1
|
Chen X, Wu C, Tang F, Zhou J, Mo L, Li Y, He J. The Immune Microenvironment: New Therapeutic Implications in Organ Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05067. [PMID: 40391706 DOI: 10.1002/advs.202505067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/28/2025] [Indexed: 05/22/2025]
Abstract
Fibrosis, characterized by abnormal deposition of structural proteins, is a major cause of tissue dysfunction in chronic diseases. The disease burden associated with progressive fibrosis is substantial, and currently approved drugs are unable to effectively reverse it. Immune cells are increasingly recognized as crucial regulators in the pathological process of fibrosis by releasing effector molecules, such as cytokines, chemokines, extracellular vesicles, metabolites, proteases, or intercellular contact. Therefore, targeting the immune microenvironment can be a potential strategy for fibrosis reduction and reversion. This review summarizes the recent advances in the understanding of the immune microenvironment in fibrosis including phenotypic and functional transformations of immune cells and the interaction of immune cells with other cells. The novel opportunities for the discovery and development of drugs for immune microenvironment remodeling and their associated challenges are also discussed.
Collapse
Affiliation(s)
- Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Wu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Tang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyue Zhou
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
2
|
Xiang J, Cui M. Neutrophil Extracellular Traps and neutrophilic asthma. Respir Med 2025:108150. [PMID: 40368066 DOI: 10.1016/j.rmed.2025.108150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/18/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025]
Abstract
There are more than 260 million asthma patients worldwide. How to provide targeted long-term standardized treatment and management still confuses clinical workers and patients. Neutrophilic asthma is a special type of asthma which is difficult to diagnose clinically and often associated with severe asthma and glucocorticoid resistance. Neutrophil Extracellular Traps (NETs) play an important role in the pathogenesis of this type of asthma particularly in children. This article explores the mechanism of NETs production, their association with neutrophilic asthma, biomarkers, and possible treatment options. A more detailed discussion is also provided on the diagnosis and treatment of children with neutrophilic asthma. Educational Aims The readers will gain an improved understanding of.
Collapse
Affiliation(s)
- Jiayi Xiang
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China.
| | - Muyan Cui
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
3
|
Deng ZQ, Si XC, Song JB, Li JY, Sun L, Dang X, Zhao M, Feng YC, Liu FX. Behavioral manifestations and underlying mechanisms of amphetamine in constructing animal models of mania: a comprehensive review. Front Neurosci 2025; 19:1544311. [PMID: 40415893 PMCID: PMC12098516 DOI: 10.3389/fnins.2025.1544311] [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: 12/20/2024] [Accepted: 04/15/2025] [Indexed: 05/27/2025] Open
Abstract
Mania is a mind disorder with heightened emotions, etc. Amphetamine (AMPH), a drug with central nervous system excitatory effects, can disrupt neurotransmitter release and metabolism, causing mania. AMPH-induced animal models of mania show increased risk and reward-seeking behaviors and excessive locomotion like mania patients, verifiable by tests like Elevated Plus Maze (EPM). It also impacts neurotransmitter balance in different brain regions, aligning with the imbalance in mania patients. Multiple signaling pathways including extracellular regulated protein kinases and others are involved, and their altered activities link to mania symptoms. In the AMPH-induced mania model, regions like the frontal cortex have increased oxidative stress and inflammatory response. Moreover, AMPH changes neurotrophin levels, potentially causing neuronal damage and cognitive impairment. In summary, the AMPH-induced mania animal model is crucial for studying mania's pathogenesis. However, further in-depth studies on neurotransmitter regulation, signaling pathway intervention, and neurotrophic factors are needed to develop more effective and personalized treatment plans.
Collapse
Affiliation(s)
- Zi-Qi Deng
- Department of Neuropsychiatric Psychology, Hospital of Encephalopathy, The First Affiliated Hospital of Traditional Henan University of Chinese Medicine, Zhengzhou, China
- Institute of Management and Science University, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiao-Chen Si
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jia-Bin Song
- College of Acupuncture, Moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jin-Yao Li
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lu Sun
- The First Clinical Medical School, Henan university of Chinese Medicine, Zhengzhou, China
| | - Xue Dang
- Traditional Chinese Medicine (Zhong Jing) School, Henan University of Chinese Medicine, Zhengzhou, China
| | - Min Zhao
- Department of Neuropsychiatric Psychology, Hospital of Encephalopathy, The First Affiliated Hospital of Traditional Henan University of Chinese Medicine, Zhengzhou, China
| | - Yan-Chen Feng
- Department of Neuropsychiatric Psychology, Hospital of Encephalopathy, The First Affiliated Hospital of Traditional Henan University of Chinese Medicine, Zhengzhou, China
- The First Clinical Medical School, Henan university of Chinese Medicine, Zhengzhou, China
| | - Fei-Xiang Liu
- Department of Neuropsychiatric Psychology, Hospital of Encephalopathy, The First Affiliated Hospital of Traditional Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
4
|
Wang L, Pan M, Dong J, He Z, Wang W, Shu J, Wang T, Wang Y. Investigating cigarette smoke-induced airway inflammation and sperm activity impairment in rats based on cilia-associated proteins. 3 Biotech 2025; 15:136. [PMID: 40260407 PMCID: PMC12009257 DOI: 10.1007/s13205-025-04302-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/02/2025] [Indexed: 04/23/2025] Open
Abstract
The aim of this study was to investigate the mechanism of smoking-induced chronic obstructive pulmonary disease (COPD) and its impact on reproductive function in male rats and its relationship with chronic lung inflammation. The study used various methodologies including lung function tests, sperm quality assessment, serum hormone level measurement, and ultrastructural observations of airway cilia and sperm flagella to elucidate the effects of smoking on the reproductive and respiratory systems of rats. The results showed that smoking significantly induced lung damage and reduced sperm quality in rats, and the trend of lung damage and decreased sperm quality became more obvious with the increased duration of smoking. Transmission electron microscopy revealed that smoking exposure led to structural abnormalities of airway cilia and sperm flagella, and exposure after a period of three months showed significant damage to cilia and flagellar structures. Western blot and immunohistochemistry results indicated that the relative expression of NE proteins was significantly higher in the rats of the CS group, whereas the expression of FOXJ1 and SPAG6 proteins was notably lower in these rats after three months of smoking. In summary, smoke causes damage to the respiratory and reproductive systems of male rats, and the mechanism may be related to the destruction of airway cilia and sperm flagellar structures and the down-regulation of the expression of key ciliary proteins by smoke.
Collapse
Affiliation(s)
- Lei Wang
- Department of Physiology and Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012 China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230061 China
| | - Min Pan
- Department of Physiology and Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Jinhui Dong
- Department of Physiology and Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Zengyang He
- Technology Center of China Tobacco Anhui Industrial Co., LTD, Hefei, 230088 China
| | - Wenbin Wang
- Technology Center of China Tobacco Anhui Industrial Co., LTD, Hefei, 230088 China
| | - Junsheng Shu
- Technology Center of China Tobacco Anhui Industrial Co., LTD, Hefei, 230088 China
| | - Tongsheng Wang
- Department of Physiology and Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012 China
| | - Yajuan Wang
- Department of Physiology and Pharmacology, Anhui University of Chinese Medicine, Hefei, 230012 China
| |
Collapse
|
5
|
Aloisi AM, Casini I. Fibromyalgia: Chronic Pain Due to a Blood Dysfunction? Int J Mol Sci 2025; 26:4153. [PMID: 40362392 PMCID: PMC12071621 DOI: 10.3390/ijms26094153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Fibromyalgia (FM) is a common chronic disorder with chronic pain. FM generally affects all ages and occurs more commonly in women. The cause of FM remains undefined, but a number of factors suggest the cardiovascular system and the blood in particular as contributors to its occurrence and maintenance. Hemograms and other blood indexes often show high percentages of values at the 'normal', low, or high limits and several values outside of the 'normal' ranges. On the other hand, vessels regulate blood arrival to tissues depending on many internal and external factors. Both aspects can interfere with tissue oxygenation and then with the numerous consequences induced by hypoxia. In this narrative review, efforts were made to highlight factors that are potentially able to affect oxygen arrival in cells, as well as other factors related to blood elements that can play a role in the chronic pain experienced by FM patients. Data strongly indicate that most of the symptoms commonly present in FM patients can find their physio-pathological basis in the blood, suggesting blood-related interventions in these patients.
Collapse
Affiliation(s)
- Anna Maria Aloisi
- Stress and Pain Neurophysiology Laboratory, Department of Medicine, Surgery and Neuroscience University of Siena, 53100 Siena, Italy;
| | | |
Collapse
|
6
|
Song Y, Zeng K, Zhang LK, Zhang JN, Zhang KL, Xin Y, Wang XR, Zhou YX, Li HX, Wang CS, Yu KJ. Urine Neutrophil Elastase: A Novel Predictor of ICU Admission for Patients with COVID-19 Infection. J Inflamm Res 2025; 18:5545-5553. [PMID: 40297545 PMCID: PMC12036589 DOI: 10.2147/jir.s503276] [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: 10/28/2024] [Accepted: 04/05/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction We aimed to explore the differences of neutrophil elastase (NE) levels between intensive care unit (ICU) and non-ICU patients with COVID-19 infection, as well as its predictive value for COVID-19 progression. Methods We enrolled the patients admitted with a primary diagnosis of COVID-19. All patients in ICU were diagnosed with the critical type upon admission. Blood was taken within 24 hours, followed by examination of the blood NE level and urine NE level. Other clinical features were recorded. A logistic regression model was used to predict ICU admission. Results A total of 83 patients were diagnosed, including 52 non-ICU cases and 31 ICU cases. The ICU group showed significantly elevated levels of Neutrophil%, Cr, D-dimer (DD), Procalcitonin (PCT), and C-reactive protein (CRP). Meanwhile, the CD3-cell, T4-cell, and Lymphocyte% levels were lower in the ICU group. Notably, the blood NE levels were similar between groups, whereas the urine NE level was highly significantly higher in the ICU group vs the non-ICU group. After dimension reduction, we constructed a logistic model (UD) using only two factors: the urine NE level and the blood DD level. The overall accuracy of was 86.1%. The urine NE has a strong efficacy in ICU prediction (AUC = 0.893), and the performance of the UD model was even better (AUC = 0.933). Conclusion Urine NE level is a useful predictor of COVID-19 progression, particularly in patients requiring ICU care. Urine NE has a significantly positive correlation with neutrophil%, DD, and PCT, as well as a negative correlation with lymphocyte levels.
Collapse
Affiliation(s)
- Yu Song
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Kai Zeng
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Li-Kun Zhang
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jian-Nan Zhang
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Kai-Li Zhang
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yu Xin
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xin-Ran Wang
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yu-Xin Zhou
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Hong-Xu Li
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Chang-Song Wang
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Kai-Jiang Yu
- Key Laboratory of Critical Care Medicine of Heilongjiang Province; Department of Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
7
|
Huckriede J, Keulen GM, van de Poll MCG, Wichapong K, Reutelingsperger CPM, Nicolaes GAF. Proteolytic neutralization of extracellular histones by neutrophil elastase is enhanced by heparin. J Thromb Haemost 2025:S1538-7836(25)00239-9. [PMID: 40221045 DOI: 10.1016/j.jtha.2025.03.039] [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: 01/14/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Release of extracellular histones during cell death or neutrophil extracellular trap formation is linked to initiation and progression of several acute inflammatory diseases. Presence and proteolysis status of extracellular histones are associated with disease severity and the risk of thromboembolic events. Targeting extracellular histones by proteolysis or complexation constitutes a potential therapeutic option. Neutrophil elastase (NE), released during neutrophil activation or neutrophil extracellular trap formation, is associated with extracellular histone plasma levels in severe intensive care unit COVID-19 patients and other acute inflammatory diseases. OBJECTIVES We aimed to investigate the ability of NE to proteolyze extracellular histones, characterize the effect of proteolyzed histones on inflammation and cytotoxicity, and explore the influence of heparin on proteolysis. METHODS Human-activated NE in the presence or absence of heparin was used to proteolyze extracellular histones. Reaction mixtures were analyzed by Western blot analysis, mass spectrometry, and in vitro cell-based systems to assess TLR4 activation with human embryonic kidney (HEK)-blue cells and cell cytotoxicity with human vascular endothelial (EA.hy926) cells. RESULTS All classes of extracellular histones could be cleaved by NE, which resulted in multiple histone fragments. Histone H3 and H4 proteolysis significantly reduced their cytotoxic potential but did not reduce TLR4 activation. Addition of heparin significantly increased the rate of proteolysis of histones by NE and significantly reduced histone-mediated cytotoxicity. CONCLUSION NE can proteolyze extracellular histones, resulting in reduced cytotoxicity and inflammation in vitro. Histone proteolysis can be enhanced by the addition of heparins, indicating a potential combinatory treatment of histone-mediated events by heparins and NE in acute inflammatory diseases.
Collapse
Affiliation(s)
- Joram Huckriede
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Gwen M Keulen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Marcel C G van de Poll
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Centre, School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht, the Netherlands
| | - Kanin Wichapong
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Chris P M Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
8
|
Chukwuanukwu RC, Agu CE, Ehiaghe A, Ezeagwuna D, Herrmann M, Udigwe G. Markers of neutrophil activation and some immune and haematological indices in malaria infection during pregnancy. BMC Immunol 2025; 26:28. [PMID: 40200152 PMCID: PMC11978171 DOI: 10.1186/s12865-025-00709-4] [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: 09/26/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Neutrophils are the first responders to pathogen invasion and are important first-line defenders. The defence mechanism of activated neutrophils includes neutrophil extracellular traps (NETs) formation that immobilize pathogens, stop their spread within the tissues, and ultimately kill them. However, their roles in the context of malaria during pregnancy are still elusive. This study was conducted to investigate markers of neutrophil activation as well as immunological and haematological cellular responses during Plasmodium infection in pregnancy. METHOD A total of 340 pregnant women aged between 19 and 42 years were recruited for this study carried out in South-east, Nigeria. All the subjects were tested for malaria parasite (MP) status. Those infected with human immunodeficiency virus (HIV) and those with any other co-morbidity were excluded from the study. A total of 45 (13.2%) of the 340 pregnant women were positive for malaria. To assess immune, haematologic and NETs markers in the MP positive group, 45 matched malaria-negative pregnant women from the malaria negative group served as controls. Thus, the final study population was grouped into two categories: 45 pregnant women infected with Plasmodium falciparum and 45 pregnant malaria-negative control group. The neutrophil elastase concentration, myeloperoxidase activity, total white blood cell counts, white blood cell differential counts, platelet counts and haematocrit were assessed via standard laboratory methods. RESULTS Findings from this study revealed lower levels of myeloperoxidase in the malaria- infected cohort (p = 0.013) than in the malaria negative cohort. The neutrophil elastase levels were also lower in the malaria negative group (p = 0.042). The total white blood cells, platelet and neutrophil counts were lower (p = 0.046, 0.012 and 0.015, respectively) in the malaria infected group when compared to the controls. Conversely, lymphocyte counts were higher in the malaria-infected group (p = 0.003). No cases with high parasitaemia were encountered among the pregnant women infected with Plasmodium falciparum. CONCLUSION Malaria infection led to alterations in immune and haematological parameters in this group, with mild and moderate malaria parasitaemia in the study cohort. Although there were some significant differences, the assessed values remained mostly within the normal range. Further studies in a larger cohort assessing pregnant women infected with placental malaria and those with fatal outcomes are important to further investigate the role of NETs in malaria infection.
Collapse
Affiliation(s)
- Rebecca Chinyelu Chukwuanukwu
- Immunology Department, Faculty of Medical Laboratory Science, Nnamdi Azikiwe University, Awka, Nigeria.
- Department of Internal Medicine 3, Uniklinikum Erlangen, Erlangen and Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Chioma Esther Agu
- Immunology Department, Faculty of Medical Laboratory Science, Nnamdi Azikiwe University, Awka, Nigeria
| | - Alfred Ehiaghe
- Immunology Department, Faculty of Medical Laboratory Science, Nnamdi Azikiwe University, Awka, Nigeria
| | - Dorothy Ezeagwuna
- Department of Parasitology & Entomology, Faculty of Bioscience, Nnamdi Azikiwe University, Awka, Nigeria
| | - Martin Herrmann
- Department of Internal Medicine 3, Uniklinikum Erlangen, Erlangen and Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Pediatric Surgery, University Medical Center Mannheim, Heidelberg, 68167, Germany
| | - Gerald Udigwe
- Department of Obstetrics & Gynaecology, Faculty of Medicine, Nnamdi Azikiwe University, Awka, Nigeria
| |
Collapse
|
9
|
Górecka A, Komosinska-Vassev K. Neutrophil Elastase and Elafin in Inflammatory Bowel Diseases: Urinary Biomarkers Reflecting Intestinal Barrier Dysfunction and Proteolytic Activity. J Clin Med 2025; 14:2466. [PMID: 40217915 PMCID: PMC11989340 DOI: 10.3390/jcm14072466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), is a chronic inflammatory disorder driven by a complex interplay of immune and proteolytic mechanisms. Neutrophil elastase (NE), released at sites of inflammation, plays a central role by promoting inflammation, degrading the extracellular matrix (ECM), and disturbing intestinal barrier integrity via NF-κB activation and E-cadherin degradation. Elafin, an endogenous NE inhibitor, mitigates proteolytic damage, reinforces the intestinal barrier, and exerts anti-inflammatory effects by suppressing NF-κB and reducing pro-inflammatory cytokines. Since the NE/elafin balance is critical in IBD, assessing their ratio may provide a more precise measure of proteolytic dysregulation. This study aimed to evaluate the diagnostic and prognostic utility of urinary NE, elafin, and their ratio in IBD patients. Methods: Urinary concentrations of NE and elafin were measured by immunoassay in 88 subjects including ulcerative colitis and Crohn's disease patients and healthy individuals. The diagnostic accuracy of these biomarkers was assessed using receiver operating characteristic (ROC) curve analysis. Results: Urinary NE levels were significantly elevated in both UC and CD patients compared to controls, with a 17-fold increase in the UC patients and a 28-fold increase in the CD patients (p < 0.0001). Elafin levels were also increased in IBD patients. The NE/elafin ratio was significantly increased in both disease groups, with a 4.5-fold increase in the UC and 5.6-fold increase in the CD patients compared to healthy controls. The ROC curve analysis demonstrated that the NE/elafin ratio is the most effective biomarker for distinguishing CD patients from healthy individuals (AUC = 0.896), with a high sensitivity (92.9%) and specificity (69.7%), making it a strong diagnostic tool. NE also showed an excellent diagnostic performance both in CD (AUC = 0.842) and UC (AUC = 0.880). The elafin urinary profile had a high diagnostic value, with a better accuracy in the UC patients (AUC = 0.772) than the CD patients (AUC = 0.674), though it was inferior to NE and NE/elafin. Conclusions: Our findings indicate that urinary NE, elafin, the and NE/elafin ratio have significant diagnostic value in differentiating IBD patients from healthy controls. The NE/elafin ratio and NE proved to be the most reliable urinary biomarkers in both CD and UC diagnosis, with a high predictive value and strong discriminatory power.
Collapse
Affiliation(s)
- Aleksandra Górecka
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland;
| | | |
Collapse
|
10
|
Ma X, Zhao X, Yang Y, Yan J, Shi X, Wu H, Liu Y, Dai M. Paeonol inhibits NETs-mediated foam cell inflammation through the CitH3/NLRP3/caspase-1 signaling pathway in atherosclerosis. Int Immunopharmacol 2025; 151:114340. [PMID: 40020464 DOI: 10.1016/j.intimp.2025.114340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by lipid streaks, which are produced by aggregates of lipid-rich foam cells. Foam cells intensify atherosclerosis by secreting a range of inflammatory mediators. Neutrophil extracellular traps produced by activated neutrophils, which are abundantly present in lipid-accumulating plaques. However, the relationship between neutrophil extracellular traps and foam cells inflammation is still unclear. Paeonol is well known for its anti-inflammatory effects in atherosclerosis. Nevertheless, the exact pharmacological mechanisms by which paeonol affects atherosclerosis are not fully understood which require further investigation. The purpose of this study is to investigate the effects of paeonol on the neutrophil extracellular traps' formation and foam cell inflammation caused by neutrophil extracellular traps, and to explore the potential mechanisms. A high-fat diet was administered to ApoE-/- mice for a period of 12 weeks to induce an atherosclerosis model. Our findings demonstrated that paeonol notably suppressed the advancement of atherosclerosis in ApoE-/- mice, curtailed the formation of neutrophil extracellular traps, and lowered inflammatory factor levels within the plaque. In vitro studies have shown that neutrophil extracellular traps could enhance the inflammation in foam cells. CitH3 played a role in the cellular communication between neutrophil extracellular traps and foam cells. Concurrently, NLRP3 acted as a key receptor in the inflammation mediated by this interaction. Paeonol is capable of regulating NE, thereby affecting the formation of neutrophil extracellular traps. Most notably, the foam cell inflammation caused by neutrophil extracellular traps was significantly mitigated by the inclusion of paeonol. Our findings suggested that paeonol inhibited foam cell inflammation which induced by neutrophil extracellular traps through the CitH3/NLRP3/caspase-1 signaling pathway, shedding new lights on its anti-atherosclerotic pharmacological mechanism.
Collapse
Affiliation(s)
- Xiaolin Ma
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xuan Zhao
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yulong Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jinjin Yan
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Xiaoyan Shi
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, 230012, China
| | - Hongfei Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, 230012, China
| | - Yarong Liu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, 230012, China.
| | - Min Dai
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China; Anhui Key Laboratory for Research and Development of Traditional Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
11
|
Schneider CV, Decraecker M, Beaufrère A, Payancé A, Coilly A, Schneider KM, Bioulac P, Blanc JF, Le Bail B, Amintas S, Bouchecareilh M. Alpha-1 antitrypsin deficiency and primary liver cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189290. [PMID: 39999944 DOI: 10.1016/j.bbcan.2025.189290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/31/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Primary liver cancers (PLCs) remain a major challenge to global health and an escalating threat to human life, with a growing incidence worldwide. PLCs are composed of hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and mixed HCC-CCA, accounting for 85 %, 10 %, and 5 % of cases, respectively. Among the numerous identified risk factors associated with liver cancers, Alpha 1-AntiTrypsin Deficiency (AATD) genetic disease emerges as an intriguing one. AATD-related liver disease may lead to chronic hepatitis, cirrhosis, and PLCs in adulthood. Although our knowledge about the natural history of AATD-liver disease has improved recently, liver cancers associated with AATD remain poorly understood and explored, while this specific population is at a 20 to 50 times higher risk of developing PLC. Thus, we review here current knowledge about AATD-associated PLCs, describing the impact of AATD genotypes on their occurrence. We also discuss emerging hypotheses regarding the AATD PiZZ genotype-related hepatic carcinogenesis process. Finally, we perform an updated analysis of the United Kingdom Biobank database that highlights and confirms AATD PiZZ genotype as an important HCC risk factor.
Collapse
Affiliation(s)
- Carolin Victoria Schneider
- Department of Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Marie Decraecker
- University of Bordeaux, CNRS, INSERM, BRIC, U1312 Bordeaux, France; Oncology Unit, Hôpital Haut Lévêque, CIC 1401, Bordeaux University Hospital, 33604 Pessac, France
| | - Aurélie Beaufrère
- AP-HP Nord, Department of Pathology, FHU MOSAIC, SIRIC InsiTu, DMU DREAM, Université Paris Cité, Beaujon Hospital, Clichy, France
| | - Audrey Payancé
- AP-HP, Hôpital Beaujon, Service d'Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Clichy, France
| | - Audrey Coilly
- Centre Hépato-Biliaire, Hôpital Paul Brousse, UMR-1193, APHP, Université Paris Saclay, Villejuif, France
| | - Kai Markus Schneider
- Departement of Medicine I, Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany; Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Paulette Bioulac
- University of Bordeaux, CNRS, INSERM, BRIC, U1312 Bordeaux, France
| | - Jean-Frédéric Blanc
- Oncology Unit, Hôpital Haut Lévêque, CIC 1401, Bordeaux University Hospital, 33604 Pessac, France
| | - Brigitte Le Bail
- University of Bordeaux, CNRS, INSERM, BRIC, U1312 Bordeaux, France; Pathology Department, Pellegrin University Hospital, CHU Bordeaux, France; French National and Bordeaux Local Liver Tumor Bank, France
| | - Samuel Amintas
- University of Bordeaux, CNRS, INSERM, BRIC, U1312 Bordeaux, France; Tumor Biology and Tumor Bank Laboratory, CHU Bordeaux, Pessac, France.
| | | |
Collapse
|
12
|
Bertolini A, Picone F, Ferrara I, Della Corte AM, Serio B, Gorrese M, Campana A, Simeon V, Luponio S, Marcucci R, Scala P, Langella M, Selleri C, Giudice V. Immature forms of low density granulocytes are increased in acute myeloid leukemia and myelodysplastic syndromes. Sci Rep 2025; 15:8661. [PMID: 40082470 PMCID: PMC11906629 DOI: 10.1038/s41598-025-92513-8] [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: 05/29/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
Neutrophils can promote or suppress tumor growth. These different immunological functions mirror a great heterogenicity of neutrophil maturation and activation status: low-density granulocytes (LDGs) and normal-density neutrophils (NDNs). LDGs participate in immune dysregulation during autoimmune disorders with an activated phenotype, while NDNs might exert immunosuppressive activities. Here, we investigated variations in distribution of LDGs and NDNs in benign and malignant hematological conditions using an optimized 10-color flow cytometry staining for immunophenotyping of the main circulating populations. A total of 102 consecutive subjects diagnosed with hematological malignancies was enrolled for immunophenotyping by flow cytometry. We showed impaired neutrophil subset distribution in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) patients compared to healthy individuals, with intermediate and mature LDGs significantly reduced, also displaying a good diagnostic sensitivity in MDS (AUC, 0.793 and P = 0.0013; and AUC, 0.7319 and P = 0.0109, respectively) and AML (AUC, 0.9059 and P = 0.0069; and AUC, 0.9176 and P = 0.00057, respectively). In conclusion, LDG and NDN subsets could be altered in AML and MDS, in favor of more immature forms, suggesting that emergency hemopoiesis could be a first mechanism to sustain peripheral blood counts, while maintaining a pro-inflammatory microenvironment.
Collapse
Affiliation(s)
- Angela Bertolini
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| | - Francesca Picone
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| | - Idalucia Ferrara
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Anna Maria Della Corte
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Bianca Serio
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Marisa Gorrese
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Annapaola Campana
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Vittorio Simeon
- Department of Mental Health and Public Medicine, Unit of Medical Statistics, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Serena Luponio
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Rossella Marcucci
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Pasqualina Scala
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Maddalena Langella
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy.
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy.
| | - Valentina Giudice
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy.
- Hematology and Transplant Center, University Hospital "San Giovanni di Dio e Ruggi d'Aragona", Salerno, Italy.
| |
Collapse
|
13
|
Santos AP, Rodrigues LS, Rother N, Mello FCDQ, Magis-Escurra C. The role of neutrophil response in lung damage and post-tuberculosis lung disease: a translational narrative review. Front Immunol 2025; 16:1528074. [PMID: 40124364 PMCID: PMC11925771 DOI: 10.3389/fimmu.2025.1528074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
It is estimated that more than 150 million individuals alive in 2020 had survived tuberculosis (TB). A portion of this large population continues to experience chronic respiratory abnormalities, with or without symptoms, due to previous active pulmonary TB. This condition known as Post-TB Lung Disease (PTLD), involves a complex interaction between pathogen, host and environmental factors. These interactions are believed to drive a hyperinflammatory process in the lungs during active TB, resulting in tissue damage, which may lead to radiological sequelae, impaired pulmonary function, clinical symptoms, such as cough, dyspnea, hemoptysis, and respiratory infections. Such complications impose significant health, financial, and social burdens, which remain poorly understood and inadequately addressed by health care systems. Given the heterogeneity of immune cells and their products infiltrating the airways and the lung parenchyma during acute and chronic inflammation caused by Mycobacterium tuberculosis infection, it is evident that TB immunopathology is multifactorial. Among the various components involved, neutrophils have recently emerged as critical contributors to the deleterious immune response against TB, leading to severe pulmonary damage. In this translational narrative review, we aim to summarize the role of neutrophils and their primary products - proteases (such as elastase), matrix metalloproteinases and neutrophils extracellular traps (NETs) - in pulmonary TB. We highlight new concepts and emerging evidence of neutrophil involvement during the active disease, translating these insights from "bench to bedside" to facilitate dialogue between fundamental researchers and clinical practitioners. Additionally, we present potential targets for future treatment strategies that could mitigate or even prevent PTLD.
Collapse
Affiliation(s)
- Ana Paula Santos
- Pulmonary Diseases Department, Pedro Ernesto University Hospital, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Thoracic Diseases Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Respiratory Diseases-TB Expert Center, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luciana Silva Rodrigues
- Department of Pathology and Laboratories, Medical Sciences Faculty, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Cecile Magis-Escurra
- Department of Respiratory Diseases-TB Expert Center, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
14
|
Šutković J. Neutrophils and COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:347-384. [PMID: 40246349 DOI: 10.1016/bs.pmbts.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Neutrophils are the first line of defense against pathogens, most effectively by forming Neutrophil Extracellular Traps (NETs). Neutrophiles are further classified into several subpopulations during their development, eliminating pathogens through various mechanisms. However, due to the chaotic and uncontrolled immune response, NETs are often severely resulting in tissue damage and lung infections. The uncontrolled and poorly acknowledged host response regarding the cytokine storm is one of the major causes of severe COVID-19 conditions. Specifically, the increased formation of low-density neutrophils (LDNs), together with neutrophil extracellular traps (NETs) is closely linked with the severity and poor prognosis in patients with COVID-19. In this review, we discuss in detail the ontogeny of neutrophils at different stages and their recruitment and activation after infections, focusing on SARS-CoV-2. In addition, this chapter summarized the research progress on potential targeted drugs (NETs and Cytokine inhibitors) for neutrophil medical therapy and hoped to provide reference for the development of related therapeutic drugs for critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Jasmin Šutković
- Department Genetics and Bioegnineering, International University of Sarajevo, Hrasnička cesta, Bosnia & Herzegovina.
| |
Collapse
|
15
|
Rahman M, Scozzi D, Eguchi N, Klein R, Sankpal NV, Sureshbabu A, Fleming T, Hachem R, Smith M, Bremner R, Mohanakumar T. Downregulation of Tumor Suppressor Gene LKB1 During Severe Primary Graft Dysfunction After Human Lung Transplantation: Implication for the Development of Chronic Lung Allograft Dysfunction. Transplantation 2025; 109:476-483. [PMID: 39228019 DOI: 10.1097/tp.0000000000005172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
BACKGROUND Severe primary graft dysfunction (PGD) after lung transplantation (LTx) is a significant risk factor for the development of bronchiolitis obliterans syndrome (BOS). Recent data from our group demonstrated that small extracellular vesicles (sEVs) isolated from the plasma of LTx recipients with BOS have reduced levels of tumor suppressor gene liver kinase B1 ( LKB1 ) and promote epithelial-to-mesenchymal transition (EMT) and fibrosis. Here, we hypothesized that early inflammatory responses associated with severe PGD (PGD2/3) can downregulate LKB1 levels in sEVs, predisposing to the development of chronic lung allograft dysfunction (CLAD). METHODS sEVs were isolated from the plasma of human participants by Exosome Isolation Kit followed by 0.20-µm filtration and characterized by NanoSight and immunoblotting analysis. Lung self-antigens (K alpha 1 tubulin, Collagen V), LKB1 , nuclear factor kappa B, and EMT markers in sEVs were compared by densitometry analysis between PGD2/3 and no-PGD participants. Neutrophil-derived factors and hypoxia/reperfusion effects on LKB1 levels and EMT were analyzed in vitro using quantitative real-time polymerase chain reaction and Western blotting. RESULTS LKB1 was significantly downregulated in PGD2/3 sEVs compared with no-PGD sEVs. Within PGD2/3 participants, lower post-LTx LKB1 was associated with CLAD development. Hypoxia/reperfusion downregulates LKB1 and is associated with markers of EMT in vitro. Finally, lower LKB1 levels in PGD2/3 are associated with increased markers of EMT. CONCLUSIONS Our results suggest that in post-LTx recipients with PGD2/3, downregulation of LKB1 protein levels in sEVs is associated with increased EMT markers and may result in the development of CLAD. Our results also suggest that ischemia/reperfusion injury during LTx may promote CLAD through the early downregulation of LKB1 .
Collapse
Affiliation(s)
- Mohammad Rahman
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Davide Scozzi
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Natsuki Eguchi
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Rachel Klein
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Narendra V Sankpal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Ramsey Hachem
- Washington University School of Medicine, St. Louis, MO
| | - Michael Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Ross Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ
| | | |
Collapse
|
16
|
Sandri A, Boschi F. Exploring Proteases as Alternative Molecular Targets to Tackle Inflammation in Cystic Fibrosis Respiratory Infections. Int J Mol Sci 2025; 26:1871. [PMID: 40076497 PMCID: PMC11899166 DOI: 10.3390/ijms26051871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Cystic fibrosis (CF) is characterized by chronic respiratory infections and excessive inflammation, driven by both host- and pathogen-derived proteases. The dysregulated activity of proteolytic enzymes such as neutrophil elastase (NE), cathepsin G, and matrix metalloproteases (MMPs) degrades lung tissue, exacerbates airway remodeling, and perpetuates inflammatory cycles. Concurrently, bacterial proteases from pathogens such as Pseudomonas aeruginosa and Staphylococcus aureus contribute to immune evasion and tissue destruction, compounding disease severity. Despite advances in antimicrobial and anti-inflammatory therapies, protease-driven lung damage remains a critical challenge. This review examines the dual role of host and bacterial proteases in CF pathophysiology, highlighting emerging protease-targeted therapies aimed at mitigating lung damage and inflammation. Strategies explored include the inhibition of NE, MMPs, and bacterial proteases, with a focus on innovative therapeutic approaches such as dual-function inhibitors, biologics, and advanced drug delivery systems. By restoring the protease-antiprotease balance, these interventions offer the potential to improve clinical outcomes and quality of life for CF patients.
Collapse
Affiliation(s)
- Angela Sandri
- Department of Diagnostics and Public Health, University of Verona, Strada Le Grazie 8-15, 37134 Verona, Italy;
- General and Upper GI Surgery Division, Azienda Ospedaliera Universitaria Integrata Verona, Piazzale Stefani 1, 37126 Verona, Italy
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, Strada Le Grazie 8-15, 37134 Verona, Italy
| |
Collapse
|
17
|
Wilske F, Eriksson O, Amini RM, Estrada S, Janols H, Khalil A, Larsson A, Lipcsey M, Mangsbo S, Sigfridsson J, Sjölin J, Skorup P, Wall A, Wilson V, Castegren M, Antoni G. Repeated positron emission tomography tracing neutrophil elastase in a porcine intensive-care sepsis model. Intensive Care Med Exp 2025; 13:14. [PMID: 39904820 PMCID: PMC11794750 DOI: 10.1186/s40635-025-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Neutrophil granulocytes are important parts of the defence against bacterial infections. Their action is a two-edged sword, the mediators killing the intruding bacteria are at the same time causing tissue damage. Neutrophil activation is part of the dysregulated immune response to infection defining sepsis and neutrophil elastase is one of the powerful proteases causing both effects and damage. Inhibition of neutrophil elastase has been tried in sepsis and ARDS, so far with inconclusive results. METHODS We used positron emission tomography (PET) combined with computed tomography (CT) and the selective and specific neutrophil elastase inhibitor PET-tracer [11C]GW457427 ([11C]NES), in an intensive care unit porcine Escherichia coli sepsis model with the primary aim to visualise the biodistribution of neutrophil elastase in the initial acute phase of the septic reaction. Repeated PET-CT investigations were performed before and after induction of sepsis. RESULTS At baseline [11C]NES uptake was found in the bone marrow, spleen and liver. The uptake in the bone marrow was markedly increased two hours into the sepsis, whereas in spleen and liver the uptake was not as markedly changed compared to baseline. At 4 h after the sepsis induction [11C]NES in the bone marrow decreased while the uptake increased in the spleen, liver and lungs. CONCLUSION The neutrophil elastase PET-tracer [11C]NES is a novel and unique instrument to study the acute innate neutrophil immune response in sepsis and associated vital organ failure. We here present images and quantitative data of the neutrophil elastase distribution the first hours of acute experimental sepsis. Surprisingly, a pronounced increase of neutrophil elastase was found in the bone marrow 2 h into the sepsis reaction followed at 4 h by increase in the liver, spleen and lungs and a concomitant reduction of the tracer uptake in bone marrow.
Collapse
Affiliation(s)
- Frida Wilske
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden.
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Rose-Marie Amini
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sergio Estrada
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Helena Janols
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Amina Khalil
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Miklós Lipcsey
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Sara Mangsbo
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Jonathan Sigfridsson
- Department of Surgical Sciences, Molecular Imaging and Medical Physics, Uppsala University, Uppsala, Sweden
| | - Jan Sjölin
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Paul Skorup
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Anders Wall
- Department of Surgical Sciences, Molecular Imaging and Medical Physics, Uppsala University, Uppsala, Sweden
| | - Viola Wilson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Markus Castegren
- Department of Medical Sciences, Infectious Diseases, Uppsala University, Uppsala, Sweden
- CLINTEC, Karolinska Institutet, Stockholm, Sweden
- Centre for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden
| | - Gunnar Antoni
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Vagstad AL, Lakis E, Csizi KS, Walls W, Richter D, Lee KS, Stocker R, Gugger M, Broderick WE, Broderick JB, Reiher M, Piel J. Mechanistic Insights Into Post-Translational α-Keto-β-Amino Acid Formation by a Radical S-Adenosyl Methionine Peptide Splicease. Angew Chem Int Ed Engl 2025; 64:e202418054. [PMID: 39688170 PMCID: PMC11879070 DOI: 10.1002/anie.202418054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Radical S-adenosyl methionine enzymes catalyze a diverse repertoire of post-translational modifications in protein and peptide substrates. Among these, an exceptional and mechanistically obscure example is the installation of α-keto-β-amino acid residues by formal excision of a tyrosine-derived tyramine unit. The responsible spliceases are key maturases in a widespread family of natural products termed spliceotides that comprise potent protease inhibitors, with the installed β-residues being crucial for bioactivity. Here, we established the in vitro activity of the model splicease PcpXY to interrogate the mechanism of non-canonical protein splicing. Identification of shunt and coproducts, deuterium labeling studies, and density functional theory energy calculations of hypothesized intermediates support a mechanism involving hydrogen abstraction at tyrosine Cα as the initial site of peptide radical formation and release of 4-hydroxybenzaldehyde as the tyrosine-derived coproduct. The data illuminate key features of this unprecedented radical-mediated biotransformation yielding ketoamide pharmacophores that are also present in peptidomimetic therapeutics.
Collapse
Affiliation(s)
- Anna L. Vagstad
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| | - Edgars Lakis
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| | - Katja-Sophia Csizi
- Institute of Molecular Physical Science, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| | - William Walls
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana, 59717, United States
| | - Daniel Richter
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| | - Kang Soo Lee
- Department of Civil, Environmental and Geomatic Engineering, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, South Korea
| | - Roman Stocker
- Department of Civil, Environmental and Geomatic Engineering, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| | - Muriel Gugger
- Institut Pasteur, Université Paris Cité, Collection of Cyanobacteria, F-75015 Paris, France
| | - William E. Broderick
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana, 59717, United States
| | - Joan B. Broderick
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, Montana, 59717, United States
| | - Markus Reiher
- Institute of Molecular Physical Science, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zurich, Switzerland
| |
Collapse
|
19
|
Brannon ER, Piegols LD, Cady G, Kupor D, Chu X, Guevara MV, Lima MR, Kanthi Y, Pinsky DJ, Uhrich KE, Eniola‐Adefeso O. Polymerized Salicylic Acid Microparticles Reduce the Progression and Formation of Human Neutrophil Extracellular Traps (NET)s. Adv Healthc Mater 2025; 14:e2400443. [PMID: 38898728 PMCID: PMC11628640 DOI: 10.1002/adhm.202400443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/02/2024] [Indexed: 06/21/2024]
Abstract
Neutrophils can contribute to inflammatory disease propagation via innate mechanisms intended for inflammation resolution. For example, neutrophil extracellular traps (NETs) are necessary for trapping pathogens but can contribute to clot formation and blood flow restriction, that is, ischemia. Currently, no therapeutics in the clinic directly target NETs despite the known involvement of NETs contributing to mortality and increased disease severity. Vascular-deployed particle-based therapeutics are a novel and robust alternative to traditional small-molecule drugs by enhancing drug delivery to cells of interest. This work designs a high-throughput assay to investigate the immunomodulatory behavior and functionality of salicylic acid-based polymer-based particle therapeutics against NETosis in human neutrophils. Briefly, this work finds that polymeric composition plays a role, and particle size can also influence rates of NETosis. Salicylate-based polymeric (Poly-SA) particles are found to functionally inhibit NETosis depending on the particle size and concentration exposed to neutrophils. This work demonstrates the high throughput method can help fast-track particle-based therapeutic optimization and design, more efficiently preparing this innovative therapeutics for the clinic.
Collapse
Affiliation(s)
- Emma R. Brannon
- Department of Chemical EngineeringUniversity of Michigan2800 Plymouth Road, NCRC B28Ann ArborMI48109USA
| | - Logan D. Piegols
- Department of Chemical EngineeringUniversity of Michigan2800 Plymouth Road, NCRC B28Ann ArborMI48109USA
| | - Gillian Cady
- Division of Cardiovascular MedicineSamuel and Jean Frankel Cardiovascular CenterUniversity of MichiganAnn ArborMI48109USA
| | - Daniel Kupor
- Department of Chemical EngineeringUniversity of Michigan2800 Plymouth Road, NCRC B28Ann ArborMI48109USA
| | - Xueqi Chu
- Department of Chemical EngineeringUniversity of Michigan2800 Plymouth Road, NCRC B28Ann ArborMI48109USA
| | - M. Valentina Guevara
- Department of Chemical EngineeringUniversity of Michigan2800 Plymouth Road, NCRC B28Ann ArborMI48109USA
| | - Mariana R.N. Lima
- Department of ChemistryUniversity of California RiversideRiversideCA92521USA
| | - Yogendra Kanthi
- Division of Cardiovascular MedicineSamuel and Jean Frankel Cardiovascular CenterUniversity of MichiganAnn ArborMI48109USA
- Section of Vascular Thrombosis & InflammationDivision of Intramural ResearchNational HeartLungand Blood InstituteBethesdaMD20892USA
| | - David J. Pinsky
- Division of Cardiovascular MedicineSamuel and Jean Frankel Cardiovascular CenterUniversity of MichiganAnn ArborMI48109USA
| | - Kathryn E. Uhrich
- Department of ChemistryUniversity of California RiversideRiversideCA92521USA
| | - Omolola Eniola‐Adefeso
- Department of Chemical EngineeringUniversity of Michigan2800 Plymouth Road, NCRC B28Ann ArborMI48109USA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
20
|
Wu L, Cen C, Xie B, Hu L, Huang J, Shen N, Dong Q. Cross-sectional study of proteomic differences between moderate and severe psoriasis. Sci Rep 2025; 15:3387. [PMID: 39870771 PMCID: PMC11772871 DOI: 10.1038/s41598-025-87252-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 01/17/2025] [Indexed: 01/29/2025] Open
Abstract
Although an ongoing understanding of psoriasis vulgaris (PV) pathogenesis, little is known about the proteomic differences between moderate and severe psoriasis. In this cross-sectional study, we evaluated the proteomic differences between moderate and severe psoriasis using data-independent acquisition mass spectrometry (DIA-MS). 173 differentially expressed proteins (DEPs) were significantly differentially expressed between the two groups. Among them, 85 proteins were upregulated, while 88 were downregulated (FC ≥ ± 1.5, P < 0.05). Eighteen DEPs were mainly enriched in the IL - 17 signalling pathway, Neutrophil extracellular trap formation, Neutrophil degranulation and NF - kappa B signalling pathway, which were associated with psoriasis pathogenesis. Ingenuity pathway Analysis (IPA) identified TNF and TDP53 as the top upstream up-regulators, while Lipopolysaccharide and YAP1 were the top potential down-regulators. The main active pathways were antimicrobial peptides and PTEN signalling, while the inhibitory pathways were the neutrophil extracellular trap pathway, neutrophil degranulation, and IL-8 signalling. 4D-parallel reaction monitoring (4D-PRM) suggested that KRT6A were downregulated in severe psoriasis. Our data identify Eighteen DEPs as biomarkers of disease severity, and are associated with IL - 17 signalling pathway, Neutrophil extracellular trap formation, NF - kappa B signalling pathway, and defence response to the bacterium. Targeting these molecules and measures to manage infection may improve psoriasis's severity and therapeutic efficacy.
Collapse
Affiliation(s)
- Lingling Wu
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China
| | - Chen Cen
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China
| | - Bibo Xie
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China
| | - Lihua Hu
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China
| | - Jia Huang
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China
| | - Ningning Shen
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China
| | - Qiang Dong
- Department of Dermatology, Dermatology Hospital of Zhejiang Province, Huzhou, 313299, China.
| |
Collapse
|
21
|
Magiera A, Kołodziejczyk-Czepas J, Olszewska MA. Antioxidant and Anti-Inflammatory Effects of Vanillic Acid in Human Plasma, Human Neutrophils, and Non-Cellular Models In Vitro. Molecules 2025; 30:467. [PMID: 39942571 PMCID: PMC11820348 DOI: 10.3390/molecules30030467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
Vanillic acid (VA) is a dietary benzoic acid derivative, flavoring agent, and food stabilizer. In this study, the antioxidant and anti-inflammatory potential of VA was explored in vitro and ex vivo in human immune cells and non-cellular models. In neutrophils, VA significantly downregulated the fMLP-induced oxidative burst and the generation of reactive oxygen species (ROS); it also suppressed the release of pro-inflammatory cytokines (TNF-α, IL-8) and the tissue-remodeling enzyme elastase-2 (ELA-2) in cells stimulated with LPS and fMLP+cytochalasin B. Additionally, VA showed good biocompatibility with human neutrophils and peripheral blood mononuclear cells (PBMCs) across the tested concentrations of 1-50 µg/mL. Furthermore, VA at 1-5 μg/mL enhanced the non-enzymatic antioxidant capacity of human plasma (NEAC) and prevented oxidative and nitrative damage to plasma proteins by protecting tyrosine moieties and thiols from peroxynitrite. VA also inhibited lipid peroxidation and the formation of thiobarbituric acid-reactive substances (at 50 μg/mL) and protein-bound carbonyls (at 5-50 μg/mL) in peroxynitrite-treated plasma. In non-cellular tests, VA acted as a hypochlorous acid and hydrogen peroxide scavenger and inhibited non-enzymatic protein glycation, outperforming the references Trolox and aminoguanidine. Along with existing data from animal models and studies on polyphenol intake, these results might support the synergic role of VA in dietary protection against chronic diseases related to oxidative stress and inflammation.
Collapse
Affiliation(s)
- Anna Magiera
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego St., 90-151 Lodz, Poland;
| | - Joanna Kołodziejczyk-Czepas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 141/143 Pomorska, 90-236 Lodz, Poland;
| | - Monika Anna Olszewska
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego St., 90-151 Lodz, Poland;
| |
Collapse
|
22
|
Lu Y, Wu H, Luo Y, Xia W, Sun D, Chen R, Miao Z, Zhang W, Yu Y, Wen A. CircIRAK3 Promotes Neutrophil Extracellular Trap Formation by Improving the Stability of ELANE mRNA in Sepsis. Inflammation 2024:10.1007/s10753-024-02206-z. [PMID: 39707013 DOI: 10.1007/s10753-024-02206-z] [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: 10/01/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/23/2024]
Abstract
Excessive formation of neutrophil extracellular traps (NETs) has been shown to exacerbate inflammatory injury and organ damage in patients with sepsis. Circular RNAs (circRNAs) abnormally expressed in immune cells of sepsis patients, and play an important role in the pathogenesis of dysregulated immune responses. However, the functions of circRNAs in NET formation during sepsis remain unknown. Here, we identified circIRAK3, a novel circRNA that was upregulated in peripheral blood neutrophils of sepsis patients. Combining clinical data, we revealed that elevated circIRAK3 was positively correlated with blood NET levels. Furthermore, knockdown and overexpression in differentiated HL-60 (dHL-60) neutrophil-like cells demonstrated that circIRAK3 promoted NET formation. In addition, we found that circIRAK3 promoted NET formation via positively regulating elastase expression in dHL-60 cells when treated with inflammatory stimuli. Mechanistically, circIRAK3 directly interacted with ELAVL1 to improve ELANE mRNA stability and consequently promote elastase protein expression. In summary, our study reveals that circIRAK3 promotes NET formation in sepsis by increasing ELANE mRNA levels.
Collapse
Affiliation(s)
- Yao Lu
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Huang Wu
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Yuanyuan Luo
- Department of Blood Transfusion Medicine, The First Medical Center of Chinese PLA General Hospital, NO 28, Fuxing Road, Haidian District, Beijing, 100853, China
| | - Wenjun Xia
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Denglian Sun
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Ruichi Chen
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Zeqing Miao
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Weiwei Zhang
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China
| | - Yang Yu
- Department of Blood Transfusion Medicine, The First Medical Center of Chinese PLA General Hospital, NO 28, Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Aiqing Wen
- Department of Blood Transfusion, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Burns and Combined Injury, Army Medical University, NO 10, Changjiang Branch Road, Daping District, Chongqing, 400042, China.
| |
Collapse
|
23
|
Zhang F, Xia Y, Su J, Quan F, Zhou H, Li Q, Feng Q, Lin C, Wang D, Jiang Z. Neutrophil diversity and function in health and disease. Signal Transduct Target Ther 2024; 9:343. [PMID: 39638788 PMCID: PMC11627463 DOI: 10.1038/s41392-024-02049-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/21/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Neutrophils, the most abundant type of granulocyte, are widely recognized as one of the pivotal contributors to the acute inflammatory response. Initially, neutrophils were considered the mobile infantry of the innate immune system, tasked with the immediate response to invading pathogens. However, recent studies have demonstrated that neutrophils are versatile cells, capable of regulating various biological processes and impacting both human health and disease. Cytokines and other active mediators regulate the functional activity of neutrophils by activating multiple receptors on these cells, thereby initiating downstream signal transduction pathways. Dysfunctions in neutrophils and disruptions in neutrophil homeostasis have been implicated in the pathogenesis of numerous diseases, including cancer and inflammatory disorders, often due to aberrant intracellular signaling. This review provides a comprehensive synthesis of neutrophil biological functions, integrating recent advancements in this field. Moreover, it examines the biological roles of receptors on neutrophils and downstream signaling pathways involved in the regulation of neutrophil activity. The pathophysiology of neutrophils in numerous human diseases and emerging therapeutic approaches targeting them are also elaborated. This review also addresses the current limitations within the field of neutrophil research, highlighting critical gaps in knowledge that warrant further investigation. In summary, this review seeks to establish a comprehensive and multidimensional model of neutrophil regulation, providing new perspectives for potential clinical applications and further research.
Collapse
Affiliation(s)
- Fengyuan Zhang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yidan Xia
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jiayang Su
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fushi Quan
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Hengzong Zhou
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qirong Li
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Qiang Feng
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Chao Lin
- School of Grain Science and Technology, Jilin Business and Technology College, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China.
| | - Ziping Jiang
- Department of Hand and Foot Surgery, Orthopedics Center, The First Hospital of Jilin University, Changchun, People's Republic of China.
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
24
|
Namin SS, Zhu YP, Croker BA, Tan Z. Turning Neutrophil Cell Death Deadly in the Context of Hypertensive Vascular Disease. Can J Cardiol 2024; 40:2356-2367. [PMID: 39326672 DOI: 10.1016/j.cjca.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/24/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Hypertensive vascular disease (HVD) is a major health burden globally and is a comorbidity commonly associated with other metabolic diseases. Many factors are associated with HVD including obesity, diabetes, smoking, chronic kidney disease, and sterile inflammation. Increasing evidence points to neutrophils as an important component of the chronic inflammatory response in HVD. Neutrophils are abundant in the circulation and can respond rapidly upon stimulation to deploy an armament of antimicrobial effector functions. One of the outcomes of neutrophil activation is the generation of neutrophil extracellular traps (NETs), a regulated extrusion of chromatin and proteases. Although neutrophils and NETs are well described as components of the innate immune response to infection, recent evidence implicates them in HVD. Endothelial cell activation can trigger neutrophil adhesion, activation, and production of NETs promoting vascular dysfunction, vessel remodelling, and loss of resistance. The regulated release of NETs can be controlled by the pore-forming activities of distinct cell death pathways. The best characterized pathways in this context are apoptosis, pyroptosis, and necroptosis. In this review, we discuss how inflammatory cell death signalling and NET formation contribute to hypertensive disease. We also examine novel therapeutic approaches to limit NET production and their future potential as therapeutic drugs for cardiovascular disorders.
Collapse
Affiliation(s)
- Sahand Salari Namin
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Yanfang Peipei Zhu
- Department of Biochemistry and Molecular Biology, Immunology Center of Georgia, Augusta University, Augusta, Georgia, USA
| | - Ben A Croker
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Zhehao Tan
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
25
|
Huth SW, Geri JB, Oakley JV, MacMillan DWC. μMap-Interface: Temporal Photoproximity Labeling Identifies F11R as a Functional Member of the Transient Phagocytic Surfaceome. J Am Chem Soc 2024; 146:32255-32262. [PMID: 39532068 DOI: 10.1021/jacs.4c11058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Phagocytosis is usually carried out by professional phagocytic cells in the context of pathogen response or wound healing. The transient surface proteins that regulate phagocytosis pose a challenging proteomics target; knowledge thereof could lead to new therapeutic insights. Herein, we describe a novel photocatalytic proximity labeling method: "μMap-Interface", allowing for spatiotemporal mapping of phagocytosis. Utilizing photocatalyst-conjugated IGG-opsonized beads and initiating phagocytosis in a synchronized manner, we capture phagocytic interactome "snapshots" at the interface of the phagocyte and its target. This allows profiling of the dynamic surface proteome of human macrophages during the engulfment process. We reveal previously known phagocytic mediators as well as potential novel interactors and validate their presence with super-resolution microscopy. This includes F11R, an important cancer target yet to be investigated in the context of phagocytosis. Further, we demonstrate that knocking down F11R leads to an increased degree of phagocytosis; this insight could contribute to explaining its oncogenic activity. Lastly, we show capture of orthogonal phagocytic surfaceomes across different cells, using a neutrophil-like model. We believe this method will enable new insights into phagocytic processes in a variety of contexts.
Collapse
Affiliation(s)
- Sean W Huth
- Merck Center for Catalysis at Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Jacob B Geri
- Merck Center for Catalysis at Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - James V Oakley
- Merck Center for Catalysis at Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - David W C MacMillan
- Merck Center for Catalysis at Princeton University, Princeton, New Jersey 08544, United States
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
26
|
Zhang S, Liu Y, Jin S, Xia T, Song H, Cao C, Liao Y, Pan R, Yan M, Chang Q. Exploration of novel human neutrophil elastase inhibitors from natural compounds: Virtual screening, in vitro, molecular dynamics simulation and in vivo study. Eur J Pharmacol 2024; 982:176825. [PMID: 39159715 DOI: 10.1016/j.ejphar.2024.176825] [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: 12/18/2023] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Human neutrophil elastase (HNE) is an important contributor to lung diseases such as acute lung injury (ALI) or acute respiratory distress syndrome. Therefore, this study aimed to identify natural HNE inhibitors with anti-inflammatory activity through machine learning algorithms, in vitro assays, molecular dynamic simulation, and an in vivo ALI assay. METHODS Based on the optimized Discovery Studio two-dimensional molecular descriptors, combined with different molecular fingerprints, six machine learning models were established using the Naïve Bayesian (NB) method to identify HNE inhibitors. Subsequently, the optimal model was utilized to screen 6925 drug-like compounds obtained from the Traditional Chinese Medicine Systems Pharmacy Database and Analysis Platform (TCMSP), followed by ADMET analysis. Finally, 10 compounds with reported anti-inflammatory activity were selected to determine their inhibitory activities against HNE in vitro, and the compounds with the best activity were selected for a 100 ns molecular dynamics simulation and its anti-inflammatory effect was evaluated using Poly (I:C)-induced ALI model. RESULTS The evaluation of the in vitro HNE inhibition efficiency of the 10 selected compounds showed that the flavonoid tricetin had the strongest inhibitory effect on HNE. The molecular dynamics simulation indicated that the binding of tricetin to HNE was relatively stable throughout the simulation. Importantly, in vivo experiments indicated that tricetin treatment substantially improved the Poly (I:C)-induced ALI. CONCLUSION The proposed NB model was proved valuable for exploring novel HNE inhibitors, and natural tricetin was screened out as a novel HNE inhibitor, which was confirmed by in vitro and in vivo assays for its inhibitory activities.
Collapse
Affiliation(s)
- Shanshan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Yongguang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Suwei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Tianji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Hongbin Song
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Chenxi Cao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Yonghong Liao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Ruile Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Mingzhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
27
|
Zhang L, Cheng HH, Krüger N, Hörnich B, Graichen L, Hahn AS, Schulz SR, Jäck HM, Stankov MV, Behrens GMN, Müller MA, Drosten C, Mörer O, Winkler MS, Qian Z, Pöhlmann S, Hoffmann M. ACE2-independent sarbecovirus cell entry can be supported by TMPRSS2-related enzymes and can reduce sensitivity to antibody-mediated neutralization. PLoS Pathog 2024; 20:e1012653. [PMID: 39536058 PMCID: PMC11559990 DOI: 10.1371/journal.ppat.1012653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, demonstrated that zoonotic transmission of animal sarbecoviruses threatens human health but the determinants of transmission are incompletely understood. Here, we show that most spike (S) proteins of horseshoe bat and Malayan pangolin sarbecoviruses employ ACE2 for entry, with human and raccoon dog ACE2 exhibiting broad receptor activity. The insertion of a multibasic cleavage site into the S proteins increased entry into human lung cells driven by most S proteins tested, suggesting that acquisition of a multibasic cleavage site might increase infectivity of diverse animal sarbecoviruses for the human respiratory tract. In contrast, two bat sarbecovirus S proteins drove cell entry in an ACE2-independent, trypsin-dependent fashion and several ACE2-dependent S proteins could switch to the ACE2-independent entry pathway when exposed to trypsin. Several TMPRSS2-related cellular proteases but not the insertion of a multibasic cleavage site into the S protein allowed for ACE2-independent entry in the absence of trypsin and may support viral spread in the respiratory tract. Finally, the pan-sarbecovirus antibody S2H97 enhanced cell entry driven by two S proteins and this effect was reversed by trypsin while trypsin protected entry driven by a third S protein from neutralization by S2H97. Similarly, plasma from quadruple vaccinated individuals neutralized entry driven by all S proteins studied, and availability of the ACE2-independent, trypsin-dependent pathway reduced neutralization sensitivity. In sum, our study reports a pathway for entry into human cells that is ACE2-independent, can be supported by TMPRSS2-related proteases and may be associated with antibody evasion.
Collapse
Affiliation(s)
- Lu Zhang
- Infection Biology Unit, German Primate Center–Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Hsiu-Hsin Cheng
- Infection Biology Unit, German Primate Center–Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Nadine Krüger
- Platform Infection Models, German Primate Center, Göttingen, Germany
| | - Bojan Hörnich
- Junior Research Group Herpesviruses, German Primate Center, Göttingen, Germany
| | - Luise Graichen
- Infection Biology Unit, German Primate Center–Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Alexander S. Hahn
- Junior Research Group Herpesviruses, German Primate Center, Göttingen, Germany
| | - Sebastian R. Schulz
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Metodi V. Stankov
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Georg M. N. Behrens
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), partner site Hannover-Braunschweig, Hannover, Germany
| | - Marcel A. Müller
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Infection Research (DZIF), partner site Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Infection Research (DZIF), partner site Berlin, Berlin, Germany
| | - Onnen Mörer
- Department of Anesthesiology, University of Göttingen Medical Center, Göttingen, Georg-August University of Göttingen, Göttingen, Germany
| | - Martin Sebastian Winkler
- Department of Anesthesiology, University of Göttingen Medical Center, Göttingen, Georg-August University of Göttingen, Göttingen, Germany
| | - ZhaoHui Qian
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center–Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center–Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, Göttingen, Germany
| |
Collapse
|
28
|
Hu C, Long L, Lou J, Leng M, Yang Q, Xu X, Zhou X. CTC-neutrophil interaction: A key driver and therapeutic target of cancer metastasis. Biomed Pharmacother 2024; 180:117474. [PMID: 39316968 DOI: 10.1016/j.biopha.2024.117474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024] Open
Abstract
Circulating tumor cells (CTCs) are cancer cells that detach from the primary tumor and enter the bloodstream, where they can seed new metastatic lesions in distant organs. CTCs are often associated with white blood cells (WBCs), especially neutrophils, the most abundant and versatile immune cells in the blood. Neutrophils can interact with CTCs through various mechanisms, such as cell-cell adhesion, cytokine secretion, protease release, and neutrophil extracellular traps (NETs) formation. These interactions can promote the survival, proliferation, invasion, and extravasation of CTCs, as well as modulate the pre-metastatic niche and the tumor microenvironment. Therefore, inhibiting CTC-neutrophils interaction could be a potential strategy to reduce tumor metastasis and improve the prognosis of cancer patients. In this review, we summarize the current literature on CTC-neutrophils interaction' role in tumor metastasis and discuss the possible therapeutic approaches to target this interaction.
Collapse
Affiliation(s)
- Chengyi Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China; Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming 650500, PR China
| | - Ling Long
- School of Pharmacy, Kunming Medical University, Kunming 650500, PR China; Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing 400054, PR China
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Mingjing Leng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Qingqing Yang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, PR China
| | - Xiang Xu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming 650500, PR China; Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China.
| | - Xing Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine & School of Rehabilitation, Kunming Medical University, Kunming 650500, PR China.
| |
Collapse
|
29
|
Zhang Y, Zeng Y, Bai H, Zhang W, Xue Z, Hu S, Lu S, Wang N. Depression of Ca V1.2 activation and expression in mast cells ameliorates allergic inflammation diseases. J Pharm Anal 2024; 14:101149. [PMID: 39720622 PMCID: PMC11667708 DOI: 10.1016/j.jpha.2024.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
Allergic inflammation is closely related to the activation of mast cells (MCs), which is regulated by its intracellular Ca2+ level, but the intake and effects of the intracellular Ca2+ remain unclear. The Ca2+ influx is controlled by members of Ca2+ channels, among which calcium voltage-gated channel subunit alpha1 C (CaV1.2) is the most robust. This study aimed to reveal the role and underlying mechanism of MC CaV1.2 in allergic inflammation. We found that CaV1.2 participated in MC activation and allergic inflammation. Nimodipine (Nim), as a strong CaV1.2-specific antagonist, ameliorated allergic inflammation in mice. Further, CaV1.2 activation in MC was triggered by phosphatizing at its Ser1928 through protein kinase C (PKC), which calcium/calmodulin-dependent protein kinase II (CaMKII) catalyzed. Overexpression or knockdown of MC CaV1.2 influenced MC activation. Importantly, CaV1.2 expression in MC had detrimental effects, while its deficiency ameliorated allergic pulmonary inflammation. Results provide novel insights into CaV1.2 function and a potential drug target for controlling allergic inflammation.
Collapse
Affiliation(s)
- Yongjing Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yingnan Zeng
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Haoyun Bai
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wen Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhuoyin Xue
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shiling Hu
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shemin Lu
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Nan Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, China
| |
Collapse
|
30
|
Wen S, Zheng X, Yin W, Liu Y, Wang R, Zhao Y, Liu Z, Li C, Zeng J, Rong M. Dental stem cell dynamics in periodontal ligament regeneration: from mechanism to application. Stem Cell Res Ther 2024; 15:389. [PMID: 39482701 PMCID: PMC11526537 DOI: 10.1186/s13287-024-04003-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024] Open
Abstract
Periodontitis, a globally prevalent chronic inflammatory disease is characterized by the progressive degradation of tooth-supporting structures, particularly the periodontal ligament (PDL), which can eventually result in tooth loss. Despite the various clinical interventions available, most focus on symptomatic relief and lack substantial evidence of supporting the functional regeneration of the PDL. Dental stem cells (DSCs), with their homology and mesenchymal stem cell (MSC) properties, have gained significant attention as a potential avenue for PDL regeneration. Consequently, multiple therapeutic strategies have been developed to enhance the efficacy of DSC-based treatments and improve clinical outcomes. This review examines the mechanisms by which DSCs and their derivatives promote PDL regeneration, and explores the diverse applications of exogenous implantation and endogenous regenerative technology (ERT) aimed at amplifying the regenerative capacity of endogenous DSCs. Additionally, the persistent challenges and controversies surrounding DSC therapies are discussed, alongside an evaluation of the limitations in current research on the underlying mechanisms and innovative applications of DSCs in PDL regeneration with the aim of providing new insights for future development. Periodontitis, a chronic inflammatory disease, represents a major global public health concern, affecting a significant proportion of the population and standing as the leading cause tooth loss in adults. The functional periodontal ligament (PDL) plays an indispensable role in maintaining periodontal health, as its structural and biological integrity is crucial for the long-term prognosis of periodontal tissues. It is widely recognized as the cornerstone of periodontal regeneration Despite the availability of various treatments, ranging from nonsurgical interventions to guided tissue regeneration (GTR) techniques, these methods have shown limited success in achieving meaningful PDL regeneration. As a result, the inability to fully restore PDL function underscores the urgent need for innovative therapeutic strategies at reconstructing this essential structure. Stem cell therapy, known for its regenerative and immunomodulatory potential, offers a promising approach for periodontal tissue repair. Their application marks a significant paradigm shift in the treatment of periodontal diseases, opening new avenues for functional PDL regeneration. However, much of the current research has primarily focused on the regeneration of alveolar bone and gingiva, as these hard and soft tissues can be more easily evaluated through visual assessment. The complexity of PDL structure, coupled with the intricate interactions among cellular and molecular components, presents significant scientific and clinical hurdles in translating DSC research into practical therapeutic applications. This review provides a thorough exploration of DSC dynamics in periodontal regeneration, detailing their origins, properties, and derived products, while also examining their potential mechanisms and applications in PDL regeneration. It offers an in-depth analysis of the current research, landscape, acknowledging both the progress made and the challenges that remain in bridging the gap between laboratory findings and clinical implementation. Finally, the need for continued investigation into the intricate mechanisms governing DSC behavior and the optimization of their use in regenerative therapies for periodontal diseases is also emphasized.
Collapse
Affiliation(s)
- Shuyi Wen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xiao Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Wuwei Yin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yushan Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Ruijie Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yaqi Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Ziyi Liu
- Department of Stomatology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, Foshan, Guangdong, 528308, China
| | - Cong Li
- Dongguan Key Laboratory of Metabolic Immunology and Oral Diseases, Dongguan Maternal and Child Health Care Hospital, Dongguan, Guangdong, 523000, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, Guangdong, 523808, China.
| | - Mingdeng Rong
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China.
| |
Collapse
|
31
|
Zhou S, Sun H. Prognostic model for gastric cancer patients with COVID-19 and network pharmacology study on treatment by lentinan. Sci Rep 2024; 14:24645. [PMID: 39428405 PMCID: PMC11491479 DOI: 10.1038/s41598-024-76783-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024] Open
Abstract
Patients with gastric cancer (GC) are more susceptible to coronavirus disease 2019 (COVID-19), which further worsens their already challenging prognosis. However, there are no effective treatment options for these patients. Lentinan is a potent bioactive component with antiviral and antitumor effects. We hypothesized that lentinan might exert powerful pharmacological effects in patients with GC and COVID-19. In this study, a prognostic model of patients with GC/COVID-19 was constructed and used to apply a network pharmacology approach to reveal biological functions, drug targets, and molecular mechanisms of the action of lentinan against GC/COVID-19. Clinical analysis revealed key prognostic genes in patients with GC/COVID-19. The results of network pharmacology analysis suggested that the therapeutic effect of lentinan on GC/COVID-19 mainly involves the modulation of several neutrophil-related biological processes, as well as the nucleotide-binding and oligomerization domain-like receptor and interleukin-17 signaling pathways. In addition, C-X-C motif chemokine ligand 8, vascular endothelial growth factor A, ribonuclease 3, and F2 were identified as key genes of lentinan against GC/COVID-19. Key prognostic genes were identified in patients with GC/COVID-19 through the construction of a prognostic model. Pharmacological functions and signaling pathways of lentinan against GC/COVID-19 were revealed. These included the regulation of neutrophils and NOD-like receptor signaling pathways. The findings provide the first published evidence of the potential value of lentinan as a complementary therapy for GC/COVID-19.
Collapse
Affiliation(s)
- Sitong Zhou
- Department of General Surgery Department, The second hospital of Jilin University, Changchun, 130041, China
| | - Hao Sun
- Department of General Surgery Department, The second hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
32
|
Muchova M, Kuehne SA, Grant MM, Smith PP, Nagi M, Chapple ILC, Hirschfeld J. Fusobacterium nucleatum elicits subspecies-specific responses in human neutrophils. Front Cell Infect Microbiol 2024; 14:1449539. [PMID: 39450334 PMCID: PMC11499235 DOI: 10.3389/fcimb.2024.1449539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/06/2024] [Indexed: 10/26/2024] Open
Abstract
Fusobacterium nucleatum as a Gram-negative anaerobe plays a key bridging role in oral biofilms. It is involved in periodontal and extraoral diseases, the most prominent being colorectal cancer. Five subspecies are recognised: animalis, fusiforme, nucleatum, polymorphum and vincentii. Subspecies interact with neutrophils constantly patrolling tissues to remove microbial intruders. Neutrophil antimicrobial activities include generation of reactive oxygen species (ROS), formation of neutrophil extracellular traps (NETs) and release of cytokines and neutrophil enzymes. Subspecies-specific differences in immunogenicity have previously been observed in a neutrophil-like cell line but were not investigated in human neutrophils. Additionally, neutrophil responses to planktonic and biofilm-grown F. nucleatum have not been studied to date. The aims of this study were to compare the immunogenicity of planktonic and biofilm-grown F. nucleatum and to investigate potential differences in human neutrophil responses when stimulated with individual F. nucleatum subspecies. Human neutrophils isolated from peripheral blood were stimulated with planktonic and biofilm-grown F. nucleatum subspecies. Generation of ROS and NET formation were quantified by luminescence and fluorescence assays, respectively. Secretion of cytokines (IL-1β, TNF-α, IL-6, IL-8), neutrophil elastase and matrix metalloproteinase-9 was quantified by enzyme-linked immunosorbent assay (ELISA). Neutrophil responses showed biofilm-grown bacteria induced a significantly higher total and intracellular ROS response, as well as shorter time to total ROS release. Biofilm-grown F. nucleatum led to significantly lower IL-1β release. We found significant differences among individual subspecies in terms of total, intracellular ROS and extracellular superoxide. Subspecies polymorphum stimulated the highest mean amount of NET release. Amounts of cytokines released differed significantly among subspecies, while no differences were found in lysosomal enzyme release. Immunogenicity of F. nucleatum in human neutrophils is highly subspecies-specific in vitro with regard to ROS release and cytokine production. Understanding subspecies-specific immunogenicity of F. nucleatum may facilitate the discovery of novel therapeutic targets in F. nucleatum-mediated diseases.
Collapse
Affiliation(s)
- Maria Muchova
- Periodontal Research Group, Birmingham School of Dentistry, Institute of Clinical Sciences, The University of Birmingham, Birmingham, United Kingdom
| | - Sarah A. Kuehne
- School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Melissa M. Grant
- Periodontal Research Group, Birmingham School of Dentistry, Institute of Clinical Sciences, The University of Birmingham, Birmingham, United Kingdom
- Birmingham Dental Hospital, Birmingham Community Health National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Birmingham National Institute for Health and Care Research (NIHR) Biomedical Research Centre (BRC) in Inflammation, Birmingham University, Birmingham, United Kingdom
| | - Peter P. Smith
- Periodontal Research Group, Birmingham School of Dentistry, Institute of Clinical Sciences, The University of Birmingham, Birmingham, United Kingdom
| | - Malee Nagi
- Periodontal Research Group, Birmingham School of Dentistry, Institute of Clinical Sciences, The University of Birmingham, Birmingham, United Kingdom
| | - Iain L. C. Chapple
- Periodontal Research Group, Birmingham School of Dentistry, Institute of Clinical Sciences, The University of Birmingham, Birmingham, United Kingdom
- Birmingham Dental Hospital, Birmingham Community Health National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Birmingham National Institute for Health and Care Research (NIHR) Biomedical Research Centre (BRC) in Inflammation, Birmingham University, Birmingham, United Kingdom
| | - Josefine Hirschfeld
- Periodontal Research Group, Birmingham School of Dentistry, Institute of Clinical Sciences, The University of Birmingham, Birmingham, United Kingdom
- Birmingham Dental Hospital, Birmingham Community Health National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Birmingham National Institute for Health and Care Research (NIHR) Biomedical Research Centre (BRC) in Inflammation, Birmingham University, Birmingham, United Kingdom
| |
Collapse
|
33
|
Li G, Guo Y, Ma A, Wang D, Zhang Q, Zhao C, Peng X, Ding L, Chen X, Qiu F. Curcumol derivatives exhibit ameliorating effects on lipopolysaccharide-induced acute lung injury: Synthesis, biological evaluation, structure-activity relationship and action mechanism. Bioorg Chem 2024; 153:107838. [PMID: 39353222 DOI: 10.1016/j.bioorg.2024.107838] [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: 06/24/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Acute lung injury (ALI) is an intricate clinical disease marked by high mortality and a sudden start. Currently, although there are no specific therapeutics for ALI, the administration of anti-inflammatory drugs is a promising treatment strategy. Curcumol, a terpenoid natural product, has demonstrated significant anti-inflammatory activity. Herein, we designed and synthesised 42 curcumol derivatives using curcumol as the core scaffold. These derivatives underwent in vitro screening for anti-inflammatory activity, and their structure-activity relationship was assessed. Among them, derivative 2 exhibited potent anti-inflammatory potential, inhibiting the expression of inflammatory markers at the nanomolar level. In addition, its water solubility was considerably improved, thereby laying the foundation for enhanced druggability. Derivative 2 also ameliorated lipopolysaccharide (LPS)-induced ALI and reduced pulmonary inflammation at a dose of 5 mg/kg. Proteomics analysis revealed that the anti-inflammatory effect of this compound primarily involved the mTOR signalling pathway. Furthermore, molecular docking and cellular thermal shift assays indicated that GSK3β is a critical target of action of derivative 2, as verified via western blotting. These findings suggest that derivative 2 can be a lead therapeutic compound for ALI, with GSK3β emerging as a promising novel target for the development of specific anti-ALI drugs.
Collapse
Affiliation(s)
- Gen Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Yajing Guo
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Anna Ma
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Dan Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Qi Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Chongyan Zhao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Xuling Peng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Liqin Ding
- Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China
| | - Xi Chen
- School of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China; State Key Laboratory of Component-based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Jinghai District, Tianjin 301617, PR China.
| |
Collapse
|
34
|
Xu S, Tan S, Romanos P, Reedy JL, Zhang Y, Mansour MK, Vyas JM, Mecsas J, Mou H, Leong JM. Blocking HXA 3-mediated neutrophil elastase release during S. pneumoniae lung infection limits pulmonary epithelial barrier disruption and bacteremia. mBio 2024; 15:e0185624. [PMID: 39120139 PMCID: PMC11389395 DOI: 10.1128/mbio.01856-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Streptococcus pneumoniae (Sp), a leading cause of community-acquired pneumonia, can spread from the lung into the bloodstream to cause septicemia and meningitis, with a concomitant threefold increase in mortality. Limitations in vaccine efficacy and a rise in antimicrobial resistance have spurred searches for host-directed therapies that target pathogenic immune processes. Polymorphonuclear leukocytes (PMNs) are essential for infection control but can also promote tissue damage and pathogen spread. The major Sp virulence factor, pneumolysin, triggers acute inflammation by stimulating the 12-lipoxygenase (12-LOX) eicosanoid synthesis pathway in epithelial cells. This pathway is required for systemic spread in a mouse pneumonia model and produces a number of bioactive lipids, including hepoxilin A3 (HXA3), a hydroxy epoxide PMN chemoattractant that has been hypothesized to facilitate breach of mucosal barriers. To understand how 12-LOX-dependent inflammation promotes dissemination during Sp lung infection and dissemination, we utilized bronchial stem cell-derived air-liquid interface cultures that lack this enzyme to show that HXA3 methyl ester (HXA3-ME) is sufficient to promote basolateral-to-apical PMN transmigration, monolayer disruption, and concomitant Sp barrier breach. In contrast, PMN transmigration in response to the non-eicosanoid chemoattractant N-formyl-L-methionyl-L-leucyl-phenylalanine (fMLP) did not lead to epithelial disruption or bacterial translocation. Correspondingly, HXA3-ME but not fMLP increased the release of neutrophil elastase (NE) from Sp-infected PMNs. Pharmacologic blockade of NE secretion or activity diminished epithelial barrier disruption and bacteremia after pulmonary challenge of mice. Thus, HXA3 promotes barrier-disrupting PMN transmigration and NE release, pathological events that can be targeted to curtail systemic disease following pneumococcal pneumonia.IMPORTANCEStreptococcus pneumoniae (Sp), a leading cause of pneumonia, can spread from the lung into the bloodstream to cause systemic disease. Limitations in vaccine efficacy and a rise in antimicrobial resistance have spurred searches for host-directed therapies that limit pathologic host immune responses to Sp. Excessive polymorphonuclear leukocyte (PMN) infiltration into Sp-infected airways promotes systemic disease. Using stem cell-derived respiratory cultures that reflect bona fide lung epithelium, we identified eicosanoid hepoxilin A3 as a critical pulmonary PMN chemoattractant that is sufficient to drive PMN-mediated epithelial damage by inducing the release of neutrophil elastase. Inhibition of the release or activity of this protease in mice limited epithelial barrier disruption and bacterial dissemination, suggesting a new host-directed treatment for Sp lung infection.
Collapse
Affiliation(s)
- Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Patricia Romanos
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Program in Biotechnology, Francisco de Vitoria University, Madrid, Spain
| | - Jennifer L. Reedy
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Yihan Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael K. Mansour
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jatin M. Vyas
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Stuart B Levy Center for the Integrated Management of Antimicrobial Resistance, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Lee HT, Lin CS, Liu CY, Chen P, Tsai CY, Wei YH. Mitochondrial Plasticity and Glucose Metabolic Alterations in Human Cancer under Oxidative Stress-From Viewpoints of Chronic Inflammation and Neutrophil Extracellular Traps (NETs). Int J Mol Sci 2024; 25:9458. [PMID: 39273403 PMCID: PMC11395599 DOI: 10.3390/ijms25179458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Oxidative stress elicited by reactive oxygen species (ROS) and chronic inflammation are involved both in deterring and the generation/progression of human cancers. Exogenous ROS can injure mitochondria and induce them to generate more endogenous mitochondrial ROS to further perpetuate the deteriorating condition in the affected cells. Dysfunction of these cancer mitochondria may possibly be offset by the Warburg effect, which is characterized by amplified glycolysis and metabolic reprogramming. ROS from neutrophil extracellular traps (NETs) are an essential element for neutrophils to defend against invading pathogens or to kill cancer cells. A chronic inflammation typically includes consecutive NET activation and tissue damage, as well as tissue repair, and together with NETs, ROS would participate in both the destruction and progression of cancers. This review discusses human mitochondrial plasticity and the glucose metabolic reprogramming of cancer cells confronting oxidative stress by the means of chronic inflammation and neutrophil extracellular traps (NETs).
Collapse
Affiliation(s)
- Hui-Ting Lee
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Chen-Sung Lin
- Division of Thoracic Surgery, Department of Surgery, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for General Education, Kainan University, Taoyuan City 338, Taiwan
| | - Chao-Yu Liu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Po Chen
- Cancer Free Biotech, Taipei 114, Taiwan
| | - Chang-Youh Tsai
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Clinical Trial Center, Division of Immunology & Rheumatology, Fu Jen Catholic University Hospital, New Taipei City 243, Taiwan
- Faculty of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
36
|
Oshins R, Greenberg Z, Tai YL, Zhao D, Wang X, Mehrad B, He M, Patel I, Khartabil L, Zhou H, Brantly M, Khodayari N. Extracellular Vesicle-Associated Neutrophil Elastase Activates Hepatic Stellate Cells and Promotes Liver Fibrogenesis via ERK1/2 Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608832. [PMID: 39229038 PMCID: PMC11370372 DOI: 10.1101/2024.08.20.608832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Liver fibrosis associated with increased mortality is caused by activation of hepatic stellate cells and excessive production and accumulation of extracellular matrix in response to fibrotic insults. It has been shown that in addition to liver inflammation, systemic inflammation also contributes to liver fibrogenesis. A deeper understanding of mechanisms that control liver fibrotic response to intra- and extra-hepatic inflammation is essential to develop novel clinical strategies against this disease. Extracellular vesicles (EV) have been recognized as immune mediators that facilitate activation of hepatic stellate cells. In inflammatory diseases, activated neutrophils release neutrophil elastase (NE) bound to EV, which has been identified as a significant contributor to inflammation by promoting immune cell activation. Here, we aimed to explore the role of inflammation derived plasma EV-associated NE in liver fibrogenesis and its potential mechanisms. We show EV-associated NE induces activation, proliferation and migration of hepatic stellate cells by promoting activation of the ERK1/2 signaling pathway. This effect did not occur through EV without surface NE, and Sivelestat, a NE inhibitor, inhibited activation of the ERK1/2 signaling pathway mediated by EV-associated NE. Moreover, we found plasma EV-associated NE increases deposition of collagen1 and α-smooth muscle actin in the liver of a mouse model of liver fibrosis (Mdr2-/-). Notably, this effect does not occur in control mice without preexisting liver disease. These data suggest that EV-associated NE is a pro-fibrogenic factor for hepatic stellate cell activation via the ERK1/2 signaling pathway in pre-existing liver injuries. Inhibition of the plasma EV-associated NE in inflammatory conditions may be a therapeutic target for liver fibrosis in patients with inflammatory diseases.
Collapse
Affiliation(s)
- Regina Oshins
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Zachary Greenberg
- Department of Pharmaceutics, College of Pharmacy; University of Florida, Gainesville, Florida, USA
| | - Yun-Ling Tai
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Derrick Zhao
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Xuan Wang
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Borna Mehrad
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy; University of Florida, Gainesville, Florida, USA
| | - Ishan Patel
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Laith Khartabil
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Huiping Zhou
- Department of Microbiology and Immunology; Virginia Commonwealth University, Richmond VA Medical Center, Richmond, Virginia, USA
| | - Mark Brantly
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| | - Nazli Khodayari
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine; University of Florida, Gainesville, Florida, USA
| |
Collapse
|
37
|
Luan N, Cao H, Wang Y, Zhang H, Lin K, Hu J, Rong M, Liu C. ShSPI Inhibits Thrombosis Formation and Ischemic Stroke In Vivo. Int J Mol Sci 2024; 25:9003. [PMID: 39201690 PMCID: PMC11354536 DOI: 10.3390/ijms25169003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Thrombotic diseases, emerging as a global public health hazard with high mortality and disability rates, pose a significant threat to human health and longevity. Although current antithrombotic therapies are effective in treating these conditions, they often carry a substantial risk of bleeding, highlighting the urgent need for safer therapeutic alternatives. Recent evidence has increasingly pointed to a connection between elastase activity and thrombosis. In the current study, we investigated the antithrombotic effects of ShSPI, an elastase inhibitor peptide derived from the venom of Scolopendra hainanum. Results showed that ShSPI significantly attenuated carrageenan-induced thrombosis in vivo. Furthermore, ShSPI effectively inhibited the carrageenan-induced decrease in serum superoxide dismutase (SOD) activity and increase in prothrombin time, fibrinogen level, and endothelial nitric oxide synthase (eNOS) activity. In addition, ShSPI reduced intracerebral thrombosis and improved functional outcomes following ischemic stroke in a transient middle cerebral artery occlusion (tMCAO) mouse model. Collectively, these findings suggest that ShSPI is a promising candidate for the development of novel thrombotic therapies.
Collapse
Affiliation(s)
- Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| | - Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| | - Haihao Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| | - Kangyang Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| | - Jingping Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| | - Mingqiang Rong
- National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (N.L.); (H.C.); (Y.W.); (H.Z.); (K.L.); (J.H.)
| |
Collapse
|
38
|
Taenaka H, Fang X, Maishan M, Trivedi A, Wick KD, Gotts JE, Martin TR, Calfee CS, Matthay MA. Neutrophil reduction attenuates the severity of lung injury in the early phase of pneumococcal pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2024; 327:L141-L149. [PMID: 38772909 PMCID: PMC11687957 DOI: 10.1152/ajplung.00113.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024] Open
Abstract
Neutrophils are the first leukocytes to be recruited to sites of inflammation in response to chemotactic factors released by activated macrophages and pulmonary epithelial and endothelial cells in bacterial pneumonia, a common cause of acute respiratory distress syndrome (ARDS). Although neutrophilic inflammation facilitates the elimination of pathogens, neutrophils also may cause bystander tissue injury. Even though the presence of neutrophils in alveolar spaces is a key feature of acute lung injury and ARDS especially from pneumonia, their contribution to the pathogenesis of lung injury is uncertain. The goal of this study was to elucidate the role of neutrophils in a clinically relevant model of bacterial pneumonia. We investigated the effect of reducing neutrophils in a mouse model of pneumococcal pneumonia treated with antibiotics. Neutrophils were reduced with anti-lymphocyte antigen 6 complex locus G6D (Ly6G) monoclonal antibody 24 h before and immediately preceding infection. Mice were inoculated intranasally with Streptococcus pneumoniae and received ceftriaxone 12 h after bacterial inoculation. Neutrophil reduction in mice treated with ceftriaxone attenuated hypoxemia, alveolar permeability, epithelial injury, pulmonary edema, and inflammatory biomarker release induced by bacterial pneumonia, even though bacterial loads in the distal air spaces of the lung were modestly increased as compared with antibiotic treatment alone. Thus, when appropriate antibiotics are administered, lung injury in the early phase of bacterial pneumonia is mediated in part by neutrophils. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.NEW & NOTEWORTHY Neutrophil accumulation is a key feature of ARDS, but their contribution to the pathogenesis is still uncertain. We investigated the effect of reducing neutrophils in a clinically relevant mouse model of pneumococcal pneumonia treated with antibiotics. When appropriate antibiotics were administered, neutrophil reduction with Ly6G antibody markedly attenuated lung injury and improved oxygenation. In the early phase of bacterial pneumonia, neutrophils contribute to the severity of lung injury, although they also participate in host defense.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Xiaohui Fang
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Mazharul Maishan
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Alpa Trivedi
- Department of Laboratory Medicine, University of California, San Francisco, California, United States
| | - Katherine D Wick
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Jeffrey E Gotts
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Thomas R Martin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States
| | - Carolyn S Calfee
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Michael A Matthay
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, United States
| |
Collapse
|
39
|
Du L, Gong Y, Zhang X, Sun J, Gao F, Shen M, Bai H, Yang T, Cheng X, Li S, Peng J, Liu Z, Ding S, Chen J, Cheng W. PD-L1 siRNA hitched polyethyleneimine-elastase constituting nanovesicle induces tumor immunogenicity and PD-L1 silencing for synergistic antitumor immunotherapy. J Nanobiotechnology 2024; 22:442. [PMID: 39068444 PMCID: PMC11282766 DOI: 10.1186/s12951-024-02700-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND PD-1/PD-L1 blockade has become a powerful method to treat malignant tumors. However, a large proportion of patients still do not benefit from this treatment, due to low tumor immunogenicity and low tumor penetration of the agents. Recently, neutrophil elastase has been shown to induce robust tumor immunogenicity, while the insufficient enzyme activity at the tumor site restricted its anti-tumor application. Here, we designed polyethyleneimine-modified neutrophil elastase (PEI-elastase) loaded with PD-L1small interfering RNA (PD-L1 siRNA) for improving enzymatic activity and delivering siRNA to tumor, which was expected to solve the above-mentioned problems. RESULTS We first demonstrated that PEI-elastase possessed high enzymatic activity, which was also identified as an excellent gene-delivery material. Then, we synthesized anti-tumor lipopolymer (P-E/S Lip) by encapsulating PEI-elastase and PD-L1siRNA with pH-responsive anionic liposomes. The P-E/S Lip could be rapidly cleaved in tumor acidic environment, leading to exposure of the PEI-elastase/PD-L1 siRNA. Consequently, PEI-elastase induced powerful tumor immunogenicity upon direct tumor killing with minimal toxicity to normal cells. In parallel, PEI-elastase delivered PD-L1siRNA into the tumor and reduced PD-L1 expression. Orthotopic tumor administration of P-E/S Lip not only attenuated primary tumor growth, but also produced systemic anti-tumor immune response to inhibit growth of distant tumors and metastasis. Moreover, intravenous administration of P-E/S Lip into mice bearing subcutaneous tumors leaded to an effective inhibition of established B16-F10 tumor and 4T1 tumor, with histological analyses indicating an absence of detectable toxicity. CONCLUSIONS In our study, a protease-based nanoplatform was used to cooperatively provoke robust tumor immunogenicity and down-regulate PD-L1 expression, which exhibited great potential as a combination therapy for precisely treating solid tumors.
Collapse
Affiliation(s)
- Li Du
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yao Gong
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoying Zhang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jide Sun
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Fengxia Gao
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Meiying Shen
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Huili Bai
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tiantian Yang
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoxue Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Siqiao Li
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Jian Peng
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhangling Liu
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Junman Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Biobank, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
40
|
Birk D, Siepmann E, Simon S, Sommerhoff CP. Human Neutrophil Elastase: Characterization of Intra- vs. Extracellular Inhibition. Int J Mol Sci 2024; 25:7917. [PMID: 39063160 PMCID: PMC11276905 DOI: 10.3390/ijms25147917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Neutrophil elastase (HNE), like other members of the so-called GASPIDs (Granule-Associated Serine Peptidases of Immune Defense), is activated during protein biosynthesis in myeloid precursors and stored enzymatically active in cytoplasmic granules of resting neutrophils until secreted at sites of host defense and inflammation. Inhibitors thus could bind to the fully formed active site of the protease intracellularly in immature progenitors, in circulating neutrophils, or to HNE secreted into the extracellular space. Here, we have compared the ability of a panel of diverse inhibitors to inhibit HNE in the U937 progenitor cell line, in human blood-derived neutrophils, and in solution. Most synthetic inhibitors and, surprisingly, even a small naturally occurring proteinaceous inhibitor inhibit HNE intracellularly, but the extent and dynamics differ markedly from classical enzyme kinetics describing extracellular inhibition. Intracellular inhibition of HNE potentially affects neutrophil functions and has side effects, but it avoids competition of inhibitors with extracellular substrates that limit its efficacy. As both intra- and extracellular inhibition have advantages and disadvantages, the quantification of intracellular inhibition, in addition to classical enzyme kinetics, will aid the design of novel, clinically applicable HNE inhibitors with targeted sites of action.
Collapse
Affiliation(s)
- Denise Birk
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Erika Siepmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Stefan Simon
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Christian P. Sommerhoff
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
- Institute of Medical Education, University Hospital, LMU Munich, 80336 Munich, Germany
| |
Collapse
|
41
|
Ross MH, Jia S. Heterogeneity in non-cystic fibrosis bronchiectasis: insights from ASPEN trial participants. ERJ Open Res 2024; 10:00372-2024. [PMID: 39081500 PMCID: PMC11288401 DOI: 10.1183/23120541.00372-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 08/02/2024] Open
Abstract
ASPEN trial participant characteristics highlight the heterogeneity of non-cystic fibrosis bronchiectasis and global variations in clinical practice patterns https://bit.ly/447XeP0.
Collapse
Affiliation(s)
- Melissa H. Ross
- Departments of Internal Medicine and Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Shijing Jia
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
42
|
Read NE, Wilson HM. Recent Developments in the Role of Protein Tyrosine Phosphatase 1B (PTP1B) as a Regulator of Immune Cell Signalling in Health and Disease. Int J Mol Sci 2024; 25:7207. [PMID: 39000313 PMCID: PMC11241678 DOI: 10.3390/ijms25137207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a non-receptor tyrosine phosphatase best known for its role in regulating insulin and leptin signalling. Recently, knowledge on the role of PTP1B as a major regulator of multiple signalling pathways involved in cell growth, proliferation, viability and metabolism has expanded, and PTP1B is recognised as a therapeutic target in several human disorders, including diabetes, obesity, cardiovascular diseases and hematopoietic malignancies. The function of PTP1B in the immune system was largely overlooked until it was discovered that PTP1B negatively regulates the Janus kinase-a signal transducer and activator of the transcription (JAK/STAT) signalling pathway, which plays a significant role in modulating immune responses. PTP1B is now known to determine the magnitude of many signalling pathways that drive immune cell activation and function. As such, PTP1B inhibitors are being developed and tested in the context of inflammation and autoimmune diseases. Here, we provide an up-to-date summary of the molecular role of PTP1B in regulating immune cell function and how targeting its expression and/or activity has the potential to change the outcomes of immune-mediated and inflammatory disorders.
Collapse
Affiliation(s)
- Neve E Read
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Heather M Wilson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
43
|
Harman RM, Sipka A, Oxford KA, Oliveira L, Huntimer L, Nydam DV, Van de Walle GR. The mammosphere-derived epithelial cell secretome modulates neutrophil functions in the bovine model. Front Immunol 2024; 15:1367432. [PMID: 38994364 PMCID: PMC11236729 DOI: 10.3389/fimmu.2024.1367432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
Background Innovative therapies against bacterial infections are needed. One approach is to focus on host-directed immunotherapy (HDT), with treatments that exploit natural processes of the host immune system. The goals of this type of therapy are to stimulate protective immunity while minimizing inflammation-induced tissue damage. We use non-traditional large animal models to explore the potential of the mammosphere-derived epithelial cell (MDEC) secretome, consisting of all bioactive factors released by the cells, to modulate host immune functions. MDEC cultures are enriched for mammary stem and progenitor cells and can be generated from virtually any mammal. We previously demonstrated that the bovine MDEC secretome, collected and delivered as conditioned medium (CM), inhibits the growth of bacteria in vitro and stimulates functions related to tissue repair in cultured endothelial and epithelial cells. Methods The immunomodulatory effects of the bovine MDEC secretome on bovine neutrophils, an innate immune cell type critical for resolving bacterial infections, were determined in vitro using functional assays. The effects of MDEC CM on neutrophil molecular pathways were explored by evaluating the production of specific cytokines by neutrophils and examining global gene expression patterns in MDEC CM-treated neutrophils. Enzyme linked immunosorbent assays were used to determine the concentrations of select proteins in MDEC CM and siRNAs were used to reduce the expression of specific MDEC-secreted proteins, allowing for the identification of bioactive factors modulating neutrophil functions. Results Neutrophils exposed to MDEC secretome exhibited increased chemotaxis and phagocytosis and decreased intracellular reactive oxygen species and extracellular trap formation, when compared to neutrophils exposed to control medium. C-X-C motif chemokine 6, superoxide dismutase, peroxiredoxin-2, and catalase, each present in the bovine MDEC secretome, were found to modulate neutrophil functions. Conclusion The MDEC secretome administered to treat bacterial infections may increase neutrophil recruitment to the site of infection, stimulate pathogen phagocytosis by neutrophils, and reduce neutrophil-produced ROS accumulation. As a result, pathogen clearance might be improved and local inflammation and tissue damage reduced.
Collapse
Affiliation(s)
- Rebecca M. Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Anja Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Kelly A. Oxford
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | | | | | - Daryl V. Nydam
- Department of Public and Ecosystem Health, Cornell University, Ithaca, NY, United States
| | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| |
Collapse
|
44
|
Xu S, Tan S, Romanos P, Reedy JL, Zhang Y, Mansour MK, Vyas JM, Mecsas J, Mou H, Leong JM. Blocking HXA 3-mediated neutrophil elastase release during S. pneumoniae lung infection limits pulmonary epithelial barrier disruption and bacteremia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600637. [PMID: 38979170 PMCID: PMC11230237 DOI: 10.1101/2024.06.25.600637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Streptococcus pneumoniae (Sp), a leading cause of community-acquired pneumonia, can spread from the lung into the bloodstream to cause septicemia and meningitis, with a concomitant three-fold increase in mortality. Limitations in vaccine efficacy and a rise in antimicrobial resistance have spurred searches for host-directed therapies that target pathogenic immune processes. Polymorphonuclear leukocytes (PMNs) are essential for infection control but can also promote tissue damage and pathogen spread. The major Sp virulence factor, pneumolysin (PLY), triggers acute inflammation by stimulating the 12-lipoxygenase (12-LOX) eicosanoid synthesis pathway in epithelial cells. This pathway is required for systemic spread in a mouse pneumonia model and produces a number of bioactive lipids, including hepoxilin A3 (HXA3), a hydroxy epoxide PMN chemoattractant that has been hypothesized to facilitate breach of mucosal barriers. To understand how 12-LOX-dependent inflammation promotes dissemination during Sp lung infection and dissemination, we utilized bronchial stem cell-derived air-liquid interface (ALI) cultures that lack this enzyme to show that HXA3 methyl ester (HXA3-ME) is sufficient to promote basolateral-to-apical PMN transmigration, monolayer disruption, and concomitant Sp barrier breach. In contrast, PMN transmigration in response to the non-eicosanoid chemoattractant fMLP did not lead to epithelial disruption or bacterial translocation. Correspondingly, HXA3-ME but not fMLP increased release of neutrophil elastase (NE) from Sp-infected PMNs. Pharmacologic blockade of NE secretion or activity diminished epithelial barrier disruption and bacteremia after pulmonary challenge of mice. Thus, HXA3 promotes barrier disrupting PMN transmigration and NE release, pathological events that can be targeted to curtail systemic disease following pneumococcal pneumonia.
Collapse
Affiliation(s)
- Shuying Xu
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Graduate Program in Immunology, Tufts Graduate School of Biomedical Sciences, Boston, MA
| | - Shumin Tan
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Patricia Romanos
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Francisco de Vitoria University, Madrid, Spain
| | - Jennifer L. Reedy
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Yihan Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA
| | - Michael K. Mansour
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Jatin M. Vyas
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA
| | - Joan Mecsas
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA
| | - John M. Leong
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA
- Stuart B Levy Center for the Integrated Management of Antimicrobial Resistance, Tufts University, Boston, MA
| |
Collapse
|
45
|
Feng Q, Shu X, Fang H, Shi X, Zhang Y, Zhang H. Discovery of pharmacological effects and targets of Citri Grandis Exocarpium based on SYSTCM and virtual screening. Food Nutr Res 2024; 68:10618. [PMID: 38974913 PMCID: PMC11227261 DOI: 10.29219/fnr.v68.10618] [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: 02/24/2024] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 07/09/2024] Open
Abstract
Citri Grandis Exocarpium (Huajuhong, CGE) is the peel of the unripe fruits of Citrus grandis 'Tomentosa' and Citrus grandis (L.) Osbeck, which is commonly used in the clinic for the treatment of cough and indigestion. The pharmacological mechanism of CGE is unclear. In this study, the pharmacological effect of CGE was predicted by System Traditional Chinese Medicine (SYSTCM), which integrated the pharmacological effect prediction approach by artificial intelligence into the systemic traditional Chinese medicine (TCM) platform. The main pharmacological effect of CGE was antiallergy, promoting bile, blood lipid regulation, cardiotonics, diuresis, and antiarrhythmia by prediction of SYSTCM. In vitro cell experiments were carried out to identify the antiallergic effect of CGE. Extracts of Citri Grandis Exocarpium (ECGE) inhibited lipopolysaccharide-induced cell injury and nitric oxide release in RAW264.7 cells. ECGE and naringin-inhibited immunoglobulin E-induced cell degranulation in RBL-2H3 cells. Target profiling, protein interaction network, and molecular docking of compounds from CGE indicated that mitogen-activated protein kinase 14 (MAPK14) and matrix metalloprotease 9 (MMP9) were key potential targets of CGE with antiallergic activity. This study identified and validated the antiallergic effect of CGE by combining SYSTCM, cell experiments, and virtual screening, which provided a new paradigm and approach for studying the pharmacological effect and mechanism of TCM.
Collapse
Affiliation(s)
- Qinqi Feng
- Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonar y Diseases, National Center for Respirator y Medicine, National Clinical Research Center for Respirator y Diseases, Institute of Respirator y Medicine, Center of Respirator y Medicine, China-Japan Friendship Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Xinyang Shu
- Department of Traditional Chinese Medicine for Pulmonar y Diseases, National Center for Respirator y Medicine, National Clinical Research Center for Respirator y Diseases, Institute of Respirator y Medicine, Center of Respirator y Medicine, China-Japan Friendship Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Hanyu Fang
- Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonar y Diseases, National Center for Respirator y Medicine, National Clinical Research Center for Respirator y Diseases, Institute of Respirator y Medicine, Center of Respirator y Medicine, China-Japan Friendship Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoxi Shi
- Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine for Pulmonar y Diseases, National Center for Respirator y Medicine, National Clinical Research Center for Respirator y Diseases, Institute of Respirator y Medicine, Center of Respirator y Medicine, China-Japan Friendship Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Yanling Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hongchun Zhang
- Department of Traditional Chinese Medicine for Pulmonar y Diseases, National Center for Respirator y Medicine, National Clinical Research Center for Respirator y Diseases, Institute of Respirator y Medicine, Center of Respirator y Medicine, China-Japan Friendship Hospital Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Xu J, Zhang C, Wu K, Qian Y, Hu W. A comparative analysis of sivelestat sodium hydrate and ulinastatin combination therapy in the treatment of sepsis with acute respiratory distress syndrome. BMC Pulm Med 2024; 24:283. [PMID: 38886709 PMCID: PMC11184757 DOI: 10.1186/s12890-024-03083-w] [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: 02/21/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE This comparative analysis aimed to investigate the efficacy of Sivelestat Sodium Hydrate (SSH) combined with Ulinastatin (UTI) in the treatment of sepsis with acute respiratory distress syndrome (ARDS). METHODS A control group and an observation group were formed with eighty-four cases of patients with sepsis with ARDS, with 42 cases in each group. The control group was intravenously injected with UTI based on conventional treatment, and the observation group was injected with SSH based on the control group. Both groups were treated continuously for 7 days, and the treatment outcomes and efficacy of both groups were observed. The Murray Lung Injury Score (MLIS), Sequential Organ Failure Assessment (SOFA), and Acute Physiology and Chronic Health Evaluation II (APACHE II) were compared. Changes in respiratory function, inflammatory factors, and oxidative stress indicators were assessed. The occurrence of adverse drug reactions was recorded. RESULTS The total effective rate in the observation group (95.24%) was higher than that in the control group (80.95%) (P < 0.05). The mechanical ventilation time, intensive care unit (ICU) hospitalization time, and duration of antimicrobial medication in the observation group were shorter and multiple organ dysfunction syndrome incidence was lower than those in the control group (P < 0.05). The mortality rate of patients in the observation group (35.71%) was lower than that in the control group (52.38%), but there was no statistically significant difference between the two groups (P > 0.05). MLIS, SOFA, and APACHE II scores in the observation group were lower than the control group (P < 0.05). After treatment, respiratory function, inflammation, and oxidative stress were improved in the observation group (P < 0.05). Adverse reactions were not significantly different between the two groups (P > 0.05). CONCLUSION The combination of SSH plus UTI improves lung injury and pulmonary ventilation function, and reduces inflammation and oxidative stress in patients with sepsis and ARDS.
Collapse
Affiliation(s)
- Jian Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Wuxi Fifth Hospital of Jiangnan University, Wuxi, 214000, Jiangsu, China
- Wuxi Medical College of Jiangnan University, No. 1215, Guangrui Road, Liangxi District, Wuxi, 214000, Jiangsu, China
| | - Chenfei Zhang
- Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Keren Wu
- Department of Respiratory and Critical Care Medicine, the 904 Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Wuxi, 214000, Jiangsu, China
| | - Yanhua Qian
- Department of Respiratory and Critical Care Medicine, Affiliated Wuxi Fifth Hospital of Jiangnan University, Wuxi, 214000, Jiangsu, China.
- Wuxi Medical College of Jiangnan University, No. 1215, Guangrui Road, Liangxi District, Wuxi, 214000, Jiangsu, China.
| | - Wei Hu
- Department of Pharmacy, the 904 Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, No. 101, Xingyuan Road, Liangxi District, Wuxi, 214000, Jiangsu, China.
| |
Collapse
|
47
|
Jia W, Mao Y, Luo Q, Wu J, Guan Q. Targeting neutrophil elastase is a promising direction for future cancer treatment. Discov Oncol 2024; 15:167. [PMID: 38750338 PMCID: PMC11096153 DOI: 10.1007/s12672-024-01010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
Neutrophil elastase (NE) is a proteolytic enzyme released extracellular during the formation of neutrophil extracellular traps (NETs) through degranulation. In addition to participating in the body's inflammatory response, NE also plays an important role in cancer. It can promote tumor proliferation, migration, and invasion, induce epithelial-mesenchymal transition (EMT), and change the tumor microenvironment (TME) to promote tumor progression. Concurrently, NE promotes systemic treatment resistance by inducing EMT. However, it can also selectively kill cancer cells and attenuate tumor development. Sivelestat is a specific NE inhibitor that can be used in the perioperative period of esophageal cancer patients to reduce the incidence of postoperative complications after esophagectomy. In addition, the combination of sivelestat and trastuzumab can enhance the efficacy of human epidermal growth factor receptor 2(HER 2) positive breast cancer patients. Meanwhile, targeting the human antibody domains and fragments of NE is also a new way to treat cancer and inflammation-related diseases. This review provides valuable insights into the role of NE in cancer treatment. Additionally, we discuss the challenges associated with the clinical application of sivelestat. By shedding light on the promising potential of NE, this review contributes to the advancement of cancer treatment strategies.
Collapse
Affiliation(s)
- Wangqiang Jia
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yudong Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qianwen Luo
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jiang Wu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Quanlin Guan
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.
- Department of Oncology Surgery, the First Hospital of Lanzhou University, No. 1, Donggang West Road, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
48
|
Caroli C, Baron G, Cappellucci G, Brighenti V, Della Vedova L, Fraulini F, Oliaro-Bosso S, Alessandrini A, Zambon A, Lusvardi G, Aldini G, Biagi M, Corsi L, Pellati F. Extraction, purification and in vitro assessment of the antioxidant and anti-inflammatory activity of policosanols from non-psychoactive Cannabis sativa L. Heliyon 2024; 10:e30291. [PMID: 38737258 PMCID: PMC11088244 DOI: 10.1016/j.heliyon.2024.e30291] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024] Open
Abstract
Policosanols (PCs) are bioactive compounds extracted from different natural waxes. In this work, the purification, characterization and assessment of the antioxidant and anti-inflammatory activity was carried out on PCs from an innovative source, i.e. a waxy material from supercritical-fluid extraction (SFE) of non-psychoactive Cannabis sativa L. (hemp) inflorescences. Starting from this material, PCs were obtained by microwave-assisted trans-esterification and hydrolysis, followed by preparative liquid chromatography under normal phase conditions. The purified product was characterized using high-performance liquid chromatography (HPLC) with an evaporative light scattering detector (ELSD). In vitro cell-free and cell-based antioxidant and anti-inflammatory assays were then performed to assess their bioactivity. HPLC-ELSED analysis of the purified mixture from hemp wax revealed C26OH and C28OH as the main compounds. In vitro assays indicated an inhibition of intracellular reactive oxygen species (ROS) production, a reduction of nuclear factor kappa B (NF-κB) activation and of the activity of the neutrophil elastase. Immunoblotting assays allowed us to hypothesize the mechanism of action of the compounds of interest, given the higher levels of MAPK-activated protein kinase 2 (MK2) and heme oxygenase-1 (HO-1) protein expression in the PC pretreated HaCaT cells. In conclusion, even if more research is needed to unveil other molecular mechanisms involved in hemp PC activity, the results of this work suggest that these compounds may have potential for use in oxinflammation processes.
Collapse
Affiliation(s)
- Clarissa Caroli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103-287, 41125, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio, Via Giuseppe Campi 287, 41125, Modena, Italy
| | - Giovanna Baron
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Giorgio Cappellucci
- Department of Physical Sciences, Earth and Environment, University of Siena, Via Laterina, 8, 53100, Siena, Italy
| | - Virginia Brighenti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103-287, 41125, Modena, Italy
| | - Larissa Della Vedova
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Francesca Fraulini
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Simonetta Oliaro-Bosso
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125, Turin, Italy
| | - Andrea Alessandrini
- Department of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, Via Giuseppe Campi 213/A, 41125, Modena, Italy
- National Institute of Biostructures e Biosystems (INBB), 00136, Roma, Italy
| | - Alfonso Zambon
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Gigliola Lusvardi
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125, Modena, Italy
| | - Giancarlo Aldini
- Department of Pharmaceutical Sciences, University of Milan, Via Mangiagalli 25, 20133, Milan, Italy
| | - Marco Biagi
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/A, 43124, Parma, Italy
| | - Lorenzo Corsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103-287, 41125, Modena, Italy
- National Institute of Biostructures e Biosystems (INBB), 00136, Roma, Italy
| | - Federica Pellati
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103-287, 41125, Modena, Italy
| |
Collapse
|
49
|
Sun J, Li J, Deng Y, Yin X, Huangfu X, Ye Z, Zhou X, Chen Y, Yuan S, Wang X. The beneficial effects of neutrophil elastase inhibitor on gastrointestinal dysfunction in sepsis. Clin Transl Sci 2024; 17:e13829. [PMID: 38769746 PMCID: PMC11106555 DOI: 10.1111/cts.13829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
To investigate the effects of neutrophil elastase inhibitor (sivelestat sodium) on gastrointestinal function in sepsis. A reanalysis of the data from previous clinical trials conducted at our center was performed. Septic patients were divided into either the sivelestat group or the non-sivelestat group. The gastrointestinal dysfunction score (GIDS), feeding intolerance (FI) incidence, serum levels of intestinal barrier function and inflammatory biomarkers were recorded. The clinical severity and outcome variables were also documented. A total of 163 septic patients were included. The proportion of patients with GIDS ≥2 in the sivelestat group was reduced relative to that in the non-sivelestat group (9.6% vs. 22.5%, p = 0.047) on the 7th day of intensive care unit (ICU) admission. The FI incidence was also remarkably reduced in the sivelestat group in contrast to that in the non-sivelestat group (21.2% vs. 37.8%, p = 0.034). Furthermore, the sivelestat group had fewer days of FI [4 (3, 4) vs. 5 (4-6), p = 0.008]. The serum levels of d-lactate (p = 0.033), intestinal fatty acid-binding protein (p = 0.005), interleukin-6 (p = 0.001), white blood cells (p = 0.007), C-reactive protein (p = 0.001), and procalcitonin (p < 0.001) of the sivelestat group were lower than those of the non-sivelestat group. The sivelestat group also demonstrated longer ICU-free days [18 (0-22) vs. 13 (0-17), p = 0.004] and ventilator-free days [22 (1-24) vs. 16 (1-19), p = 0.002] compared with the non-sivelestat group. In conclusion, sivelestat sodium administration appears to improve gastrointestinal dysfunction, mitigate dysregulated inflammation, and reduce disease severity in septic patients.
Collapse
Affiliation(s)
- Jia‐Kui Sun
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Jing‐Jing Li
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Yi‐Hang Deng
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xiang Yin
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xiao‐Tian Huangfu
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Zi‐Yu Ye
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xue‐Hui Zhou
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Yong‐Ming Chen
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Shou‐Tao Yuan
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xiang Wang
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
50
|
Liu YG, Zhang SS, Jin SW, Xia TJ, Liao YH, Pan RL, Yan MZ, Chang Q. Anti-inflammatory effect and pharmacokinetics of dehydroandrographolide, an active component of Andrographis paniculata, on Poly(I:C)-induced acute lung injury. Biomed Pharmacother 2024; 174:116456. [PMID: 38552441 DOI: 10.1016/j.biopha.2024.116456] [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: 11/25/2023] [Revised: 02/29/2024] [Accepted: 03/15/2024] [Indexed: 05/01/2024] Open
Abstract
Acute lung injury (ALI) is a common and critical respiratory disorder caused by various factors, with viral infection being the leading contributor. Dehydroandrographolide (DAP), a constituent of the Chinese herbal plant Andrographis paniculata, exhibits a range of activities including anti-inflammatory, in vitro antiviral and immune-enhancing effects. This study evaluated the anti-inflammatory effects and pharmacokinetics (PK) profile of DAP in ALI mice induced by intratracheal instillation of Poly(I:C) (PIC). The results showed that oral administration of DAP (10-40 mg/kg) effectively suppressed the increase in lung wet-dry weight ratio, total cells, total protein content, accumulation of immune cells, inflammatory cytokines and neutrophil elastase levels in bronchoalveolar lavage fluid of PIC-treated mice. DAP concentrations, determined by an LC-MS/MS method, in plasma after receiving DAP (20 mg/kg) were unchanged compared to those in normal mice. However, DAP concentrations and relative PK parameters in the lungs were significantly altered in PIC-treated mice, exhibiting a relatively higher maximum concentration, larger AUC, and longer elimination half-life than those in the lungs of normal mice. These results demonstrated that DAP could improve lung edema and inflammation in ALI mice, and suggested that lung injury might influence the PK properties of DAP, leading to increased lung distribution and residence. Our study provides evidence that DAP displays significant anti-inflammatory activity against viral lung injury and is more likely to distribute to damaged lung tissue.
Collapse
Affiliation(s)
- Yong-Guang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Shan-Shan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Su-Wei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Tian-Ji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Yong-Hong Liao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Rui-Le Pan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China
| | - Ming-Zhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|