivo and in vitro. RNA sequencing and co-immunoprecipitation/mass spectrometry (Co-IP/MS) analyses identified that Adipsin promoted angiogenesis by interacting with SERBP1, which disrupted the binding of SERBP1 to SERPINE1 mRNA, resulting in reduced SERPINE1 expression and the subsequent activation of the VEGFR2 signaling cascade. CONCLUSIONS Adipsin promotes angiogenesis and facilitates blood perfusion recovery in diabetic mice with HLI by downregulating SERPINE1 through interaction with SERBP1. These findings elucidate a novel therapeutic potential for Adipsin in the management of PAD in diabetic patients, highlighting its role in enhancing angiogenesis and tissue repair.
Collapse
MESH Headings
- Animals
- Ischemia/physiopathology
- Ischemia/metabolism
- Ischemia/genetics
- Hindlimb
- Neovascularization, Physiologic
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/complications
- Humans
- Male
- Mice, Inbred C57BL
- Regional Blood Flow
- Human Umbilical Vein Endothelial Cells/metabolism
- Complement Factor D/metabolism
- Complement Factor D/genetics
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Signal Transduction
- Mice, Transgenic
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/genetics
- Diabetic Angiopathies/etiology
- Cell Proliferation
- Recovery of Function
- Mice
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Cell Movement
- Angiogenesis Inducing Agents/pharmacology
- Cells, Cultured
- Microvascular Density
- Angiogenesis
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Mengyuan Jiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xuebin Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yixuan Zuo
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Huanle Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Tingting Zhang
- Xijing 986 Hospital Department, Air Force Medical University, Xi'an, China
| | - Liyu Yang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jie Lin
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yan Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xinchun Dai
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wen Ge
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chuang Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Fang Yang
- Basic Medical Teaching Experiment Center, Basic Medical College, Air Force Medical University, Xi'an, China
| | - Jiye Zhang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yue Liu
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yangyang Wang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Huanhuan Qiang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiaojie Yang
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
4
|
Kriegshäuser G, Hayrapetyan H, Oberkanins C, Sarkisian T. Plasminogen activator inhibitor-1 genotype 4G/5G associates with skin involvement in Armenian familial Mediterranean fever patients. Rheumatol Int 2024; 44:2555-2559. [PMID: 38976029 DOI: 10.1007/s00296-024-05653-x] [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: 03/29/2024] [Accepted: 06/18/2024] [Indexed: 07/09/2024]
Abstract
There is little and conflicting data on the role of the plasminogen activator inhibitor-1 (PAI-1, SERPINE1) 4G/5G polymorphism in familial Mediterranean fever (FMF). Therefore this study aimed at evaluating the impact of this polymorphism on the disease course in a cohort of 303 Armenian FMF patients. Genotyping for 12 Mediterranean fever (MEFV) gene mutations and the PAI-1 4G/5G (rs1799762) polymorphism were performed by PCR/reverse-hybridization (StripAssay) and real-time PCR, respectively. PAI-1 genotypes 4G/4G, 4G/5G, and 5G/5G could be identified in 4 (5.88%), 30 (18.63%) and 9 (12.16%) patients with erysipelas-like erythema (ELE), while this was the case for 64 (94.12%), 131 (81.37%), and 65 (87.84%) patients without ELE, respectively (P < 0.033). We have identified a significant relationship between the PAI-1 4G/5G genotype and the occurence of ELE in a relatively large cohort of Armenian FMF patients. Because of conflicting results concerning the impact of this polymorphism on the clinical course of FMF in different populations, further studies are desirable to substantiate the findings reported here.
Collapse
Affiliation(s)
- Gernot Kriegshäuser
- Department of Medical Genetics, Yerevan State Medical University, Koryun 2, Yerevan, 0025, Armenia.
| | - Hasmik Hayrapetyan
- Department of Medical Genetics, Yerevan State Medical University, Koryun 2, Yerevan, 0025, Armenia
- Center of Medical Genetics and Primary Health Care, Yerevan, Armenia
| | | | - Tamara Sarkisian
- Department of Medical Genetics, Yerevan State Medical University, Koryun 2, Yerevan, 0025, Armenia
- Center of Medical Genetics and Primary Health Care, Yerevan, Armenia
| |
Collapse
|
5
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [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: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
6
|
Aji N, Wang L, Wang S, Pan T, Song J, Chen C, Wang L, Feng N, Tang X, Song Y. PAI-1 Deficiency Promotes NET-mediated Pyroptosis and Ferroptosis during Pseudomonas Aeruginosa-induced Acute Lung Injury by Regulating the PI3K/MAPK/AKT Axis. Inflammation 2024:10.1007/s10753-024-02102-6. [PMID: 39060815 DOI: 10.1007/s10753-024-02102-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Circulating neutrophil extracellular trap (NET) formation is an adaptive process during acute lung injury (ALI). The important role of plasminogen activator inhibitor (PAI)-1 in NET formation during ALI remains unclear. This research intends to examine the impacts of the decrease in PAI-1 levels on NET formation and the underlying mechanism. We found a relative association between the increase in plasma NET levels and thromboinflammation-induced lung damage in patients with ARDS. PAI-1 knockout (KO) mice exhibited significant increases in Pseudomonas aeruginosa (PAO1 strain)-induced ALI, inflammation, inflammatory cell accumulation, and proinflammatory cytokine secretion, and wild-type mice exhibited the opposite changes. During PAO1-induced ALI, PAI-1 KO increased NET release and the levels of prothrombotic markers in mice. PAI-1 deficiency also promoted NET formation and NET-mediated pyroptosis and ferroptosis by activating the PI3K/MAPK/AKT pathway in a PAO1-induced ALI mouse model. In conclusion, PAI-1 KO exacerbated PAO1-induced pneumonia-associated injury and contributed to NET-mediated pyroptosis and ferroptosis through PI3K/MAPK/AKT pathway activation. Thus, targeting PAI-1 and NETs may be a promising therapeutic approach for ameliorating pneumonia and thromboinflammation-associated ALI.
Collapse
Affiliation(s)
- Nurbiya Aji
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Linlin Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Sijiao Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ting Pan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Juan Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 200032, China
- Shanghai Respiratory Research Institute, Shanghai, 200032, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China
- Department of Pulmonary Medicine, Jinshan Hospital of Fudan University, Shanghai, 201508, China
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, China
| | - Cuicui Chen
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | | | - Nana Feng
- Department of Respiratory and Critical Medicine, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, China.
| | - Xinjun Tang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yuanlin Song
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary and Critical Medicine Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, 200032, China.
- Shanghai Respiratory Research Institute, Shanghai, 200032, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Department of Pulmonary Medicine, Jinshan Hospital of Fudan University, Shanghai, 201508, China.
| |
Collapse
|
7
|
Quinn M, Zhang RYK, Bello I, Rye KA, Thomas SR. Myeloperoxidase as a Promising Therapeutic Target after Myocardial Infarction. Antioxidants (Basel) 2024; 13:788. [PMID: 39061857 PMCID: PMC11274265 DOI: 10.3390/antiox13070788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) remain leading causes of death and disability worldwide. CAD begins with the formation of atherosclerotic plaques within the intimal layer of the coronary arteries, a process driven by persistent arterial inflammation and oxidation. Myeloperoxidase (MPO), a mammalian haem peroxidase enzyme primarily expressed within neutrophils and monocytes, has been increasingly recognised as a key pro-inflammatory and oxidative enzyme promoting the development of vulnerable coronary atherosclerotic plaques that are prone to rupture, and can precipitate a MI. Mounting evidence also implicates a pathogenic role for MPO in the inflammatory process that follows a MI, which is characterised by the rapid infiltration of activated neutrophils into the damaged myocardium and the release of MPO. Excessive and persistent cardiac inflammation impairs normal cardiac healing post-MI, resulting in adverse cardiac outcomes and poorer long-term cardiac function, and eventually heart failure. This review summarises the evidence for MPO as a significant oxidative enzyme contributing to the inappropriate inflammatory responses driving the progression of CAD and poor cardiac healing after a MI. It also details the proposed mechanisms underlying MPO's pathogenic actions and explores MPO as a novel therapeutic target for the treatment of unstable CAD and cardiac damage post-MI.
Collapse
Affiliation(s)
| | | | | | | | - Shane R. Thomas
- Cardiometabolic Disease Research Group, School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
8
|
Tang Y, Yuan Z, Lu X, Song Y, Zhu S, Qiu C, zhang Q, Fu B, Jia C, Li H. RAMP1 Protects Hepatocytes against Ischemia-reperfusion Injury by Inhibiting the ERK/YAP Pathway. J Clin Transl Hepatol 2024; 12:357-370. [PMID: 38638379 PMCID: PMC11022058 DOI: 10.14218/jcth.2023.00339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/03/2024] [Accepted: 02/02/2024] [Indexed: 04/20/2024] Open
Abstract
Background and Aims Hepatic ischemia-reperfusion injury (HIRI) is a prevalent complication of liver transplantation, partial hepatectomy, and severe infection, necessitating the development of more effective clinical strategies. Receptor activity-modifying protein 1 (RAMP1), a member of the G protein-coupled receptor adapter family, has been implicated in numerous physiological and pathological processes. The study aimed to investigate the pathogenesis of RAMP1 in HIRI. Methods We established a 70% liver ischemia-reperfusion model in RAMP1 knockout (KO) and wild-type mice. Liver and blood samples were collected after 0, 6, and 24 h of hypoxia/reperfusion. Liver histological and serological analyses were performed to evaluate liver damage. We also conducted in-vitro and in-vivo experiments to explore the molecular mechanism underlying RAMP1 function. Results Liver injury was exacerbated in RAMP1-KO mice compared with the sham group, as evidenced by increased cell death and elevated serum transaminase and inflammation levels. HIRI was promoted in RAMP1-KO mice via the induction of hepatocyte apoptosis and inhibition of proliferation. The absence of RAMP1 led to increased activation of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway and yes-associated protein (YAP) phosphorylation, ultimately promoting apoptosis. SCH772984, an ERK/MAPK phosphorylation inhibitor, and PY-60, a YAP phosphorylation inhibitor, reduced apoptosis in in-vitro and in-vivo experiments. Conclusions Our findings suggest that RAMP1 protects against HIRI by inhibiting ERK and YAP phosphorylation signal transduction, highlighting its potential as a therapeutic target for HIRI and providing a new avenue for intervention.
Collapse
Affiliation(s)
- Yongsheng Tang
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zenan Yuan
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xu Lu
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yingqiu Song
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuguang Zhu
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chunhui Qiu
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qi zhang
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Binsheng Fu
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Changchang Jia
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hua Li
- Department of Hepatic Surgery, Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Jiang S, Wang Y, Chen L, Mu H, Meaney C, Fan Y, Pillay J, Wang H, Zhang J, Pan S, Gao C. PAI-1 genetic polymorphisms influence septic patients' outcomes by regulating neutrophil activity. Chin Med J (Engl) 2023; 136:1959-1966. [PMID: 37439338 PMCID: PMC10431590 DOI: 10.1097/cm9.0000000000002316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1) plays an important role in the pathophysiology of sepsis, but the exact mechanism remains debatable. In this study, we investigated the associations among the serum levels of PAI-1, the incidence of 4G/5G promoter PAI-1 gene polymorphisms, immunological indicators, and clinical outcomes in septic patients. METHODS A total of 181 patients aged 18-80 years with sepsis between November 2016 and August 2018 in the intensive care unit in the Xinhua Hospital were recruited in this retrospective study, with 28-day mortality as the primary outcome. The initial serum level of PAI-1 and the presence of rs1799768 single nucleotide polymorphisms (SNPs) were examined. Univariate logistic regression and multivariate analyses were performed to determine the factors associated with different genotypes of PAI-1, serum level of PAI-1, and 28-day mortality. RESULTS The logistic analysis suggested that a high serum level of PAI-1 was associated with the rs1799768 SNP of PAI-1 (4G/4G and 4G/5G) (Odds ratio [OR]: 2.49; 95% confidence interval [CI]: 1.09, 5.68). Furthermore, a high serum level of PAI-1 strongly influenced 28-day mortality (OR 3.36; 95% CI 1.51, 7.49). The expression and activation of neutrophils (OR 0.96; 95% CI 0.93, 0.99), as well as the changes in the expression patterns of cytokines and chemokine-associated neutrophils (OR: 1.00; 95% CI: 1.00, 1.00), were both regulated by the genotype of PAI-1. CONCLUSIONS Genetic polymorphisms of PAI-1 can influence the serum levels of PAI-1, which might contribute to mortality by affecting neutrophil activity. Thus, patients with severe sepsis might clinically benefit from enhanced neutrophil clearance and the resolution of inflammation via the regulation of PAI-1 expression and activity.
Collapse
Affiliation(s)
- Shaowei Jiang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yang Wang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Liang Chen
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Honghua Mu
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Connor Meaney
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Yiwen Fan
- Department of Pathology Medicine Biology, University Medical Center Groningen, The Netherlands
| | - Janesh Pillay
- Department of Critical Care, University Medical Center Groningen, The Netherlands
| | - Hairong Wang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jincheng Zhang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
10
|
Jin Z, Dou M, Peng W, Xiao B, Liu J, Meng W, Liu W. Identification of distinct immune infiltration and potential biomarkers in patients with liver ischemia-reperfusion injury. Life Sci 2023:121726. [PMID: 37105441 DOI: 10.1016/j.lfs.2023.121726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
AIMS To identify alterations of specific gene expression, immune infiltration components, and potential biomarkers in liver ischemia-reperfusion injury (IRI) following liver transplantation (LT). MATERIALS AND METHODS GSE23649 and GSE151648 datasets were obtained from the Gene Expression Omnibus (GEO) database. To determine the differentially expressed genes (DEGs), we utilized the R package "limma". We also identify the infiltration of different immune cells through single-sample gene-set enrichment analysis (ssGSEA). Furthermore, we utilized LASSO logistic regression to select feature genes and Spearman's rank correlation analysis to determine the correlation between these genes and infiltrating immune cells. Finally, the significance of these feature genes was confirmed using a mouse model of hepatic IRI. KEY FINDINGS A total of 17 DEGs were acquired, most of which were associated with inflammation, apoptosis, cell proliferation, immune disorders, stress response, and angiogenesis. 28 immune cell types were determined using ssGSEA. 5 feature genes (ADM, KLF6, SERPINE1, SLC20A1, and HBB) were screened using LASSO analysis, but the HBB gene was ultimately excluded due to the lack of statistical significance in the GSE151648 dataset. These 4 feature genes were predominantly related to immune cells. Finally, 15 significantly distinctive types of immune cells between the control and IRI groups were verified. SIGNIFICANCE We unveiled that macrophages, dendritic cells (DCs), neutrophils, CD4 T cells, and other immune cells infiltrated the IRI that occurred after LT. Moreover, we identified ADM, KLF6, SERPINE1, and SLC20A1 as potential biological biomarkers underlying IRI post-transplant, which may improve the diagnosis and prognosis of this condition.
Collapse
Affiliation(s)
- Zhangliu Jin
- Department of General Surgery, Division of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Meng Dou
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shangxi 710000, China
| | - Weihui Peng
- Department of General Surgery, Division of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Boen Xiao
- Department of General Surgery, Division of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Jinjin Liu
- Department of General Surgery, Division of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Wen Meng
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Cardiometabolic Medicine of Hunan Province, Metabolic Syndrome Research Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Wei Liu
- Department of General Surgery, Division of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
11
|
Li Y, Miao L, Guo R, He L, Sun M, Pan Y, Lin L, Ren J, Liu J. To explore the regulatory effect of Buyang Huanwu Decoction on cerebral infarction based on quantitative proteomics. J Proteomics 2023; 277:104850. [PMID: 36813112 DOI: 10.1016/j.jprot.2023.104850] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023]
Abstract
Buyang Huanwu Decoction (BYHW) contains chemical components such as ligustrazine, oxypaeoniflora, chlorogenic acid, and others. To explore the neuroprotective effect and potential target protein of BYHW in cerebral infarction (CI). A double-blind, randomized controlled trial was established and patients with CI were divided into the BYHW group (n = 35) and the control group (n = 30). To evaluate the efficacy by TCM syndrome score and clinical indicators, and to explore the changes of serum proteins by proteomics technology, so as to explore the mechanism of BYHW and potential target proteins. The study found that compared with the control group, the TCM syndrome score, including Deficiency of Vital Energy (DVE), Blood Stasis (BS), and NIHSS in the BYHW group decreased significantly (p < 0.05), and the Barthel Index (BI) score was significantly higher. A total of 99 differential regulatory proteins were identified by proteomics, which act on lipids and atherosclerosis, complement and coagulation cascade, and TNF-α signaling pathway. In addition, Elisa verified the results of proteomics and found that BYHW can reduce the neurological impairments focus on IL-1β, IL-6, TNF-α, MCP-1, MMP-9, and PAI-1. Significance: In this study, quantitative proteomics was used in combination with liquid chromatography-mass spectrometry (LC-MS/MS) to study the therapeutic effect of BYHW on cerebral infarction (CI) and potential changes in serum proteomics. In addition, the public proteomics database was used for bioinformatics analysis, and Elisa experiment verified the results of proteomics, further clarifying the potential protection mechanism of BYHW on CI.
Collapse
Affiliation(s)
- Ying Li
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, National clinical research center of cardiovascular disease of traditional Chinese Medicine, Beijing, 100000, China
| | - Lan Miao
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, National clinical research center of cardiovascular disease of traditional Chinese Medicine, Beijing, 100000, China
| | - Rongjuan Guo
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Lijuan He
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Mingqian Sun
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, National clinical research center of cardiovascular disease of traditional Chinese Medicine, Beijing, 100000, China
| | - Yinghong Pan
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Li Lin
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, National clinical research center of cardiovascular disease of traditional Chinese Medicine, Beijing, 100000, China.
| | - Junguo Ren
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, National clinical research center of cardiovascular disease of traditional Chinese Medicine, Beijing, 100000, China.
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, National clinical research center of cardiovascular disease of traditional Chinese Medicine, Beijing, 100000, China.
| |
Collapse
|
12
|
Uhl B, Haring F, Slotta-Huspenina J, Luft J, Schneewind V, Hildinger J, Wu Z, Steiger K, Smiljanov B, Batcha AMN, Keppler OT, Hellmuth JC, Lahmer T, Stock K, Weiss BG, Canis M, Stark K, Bromberger T, Moser M, Schulz C, Weichert W, Zuchtriegel G, Reichel CA. Vitronectin promotes immunothrombotic dysregulation in the venular microvasculature. Front Immunol 2023; 14:1078005. [PMID: 36845099 PMCID: PMC9945350 DOI: 10.3389/fimmu.2023.1078005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/05/2023] [Indexed: 02/10/2023] Open
Abstract
Microvascular immunothrombotic dysregulation is a critical process in the pathogenesis of severe systemic inflammatory diseases. The mechanisms controlling immunothrombosis in inflamed microvessels, however, remain poorly understood. Here, we report that under systemic inflammatory conditions the matricellular glycoproteinvitronectin (VN) establishes an intravascular scaffold, supporting interactions of aggregating platelets with immune cells and the venular endothelium. Blockade of the VN receptor glycoprotein (GP)IIb/IIIa interfered with this multicellular interplay and effectively prevented microvascular clot formation. In line with these experimental data, particularly VN was found to be enriched in the pulmonary microvasculature of patients with non-infectious (pancreatitis-associated) or infectious (coronavirus disease 2019 (COVID-19)-associated) severe systemic inflammatory responses. Targeting the VN-GPIIb/IIIa axis hence appears as a promising, already feasible strategy to counteract microvascular immunothrombotic dysregulation in systemic inflammatory pathologies.
Collapse
Affiliation(s)
- Bernd Uhl
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany,*Correspondence: Bernd Uhl,
| | - Florian Haring
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | | | - Joshua Luft
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Vera Schneewind
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Jonas Hildinger
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Zhengquan Wu
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Katja Steiger
- Department of Pathology, Technical University of Munich, Munich, Germany
| | - Bojan Smiljanov
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Aarif M. N. Batcha
- Institute of Medical Data Processing, Biometrics, and Epidemiology (IBE), University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany,Data Integration for Future Medicine (DiFuture), University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Oliver T. Keppler
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany,German Centre for Infection Research (DZIF), Partner Site München, Munich, Germany
| | - Johannes C. Hellmuth
- Department of Medicine III, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Munich, Germany,COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Tobias Lahmer
- Department of Internal Medicine II, Technical University of Munich, Munich, Germany
| | - Konrad Stock
- Department of Nephrology, Technical University of Munich, Munich, Germany
| | - Bernhard G. Weiss
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Konstantin Stark
- Department of Cardiology, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Thomas Bromberger
- Institute of Experimental Hematology, Technical University of Munich, Munich, Germany
| | - Markus Moser
- Institute of Experimental Hematology, Technical University of Munich, Munich, Germany
| | - Christian Schulz
- Department of Cardiology, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Wilko Weichert
- Department of Pathology, Technical University of Munich, Munich, Germany
| | - Gabriele Zuchtriegel
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| | - Christoph A. Reichel
- Department of Otorhinolaryngology, University Hospital, Ludwig-Maximilians-Universität München (LMU), Munich, Germany,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians-Universität München (LMU) Munich, Munich, Germany
| |
Collapse
|
13
|
Fields AT, Andraska EA, Kaltenmeier C, Matthay ZA, Herrera K, Nuñez-Garcia B, Jones CM, Wick KD, Liu S, Luo JH, Yu YP, Matthay MA, Hendrickson CM, Bainton RJ, Barrett TJ, Berger JS, Neal MD, Kornblith LZ. Effects of the circulating environment of COVID-19 on platelet and neutrophil behavior. Front Immunol 2023; 14:1130288. [PMID: 36999030 PMCID: PMC10043426 DOI: 10.3389/fimmu.2023.1130288] [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: 12/23/2022] [Accepted: 02/23/2023] [Indexed: 04/01/2023] Open
Abstract
Introduction Thromboinflammatory complications are well described sequalae of Coronavirus Disease 2019 (COVID-19), and there is evidence of both hyperreactive platelet and inflammatory neutrophil biology that contributes to the thromoinflammatory milieu. It has been demonstrated in other thromboinflammatory diseases that the circulating environment may affect cellular behavior, but what role this environment exerts on platelets and neutrophils in COVID-19 remains unknown. We tested the hypotheses that 1) plasma from COVID-19 patients can induce a prothrombotic platelet functional phenotype, and 2) contents released from platelets (platelet releasate) from COVID-19 patients can induce a proinflammatory neutrophil phenotype. Methods We treated platelets with COVID-19 patient and disease control plasma, and measured their aggregation response to collagen and adhesion in a microfluidic parallel plate flow chamber coated with collagen and thromboplastin. We exposed healthy neutrophils to platelet releasate from COVID-19 patients and disease controls and measured neutrophil extracellular trap formation and performed RNA sequencing. Results We found that COVID-19 patient plasma promoted auto-aggregation, thereby reducing response to further stimulation ex-vivo. Neither disease condition increased the number of platelets adhered to a collagen and thromboplastin coated parallel plate flow chamber, but both markedly reduced platelet size. COVID-19 patient platelet releasate increased myeloperoxidasedeoxyribonucleic acid complexes and induced changes to neutrophil gene expression. Discussion Together these results suggest aspects of the soluble environment circulating platelets, and that the contents released from those neutrophil behavior independent of direct cellular contact.
Collapse
Affiliation(s)
- Alexander T. Fields
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Elizabeth A. Andraska
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Christof Kaltenmeier
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Zachary A. Matthay
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Kimberly Herrera
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Brenda Nuñez-Garcia
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Chayse M. Jones
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Katherine D. Wick
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Silvia Liu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jian-Hua Luo
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yan-Ping Yu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael A. Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, United States
| | - Carolyn M. Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Roland J. Bainton
- Department of Anesthesia and Perioperative Care, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Tessa J. Barrett
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Jeffrey S. Berger
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University (NYU) Grossman School of Medicine, New York, NY, United States
- New York University (NYU) Center for the Prevention of Cardiovascular Disease, New York University (NYU) Langone Health, New York, NY, United States
- Division of Vascular Surgery, Department of Surgery, New York University (NYU) Grossman School of Medicine, New York, NY, United States
| | - Matthew D. Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lucy Z. Kornblith
- Department of Surgery, University of California, San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
- *Correspondence: Lucy Z. Kornblith,
| |
Collapse
|
14
|
Neutrophils: As a Key Bridge between Inflammation and Thrombosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1151910. [PMID: 36408343 PMCID: PMC9668459 DOI: 10.1155/2022/1151910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/14/2022] [Accepted: 09/29/2022] [Indexed: 11/10/2022]
Abstract
Immunothrombosis is a mechanism of defense of the organism against pathogenic microorganisms that increases their recognition, limitation, and clearance and is part of the innate immune defense. Physiological immunothrombosis is beneficial to the body against the invasion of pathogenic microorganisms, but when immunothrombosis is out of control, it is easy to cause thrombotic diseases, thus, causing unpredictable consequences to the body. Neutrophils play a pivotal role in this process. Understanding the mechanism of neutrophils in immune thrombosis and out-of-control is particularly important for the treatment of related thrombotic diseases. In this review, we studied the role of neutrophils in immune thrombosis and each link out of control (including endothelial cell dysfunction; activation of platelets; activation of coagulation factor; inhibition of the anticoagulation system; and inhibition of the fibrinolysis system).
Collapse
|
15
|
Falcinelli E, Petito E, Gresele P. The role of platelets, neutrophils and endothelium in COVID-19 infection. Expert Rev Hematol 2022; 15:727-745. [PMID: 35930267 DOI: 10.1080/17474086.2022.2110061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION COVID-19 is associated to an increased risk of thrombosis, as a result of a complex process that involves the activation of vascular and circulating cells, the release of soluble inflammatory and thrombotic mediators and blood clotting activation. AREAS COVERED This article reviews the pathophysiological role of platelets, neutrophils and the endothelium, and of their interactions, in the thrombotic complications of COVID-19 patients, and the current and future therapeutic approaches targeting these cell types. EXPERT OPINION Virus-induced platelet, neutrophil and endothelial cell changes are crucial triggers of the thrombotic complications and of the adverse evolution of COVID-19. Both the direct interaction with the virus and the associated cytokine storm concur to trigger cell activation in a classical thromboinflammatory vicious circle. Although heparin has proven to be an effective prophylactic and therapeutic weapon for the prevention and treatment of COVID-19-associated thrombosis, it acts downstream of the cascade of events triggered by SARS-CoV-2. The identification of specific molecular targets interrupting the thromboinflammatory cascade upstream, and more specifically acting either on the interaction of SARS-CoV-2 with blood and vascular cells or on the specific signalling mechanisms associated with their COVID-19-associated activation, might theoretically offer greater protection with potentially lesser side effects.
Collapse
Affiliation(s)
- E Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - E Petito
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - P Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
16
|
Colic J, Pruner I, Damjanov N, Pekmezovic T, Sefik-Bukilica M, Antovic A. Impaired Fibrinolysis Is Linked With Digital Vasculopathy and Onset of New Digital Ulcers in Systemic Sclerosis. J Rheumatol 2022; 49:598-606. [PMID: 35169064 DOI: 10.3899/jrheum.210931] [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] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess thrombin generation, fibrin formation, and structure together with the fibrinolytic status in patients with systemic sclerosis (SSc) in relation to the occurrence of digital ulcers (DUs) during the course of disease. METHODS We studied variables of endothelial dysfunction, thrombin generation, overall hemostatic potential, and fibrin clot turbidity in plasma from 58 patients with SSc (39 with DU history and 19 DU-naïve) and 46 matched healthy controls (HCs). Fibrin structure was visualized using scanning electron microscopy (SEM). Finally, 39 patients with a history of DUs were followed for 1.5 years and the predictive value of all investigated markers for new DU onset was explored. RESULTS Significantly enhanced endogenous thrombin potential (ETP) and prolonged clot lysis time (CLT) were found in patients with DUs compared to HCs. CLT was prolonged in patients with DUs compared to those without, showing good validity in identifying DUs with an area under the curve of 0.7 (95% CI 0.6-0.8). The levels of ETP and intercellular adhesion molecule 1 were independently associated with CLT. Over the follow-up period, 20 patients developed new DUs. CLT was prolonged (P < 0.001) in patients with new DU episodes, especially those with recurrent DUs. Regression analysis showed that the Raynaud phenomenon visual analog scale and CLT were predictors of new DUs (OR 1.1, 95% CI 1.0-1.1 and OR 1.2, 95% CI 1.1-1.3, respectively). SEM confirmed denser fibrin clots in patients with new DUs. CONCLUSION Our results suggest that impaired fibrinolysis might have an emerging role in underlying digital vasculopathy and its progression in SSc.
Collapse
Affiliation(s)
- Jelena Colic
- J. Colic, MD, PhDc, Institute of Rheumatology, Belgrade, Serbia;
| | - Iva Pruner
- I. Pruner, PhD, Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden
| | - Nemanja Damjanov
- N. Damjanov, MD, PhD, M. Sefik-Bukilica, MD, PhD, Institute of Rheumatology, Belgrade, Serbia School of Medicine University of Belgrade, Belgrade, Serbia
| | - Tatjana Pekmezovic
- T. Pekmezovic, MD, PhD, Institute of Epidemiology, School of Medicine University of Belgrade, Belgrade, Serbia
| | - Mirjana Sefik-Bukilica
- N. Damjanov, MD, PhD, M. Sefik-Bukilica, MD, PhD, Institute of Rheumatology, Belgrade, Serbia School of Medicine University of Belgrade, Belgrade, Serbia
| | - Aleksandra Antovic
- A. Antovic, MD, PhD, Karolinska Institutet, Department of Medicine Division Solna of Rheumatology, Karolinska University Hospital, Rheumatology, Stockholm, Sweden
| |
Collapse
|
17
|
Badran M, Gozal D. PAI-1: A Major Player in the Vascular Dysfunction in Obstructive Sleep Apnea? Int J Mol Sci 2022; 23:5516. [PMID: 35628326 PMCID: PMC9141273 DOI: 10.3390/ijms23105516] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Obstructive sleep apnea is a chronic and prevalent condition that is associated with endothelial dysfunction, atherosclerosis, and imposes excess overall cardiovascular risk and mortality. Despite its high prevalence and the susceptibility of CVD patients to OSA-mediated stressors, OSA is still under-recognized and untreated in cardiovascular practice. Moreover, conventional OSA treatments have yielded either controversial or disappointing results in terms of protection against CVD, prompting the need for the identification of additional mechanisms and associated adjuvant therapies. Plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of tissue-type plasminogen activator (tPA) and urinary-type plasminogen activator (uPA), is a key regulator of fibrinolysis and cell migration. Indeed, elevated PAI-1 expression is associated with major cardiovascular adverse events that have been attributed to its antifibrinolytic activity. However, extensive evidence indicates that PAI-1 can induce endothelial dysfunction and atherosclerosis through complex interactions within the vasculature in an antifibrinolytic-independent matter. Elevated PAI-1 levels have been reported in OSA patients. However, the impact of PAI-1 on OSA-induced CVD has not been addressed to date. Here, we provide a comprehensive review on the mechanisms by which OSA and its most detrimental perturbation, intermittent hypoxia (IH), can enhance the transcription of PAI-1. We also propose causal pathways by which PAI-1 can promote atherosclerosis in OSA, thereby identifying PAI-1 as a potential therapeutic target in OSA-induced CVD.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
| | - David Gozal
- Department of Child Health and Child Health Research Institute, School of Medicine, University of Missouri, 400 N Keene St, Suite 010, Columbia, MO 65201, USA;
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
18
|
Nordeng J, Solheim S, Åkra S, Schandiz H, Hoffmann P, Roald B, Bendz B, Arnesen H, Helseth R, Seljeflot I. Gene expression of fibrinolytic markers in coronary thrombi. Thromb J 2022; 20:23. [PMID: 35488283 PMCID: PMC9052700 DOI: 10.1186/s12959-022-00383-1] [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: 02/23/2022] [Accepted: 04/17/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The fibrinolytic system plays an important role in coronary artery atherothrombosis, and especially circulating plasminogen-activator inhibitor (PAI) type 1 (PAI-1) associates with increased mortality, infarct size and heart failure in patients with myocardial infarction (MI). In a cross-sectional study, we aimed to study whether genes encoding tissue plasminogen activator (tPA), urinary-type plasminogen activator (uPA), PAI-1 and PAI-2 are expressed in coronary thrombi from acute ST-elevation MI (STEMI) patients. Any relations to myocardial injury measured by peak troponin T, time from symptom onset to Percutaneous Coronary Intervention (PCI), and to different cell types present in the thrombi were also explored. METHODS Intracoronary thrombi were aspirated from 33 STEMI patients treated with primary PCI. The thrombi were snap-frozen for gene expression analyses, relatively quantified by RT PCR. Peripheral blood samples were drawn. Correlations were performed by Spearmans rho. RESULTS The genes were present in 74-94% of the thrombi. Median peak troponin T was 3434 μ/L and median ischemic time 152 min. There were no significant correlations between the measured genes and troponin T, or ischemic time. Genes encoding tPA, u-PA, PAI-1 and PAI-2 all correlated significantly to the presence of monocytes/macrophages (CD68) in the thrombi (p = 0.028, p < 0.001, p = 0.003, p < 0.001). PAI-1 and PAI-2 also correlated to endothelial cells (CD31) (p = 0.002, p = 0.016). uPA associated with neutrophil granulocytes (CD 66b) (p = 0.019). CONCLUSION Genes encoding tPA, uPA, PAI-1 and PAI-2 were highly expressed in human coronary thrombi from STEMI patients, indicating fibrinolytic regulators playing active roles in the thrombi, although not related to myocardial injury. All markers related to the presence of monocytes/macrophages, indicating connection to local inflammatory cells. TRIAL REGISTRATION The study is registered at clinicaltrials.gov with identification number NCT02746822 .
Collapse
Affiliation(s)
- Jostein Nordeng
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Faculty of Medicine, University of Oslo, Klaus Torgårds vei 3, Pb 1078 Blindern, 0316 Oslo, Norway
| | - Svein Solheim
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
| | - Sissel Åkra
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
| | - Hossein Schandiz
- Department of Pathology, Akershus University Hospital, Sykehusveien 25, Pb1000, 1478 Lørenskog, Norway
| | - Pavel Hoffmann
- Section for Interventional Cardiology, Department of Cardiology, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
| | - Borghild Roald
- Faculty of Medicine, University of Oslo, Klaus Torgårds vei 3, Pb 1078 Blindern, 0316 Oslo, Norway
- Department of Pathology, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
| | - Bjørn Bendz
- Faculty of Medicine, University of Oslo, Klaus Torgårds vei 3, Pb 1078 Blindern, 0316 Oslo, Norway
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Sognsvannsveien 20, Pb 4950 Nydalen, N-0424 Oslo, Norway
| | - Harald Arnesen
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Faculty of Medicine, University of Oslo, Klaus Torgårds vei 3, Pb 1078 Blindern, 0316 Oslo, Norway
| | - Ragnhild Helseth
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Department of Cardiology, Oslo University Hospital Ullevål, Kirkeveien 166, Pb 4950 Nydalen, N-0424 Oslo, Norway
- Faculty of Medicine, University of Oslo, Klaus Torgårds vei 3, Pb 1078 Blindern, 0316 Oslo, Norway
| |
Collapse
|
19
|
Wang G, Zhang C, Kambara H, Dambrot C, Xie X, Zhao L, Xu R, Oneglia A, Liu F, Luo HR. Identification of the Transgene Integration Site and Host Genome Changes in MRP8-Cre/ires-EGFP Transgenic Mice by Targeted Locus Amplification. Front Immunol 2022; 13:875991. [PMID: 35464448 PMCID: PMC9020256 DOI: 10.3389/fimmu.2022.875991] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 01/01/2023] Open
Abstract
The MRP8-Cre-ires/EGFP transgenic mouse (Mrp8creTg, on C57BL/6J genetic background) is popular in immunological and hematological research for specifically expressing Cre recombinase and an EGFP reporter in neutrophils. It is often crossed with other transgenic lines carrying loxP-flanked genes to achieve restricted gene knockout in neutrophils. However, due to the way in which the line was created, basic knowledge about the MRP8-Cre-ires/EGFP transgene in the host genome, such as its integration site(s) and flanking sequences, remains largely unknown, hampering robust experimental design and data interpretation. Here we used a recently developed technique, targeted locus amplification (TLA) sequencing, to fill these knowledge gaps. We found that the MRP8-Cre-ires/EGFP transgene was integrated into chromosome 5 (5qG2) of the host mouse genome. This integration led to a 44 kb deletion of the host genomic sequence, resulting in complete deletion of Serpine1 and partial deletion of Ap1s1. Having determined the flanking sequences of the transgene, we designed a new genotyping protocol that can distinguish homozygous, heterozygous, and wildtype Mrp8creTg mice. To our surprise, crossing heterozygous mice produced no homozygous Mrp8creTg mice, most likely due to prenatal lethality resulting from disrupted Ap1s1 gene expression.
Collapse
Affiliation(s)
- Guan Wang
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, United States.,Department of Laboratory Medicine, Enders Research Building, Boston Children's Hospital, Boston, MA, United States
| | - Cunling Zhang
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hiroto Kambara
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, United States.,Department of Laboratory Medicine, Enders Research Building, Boston Children's Hospital, Boston, MA, United States
| | | | - Xuemei Xie
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, United States.,Department of Laboratory Medicine, Enders Research Building, Boston Children's Hospital, Boston, MA, United States
| | - Li Zhao
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, United States.,Department of Laboratory Medicine, Enders Research Building, Boston Children's Hospital, Boston, MA, United States
| | - Rong Xu
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, United States.,Department of Laboratory Medicine, Enders Research Building, Boston Children's Hospital, Boston, MA, United States
| | | | - Fei Liu
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Hongbo R Luo
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Harvard Medical School, Boston, MA, United States.,Department of Laboratory Medicine, Enders Research Building, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
20
|
LncRNA SERPINB9P1 expression and polymorphisms are associated with ischemic stroke in a Chinese Han population. Neurol Sci 2022; 43:1143-1154. [PMID: 34273050 DOI: 10.1007/s10072-021-05418-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/17/2021] [Indexed: 10/20/2022]
Abstract
Long noncoding RNAs (lncRNAs) were reported to play important roles in the pathogenesis of ischemic stroke (IS). Our study aimed to investigate the role of lncRNA SERPINB9P1 expression in ischemic stroke and the association between SERPINB9P1 polymorphisms and IS risk, as well as examine the correlation of SERPINB9P1 expression and variants with clinical parameters of IS. The SERPINB9P1 levels in human participants and oxygen-glucose deprivation (OGD)-treated human A172 cells were measured by qRT-PCR. The SERPINB9P1 polymorphisms (rs375556 and rs318429) were genotyped by the MassARRAY platform. We found that the SERPINB9P1 expression was significantly downregulated in patients with IS compared with that in healthy controls. On the 14th day in the hospital, the SERPINB9P1 level in patients with moderate and severe stroke was significantly downregulated compared with the normal group. After stratification by gender, the rs375556 polymorphism was significantly associated with susceptibility to female IS in the recessive model, and the significant association remained after adjusting for age. After adjusting for gender and age, rs318429 was significantly associated with FPG and D-D levels, and rs375556 was significantly associated with INR and PTA levels in IS cases. Besides, the lncRNA SERPINB9P1 expressed downregulated in OGD/reoxygenation-treated human A172 cells. In conclusion, the lncRNA SERPINB9P1 may protect against cerebral ischemia-reperfusion injury and neurological impairment after IS. The SERPINB9P1 rs375556 polymorphism was associated with susceptibility to female IS, and SERPINB9P1 polymorphisms may influence the metabolism of blood glucose and regulation of coagulation function in patients with IS.
Collapse
|
21
|
Zuchtriegel G, Uhl B, Pick R, Ramsauer M, Dominik J, Mittmann LA, Canis M, Kanse S, Sperandio M, Krombach F, Reichel CA. Vitronectin stabilizes intravascular adhesion of neutrophils by coordinating β2 integrin clustering. Haematologica 2021; 106:2641-2653. [PMID: 32703799 PMCID: PMC8485676 DOI: 10.3324/haematol.2019.226241] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Indexed: 11/09/2022] Open
Abstract
The recruitment of neutrophils from the microvasculature to the site of injury or infection represents a key event in the inflammatory response. Vitronectin (VN) is a multifunctional macromolecule abundantly present in blood and extracellular matrix. The role of this glycoprotein in the extravasation process of circulating neutrophils remains elusive. Employing advanced in vivo/ex vivo imaging techniques in different mouse models as well as in vitro methods, we uncovered a previously unrecognized function of VN in the transition of dynamic to static intravascular interactions of neutrophils with microvascular endothelial cells. These distinct properties of VN require the heteromerization of this glycoprotein with plasminogen activator inhibitor-1 (PAI- 1) on the activated venular endothelium and subsequent interactions of this protein complex with the scavenger receptor low-density lipoprotein receptor-related protein-1 on intravascularly adhering neutrophils. This induces p38 mitogen-activated protein kinases-dependent intracellular signaling events which, in turn, regulates the proper clustering of the b2 integrin lymphocyte function associated antigen-1 on the surface of these immune cells. As a consequence of this molecular interplay, neutrophils become able to stabilize their adhesion to the microvascular endothelium and, subsequently, to extravasate to the perivascular tissue. Hence, endothelial-bound VN-PAI-1 heteromers stabilize intravascular adhesion of neutrophils by coordinating b2 integrin clustering on the surface of these immune cells, thereby effectively controlling neutrophil trafficking to inflamed tissue. Targeting this protein complex might be beneficial for the prevention and treatment of inflammatory pathologies.
Collapse
Affiliation(s)
- Gabriele Zuchtriegel
- Walter Brendel Centre of Experimental Medicine and Klinikum der Universität München, Germany
| | - Bernd Uhl
- Walter Brendel Centre of Experimental Medicine and Klinikum der Universität München, Germany
| | - Robert Pick
- Dept. of Otorhinolaryngology, Klinikum der Universität Munchen, Munich, Germany
| | - Michaela Ramsauer
- Walter Brendel Centre of Experimental Medicine and Klinikum der Universität Munchen, Germany
| | - Julian Dominik
- Dept. of Otorhinolaryngology, Klinikum der Universität Munchen, Munich, Germany
| | - Laura A Mittmann
- Walter Brendel Centre of Experimental Medicine and Klinikum der Universität Munchen, Germany
| | | | - Sandip Kanse
- Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Markus Sperandio
- Dept. of Otorhinolaryngology, Klinikum der Universität Munchen, Munich, Germany
| | - Fritz Krombach
- Walter Brendel Centre of Experimental Medicine, Munich, Germany
| | - Christoph A Reichel
- Walter Brendel Centre of Experimental Medicine and Klinikum der Universität Munchen, Germany
| |
Collapse
|
22
|
Warner J, Hardesty J, Song Y, Sun R, Deng Z, Xu R, Yin X, Zhang X, McClain C, Warner D, Kirpich I. Fat-1 Transgenic Mice With Augmented n3-Polyunsaturated Fatty Acids Are Protected From Liver Injury Caused by Acute-On-Chronic Ethanol Administration. Front Pharmacol 2021; 12:711590. [PMID: 34531743 PMCID: PMC8438569 DOI: 10.3389/fphar.2021.711590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Alcohol-associated liver disease (ALD) is the leading cause of liver disease worldwide, and alcohol-associated hepatitis (AH), a severe form of ALD, is a major contributor to the mortality and morbidity due to ALD. Many factors modulate susceptibility to ALD development and progression, including nutritional factors such as dietary fatty acids. Recent work from our group and others showed that modulation of dietary or endogenous levels of n6-and n3-polyunsaturated fatty acids (PUFAs) can exacerbate or attenuate experimental ALD, respectively. In the current study, we interrogated the effects of endogenous n3-PUFA enrichment in a mouse model which recapitulates features of early human AH using transgenic fat-1 mice which endogenously convert n6-PUFAs to n3-PUFAs. Male wild type (WT) and fat-1 littermates were provided an ethanol (EtOH, 5% v/v)-containing liquid diet for 10 days, then administered a binge of EtOH (5 g/kg) by oral gavage on the 11th day, 9 h prior to sacrifice. In WT mice, EtOH treatment resulted in liver injury as determined by significantly elevated plasma ALT levels, whereas in fat-1 mice, EtOH caused no increase in this biomarker. Compared to their pair-fed controls, a significant EtOH-mediated increase in liver neutrophil infiltration was observed also in WT, but not fat-1 mice. The hepatic expression of several cytokines and chemokines, including Pai-1, was significantly lower in fat-1 vs WT EtOH-challenged mice. Cultured bone marrow-derived macrophages isolated from fat-1 mice expressed less Pai-1 and Cxcl2 (a canonical neutrophil chemoattractant) mRNA compared to WT when stimulated with lipopolysaccharide. Further, we observed decreased pro-inflammatory M1 liver tissue-resident macrophages (Kupffer cells, KCs), as well as increased liver T regulatory cells in fat-1 vs WT EtOH-fed mice. Taken together, our data demonstrated protective effects of endogenous n3-PUFA enrichment on liver injury caused by an acute-on-chronic EtOH exposure, a paradigm which recapitulates human AH, suggesting that n3-PUFAs may be a viable nutritional adjuvant therapy for this disease.
Collapse
Affiliation(s)
- Jeffrey Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Josiah Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Ying Song
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Rui Sun
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Zhongbin Deng
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Surgery, University of Louisville, Louisville, KY, United States.,University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Raobo Xu
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States.,Department of Chemistry, University of Louisville, Louisville, KY, United States.,Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, United States
| | - Xinmin Yin
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States.,Department of Chemistry, University of Louisville, Louisville, KY, United States.,Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, United States
| | - Xiang Zhang
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States.,Department of Chemistry, University of Louisville, Louisville, KY, United States.,Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, United States
| | - Craig McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States.,Robley Rex Veterans Affairs Medical Center, Louisville, KY, United States
| | - Dennis Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Irina Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, United States.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, United States.,University of Louisville Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
23
|
Zhang S, Cao Y, Du J, Liu H, Chen X, Li M, Xiang M, Wang C, Wu X, Liu L, Wang C, Wu Y, Li Z, Fang S, Shi J, Wang L. Neutrophil extracellular traps contribute to tissue plasminogen activator resistance in acute ischemic stroke. FASEB J 2021; 35:e21835. [PMID: 34449927 DOI: 10.1096/fj.202100471rr] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/08/2023]
Abstract
Circulating neutrophil extracellular traps (NETs) resistant to t-PA have not been studied completely although NETs in thrombi may contribute to tissue plasminogen activator (t-PA) resistance. This research intended to elucidate whether circulating NETs are associated with t-PA resistance and the underlying mechanism. The levels of NETs were detected in the circulating neutrophils, ischemic brain tissue of acute ischemic stroke (AIS) patients, and transient middle cerebral artery occlusion (tMCAO) models. NET formation in blood, thrombi, and ischemic brain tissue of mice were analyzed by immunofluorescence. Exposed phosphatidylserine (PS) was assessed using flow cytometry and confocal microscopy. Procoagulant activity (PCA) was evaluated using fibrin formation assays, thrombin, and purified coagulation complex. The plasma levels of NETs in AIS patients were significantly higher than those in healthy individuals. After thrombolysis, a significant increase was noted in NET markers in no-improvement patients, while the changes in improvement patients were not significant. Importantly, NETs were decorated with von Willebrand factor (vWF) and plasminogen activator inhibitor-1 (PAI-1) in the blood and thrombi, which could reverse the fibrinolytic effects. In addition, NETs activated platelets (PLTs) and endothelial cells (ECs), stimulating a procoagulant phenotype and facilitating vWF and PAI-1 release. DNase I, activated protein C (APC), and sivelestat markedly inhibited these effects. Furthermore, targeting NETs protected mice from tMCAO-induced cerebral ischemia, possibly by regulating vWF and PAI-1. In summary, NETs may contribute to t-PA resistance in AIS through activation of PLTs and ECs. Strategies against NETs may present a promising therapeutic approach to improve the thrombolysis efficiency of t-PA in AIS patients.
Collapse
Affiliation(s)
- Shuoqi Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Yuze Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingwen Du
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Huan Liu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaojing Chen
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mengdi Li
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mengqi Xiang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chengyue Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Langjiao Liu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chunli Wang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yinsong Wu
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhuxin Li
- Department of Acupuncture and Moxibustion, College of Acupuncture and Moxibustion, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shaohong Fang
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin, China
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China.,Department of Medicine, Brigham and Women's Hospital, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
24
|
Veluswamy P, Wacker M, Stavridis D, Reichel T, Schmidt H, Scherner M, Wippermann J, Michels G. The SARS-CoV-2/Receptor Axis in Heart and Blood Vessels: A Crisp Update on COVID-19 Disease with Cardiovascular Complications. Viruses 2021; 13:1346. [PMID: 34372552 PMCID: PMC8310117 DOI: 10.3390/v13071346] [Citation(s) in RCA: 9] [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: 06/02/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/08/2023] Open
Abstract
The SARS-CoV-2 virus causing COVID-19 disease has emerged expeditiously in the world and has been declared pandemic since March 2020, by World Health Organization (WHO). The destructive effects of SARS-CoV-2 infection are increased among the patients with pre-existing chronic conditions and, in particular, this review focuses on patients with underlying cardiovascular complications. The expression pattern and potential functions of SARS-CoV-2 binding receptors and the attributes of SARS-CoV-2 virus tropism in a physio-pathological state of heart and blood vessel are precisely described. Of note, the atheroprotective role of ACE2 receptors is reviewed. A detailed description of the possible detrimental role of SARS-CoV-2 infection in terms of vascular leakage, including endothelial glycocalyx dysfunction and bradykinin 1 receptor stimulation is concisely stated. Furthermore, the potential molecular mechanisms underlying SARS-CoV-2 induced clot formation in association with host defense components, including activation of FXIIa, complements and platelets, endothelial dysfunction, immune cell responses with cytokine-mediated action are well elaborated. Moreover, a brief clinical update on patient with COVID-19 disease with underlying cardiovascular complications and those who had new onset of cardiovascular complications post-COVID-19 disease was also discussed. Taken together, this review provides an overview of the mechanistic aspects of SARS-CoV-2 induced devastating effects, in vital organs such as the heart and vessels.
Collapse
Affiliation(s)
- Priya Veluswamy
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Max Wacker
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Dimitrios Stavridis
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Thomas Reichel
- Department of Cardiology, Diabetology and Infectiology, Klinikum Magdeburg, 39130 Magdeburg, Germany; (T.R.); (H.S.)
| | - Hendrik Schmidt
- Department of Cardiology, Diabetology and Infectiology, Klinikum Magdeburg, 39130 Magdeburg, Germany; (T.R.); (H.S.)
| | - Maximilian Scherner
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Jens Wippermann
- Heart Surgery Research, Department of Cardiothoracic Surgery, Faculty of Medicine, Otto-von-Guericke University, 39120 Magdeburg, Germany; (M.W.); (D.S.); (M.S.); (J.W.)
| | - Guido Michels
- Department of Acute and Emergency Care, Sankt Antonius-Hospital Eschweiler, 52249 Eschweiler, Germany;
| |
Collapse
|
25
|
Warner DR, Warner JB, Hardesty JE, Song YL, Chen CY, Chen Z, Kang JX, McClain CJ, Kirpich IA. Beneficial effects of an endogenous enrichment in n3-PUFAs on Wnt signaling are associated with attenuation of alcohol-mediated liver disease in mice. FASEB J 2021; 35:e21377. [PMID: 33481293 DOI: 10.1096/fj.202001202r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022]
Abstract
Alcohol-associated liver disease (ALD) is a major human health issue for which there are limited treatment options. Experimental evidence suggests that nutrition plays an important role in ALD pathogenesis, and specific dietary fatty acids, for example, n6 or n3-PUFAs, may exacerbate or attenuate ALD, respectively. The purpose of the current study was to determine whether the beneficial effects of n3-PUFA enrichment in ALD were mediated, in part, by improvement in Wnt signaling. Wild-type (WT) and fat-1 transgenic mice (that endogenously convert n6-PUFAs to n3) were fed ethanol (EtOH) for 6 weeks followed by a single LPS challenge. fat-1 mice had less severe liver damage than WT littermates as evidenced by reduced plasma alanine aminotransferase, hepatic steatosis, liver tissue neutrophil infiltration, and pro-inflammatory cytokine expression. WT mice had a greater downregulation of Axin2, a key gene in the Wnt pathway, than fat-1 mice in response to EtOH and LPS. Further, there were significant differences between WT and fat-1 EtOH+LPS-challenged mice in the expression of five additional genes linked to the Wnt signaling pathway, including Apc, Fosl1/Fra-1, Mapk8/Jnk-1, Porcn, and Nkd1. Compared to WT, primary hepatocytes isolated from fat-1 mice exhibited more effective Wnt signaling and were more resistant to EtOH-, palmitic acid-, or TNFα-induced cell death. Further, we demonstrated that the n3-PUFA-derived lipid mediators, resolvins D1 and E1, can regulate hepatocyte expression of several Wnt-related genes that were differentially expressed between WT and fat-1 mice. These data demonstrate a novel mechanism by which n3-PUFAs can ameliorate ALD.
Collapse
Affiliation(s)
- Dennis R Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jeffrey B Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Josiah E Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Ying L Song
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Chi-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zoe Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.,University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, USA.,Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA.,Robley Rex Veterans Medical Center, Louisville, KY, USA
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.,University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, USA.,Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
26
|
Uhl B, A Mittmann L, Dominik J, Hennel R, Smiljanov B, Haring F, B Schaubächer J, Braun C, Padovan L, Pick R, Canis M, Schulz C, Mack M, Gutjahr E, Sinn P, Heil J, Steiger K, Kanse SM, Weichert W, Sperandio M, Lauber K, Krombach F, Reichel CA. uPA-PAI-1 heteromerization promotes breast cancer progression by attracting tumorigenic neutrophils. EMBO Mol Med 2021; 13:e13110. [PMID: 33998175 PMCID: PMC8185543 DOI: 10.15252/emmm.202013110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 04/28/2021] [Accepted: 03/25/2021] [Indexed: 01/13/2023] Open
Abstract
High intratumoral levels of urokinase-type plasminogen activator (uPA)-plasminogen activator inhibitor-1 (PAI-1) heteromers predict impaired survival and treatment response in early breast cancer. The pathogenetic role of this protein complex remains obscure. Here, we demonstrate that heteromerization of uPA and PAI-1 multiplies the potential of the single proteins to attract pro-tumorigenic neutrophils. To this end, tumor-released uPA-PAI-1 utilizes very low-density lipoprotein receptor and mitogen-activated protein kinases to initiate a pro-inflammatory program in perivascular macrophages. This enforces neutrophil trafficking to cancerous lesions and skews these immune cells toward a pro-tumorigenic phenotype, thus supporting tumor growth and metastasis. Blockade of uPA-PAI-1 heteromerization by a novel small-molecule inhibitor interfered with these events and effectively prevented tumor progression. Our findings identify a therapeutically targetable, hitherto unknown interplay between hemostasis and innate immunity that drives breast cancer progression. As a personalized immunotherapeutic strategy, blockade of uPA-PAI-1 heteromerization might be particularly beneficial for patients with highly aggressive uPA-PAI-1high tumors.
Collapse
Affiliation(s)
- Bernd Uhl
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Laura A Mittmann
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Julian Dominik
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Roman Hennel
- Department of Radiation OncologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Bojan Smiljanov
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Florian Haring
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Johanna B Schaubächer
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Constanze Braun
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Lena Padovan
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Robert Pick
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Martin Canis
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christian Schulz
- Department of CardiologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Matthias Mack
- Department of Internal MedicineUniversity of RegensburgRegensburgGermany
| | - Ewgenija Gutjahr
- Institute for PathologyUniversity of HeidelbergHeidelbergGermany
| | - Peter Sinn
- Institute for PathologyUniversity of HeidelbergHeidelbergGermany
| | - Jörg Heil
- Department of Gynecology and ObstetricsUniversity of HeidelbergHeidelbergGermany
| | - Katja Steiger
- Department of PathologyTechnical University of MunichMunichGermany
| | - Sandip M Kanse
- Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Wilko Weichert
- Department of PathologyTechnical University of MunichMunichGermany
- German Cancer Consortium (DKTK), partner site MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and PathophysiologyLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Kirsten Lauber
- Department of Radiation OncologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Fritz Krombach
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christoph A Reichel
- Department of OtorhinolaryngologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Walter Brendel Centre of Experimental MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| |
Collapse
|
27
|
Chen J, Su Y, Pi S, Hu B, Mao L. The Dual Role of Low-Density Lipoprotein Receptor-Related Protein 1 in Atherosclerosis. Front Cardiovasc Med 2021; 8:682389. [PMID: 34124208 PMCID: PMC8192809 DOI: 10.3389/fcvm.2021.682389] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Low-density lipoprotein receptor–related protein-1 (LRP1) is a large endocytic and signaling receptor belonging to the LDL receptor (LDLR) gene family and that is widely expressed in several tissues. LRP1 comprises a large extracellular domain (ECD; 515 kDa, α chain) and a small intracellular domain (ICD; 85 kDa, β chain). The deletion of LRP1 leads to embryonic lethality in mice, revealing a crucial but yet undefined role in embryogenesis and development. LRP1 has been postulated to participate in numerous diverse physiological and pathological processes ranging from plasma lipoprotein homeostasis, atherosclerosis, tumor evolution, and fibrinolysis to neuronal regeneration and survival. Many studies using cultured cells and in vivo animal models have revealed the important roles of LRP1 in vascular remodeling, foam cell biology, inflammation and atherosclerosis. However, its role in atherosclerosis remains controversial. LRP1 not only participates in the removal of atherogenic lipoproteins and proatherogenic ligands in the liver but also mediates the uptake of aggregated LDL to promote the formation of macrophage- and vascular smooth muscle cell (VSMC)-derived foam cells, which causes a prothrombotic transformation of the vascular wall. The dual and opposing roles of LRP1 may also represent an interesting target for atherosclerosis therapeutics. This review highlights the influence of LRP1 during atherosclerosis development, focusing on its dual role in vascular cells and immune cells.
Collapse
Affiliation(s)
- Jiefang Chen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Su
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shulan Pi
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Mao
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Pluijmert NJ, Atsma DE, Quax PHA. Post-ischemic Myocardial Inflammatory Response: A Complex and Dynamic Process Susceptible to Immunomodulatory Therapies. Front Cardiovasc Med 2021; 8:647785. [PMID: 33996944 PMCID: PMC8113407 DOI: 10.3389/fcvm.2021.647785] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/02/2021] [Indexed: 01/04/2023] Open
Abstract
Following acute occlusion of a coronary artery causing myocardial ischemia and implementing first-line treatment involving rapid reperfusion, a dynamic and balanced inflammatory response is initiated to repair and remove damaged cells. Paradoxically, restoration of myocardial blood flow exacerbates cell damage as a result of myocardial ischemia-reperfusion (MI-R) injury, which eventually provokes accelerated apoptosis. In the end, the infarct size still corresponds to the subsequent risk of developing heart failure. Therefore, true understanding of the mechanisms regarding MI-R injury, and its contribution to cell damage and cell death, are of the utmost importance in the search for successful therapeutic interventions to finally prevent the onset of heart failure. This review focuses on the role of innate immunity, chemokines, cytokines, and inflammatory cells in all three overlapping phases following experimental, mainly murine, MI-R injury known as the inflammatory, reparative, and maturation phase. It provides a complete state-of-the-art overview including most current research of all post-ischemic processes and phases and additionally summarizes the use of immunomodulatory therapies translated into clinical practice.
Collapse
Affiliation(s)
- Niek J Pluijmert
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Douwe E Atsma
- Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
29
|
Liu Y, Wang M, Wang D, Fay WP, Korthuis RJ, Sowa G. Elevated postischemic tissue injury and leukocyte-endothelial adhesive interactions in mice with global deficiency in caveolin-2: role of PAI-1. Am J Physiol Heart Circ Physiol 2021; 320:H1185-H1198. [PMID: 33416452 PMCID: PMC8362680 DOI: 10.1152/ajpheart.00682.2020] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 01/13/2023]
Abstract
Ischemia/reperfusion (I/R)-induced rapid inflammation involving activation of leukocyte-endothelial adhesive interactions and leukocyte infiltration into tissues is a major contributor to postischemic tissue injury. However, the molecular mediators involved in this pathological process are not fully known. We have previously reported that caveolin-2 (Cav-2), a protein component of plasma membrane caveolae, regulated leukocyte infiltration in mouse lung carcinoma tumors. The goal of the current study was to examine if Cav-2 plays a role in I/R injury and associated acute leukocyte-mediated inflammation. Using a mouse small intestinal I/R model, we demonstrated that I/R downregulates Cav-2 protein levels in the small bowel. Further study using Cav-2-deficient mice revealed aggravated postischemic tissue injury determined by scoring of villi length in H&E-stained tissue sections, which correlated with increased numbers of MPO-positive tissue-infiltrating leukocytes determined by IHC staining. Intravital microscopic analysis of upstream events relative to leukocyte transmigration and tissue infiltration revealed that leukocyte-endothelial cell adhesive interactions in postcapillary venules, namely leukocyte rolling and adhesion were also enhanced in Cav-2-deficient mice. Mechanistically, Cav-2 deficiency increased plasminogen activator inhibitor-1 (PAI-1) protein levels in the intestinal tissue and a pharmacological inhibition of PAI-1 had overall greater inhibitory effect on both aggravated I/R tissue injury and enhanced leukocyte-endothelial interactions in postcapillary venules in Cav-2-deficient mice. In conclusion, our data suggest that Cav-2 protein alleviates tissue injury in response to I/R by dampening PAI-1 protein levels and thereby reducing leukocyte-endothelial adhesive interactions.NEW & NOTEWORTHY The role of caveolin-2 in regulating ischemia/reperfusion (I/R) tissue injury and the mechanisms underlying its effects are unknown. This study uses caveolin-2-deficient mouse and small intestinal I/R injury models to examine the role of caveolin-2 in the leukocyte-dependent reperfusion injury. We demonstrate for the first time that caveolin-2 plays a protective role from the I/R-induced leukocyte-dependent reperfusion injury by reducing PAI-1 protein levels in intestinal tissue and leukocyte-endothelial adhesive interactions in postcapillary venules.
Collapse
Affiliation(s)
- Yajun Liu
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Meifang Wang
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Derek Wang
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - William P Fay
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Medicine, University of Missouri, Columbia, Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- The Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Grzegorz Sowa
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
30
|
Exosomes Released from CaSR-Stimulated PMNs Reduce Ischaemia/Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3010548. [PMID: 33505580 PMCID: PMC7815400 DOI: 10.1155/2021/3010548] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/30/2020] [Accepted: 12/25/2020] [Indexed: 12/31/2022]
Abstract
Ischemia-reperfusion (I/R) injury caused by acute myocardial infarction (AMI) can initiate a strong inflammatory response. Polymorphonuclear cells (PMNs) are the most important inflammatory cells. Our previous studies found that the calcium-sensing receptor (CaSR) regulates the proinflammatory effects of PMNs. However, the role and mechanism of CaSR-regulated PMNs in I/R injury remain uncertain. A rat AMI model was developed in this study and showed that the expression of CaSR on PMNs increased in AMI; however, the levels of Bcl-xl and SOD in myocardial tissue decreased, while Bax and MDA levels increased. Then, after coculture with CaSR-stimulated PMNs, the expression of Bcl-xl in cardiomyocytes significantly increased, Bax expression and the apoptotic rate decreased, and ROS production was significantly inhibited. At the same time, the cardiomyocyte damage caused by hypoxia-reoxygenation was reduced. Furthermore, we found that exosomes derived from PMNs could be taken up by cardiomyocytes. Additionally, the exosomes secreted by CaSR-stimulated PMNs had the same effect on cardiomyocytes as CaSR-stimulated PMNs, while the increased phosphorylation level of AKT in cardiomyocytes could be revered by AKT transduction pathway inhibitors. Subsequently, we identified the exosomes derived from CaSR-stimulated PMNs by second-generation sequencing technology, and increased expression of lncRNA ENSRNOT00000039868 was noted. The data show that this lncRNA can prevent the hypoxia-reoxygenation injury by upregulating the expression of PDGFD in cardiomyocytes. In vivo, exosomes from CaSR-stimulated PMNs played a significant role against AMI and reperfusion injury in myocardial tissue. Thus, we propose that exosomes derived from CaSR-stimulated PMNs can reduce I/R injury in AMI, and this effect may be related to the AKT signaling pathway.
Collapse
|
31
|
Wang T, Lu H, Li D, Huang W. TGF-β1-Mediated Activation of SERPINE1 is Involved in Hemin-Induced Apoptotic and Inflammatory Injury in HT22 Cells. Neuropsychiatr Dis Treat 2021; 17:423-433. [PMID: 33603380 PMCID: PMC7884960 DOI: 10.2147/ndt.s293772] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a severe subtype of stroke with high mortality and morbidity. Serpin Family E Member 1 (SERPINE1) has been documented to be upregulated following ICH, however, the participation of SERPINE1 in the development of ICH has never been studied. METHODS Hemin was utilized to develop an in vitro model of ICH. Gene levels were evaluated by the use of quantitative reverse transcription polymerase chain reaction, Western blot, as well as enzyme-linked immunoassay assay. The activity of caspase-3 was determined using a commercial kit. Cell viability and apoptosis were assessed using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and Terminal deoxynucleotidyl transferase (TdT) d UTP Nick-End Labeling assay. RESULTS SERPINE1 was upregulated in hemin-treated HT22 cells. Silencing of SERPINE1 attenuated hemin-induced inhibition of cell viability. Moreover, knockdown of SERPINE1 repressed hemin-induced apoptosis in HT22 cells, as evidenced by the decrease in the number of TUNEL positive cells, caspase-3 activity, and Bax expression, and the increase in Bcl-2 expression. Meanwhile, knockdown of SERPINE1 repressed hemin-induced inflammation in HT22 cells, as indicated by reduced levels of tumor necrosis factor-α, interleukin-6 (IL-6), IL-1β, and inducible nitric oxide synthase. We also found that transforming growth factor-beta 1 (TGF-β1) induced SERPINE1 expression in a dose-dependent manner. Besides, SERPINE1 knockdown attenuated the effects of TGF-β1 on hemin-induced neuronal damage. CONCLUSION TGF-β1-induced SERPINE1 activation exacerbated hemin-induced apoptosis and inflammation in HT22 cells, manifesting a novel mechanism for ICH progression.
Collapse
Affiliation(s)
- Tinggang Wang
- Emergency Department, Affiliated Hospital of Zunyi Medical University, Zunyi, People's Republic of China
| | - Haibin Lu
- Department of Critical Care Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Deqiang Li
- Department of Critical Care Medicine, Daping Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Weichun Huang
- Radiology Department, First Affiliated Hospital of Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
32
|
Immler R, Lange-Sperandio B, Steffen T, Beck H, Rohwedder I, Roth J, Napoli M, Hupel G, Pfister F, Popper B, Uhl B, Mannell H, Reichel CA, Vielhauer V, Scherberich J, Sperandio M, Pruenster M. Extratubular Polymerized Uromodulin Induces Leukocyte Recruitment and Inflammation In Vivo. Front Immunol 2020; 11:588245. [PMID: 33414784 PMCID: PMC7783395 DOI: 10.3389/fimmu.2020.588245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Uromodulin (UMOD) is produced and secreted by tubular epithelial cells. Secreted UMOD polymerizes (pUMOD) in the tubular lumen, where it regulates salt transport and protects the kidney from bacteria and stone formation. Under various pathological conditions, pUMOD accumulates within the tubular lumen and reaches extratubular sites where it may interact with renal interstitial cells. Here, we investigated the potential of extratubular pUMOD to act as a damage associated molecular pattern (DAMP) molecule thereby creating local inflammation. We found that intrascrotal and intraperitoneal injection of pUMOD induced leukocyte recruitment in vivo and led to TNF-α secretion by F4/80 positive macrophages. Additionally, pUMOD directly affected vascular permeability and increased neutrophil extravasation independent of macrophage-released TNF-α. Interestingly, pUMOD displayed no chemotactic properties on neutrophils, did not directly activate β2 integrins and did not upregulate adhesion molecules on endothelial cells. In obstructed neonatal murine kidneys, we observed extratubular UMOD accumulation in the renal interstitium with tubular atrophy and leukocyte infiltrates. Finally, we found extratubular UMOD deposits associated with peritubular leukocyte infiltration in kidneys from patients with inflammatory kidney diseases. Taken together, we identified extratubular pUMOD as a strong inducer of leukocyte recruitment, underlining its critical role in mounting an inflammatory response in various kidneys pathologies.
Collapse
Affiliation(s)
- Roland Immler
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Bärbel Lange-Sperandio
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Tobias Steffen
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Heike Beck
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Ina Rohwedder
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Jonas Roth
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Matteo Napoli
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Georg Hupel
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Frederik Pfister
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Bastian Popper
- Core facility animal models, Biomedical Center, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Bernd Uhl
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hanna Mannell
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Christoph A. Reichel
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Volker Vielhauer
- Medizinische Klinik und Poliklinik IV, Nephrologisches Zentrum, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jürgen Scherberich
- Klinikum Harlaching, teaching hospital of the Ludwig-Maximilians University Munich, Munich, Germany
| | - Markus Sperandio
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| | - Monika Pruenster
- Walter Brendel Centre of Experimental Medicine, Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Ludwig-Maximilians-University Munich, Planegg-Martinsried, Germany
| |
Collapse
|
33
|
NFAT5 directs hyperosmotic stress-induced fibrin deposition and macrophage infiltration via PAI-1 in endothelium. Aging (Albany NY) 2020; 13:3661-3679. [PMID: 33410782 PMCID: PMC7906158 DOI: 10.18632/aging.202330] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022]
Abstract
Although stress can significantly promote atherosclerosis, the underlying mechanisms are still not completely understood. Here we successfully unveiled that high salt-induced nuclear factor of activated T cells 5 (NFAT5) control the endothelial-dependent fibrinolytic activity and the inflammatory adhesion-related molecules expression through regulation of plasminogen activator inhibitor-1 (PAI-1). We first observed that high salt diets instigated the expression of NFAT5 and PAI-1 in the endothelium which brought about the fibrin deposition and macrophage infiltration in the atherosclerotic arteries of ApoE-/- mice. Overexpression of NFAT5 increased PAI-1-mediated antifibrinolytic activity and activated inflammatory adhesion-related genes in endothelial cells. Knockdown of NFAT5 by siRNA inhibited the expression of PAI-1, antifibrinolytic and adhesive molecules. Moreover, chromatin immunoprecipitation assay demonstrated that high salt intake significantly promoted the binding of NFAT5 to PAI-1 promoter (TGGAATTATTT) in endothelial cells. Our study identified that NFAT5 has great potential to activate the PAI-1-mediated fibrinolytic dysfunction and inflammatory cell adhesion, thus promoting high salt-induced atherosclerosis disease.
Collapse
|
34
|
Chen X, Zhang J, Xia L, Wang L, Li H, Liu H, Zhou J, Feng Z, Jin H, Yang J, Yang Y, Wu B, Zhang L, Chen G, Wang G. β-Arrestin-2 attenuates hepatic ischemia-reperfusion injury by activating PI3K/Akt signaling. Aging (Albany NY) 2020; 13:2251-2263. [PMID: 33323551 PMCID: PMC7880335 DOI: 10.18632/aging.202246] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Hepatic ischemia-reperfusion injury (IRI) remains a common complication during liver transplantation (LT), partial hepatectomy and hemorrhagic shock in patients. As a member of the G protein-coupled receptors adaptors, ARRB2 has been reported to be involved in a variety of physiological and pathological processes. However, whether β-arrestin-2 affects the pathogenesis of hepatic IRI remains unknown. The goal of the present study was to determine whether ARRB2 protects against hepatic IR injury and elucidate the underlying mechanisms. To this end, 70% hepatic IR models were established in ARRB2 knockdown mice and wild-type littermates, with blood and liver samples collected at 1, 6 and 12 h after reperfusion to evaluate liver injury. The effect of ARBB2 on PI3K/Akt signaling during IR injury was evaluated in vivo, and PI3K/Akt pathway regulation by ARRB2 was further assessed in vitro. Our results showed that ARRB2 knockdown aggravates hepatic IR injury by promoting the apoptosis of hepatocytes and inhibiting their proliferation. In addition, ARRB2 deficiency inhibited PI3K/Akt pathway activation, while the administration of the PI3K/Akt inhibitor PX866 resulted in severe IR injury in mice. Furthermore, the liver-protecting effect of ARRB2 was shown to depend on PI3K/Akt pathway activation. In summary, our results suggest that β-Arrestin-2 protects against hepatic IRI by activating PI3K/Akt signaling, which may provide a novel therapeutic strategy for treating liver ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xiaolong Chen
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Junbin Zhang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Long Xia
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Li Wang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Hui Li
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Huilin Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China.,Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Jing Zhou
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Zhiying Feng
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Hai Jin
- Department of Medical Ultrasonics, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou 510630, Guangdong Province, P. R. China
| | - JianXu Yang
- Department of Intensive Care Unit, Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, P. R. China
| | - Yang Yang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Bin Wu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China.,Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China.,Department of Biliary-Pancreatic Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China
| | - Guihua Chen
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| | - Genshu Wang
- Department of Hepatic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, P. R. China.,Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou 510630, Guangdong Province, P. R. China
| |
Collapse
|
35
|
Stress-induced RNA-chromatin interactions promote endothelial dysfunction. Nat Commun 2020; 11:5211. [PMID: 33060583 PMCID: PMC7566596 DOI: 10.1038/s41467-020-18957-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/11/2020] [Indexed: 01/06/2023] Open
Abstract
Chromatin-associated RNA (caRNA) has been proposed as a type of epigenomic modifier. Here, we test whether environmental stress can induce cellular dysfunction through modulating RNA-chromatin interactions. We induce endothelial cell (EC) dysfunction with high glucose and TNFα (H + T), that mimic the common stress in diabetes mellitus. We characterize the H + T-induced changes in gene expression by single cell (sc)RNA-seq, DNA interactions by Hi-C, and RNA-chromatin interactions by iMARGI. H + T induce inter-chromosomal RNA-chromatin interactions, particularly among the super enhancers. To test the causal relationship between H + T-induced RNA-chromatin interactions and the expression of EC dysfunction-related genes, we suppress the LINC00607 RNA. This suppression attenuates the expression of SERPINE1, a critical pro-inflammatory and pro-fibrotic gene. Furthermore, the changes of the co-expression gene network between diabetic and healthy donor-derived ECs corroborate the H + T-induced RNA-chromatin interactions. Taken together, caRNA-mediated dysregulation of gene expression modulates EC dysfunction, a crucial mechanism underlying numerous diseases. Global interaction of chromatin-associated RNAs and DNA can be identified in situ. Here the authors report the genome-wide increase of interchromosomal RNA-DNA interactions and demonstrate the importance of such RNA-DNA contacts exemplified by LINC00607 RNA and SERPINE1 gene’s super enhancer in dysfunctional endothelial cell models.
Collapse
|
36
|
Liberale L, Bertolotto M, Minetti S, Contini P, Verzola D, Ameri P, Ghigliotti G, Pende A, Camici GG, Carbone F, Montecucco F. Recombinant Tissue Plasminogen Activator (r-tPA) Induces In-Vitro Human Neutrophil Migration via Low Density Lipoprotein Receptor-Related Protein 1 (LRP-1). Int J Mol Sci 2020; 21:7014. [PMID: 32977685 PMCID: PMC7582901 DOI: 10.3390/ijms21197014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023] Open
Abstract
Thrombolysis is the gold standard treatment for acute ischemic stroke. Besides its fibrinolytic role, recombinant tissue plasminogen activator (r-tPA) holds several non-fibrinolytic functions. Here, we investigated the potential role of r-tPA on human primary neutrophil migration in vitro. By means of modified Boyden chamber migration assay and checkerboard analysis we showed a dose-dependent chemotactic effect of r-TPA with a maximum effect reached by 0.03 mg/mL (0.003-1 mg/mL). Pre-incubation with MAP kinases inhibitors allowed the identification of PI3K/Akt, but not ERK1/2 as the intracellular pathway mediating the observed effects. Furthermore, by means of real-time PCR, immunocytochemistry and cytofluorimetry we demonstrated that the r-tPA receptor low density lipoprotein receptor-related protein 1 (LRP-1) is synthetized and expressed by neutrophils in response to r-tPA and TNF-α. Inhibition of LRP-1 by receptor-associated protein (RAP), prevented r-tPA-mediated F-actin polymerization, migration and signal through Akt but not ERK1/2. Lastly, also neutrophil degranulation in response to r-tPA seems to be mediated by LRP-1 under adhesion conditions. In conclusion, we show that r-tPA induces neutrophil chemotaxis through LRP-1/Akt pathway. Blunting r-tPA-mediated neutrophil activation might be beneficial as an adjuvant therapy to thrombolysis in this setting.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; (L.L.); (G.G.C.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Maria Bertolotto
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Silvia Minetti
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
| | - Paola Contini
- Clinical Immunology, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy;
| | - Daniela Verzola
- Division of Nephrology, Dialysis and Transplantation, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy;
| | - Pietro Ameri
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Laboratory of Cardiovascular Biology, IRCCS Ospedale Policlinico San Martino & Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Giorgio Ghigliotti
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Laboratory of Cardiovascular Biology, IRCCS Ospedale Policlinico San Martino & Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Aldo Pende
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
- Clinic of Emergency Medicine, Department of Emergency Medicine, University of Genoa, 16126 Genoa, Italy
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952 Schlieren, Switzerland; (L.L.); (G.G.C.)
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy; (M.B.); (S.M.); (F.C.)
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy; (P.A.); (G.G.); (A.P.)
| |
Collapse
|
37
|
Yu H, Liu Y, Wang M, Restrepo RJ, Wang D, Kalogeris TJ, Neumann WL, Ford DA, Korthuis RJ. Myeloperoxidase instigates proinflammatory responses in a cecal ligation and puncture rat model of sepsis. Am J Physiol Heart Circ Physiol 2020; 319:H705-H721. [PMID: 32762560 DOI: 10.1152/ajpheart.00440.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myeloperoxidase (MPO)-derived hypochlorous (HOCl) reacts with membrane plasmalogens to yield α-chlorofatty aldehydes such as 2-chlorofatty aldehyde (2-ClFALD) and its metabolite 2-chlorofatty acid (2-ClFA). Recent studies showed that 2-ClFALD and 2-ClFA serve as mediators of the inflammatory responses to sepsis by as yet unknown mechanisms. Since no scavenger for chlorinated lipids is available and on the basis of the well-established role of the MPO/HOCl/chlorinated lipid axis in inflammatory responses, we hypothesized that treatment with MPO inhibitors (N-acetyl lysyltyrosylcysteine amide or 4-aminobenzoic acid hydrazide) would inhibit inflammation and proinflammatory mediator expression induced by cecal ligation and puncture (CLP). We used intravital microscopy to quantify in vivo inflammatory responses in Sham and CLP rats with or without MPO inhibition. Small intestines, mesenteries, and lungs were collected to assess changes in MPO-positive staining and lung injury, respectively, as well as free 2-ClFA and proinflammatory mediators levels. CLP caused neutrophil infiltration, 2-ClFA generation, acute lung injury, leukocyte-/platelet-endothelium interactions, mast cell activation (MCA), plasminogen activator inhibitor-1 (PAI-1) production, and the expression of several cytokines, chemokines, and vascular endothelial growth factor, changes that were reduced by MPO inhibition. Pretreatment with a PAI-1 inhibitor or MC stabilizer prevented CLP-induced leukocyte-endothelium interactions and MCA, and abrogated exogenous 2-ClFALD-induced inflammatory responses. Thus, we provide evidence that MPO instigates these inflammatory changes in CLP and that chlorinated lipids may serve as a mechanistic link between the enzymatic activity of MPO and PAI-1- and mast cell-dependent adhesive interactions, providing a rationale for new therapeutic interventions in sepsis.NEW & NOTEWORTHY Using two distinct myeloperoxidase (MPO) inhibitors, we show for the first time that MPO plays an important role in producing increases in free 2-chlorofatty aldehyde (2-ClFALD)-a powerful proinflammatory chlorinated lipid in plasma and intestine-a number of cytokines and other inflammatory mediators, leukocyte and platelet rolling and adhesion in postcapillary venules, and lung injury in a cecal ligation and puncture model of sepsis. In addition, the use of a plasminogen activator inhibitor-1 (PAI-1) inhibitor or a mast cell stabilizer prevented inflammatory responses in CLP-induced sepsis. PAI-1 inhibition also prevented the proinflammatory responses to exogenous 2-ClFALD superfusion. Thus, our study provides some of the first evidence that MPO-derived free 2-ClFA plays an important role in CLP-induced sepsis by a PAI-1- and mast cell-dependent mechanism.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Yajun Liu
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Meifang Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Ricardo J Restrepo
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Derek Wang
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Theodore J Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - William L Neumann
- Department of Pharmaceutical Sciences, Edwardsville School of Pharmacy, Southern Illinois University, Edwardsville, Illinois
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Center for Cardiovascular Research, Saint Louis University School of Medicine, Saint Louis, Missouri
| | - Ronald J Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
38
|
Werner MJM, de Meijer VE, Adelmeijer J, de Kleine RHJ, Scheenstra R, Bontemps STH, Reyntjens KMEM, Hulscher JBF, Lisman T, Porte RJ. Evidence for a rebalanced hemostatic system in pediatric liver transplantation: A prospective cohort study. Am J Transplant 2020; 20:1384-1392. [PMID: 31841272 PMCID: PMC7216958 DOI: 10.1111/ajt.15748] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/12/2019] [Accepted: 12/04/2019] [Indexed: 01/25/2023]
Abstract
In adults with end-stage liver disease concurrent changes in pro- and antihemostatic pathways result in a rebalanced hemostasis. Children though, have a developing hemostatic system, different disease etiologies, and increased risk of thrombosis. This study aimed to assess the hemostatic state of children during and after liver transplantation. Serial blood samples were obtained from 20 children (≤16 years) undergoing primary liver transplantation (September 2017-October 2018). Routine hemostasis tests, thrombomodulin-modified thrombin generation, clot lysis times, and hemostatic proteins were measured. Reference values were established using an age-matched control group of 30 children. Thrombocytopenia was present in study patients. Von Willebrand factors were doubled and ADAMTS13 levels decreased during and after transplantation up until day 30, when platelet count had normalized. Whereas prothrombin time and activated partial thromboplastin time were prolonged during transplantation, thrombin generation was within normal ranges, except during perioperative heparin administration. Fibrinogen, factor VIII levels, and clot lysis time were elevated up until day 30. In conclusion, children with end-stage liver disease are in tight hemostatic balance. During transplantation a temporary heparin-dependent hypocoagulable state is present, which rapidly converts to a hemostatic balance with distinct hypercoagulable features that persist until at least day 30. This hypercoagulable state may contribute to the risk of posttransplant thrombosis.
Collapse
Affiliation(s)
- Maureen J. M. Werner
- Department of SurgerySection of Hepatobiliary Surgery and Liver TransplantationUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands,Department of SurgerySurgical Research LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Vincent E. de Meijer
- Department of SurgerySection of Hepatobiliary Surgery and Liver TransplantationUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Jelle Adelmeijer
- Department of SurgerySurgical Research LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Ruben H. J. de Kleine
- Department of SurgerySection of Hepatobiliary Surgery and Liver TransplantationUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - René Scheenstra
- Department of Pediatric Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Sander T. H. Bontemps
- Department of Pediatric Intensive CareUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Koen M. E. M. Reyntjens
- Department of AnesthesiologyUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Jan B. F. Hulscher
- Department of SurgerySection of Pediatric SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Ton Lisman
- Department of SurgerySection of Hepatobiliary Surgery and Liver TransplantationUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands,Department of SurgerySurgical Research LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| | - Robert J. Porte
- Department of SurgerySection of Hepatobiliary Surgery and Liver TransplantationUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands,Department of SurgerySurgical Research LaboratoryUniversity of GroningenUniversity Medical Center GroningenGroningenthe Netherlands
| |
Collapse
|
39
|
Alehagen U, Alexander J, Aaseth J, Larsson A, Lindahl TL. Significant decrease of von Willebrand factor and plasminogen activator inhibitor-1 by providing supplementation with selenium and coenzyme Q10 to an elderly population with a low selenium status. Eur J Nutr 2020; 59:3581-3590. [PMID: 32078064 PMCID: PMC7669787 DOI: 10.1007/s00394-020-02193-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
Purpose Endothelial dysfunction and inflammation are conditions which fuel atherosclerosis and ischaemic heart disease. We have previously reported reduced cardiovascular (CV) mortality following supplementation with selenium and coenzyme Q10 to 443 elderly individuals with low selenium status (mean 67 μg/L) for 4 years. Here, we wanted to evaluate a possible association between the supplementation and the plasma concentrations of the von Willebrand factor (vWf), and the plasminogen activator inhibitor-1 (PAI-1), as they, besides other functions, are also strongly associated with endothelial function. Methods In this sub-study, 308 individuals (active substance: 157, placebo: 151) were included. Blood samples were drawn after 6 and 36 months and vWf and PAI-1 were determined in plasma by ELISA. Changes in concentrations of the biomarkers were evaluated by the use of T tests, repeated measures of variance, and ANCOVA analyses. Results The active treatment group presented a lower level of vWf after 36 months compared with the placebo group (1.08 U/mL vs. 5.10 U/mL; p = 0.0007). The results were validated through the repeated measures of variance evaluation. The PAI-1 levels showed an equally significant decrease in the active group (26.2 ng/mL vs. 49.2 ng/mL; p = 0.0002) and were also validated through repeated measures of variance evaluation. Conclusion In this sub-study on elderly receiving selenium and coenzyme Q10, or placebo we found significantly lower levels of vWf and PAI-1 in the active treatment group as compared to the placebo group. We interpret this as a better endothelial function because of the intervention, which accords with a previous finding of reduced CV mortality.
Collapse
Affiliation(s)
- Urban Alehagen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, 581 85, Linköping, Sweden.
| | - J Alexander
- Norwegian Institute of Public Health, 0403, Oslo, Norway
| | - J Aaseth
- Research Department, Innlandet Hospital Trust, 2381, Brumunddal, Norway
| | - A Larsson
- Department of Medical Sciences, Uppsala University, 751 85, Uppsala, Sweden
| | - T L Lindahl
- Division of Clinical Chemistry, Department of Experimental and Clinical Medicine, Linköping University, 581 85, Linköping, Sweden
| |
Collapse
|
40
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Annual Report on Sex in Preclinical Studies: Arteriosclerosis, Thrombosis, and Vascular Biology Publications in 2018. Arterioscler Thromb Vasc Biol 2019; 40:e1-e9. [PMID: 31869272 DOI: 10.1161/atvbaha.119.313556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC (N.M.)
| | - Daniel J Rader
- Departments of Medicine and Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.)
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität (LMU) and German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
41
|
Puhr-Westerheide D, Schink SJ, Fabritius M, Mittmann L, Hessenauer MET, Pircher J, Zuchtriegel G, Uhl B, Holzer M, Massberg S, Krombach F, Reichel CA. Neutrophils promote venular thrombosis by shaping the rheological environment for platelet aggregation. Sci Rep 2019; 9:15932. [PMID: 31685838 PMCID: PMC6828708 DOI: 10.1038/s41598-019-52041-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/12/2019] [Indexed: 12/24/2022] Open
Abstract
In advanced inflammatory disease, microvascular thrombosis leads to the interruption of blood supply and provokes ischemic tissue injury. Recently, intravascularly adherent leukocytes have been reported to shape the blood flow in their immediate vascular environment. Whether these rheological effects are relevant for microvascular thrombogenesis remains elusive. Employing multi-channel in vivo microscopy, analyses in microfluidic devices, and computational modeling, we identified a previously unanticipated role of leukocytes for microvascular clot formation in inflamed tissue. For this purpose, neutrophils adhere at distinct sites in the microvasculature where these immune cells effectively promote thrombosis by shaping the rheological environment for platelet aggregation. In contrast to larger (lower-shear) vessels, this process in high-shear microvessels does not require fibrin generation or extracellular trap formation, but involves GPIbα-vWF and CD40-CD40L-dependent platelet interactions. Conversely, interference with these cellular interactions substantially compromises microvascular clotting. Thus, leukocytes shape the rheological environment in the inflamed venular microvasculature for platelet aggregation thereby effectively promoting the formation of blood clots. Targeting this specific crosstalk between the immune system and the hemostatic system might be instrumental for the prevention and treatment of microvascular thromboembolic pathologies, which are inaccessible to invasive revascularization strategies.
Collapse
Affiliation(s)
- Daniel Puhr-Westerheide
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Radiology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Severin J Schink
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Fabritius
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Radiology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Laura Mittmann
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Otorhinolaryngology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maximilian E T Hessenauer
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Plastic and Hand Surgery, Friedrich Alexander University Erlangen Nuernberg, Erlangen, Germany
| | - Joachim Pircher
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Cardiology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gabriele Zuchtriegel
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Otorhinolaryngology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Bernd Uhl
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Otorhinolaryngology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Holzer
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.,Department of Otorhinolaryngology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Massberg
- Department of Cardiology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fritz Krombach
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christoph A Reichel
- Walter Brendel Centre of Experimental Medicine, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany. .,Department of Otorhinolaryngology, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
42
|
Peng Z, Shu B, Zhang Y, Wang M. Endothelial Response to Pathophysiological Stress. Arterioscler Thromb Vasc Biol 2019; 39:e233-e243. [PMID: 31644356 DOI: 10.1161/atvbaha.119.312580] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Located in the innermost layer of the vasculature and directly interacting with blood flow, endothelium integrates various biochemical and biomechanical signals to maintain barrier function with selective permeability, vascular tone, blood fluidity, and vascular formation. Endothelial cells respond to laminar and disturbed flow by structural and functional adaption, which involves reprogramming gene expression, cell proliferation and migration, senescence, autophagy and cell death, as well as synthesizing signal molecules (nitric oxide and prostanoids, etc) that act in manners of autocrine, paracrine, or juxtacrine. Inflammation occurs after infection or tissue injury. Dysregulated inflammatory response participates in pathogenesis of many diseases. Endothelial cells exposed to inflammatory stimuli from the circulation or the microenvironment exhibit impaired vascular tone, increased permeability, elevated procoagulant activity, and dysregulated vascular formation, collectively contributing to the development of vascular diseases. Understanding the endothelial response to pathophysiological stress of hemodynamics and inflammation provides mechanistic insights into cardiovascular diseases, as well as therapeutic opportunities.
Collapse
Affiliation(s)
- Zekun Peng
- From the State Key Laboratory of Cardiovascular Disease (Z.P., B.S., Y.Z., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingyan Shu
- From the State Key Laboratory of Cardiovascular Disease (Z.P., B.S., Y.Z., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yurong Zhang
- From the State Key Laboratory of Cardiovascular Disease (Z.P., B.S., Y.Z., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao Wang
- From the State Key Laboratory of Cardiovascular Disease (Z.P., B.S., Y.Z., M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
43
|
Slack MA, Gordon SM. Protease Activity in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:e210-e218. [PMID: 31553665 PMCID: PMC6764587 DOI: 10.1161/atvbaha.119.312413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Megan A. Slack
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
44
|
Chan SL, Bishop N, Li Z, Cipolla MJ. Inhibition of PAI (Plasminogen Activator Inhibitor)-1 Improves Brain Collateral Perfusion and Injury After Acute Ischemic Stroke in Aged Hypertensive Rats. Stroke 2019; 49:1969-1976. [PMID: 29991657 DOI: 10.1161/strokeaha.118.022056] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background and Purpose- Aging and hypertension, comorbidities prevalent in the stroke population, are associated with poor collateral status and worsened stroke outcome. However, underlying mechanisms by which these conditions affect stroke outcome are not clear. We studied the role of PAI (plasminogen activator inhibitor)-1 that is increased in aging and hypertension on brain and vascular expression of inflammatory factors and perfusion that may contribute to worse stroke outcomes. Methods- Aged (≈50 weeks) and young (≈18 weeks) spontaneously hypertensive rats (SHR) were subjected to ischemia by middle cerebral artery occlusion (2 hours) and reperfusion (2 hours) with or without treatment with the PAI-1 inhibitor TM5441. Changes in middle cerebral artery and collateral perfusion territories were measured by multisite laser Doppler. Reactivity to TM5441 was studied using isolated and pressurized leptomeningeal anastomotic arterioles. Brain injury was determined by 2,3,5-triphenyltetrazolium staining and quantitative immunohistochemistry of amyloid-β-42, PAI-1, and hemoglobin. Circulating inflammatory factors were measured by ELISA. Results- Changes in cerebral blood flow during middle cerebral artery occlusion were similar between groups, with both having poor collateral perfusion and incomplete reperfusion. However, aged SHR had greater brain injury versus young (41±2 versus 23±2%, P<0.05) as well as increased brain deposition of amyloid-β-42 and circulating oxLDL (oxidized low-density lipoprotein). Erythrocyte aggregation and hemorrhage within the injured brain was observed in 50% of aged but no young SHR, with increased circulating PAI-1 in this subgroup of aged SHR (16±3 versus 6±2 ng/mL, P<0.05). PAI-1 inhibition with TM5441 improved brain injury but did not affect hemorrhage. TM5441 increased collateral perfusion by 38±7% and dilated leptomeningeal anastomotic arterioles by 44±10%, which was abolished by nitric oxide synthase inhibition. Conclusions- Increased injury in aged SHR seemed to be related to poor collateral perfusion, hemorrhagic transformation, increased amyloid-β-42, and oxidative stress. PAI-1 inhibition reduced infarction in both groups of SHR that possibly due, in part, to increased collateral perfusion.
Collapse
Affiliation(s)
- Siu-Lung Chan
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Nicole Bishop
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Zhaojin Li
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.)
| | - Marilyn J Cipolla
- From the Departments of Neurological Sciences (S.-L.C., N.B., Z.L., M.J.C.).,Obstetrics, Gynecology and Reproductive Sciences (M.J.C.).,Pharmacology (M.J.C.), University of Vermont Larner College of Medicine, Burlington
| |
Collapse
|
45
|
Palasubramaniam J, Wang X, Peter K. Myocardial Infarction-From Atherosclerosis to Thrombosis. Arterioscler Thromb Vasc Biol 2019; 39:e176-e185. [PMID: 31339782 DOI: 10.1161/atvbaha.119.312578] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Jathushan Palasubramaniam
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia (J.P., X.W., K.P.).,Department of Medicine, Monash University, Melbourne, Australia (J.P., X.W., K.P.).,Department of Cardiology, Alfred Hospital, Melbourne, Australia (J.P., K.P.)
| | - Xiaowei Wang
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia (J.P., X.W., K.P.).,Department of Medicine, Monash University, Melbourne, Australia (J.P., X.W., K.P.)
| | - Karlheinz Peter
- From the Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia (J.P., X.W., K.P.).,Department of Medicine, Monash University, Melbourne, Australia (J.P., X.W., K.P.).,Department of Cardiology, Alfred Hospital, Melbourne, Australia (J.P., K.P.)
| |
Collapse
|
46
|
Wang M, Hao H, Leeper NJ, Zhu L. Thrombotic Regulation From the Endothelial Cell Perspectives. Arterioscler Thromb Vasc Biol 2019; 38:e90-e95. [PMID: 29793992 DOI: 10.1161/atvbaha.118.310367] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Miao Wang
- From the State Key Laboratory of Cardiovascular Disease (M.W., H.H., L.Z.) .,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| | - Huifeng Hao
- From the State Key Laboratory of Cardiovascular Disease (M.W., H.H., L.Z.)
| | | | - Liyuan Zhu
- From the State Key Laboratory of Cardiovascular Disease (M.W., H.H., L.Z.)
| | | |
Collapse
|
47
|
Andreadou I, Cabrera-Fuentes HA, Devaux Y, Frangogiannis NG, Frantz S, Guzik T, Liehn EA, Gomes CPC, Schulz R, Hausenloy DJ. Immune cells as targets for cardioprotection: new players and novel therapeutic opportunities. Cardiovasc Res 2019; 115:1117-1130. [PMID: 30825305 PMCID: PMC6529904 DOI: 10.1093/cvr/cvz050] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 02/24/2019] [Indexed: 12/22/2022] Open
Abstract
New therapies are required to reduce myocardial infarct (MI) size and prevent the onset of heart failure in patients presenting with acute myocardial infarction (AMI), one of the leading causes of death and disability globally. In this regard, the immune cell response to AMI, which comprises an initial pro-inflammatory reaction followed by an anti-inflammatory phase, contributes to final MI size and post-AMI remodelling [changes in left ventricular (LV) size and function]. The transition between these two phases is critical in this regard, with a persistent and severe pro-inflammatory reaction leading to adverse LV remodelling and increased propensity for developing heart failure. In this review article, we provide an overview of the immune cells involved in orchestrating the complex and dynamic inflammatory response to AMI-these include neutrophils, monocytes/macrophages, and emerging players such as dendritic cells, lymphocytes, pericardial lymphoid cells, endothelial cells, and cardiac fibroblasts. We discuss potential reasons for past failures of anti-inflammatory cardioprotective therapies, and highlight new treatment targets for modulating the immune cell response to AMI, as a potential therapeutic strategy to improve clinical outcomes in AMI patients. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, Athens, Greece
| | - Hector A Cabrera-Fuentes
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Institute of Biochemistry, Medical School, Justus-Liebig University, Ludwigstrasse 23, Giessen, Germany
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Av. Eugenio Garza Sada 2501 Sur, Nuevo Leon, Mexico
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Kremlyovskaya St, 18, Kazan, Respublika Tatarstan, Russia
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Thomas Edison, Strassen, Luxembourg
| | - Nikolaos G Frangogiannis
- Wilf Family Cardiovascular Research Institute Department of Medicine (Cardiology) Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B Bronx NY USA
| | - Stefan Frantz
- Department of Internal Medicine I, University Hospital Würzburg, Oberdürrbacher Str. 6, Würzburg, Germany
| | - Tomasz Guzik
- Department of Internal and Agricultural Medicine, Jagiellonian University Medical College, Świętej Anny 12, Kraków, Poland
- Institute of Cardiovascular and Medical Sciences, University ofGlasgow, University Avenue, Glasgow, UK
| | - Elisa A Liehn
- Institute for Molecular Cardiovascular Research, Rheinisch Westfälische Technische Hochschule Aachen University,Templergraben 55, Aachen, Germany
- Human Genomics Laboratory, University of Medicine and Pharmacy Craiova, Strada Petru Rareș 2, Craiova, Romania
- Department of Cardiology, Pulmonology, Angiology and Intensive Care, University Hospital, Rheinisch Westfälische Technische Hochschule,Templergraben 55, Aachen, Germany
| | - Clarissa P C Gomes
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, 1A-B rue Thomas Edison, Strassen, Luxembourg
| | - Rainer Schulz
- Physiologisches Institut Fachbereich Medizin der Justus-Liebig-Universität, Aulweg 129, Giessen, Germany
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Av. Eugenio Garza Sada 2501 Sur, Nuevo Leon, Mexico
- Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, Singapore
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, Maple House 1st floor, 149 Tottenham Court Road, London, UK
| |
Collapse
|
48
|
Huțanu A, Iancu M, Maier S, Bălaşa R, Dobreanu M. Plasma Biomarkers as Potential Predictors of Functional Dependence in Daily Life Activities after Ischemic Stroke: A Single Center Study. Ann Indian Acad Neurol 2019; 23:496-503. [PMID: 33223667 PMCID: PMC7657279 DOI: 10.4103/aian.aian_74_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 02/26/2019] [Accepted: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
Objective: Despite advances made in the treatment of ischemic stroke, it still remains one of the leading causes of mortality and disability worldwide. The main objective of this study was to identify from a panel of 10 inflammatory markers and chemokines those biomarkers that have a potential predictive role in the evolution of disability and functional dependence in daily activities after an ischemic stroke. Methods: The study included 116 patients with ischemic stroke and 40 healthy volunteers matched for gender and age. Stroke severity was assessed by the National Institute of Health Stroke Scale (NIHSS) on admission and during hospitalization and functional mobility in daily activities by Barthel index (BI). Multiplex panel with 10 biomarkers [brain-derived neurotrophic factor (BDNF), platelet-derived growth factor (PDGF)-AA, PDGF-AB/BB, neural cell adhesion molecule (NCAM), cathepsin D, soluble vascular cell adhesion molecule (sVCAM), soluble intercellular cell adhesion molecule (sICAM), myeloperoxidase (MPO), regulated on activation, normal T cell expressed and secreted (RANTES), plasminogen activator inhibitor (PAI)-1] was analyzed on days 1 and 5 after admission using the xMAP technology. Results: Plasma concentrations of RANTES and NCAM were significantly lower in patients with ischemic stroke compared with healthy controls, while MPO and sICAM were significantly higher in patients versus controls. Plasma concentrations of sICAM, sVCAM, and RANTES significantly decreased during the analyzed period. For the first-day measurement, the bivariate analysis revealed the association of NIHSS on admission with sVCAM, and on discharge negative association with PDGF-AA, PDGR-AB/BB, BDNF, and RANTES. Plasma levels of PDGF-AA, PDGF-AB/BB, BDNF, and RANTES were found to be significantly lower in patients with BI ≤ 80, on day 5 after disease onset. PDGF-AA, PDGF-AB/BB, and BDNF were univariate and multivariate predictors for functional dependence in daily life activity (BI ≤ 80), having a protective effect (odds ratio < 1). Conclusion: Plasma levels of BDNF, PDGF-AA, and PDGF-AB/BB are independent predictors for functional dependency in daily life activities and may be useful prognostic markers in the evaluation of ischemic stroke patients.
Collapse
Affiliation(s)
- Adina Huțanu
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Sciences and Technology Tîrgu-Mureş, Romania.,Department of Laboratory Medicine, University of Medicine, Pharmacy, Sciences and Technology Tîrgu-Mureş, Romania
| | - Mihaela Iancu
- University of Medicine and Pharmacy "Iuliu Hațieganu", Department of Medical Informatics and Biostatistics, Cluj-Napoca, Romania
| | - Smaranda Maier
- Department of Neurology, Clinic, Emergency County Hospital Targu Mures, University of Medicine, Pharmacy, Sciences and Technology Tîrgu Mureş
| | - Rodica Bălaşa
- Department of Neurology, Clinic, Emergency County Hospital Targu Mures, University of Medicine, Pharmacy, Sciences and Technology Tîrgu Mureş
| | - Minodora Dobreanu
- Center for Advanced Medical and Pharmaceutical Research, University of Medicine, Pharmacy, Sciences and Technology Tîrgu-Mureş, Romania.,Department of Laboratory Medicine, University of Medicine, Pharmacy, Sciences and Technology Tîrgu-Mureş, Romania
| |
Collapse
|
49
|
Puhl SL, Steffens S. Neutrophils in Post-myocardial Infarction Inflammation: Damage vs. Resolution? Front Cardiovasc Med 2019; 6:25. [PMID: 30937305 PMCID: PMC6431642 DOI: 10.3389/fcvm.2019.00025] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 02/26/2019] [Indexed: 01/08/2023] Open
Abstract
Inflammation not only plays a crucial role in acute ischemic cardiac injury, but also contributes to post-infarction repair and remodeling. Traditionally, neutrophils have been merely considered as detrimental in the setting of an acute myocardial infarction. However, recently published studies demonstrated that neutrophils might also play an important role in cardiac repair by regulating reparative processes. An emerging concept is that different neutrophil subsets exist, which might exhibit separate functional properties. In support of the existence of distinct neutrophil subsets in the ischemic heart, transcriptional changes in cardiac neutrophils have been reported within the first few days after myocardial infarction. In addition, there is an increasing awareness of sex-specific differences in many physiological and pathophysiological responses, including cardiovascular parameters and inflammation. Of particular interest in this context are recent experimental data dissecting sex-specific differences in neutrophil signaling after myocardial infarction. Unraveling the distinct and possibly stage-dependent properties of neutrophils in cardiac repair may provide new therapeutic strategies in order to improve the clinical outcome for myocardial infarction patients. This review will briefly discuss recent advances in our understanding of the neutrophil functional repertoire and emerging insights of sex-specific differences in post-myocardial infarction inflammatory responses.
Collapse
Affiliation(s)
- Sarah-Lena Puhl
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University (LMU), Munich, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
50
|
Fay WP, Korthuis RJ. No Sweetie Pie: Newly Uncovered Role for PAI (Plasminogen Activator Inhibitor)-1 in Inflammatory Responses to Ischemia/Reperfusion. Arterioscler Thromb Vasc Biol 2018; 38:695-697. [PMID: 29563114 DOI: 10.1161/atvbaha.118.310824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- William P Fay
- From the Departments of Medical Pharmacology and Physiology and Medicine, University of Missouri School of Medicine, Columbia (W.P.F., R.J.K.); Dalton Cardiovascular Research Center, Columbia, MO (W.P.F., R.J.K.); and Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (W.P.F.).
| | - Ronald J Korthuis
- From the Departments of Medical Pharmacology and Physiology and Medicine, University of Missouri School of Medicine, Columbia (W.P.F., R.J.K.); Dalton Cardiovascular Research Center, Columbia, MO (W.P.F., R.J.K.); and Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO (W.P.F.)
| |
Collapse
|