1
|
Kim C, Kim H, Sim WS, Jung M, Hong J, Moon S, Park JH, Kim JJ, Kang M, Kwon S, Kim MJ, Ban K, Park HJ, Kim BS. Spatiotemporal control of neutrophil fate to tune inflammation and repair for myocardial infarction therapy. Nat Commun 2024; 15:8481. [PMID: 39353987 PMCID: PMC11445496 DOI: 10.1038/s41467-024-52812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
Neutrophils are critical mediators of both the initiation and resolution of inflammation after myocardial infarction (MI). Overexuberant neutrophil signaling after MI exacerbates cardiomyocyte apoptosis and cardiac remodeling while neutrophil apoptosis at the injury site promotes macrophage polarization toward a pro-resolving phenotype. Here, we describe a nanoparticle that provides spatiotemporal control over neutrophil fate to both stymie MI pathogenesis and promote healing. Intravenous injection of roscovitine/catalase-loaded poly(lactic-co-glycolic acid) nanoparticles after MI leads to nanoparticle uptake by circulating neutrophils migrating to the infarcted heart. Activated neutrophils at the infarcted heart generate reactive oxygen species, triggering intracellular release of roscovitine, a cyclin-dependent kinase inhibitor, from the nanoparticles, thereby inducing neutrophil apoptosis. Timely apoptosis of activated neutrophils at the infarcted heart limits neutrophil-driven inflammation, promotes macrophage polarization toward a pro-resolving phenotype, and preserves heart function. Modulating neutrophil fate to tune both inflammatory and reparatory processes may be an effective strategy to treat MI.
Collapse
Affiliation(s)
- Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Mikyung Kang
- School of Health and Environmental Science, Korea University, Seoul, Republic of Korea
| | - Sungpil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea
| | - Mi-Jeong Kim
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul Saint Mary's Hospital, Seoul, Republic of Korea.
- Cell Death Disease Research Center, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, Republic of Korea.
- Institute of Chemical Processes, Seoul National University, Seoul, Republic of Korea.
- Institute of Engineering Research, Seoul National University, Seoul, Republic of Korea.
- Bio-MAX Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Yang H, Shi P, Li M, Liu S, Mou B, Xia Y, Sun J. Plasma proteome mediate the impact of PM 2.5 on stroke: A 2-step Mendelian randomization study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116624. [PMID: 38908058 DOI: 10.1016/j.ecoenv.2024.116624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The objectives of this study were to measure the mediation effect of plasma proteins and to clarify their mediating role in the relationship between stroke risk and particulate matter 2.5 (PM2.5) exposure. The possible mediating role of plasma proteins on the causative link between PM2.5 exposure and stroke incidence were examined using a two-step Mendelian randomization (MR) approach based on two-sample Mendelian randomization (TSMR). The findings revealed a significant positive causal relationship between PM2.5 exposure and stroke, with an inverse variance weighted odds ratio of 1.219 (95 % CI: 1.002 - 1.482, P < 0.05). Additionally, a positive causal association was identified between PM2.5 exposure and several plasma proteins, including FAM134B, SAP, ITGB7, Elafin, and DCLK3. Among these, FAM134B, ITGB7, Elafin, and DCLK3 also demonstrated a positive causal association with stroke, whereas only SAP was found to be negatively causally associated with stroke. Remarkably, four plasma proteins, namely DCLK3, FAM134B, Elafin, and ITGB7, were identified as mediators, accounting for substantial proportions (14.5 %, 13.6 %, 11.1 %, and 9.9 %) of the causal association between PM2.5 and stroke. These results remained robust across various sensitivity analyses. Consequently, the study highlights the significant and independent impact of PM2.5 on stroke risk and identifies specific plasma proteins as potential targets for preventive interventions against PM2.5-induced stroke.
Collapse
Affiliation(s)
- Huajie Yang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Peng Shi
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Mingzheng Li
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China; Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Shuailing Liu
- Department of Child and Adolescent Health, School of Public Health, China Medical University, Shenyang 110122, China
| | - Baohua Mou
- First Affiliated Hospital of Dalian Medical University, Dalian 116000, China
| | - Yinglan Xia
- Zhejiang Greentown Cardiovascular Hospital, Hangzhou 310000, China
| | - Jiaxing Sun
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
3
|
Zhu W, Ou Y, Wang C, An R, Lai J, Shen Y, Ye X, Wang H. A neutrophil elastase inhibitor, sivelestat, attenuates sepsis-induced acute kidney injury by inhibiting oxidative stress. Heliyon 2024; 10:e29366. [PMID: 38638960 PMCID: PMC11024609 DOI: 10.1016/j.heliyon.2024.e29366] [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: 05/22/2023] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/20/2024] Open
Abstract
Background Sivelestat, a selective inhibitor of neutrophil elastase (NE), can mitigate sepsis-related acute lung injury. However, the role of sivelestat in inhibiting oxidative stress and attenuating sepsis-related acute kidney injury (AKI) remains unclear. Here, we reported the effects of sivelestat against oxidative stress-induced AKI by suppressing the production of oxidative stress indicators. Materials and methods A male Sprague-Dawley rat model of sepsis was established by cecal ligation and puncture (CLP). Sivelestat or normal saline was administered into jugular vein with a sustained-release drug delivery system. Indicators of inflammation and AKI, including white blood cells (WBC), neutrophils, lymphocytes, C-reactive proteins (CRP), procalcitonin (PCT), blood urea nitrogen (BUN), creatinine (Cr) and uric acid (UA), were assessed at 24 h post-sivelestat treatment. Indicators of liver injury, including direct bilirubin (DBIL), indirect bilirubin (IBIL), aspartate aminotransferase (AST) and alanine aminotransferase (ALT), were also assessed at 24 h post-sivelestat treatment. Indicators of oxidative stress, including superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px), were assessed at 12 h and 24 h post-sivelestat treatment. At 24 h post-sivelestat treatment, H&E staining of kidney and liver tissue was performed to observe pathological alterations. Results At 24 h post normal saline or sivelestat (0.2 g/kg body weight) treatment, WBC, neutrophil, CRP, PCT, MDA, BUN, Cr, UA, AST, ALT, DBIL and IBIL were increased, while SOD and GSH-Px were decreased, in septic rats treated with normal saline compared with that in non-septic rats treated with normal saline (all p < 0.05). The changes of these indicators were reversed in septic rats treated with sivelestat compared with that in septic rats treated with normal saline (all p < 0.05). Similar results were found regarding the levels of oxidative stress indicators at 12 h post-sivelestat treatment. The degenerative histopathological changes in both kidney and liver tissues were ameliorated upon sivelestat treatment. Conclusions Sivelestat plays a protective role in sepsis-related AKI by inhibiting oxidative stress. Our study reveals a possible therapeutic potential of sivelestat for oxidative stress-induced AKI.
Collapse
Affiliation(s)
- Wei Zhu
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Yingwei Ou
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Chunnian Wang
- Ningbo Clinical Pathology Diagnosis Center, Ningbo 315000, Zhejiang, China
| | - Rongcheng An
- Emergency and Critical Care Center, Department of Emergency Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Junmei Lai
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Ye Shen
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Xiangming Ye
- Rehabilitation Medicine Center, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Intensive rehabilitation unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Haochu Wang
- Rehabilitation Medicine Center, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
4
|
Aimond G, Nicolle S, Debret R, Oréa V, Josset-Lamaugarny A, Palierne JF, Sommer P, Sigaudo-Roussel D, Fromy B. Dill Extract Preserves Dermal Elastic Fiber Network and Functionality: Implication of Elafin. Skin Pharmacol Physiol 2023; 36:249-258. [PMID: 37788642 DOI: 10.1159/000534248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/19/2023] [Indexed: 10/05/2023]
Abstract
INTRODUCTION Elastic skin fibers lose their mechanical properties during aging due to enzymatic degradation, lack of maturation, or posttranslational modifications. Dill extract has been observed to increase elastin protein expression and maturation in a 3D skin model, to improve mechanical properties of the skin, to increase elastin protein expression in vascular smooth muscle cells, to preserve aortic elastic lamella, and to prevent glycation. OBJECTIVE The aim of the study was to highlight dill actions on elastin fibers during aging thanks to elastase digestion model and the underlying mechanism. METHODS In this study, elastic fibers produced by dermal fibroblasts in 2D culture model were injured by elastase, and we observed the action of dill extract on elastic network by elastin immunofluorescence. Then action of dill extract was examined on mice skin by injuring elastin fibers by intradermal injection of elastase. Then elastin fibers were observed by second harmonic generation microscopy, and their functionality was evaluated by oscillatory shear stress tests. In order to understand mechanism by which dill acted on elastin fibers, enzymatic tests and real-time qPCR on cultured fibroblasts were performed. RESULTS We evidence in vitro that dill extract is able to prevent elastin from elastase digestion. And we confirm in vivo that dill extract treatment prevents elastase digestion, allowing preservation of the cutaneous elastic network in mice and preservation of the cutaneous elastic properties. Although dill extract does not directly inhibit elastase activity, our results show that dill extract treatment increases mRNA expression of the endogenous inhibitor of elastase, elafin. CONCLUSION Dill extract can thus be used to counteract the negative effects of elastase on the cutaneous elastic fiber network through modulation of PI3 gene expression.
Collapse
Affiliation(s)
- Géraldine Aimond
- LBTI UMR5305, CNRS/Univ Lyon/Université Claude Bernard Lyon 1, Lyon, France
| | - Stéphane Nicolle
- LBMC UMR_T9406 /Univ Lyon/ Université Claude Bernard Lyon 1/Université Gustave Eiffel, Lyon, France
| | - Romain Debret
- LBTI UMR5305, CNRS/Univ Lyon/Université Claude Bernard Lyon 1, Lyon, France
| | - Valérie Oréa
- ANIPHY Platform SFR Santé Lyon-Est UCBL, UAR3453/US7/Faculté De Médecine, Lyon, France
| | | | - Jean-François Palierne
- Laboratoire De Physique, ENS De Lyon, CNRS/ Univ Lyon/ Univ Claude Bernard, Lyon, France
| | - Pascal Sommer
- Hôpital Sainte Marguerite/Aix Marseille Université, Marseille, France
| | | | - Bérengère Fromy
- LBTI UMR5305, CNRS/Univ Lyon/Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
5
|
Chen WC, Wu CC, Liu YP, Zhuo GY, Wang YK, Chen YH, Chen CC, Wang YH, Wu MT, Wu IC. Elafin as a Prognostic Marker in Esophageal Squamous Cell Carcinoma: A Pilot Study Using Three-Dimensional Imaging and Genomic Profiling. Cancers (Basel) 2023; 15:3825. [PMID: 37568641 PMCID: PMC10417143 DOI: 10.3390/cancers15153825] [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: 06/28/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Esophageal cancers are globally the sixth deadliest malignancy, with limited curative options. The association of high serum elafin levels, a molecule produced by epithelial cells, with esophageal squamous cell carcinoma (ESCC) risk is established, but its link to poor ESCC prognosis remains unclear. To explore this question, we first used three-dimensional confocal imaging to create a model of the spatial distribution of elafin inside locoregional ESCC tissues. Then, after analyzing data obtained from whole-genome microarrays for ESCC cell lines and their more invasive sublines, we performed in vitro experiments using RNA sequencing to identify possible elafin-related pathways. Three-dimensional tissue imaging showed elafin distributed as an interweaved-like fibrous structure in the stroma of tissue obtained from patients with high serum levels of elafin and poorer prognoses. By contrast, the signal was confined inside or around the tumor nest in patients who had lower serum levels and better survival. The analysis of a TCGA dataset revealed that higher levels of elafin mRNA in stage I-IIIA ESCC patients were associated with shorter survival. The in vitro studies revealed that elafin promoted ESCC cell proliferation, migration, and invasion via the epithelial-mesenchymal transition pathway. Thus, elafin inhibition could potentially be used therapeutically to improve survival in patients with locoregional ESCC.
Collapse
Affiliation(s)
- Wei-Chung Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (M.-T.W.)
| | - Chun-Chieh Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (Y.-K.W.)
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Peng Liu
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Guan-Yu Zhuo
- Institute of New Drug Development, China Medical University, Taichung 404, Taiwan;
| | - Yao-Kuang Wang
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (Y.-K.W.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Yi-Hsun Chen
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Chu-Chih Chen
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli 350, Taiwan; (C.-C.C.); (Y.-H.W.)
| | - Yin-Han Wang
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli 350, Taiwan; (C.-C.C.); (Y.-H.W.)
| | - Ming-Tsang Wu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (M.-T.W.)
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - I-Chen Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (Y.-K.W.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
6
|
Antioxidant, Tyrosinase, α-Glucosidase, and Elastase Enzyme Inhibition Activities of Optimized Unripe Ajwa Date Pulp ( Phoenix dactylifera) Extracts by Response Surface Methodology. Int J Mol Sci 2023; 24:ijms24043396. [PMID: 36834805 PMCID: PMC9966286 DOI: 10.3390/ijms24043396] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
The Ajwa date (Phoenix dactylifera L., Arecaceae family) is a popular edible fruit consumed all over the world. The profiling of the polyphenolic compounds of optimized unripe Ajwa date pulp (URADP) extracts is scarce. The aim of this study was to extract polyphenols from URADP as effectively as possible by using response surface methodology (RSM). A central composite design (CCD) was used to optimize the extraction conditions with respect to ethanol concentration, extraction time, and temperature and to achieve the maximum amount of polyphenolic compounds. High-resolution mass spectrometry was used to identify the URADP's polyphenolic compounds. The DPPH-, ABTS-radical scavenging, α-glucosidase, elastase and tyrosinase enzyme inhibition of optimized extracts of URADP was also evaluated. According to RSM, the highest amounts of TPC (24.25 ± 1.02 mgGAE/g) and TFC (23.98 ± 0.65 mgCAE/g) were obtained at 52% ethanol, 81 min time, and 63 °C. Seventy (70) secondary metabolites, including phenolic, flavonoids, fatty acids, and sugar, were discovered using high-resolution mass spectrometry. In addition, twelve (12) new phytoconstituents were identified for the first time in this plant. Optimized URADP extract showed inhibition of DPPH-radical (IC50 = 87.56 mg/mL), ABTS-radical (IC50 = 172.36 mg/mL), α-glucosidase (IC50 = 221.59 mg/mL), elastase (IC50 = 372.25 mg/mL) and tyrosinase (IC50 = 59.53 mg/mL) enzymes. The results revealed a significant amount of phytoconstituents, making it an excellent contender for the pharmaceutical and food industries.
Collapse
|
7
|
Farag M, Peverelli M, Spinthakis N, Gue YX, Egred M, Gorog DA. Spontaneous Reperfusion in Patients with Transient ST-Elevation Myocardial Infarction-Prevalence, Importance and Approaches to Management. Cardiovasc Drugs Ther 2023; 37:169-180. [PMID: 34245445 DOI: 10.1007/s10557-021-07226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 01/19/2023]
Abstract
Patients with transient ST-elevation myocardial infarction (STEMI) or spontaneous resolution (SpR) of the ST-segment elevation on electrocardiogram could potentially represent a unique group of patients posing a therapeutic management dilemma. In this review, we discuss the potential mechanisms underlying SpR, its relation to clinical outcomes and the proposed management options for patients with transient STEMI with a focus on immediate versus early percutaneous coronary intervention. We performed a structured literature search of PubMed and Cochrane Library databases from inception to December 2020. Studies focused on SpR in patients with acute coronary syndrome were selected. Available data suggest that deferral of angiography and revascularization within 24-48 h in these patients is reasonable and associated with similar or perhaps better outcomes than immediate angiography. Further randomized trials are needed to elucidate the best pharmacological and invasive strategies for this cohort.
Collapse
Affiliation(s)
- Mohamed Farag
- Cardiothoracic Department, Freeman Hospital, Newcastle Upon Tyne, UK.
- School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire, UK.
| | - Marta Peverelli
- Department of Cardiology, Royal Papworth Hospital, Cambridge, UK
| | - Nikolaos Spinthakis
- School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire, UK
| | - Ying X Gue
- School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire, UK
| | - Mohaned Egred
- Cardiothoracic Department, Freeman Hospital, Newcastle Upon Tyne, UK
| | - Diana A Gorog
- School of Life and Medical Sciences, University of Hertfordshire, Hertfordshire, UK
| |
Collapse
|
8
|
Liu ML, Lyu X, Werth VP. Recent progress in the mechanistic understanding of NET formation in neutrophils. FEBS J 2022; 289:3954-3966. [PMID: 34042290 PMCID: PMC9107956 DOI: 10.1111/febs.16036] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 01/03/2023]
Abstract
Neutrophils are the most abundant circulating white blood cells and one of the major cell types of the innate immune system. Neutrophil extracellular traps (NETs) are a result of the extracellular release of nuclear chromatin from the ruptured nuclear envelope and plasma membrane. The externalized chromatin is an ancient defense weapon for animals to entrap and kill microorganisms in the extracellular milieu, thus protecting animals ranging from lower invertebrates to higher vertebrates. Although the externalized chromatin has the advantage of acting as anti-infective to protect against infections, extracellular chromatin might be problematic in higher vertebrate animals as they have an adaptive immune system that can trigger further immune or autoimmune responses. NETs and their associated nuclear and/or cytoplasmic components may induce sterile inflammation, immune, and autoimmune responses, leading to various human diseases. Though important in human pathophysiology, the cellular and molecular mechanisms of NET formation (also called NETosis) are not well understood. Given that nuclear chromatin forms the backbone of NETs, the nucleus is the root of the nuclear DNA extracellular traps. Thus, nuclear chromatin decondensation, along with the rupture of nuclear envelope and plasma membrane, is required for nuclear chromatin extracellular release and NET formation. So far, most of the literature focuses on certain signaling pathways, which are involved in NET formation but without explanation of cellular events and morphological changes described above. Here, we have summarized emerging evidence and discuss new mechanistic understanding, with our perspectives, in NET formation in neutrophils.
Collapse
Affiliation(s)
- Ming-Lin Liu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xing Lyu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA,Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Victoria P. Werth
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
9
|
Dakhel A, Memon AA, Zarrouk M, Ågren-Witteschus S, Sundquist J, Sundquist K, Gottsäter A. Novel cardiovascular biomarkers associated with peripheral arterial disease in men screened for abdominal aortic aneurysm. VASA 2022; 51:167-173. [DOI: 10.1024/0301-1526/a000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Summary: Background: Peripheral arterial disease (PAD) is a common atherosclerotic disease with severity ranging from asymptomatic to chronic limb threatening ischemia. The aim of the present cross-sectional study was to identify novel biomarkers associated with PAD. Patients and methods: Levels of 91 cardiovascular specific proteins in plasma samples were measured by the Proseek Multiplex CVD III96x96 panel from a cohort consisting of 267 65-year-old men recruited from a screening program for abdominal aortic aneurysm (AAA) Levels of protein biomarkers were compared in men with and without PAD (defined as an ankle brachial index of <0.9) and their diagnostic potential was calculated by receiver-operating characteristic analysis. Results: The prevalence of PAD was 14.2% (38/267). After adjustment for multiple comparisons, levels of the following 11 biomarkers remained significantly higher ( p<0.0001) in patients with PAD: secretoglobin family 3A member 2, osteoprotegerin, urokinase-type plasminogen activator surface receptor, serum macrophage chemokine ligand 16, matrix metalloproteinase 9, p-selectin, growth differentiation factor 15, elafin, cystatin B, trefoil factor 3, and fatty acid-binding protein 4. Multivariable logistic regression analysis (adjusted for smoking, use of antihypertensive and lipid-lowering medication, and metformin) showed that 11 biomarkers were significantly associated with higher risk of PAD with odds ratios ranging from 1.6 to 2.4. Area under curve calculated by receiver operating characteristic curve analysis (diagnostic value) for each protein biomarker ranged from 0.63 to 0.74. Conclusions: We have identified multiple proteins with a potential to be diagnostic biomarkers for PAD, and further research is warranted to clarify their potential predictive and prognostic value.
Collapse
Affiliation(s)
- Ardwan Dakhel
- Department of Vascular Diseases, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Ashfaque A. Memon
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Moncef Zarrouk
- Department of Vascular Diseases, Skåne University Hospital, Lund University, Malmö, Sweden
| | | | - Jan Sundquist
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Kristina Sundquist
- Wallenberg Laboratory, Center for Primary Health Care Research, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Anders Gottsäter
- Department of Acute and Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
10
|
Zaghi A, Holm H, Korduner J, Dieden A, Molvin J, Bachus E, Jujic A, Magnusson M. Physical Inactivity Is Associated With Post-discharge Mortality and Re-hospitalization Risk Among Swedish Heart Failure Patients—The HARVEST-Malmö Study. Front Cardiovasc Med 2022; 9:843029. [PMID: 35265689 PMCID: PMC8899472 DOI: 10.3389/fcvm.2022.843029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/26/2022] [Indexed: 11/16/2022] Open
Abstract
Background Several studies have examined the role of physical activity as a predictor of heart failure (HF) mortality and morbidity. Here, we aimed to evaluate the role of self-reported physical activity as an independent risk factor of post-discharge mortality and re-hospitalization in patients hospitalized for HF, as well as study the association between physical activity and 92 plasma proteins associated with cardiovascular disease (CVD). Methods Four-hundred-and-thirty-four patients hospitalized for HF (mean age 75 years; 32% women) were screened for physical activity derived from questionnaires in the Swedish national public health survey. The median follow-up time to death and re-hospitalization was 835 (interquartile range, 390–1,432) and 157 (43–583) days, respectively. Associations between baseline reported physical activity, mortality and re-hospitalization risk were analyzed using multivariable Cox regression analysis. Plasma samples from 295 study participants were analyzed with a proximity extension assay consisting of 92 proteins. Associations between proteins and physical activity were explored using a false discovery rate of <5%, and significant associations were taken forward to multivariate analyses. Results In the multivariate Cox regression model, physical inactivity, defined as physical activity time <1 h throughout the week was associated with increased risk of all-cause mortality (HR 1.71; CI95% 1.26–2.31; p = 5.9 × 10−4) as well as all-cause re-hospitalization (HR 1.27; CI95% 1.01–1.60; p = 0.038). Further, physical inactivity was associated with elevated plasma levels of Metalloproteinase inhibitor 4, Soluble interleukin 1 receptor-like 1, Elafin and Transferrin receptor protein 1, which are implicated in myocardial fibrosis, migration and apoptosis. Conclusions Self-reported low weekly physical activity is associated with increased risk of mortality and re-hospitalization in patients hospitalized for HF independent of traditional risk factors. Furthermore, physical inactivity was associated with elevated levels of 4 proteins linked to cardiovascular disease.
Collapse
Affiliation(s)
- Amir Zaghi
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- *Correspondence: Amir Zaghi
| | - Hannes Holm
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skane University Hospital, Lund University, Malmö, Sweden
| | - Johan Korduner
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skane University Hospital, Lund University, Malmö, Sweden
| | - Anna Dieden
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Biomedical Science, Malmö University, Malmö, Sweden
- Biofilms-Research Centre for Biointerfaces, Malmö University, Malmö, Sweden
| | - John Molvin
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skane University Hospital, Lund University, Malmö, Sweden
| | - Erasmus Bachus
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Amra Jujic
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skane University Hospital, Lund University, Malmö, Sweden
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Martin Magnusson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skane University Hospital, Lund University, Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Hypertension in Africa Research Team, North-West University, Potchefstroom, South Africa
| |
Collapse
|
11
|
Colakerol A, Suzan S, Temiz MZ, Gonultas S, Aykan S, Ozsoy S, Kucuk SH, Yuruk E, Kandırali E, Semercioz A. Tissue neutrophil elastase contributes to extracorporeal shock wave lithotripsy-induced kidney damage and the neutrophil elastase inhibitor, sivelestat, attenuates kidney damage with gratifying immunohistopathological and biochemical findings: an experimental study. Urolithiasis 2022; 50:103-112. [PMID: 34778918 DOI: 10.1007/s00240-021-01287-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/20/2021] [Indexed: 02/07/2023]
Abstract
Although the efficacy of extracorporeal shock wave lithotripsy (ESWL) has been well established within the literature, debate continues on the safety of the procedure while focusing on cellular injury and its long-term consequences. Here, we describe the role of neutrophil elastase (NE) in ESWL-related rat kidney damage and investigate the protective effects of sivelestat, an inhibitor of NE, during the early and late phases. Four groups including control, ESWL alone, ESWL with sivelestat 50 mg/kg and ESWL with treatment of 100 mg/kg, each consisting of ten rats were created. Biochemical parameters of kidney function and damage and immunohistopathological findings were compared in the early (72 h after ESWL) and late (1 week after ESWL) periods between the groups. During the early period, serum and urine creatinine levels and urine kidney injury molecule-1 (KIM-1) levels and the KIM-1/creatinine ratio increased in rats treated with ESWL compared to the control group. Furthermore, increased tissue inflammation, ductal dilatation and hemorrhage, and glomerular, tubular, and interstitial damage with increased NE staining were also detected in the ESWL treatment group. During the late phase, although urine KIM-1 levels remained stable at high levels, other parameters showed significant improvements. On the other hand, the administration of sivelestat 50 mg/kg decreased serum creatinine and urine KIM-1 and KIM-1/creatinine levels significantly in rats treated with ESWL, during the early and late periods. Significant decreases in tissue inflammation, tubular, and interstitial tissue damage were also observed during the early period. In conclusion, ESWL-related kidney tissue damage occurs primarily during the early period, and NE is involved in this process. On the other hand, the NE inhibitor sivelestat attenuated this ESWL-induced kidney damage.
Collapse
Affiliation(s)
- Aykut Colakerol
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Serhat Suzan
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Mustafa Zafer Temiz
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Serkan Gonultas
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Serdar Aykan
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Sule Ozsoy
- Department of Pathology, Bagcilar Training and Research Hospital, Bagcilar, Istanbul, Turkey
| | - Suat Hayri Kucuk
- Department of Biochemistry, Bagcilar Training and Research Hospital, Bagcilar, Istanbul, Turkey
| | - Emrah Yuruk
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Engin Kandırali
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey
| | - Atilla Semercioz
- Department of Urology, Bagcilar Training and Research Hospital, Merkez Mah., Dr. Sadik Ahmet Cad., Bagcilar, Istanbul, Turkey.
| |
Collapse
|
12
|
Wang X, Kaiser H, Kvist-Hansen A, McCauley BD, Skov L, Hansen PR, Becker C. IL-17 Pathway Members as Potential Biomarkers of Effective Systemic Treatment and Cardiovascular Disease in Patients with Moderate-to-Severe Psoriasis. Int J Mol Sci 2022; 23:ijms23010555. [PMID: 35008981 PMCID: PMC8745093 DOI: 10.3390/ijms23010555] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Psoriasis is a chronic inflammatory condition associated with atherosclerotic cardiovascular disease (CVD). Systemic anti-psoriatic treatments mainly include methotrexate and biological therapies targeting TNF, IL-12/23 and IL-17A. We profiled plasma proteins from patients with moderate-to-severe psoriasis to explore potential biomarkers of effective systemic treatment and their relationship to CVD. We found that systemically well-treated patients (PASI < 3.0, n = 36) had lower circulating levels of IL-17 pathway proteins compared to untreated patients (PASI > 10, n = 23). Notably, IL-17C and PI3 were decreased with all four examined systemic treatment types. Furthermore, in patients without CVD, we observed strong correlations among IL-17C/PI3/PASI (r ≥ 0.82, p ≤ 1.5 × 10−12) pairs or between IL-17A/PASI (r = 0.72, p = 9.3 × 10−8). In patients with CVD, the IL-17A/PASI correlation was abolished (r = 0.2, p = 0.24) and the other correlations were decreased, e.g., IL-17C/PI3 (r = 0.61, p = 4.5 × 10−5). Patients with moderate-to-severe psoriasis and CVD had lower levels of IL-17A compared to those without CVD (normalized protein expression [NPX] 2.02 vs. 2.55, p = 0.013), and lower IL-17A levels (NPX < 2.3) were associated with higher incidence of CVD (OR = 24.5, p = 0.0028, 95% CI 2.1–1425.1). As a result, in patients with moderate-to-severe psoriasis, we propose circulating IL-17C and PI3 as potential biomarkers of effective systemic anti-psoriatic treatment, and IL-17A as potential marker of CVD.
Collapse
Affiliation(s)
- Xing Wang
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (X.W.); (B.D.M.)
| | - Hannah Kaiser
- Department of Cardiology, University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark; (H.K.); (A.K.-H.); (P.R.H.)
- Department of Dermatology and Allergy, University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Amanda Kvist-Hansen
- Department of Cardiology, University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark; (H.K.); (A.K.-H.); (P.R.H.)
- Department of Dermatology and Allergy, University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark;
| | - Benjamin D. McCauley
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (X.W.); (B.D.M.)
| | - Lone Skov
- Department of Dermatology and Allergy, University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark;
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Peter Riis Hansen
- Department of Cardiology, University Hospital—Herlev and Gentofte, 2900 Hellerup, Denmark; (H.K.); (A.K.-H.); (P.R.H.)
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christine Becker
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (X.W.); (B.D.M.)
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence:
| |
Collapse
|
13
|
Classical swine fever virus NS4B protein interacts with MAVS and inhibits IL-8 expression in PAMs. Virus Res 2022; 307:198622. [PMID: 34762991 DOI: 10.1016/j.virusres.2021.198622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/27/2022]
Abstract
Classical swine fever virus (CSFV) infection causes a severe disease of pigs, resulting in significant economic losses. The CSFV NS4B protein is crucial for viral replication and pathogenicity. Interleukin 8 (IL-8), a main chemokine, is induced by multiple cell types and plays an essential role in host defense mechanisms against numerous viruses. It has been reported that NS4A of CSFV is involved in the induction of IL-8 expression in swine umbilical vein endothelial cells. However, the effect of CSFV NS4B on IL-8 expression is unknown. In this study, we showed that CSFV NS4B inhibited IL-8 expression in porcine alveolar macrophages (PAMs), and NS4B inhibited mitochondrial antiviral signaling protein (MAVS)-induced IL-8 expression. Moreover, CSFV NS4B interacted with MAVS. However, NS4B did not alter MAVS expression. Subsequently, we demonstrated that IRF3 knockdown or NF-κB inhibition reduced MAVS-induced IL-8 expression. Furthermore, the IRF3 and NF-κB pathways were activated by MAVS expression. However, CSFV NS4B inhibited MAVS-mediated NF-κB activation and IRF3 expression. Finally, CSFV NS4B inhibited IRF3 expression. Our findings reveal that CSFV NS4B interacts with MAVS and inhibits IL-8 expression by blocking the activation of IRF3 and NF-κB. Taken together, this study provides insights into the mechanism of NS4B-inhibited IL-8 expression.
Collapse
|
14
|
Molecular Pathogenesis of Psoriasis and Biomarkers Reflecting Disease Activity. J Clin Med 2021; 10:jcm10153199. [PMID: 34361983 PMCID: PMC8346978 DOI: 10.3390/jcm10153199] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease induced by multifactorial causes and is characterized by bothersome, scaly reddish plaques, especially on frequently chafed body parts, such as extensor sites of the extremities. The latest advances in molecular-targeted therapies using biologics or small-molecule inhibitors help to sufficiently treat even the most severe psoriatic symptoms and the extra cutaneous comorbidities of psoriatic arthritis. The excellent clinical effects of these therapies provide a deeper understanding of the impaired quality of life caused by this disease and the detailed molecular mechanism in which the interleukin (IL)-23/IL-17 axis plays an essential role. To establish standardized therapeutic strategies, biomarkers that define deep remission are indispensable. Several molecules, such as cytokines, chemokines, antimicrobial peptides, and proteinase inhibitors, have been recognized as potent biomarker candidates. In particular, blood protein markers that are repeatedly measurable can be extremely useful in daily clinical practice. Herein, we summarize the molecular mechanism of psoriasis, and we describe the functions and induction mechanisms of these biomarker candidates.
Collapse
|
15
|
Wu IC, Wang YK, Chen YH, Wu CC, Wu MC, Chen WC, Wang WL, Lin HS, Chen CC, Chou SH, Liu YP, Wu MT. High Serum Elafin Prediction of Poor Prognosis of Locoregional Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13123082. [PMID: 34205756 PMCID: PMC8233752 DOI: 10.3390/cancers13123082] [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: 05/14/2021] [Revised: 06/17/2021] [Accepted: 06/17/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Conventional serum markers such as carcinoembryonic antigen (CEA), squamous cell carcinoma antigen (SCC), and tissue polypeptide antigen (TPA) have a low sensitivity in predicting the prognosis of locoregional esophageal squamous cell carcinoma cell (ESCC). In our clinical study, we found high serum elafin to be an independent outcome predictor for stage I-IIIA ESCC, considering T, N, overall stage, and treatment. In vitro experiments showed that adding recombinant elafin drove ESCC cell proliferation, migration and invasion, while shRNA attenuated elafin levels, abrogating those effects. Our results suggested serum elafin might be a noninvasive biomarker to predict the outcome of locoregional ESCC and could potentially be used as a therapeutic target. Abstract Esophageal squamous cell carcinoma (ESCC) is a highly aggressive tumor known to have locally advanced and metastatic features which cause a dismal prognosis. We sought to determine whether elafin, a non-invasive and secretory small-molecule marker, could be used to predict prognosis in locoregional ESCC patients in human and in vitro studies. In our human study, 119 subjects were identified as having incident and pathologically-proved ESCC with stage I-IIIA tumors from southern Taiwan between 2000 and 2016. We measured their serum elafin levels at baseline and followed them until the date of cancer death or until January 2020, the end of this study. Those with high serum elafin levels were found to have a 1.99-fold risk (95% confidence interval: 1.17–3.38) shorter survival than those who did not. In our in vitro experiments, elevated elafin levels were found to drive ESCC cell proliferation, migration and invasion, while attenuation of elafin level by shRNA abrogated those effects. We concluded that elafin promotes ESCC motility and invasion and leads to a worse clinical prognosis in ESCC patients without distant metastasis.
Collapse
Affiliation(s)
- I-Chen Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (I.-C.W.); (Y.-K.W.); (Y.-H.C.)
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
| | - Yao-Kuang Wang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (I.-C.W.); (Y.-K.W.); (Y.-H.C.)
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
| | - Yi-Hsun Chen
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (I.-C.W.); (Y.-K.W.); (Y.-H.C.)
| | - Chun-Chieh Wu
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Meng-Chieh Wu
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 807, Taiwan;
| | - Wei-Chung Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wen-Lun Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-DA Hospital/I-Shou University, Kaohsiung 824, Taiwan;
| | - Hung-Shun Lin
- Department of Laboratory Medicine & Department of Research, Education & Training, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Public Health, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chou-Cheng Chen
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
| | - Shah-Hwa Chou
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.W.); (S.-H.C.)
- Division of Chest Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Peng Liu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (Y.-P.L.); (M.-T.W.); Tel.: +886-7-3121101 (ext. 5092-424) (Y.-P.L.); +886-7-3121101 (ext. 2315) (M.-T.W.)
| | - Ming-Tsang Wu
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (W.-C.C.); (C.-C.C.)
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Public Health, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Rapid Screening Research Center for Toxicology and Biomedicine, National Sun Yat-Sen University, Kaohsiung 807, Taiwan
- Correspondence: (Y.-P.L.); (M.-T.W.); Tel.: +886-7-3121101 (ext. 5092-424) (Y.-P.L.); +886-7-3121101 (ext. 2315) (M.-T.W.)
| |
Collapse
|
16
|
Guéant J, Guéant‐Rodriguez R, Fromonot J, Oussalah A, Louis H, Chery C, Gette M, Gleye S, Callet J, Raso J, Blanchecotte F, Lacolley P, Guieu R, Regnault V. Elastase and exacerbation of neutrophil innate immunity are involved in multi-visceral manifestations of COVID-19. Allergy 2021; 76:1846-1858. [PMID: 33484168 PMCID: PMC8014109 DOI: 10.1111/all.14746] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Many arguments suggest that neutrophils could play a prominent role in COVID-19. However, the role of key components of neutrophil innate immunity in severe forms of COVID-19 has deserved insufficient attention. We aimed to evaluate the involvement of neutrophil elastase, histone-DNA, and DNases in systemic and multi-organ manifestations of COVID-19. METHODS We performed a multicenter study of markers of neutrophil innate immunity in 155 cases consecutively recruited in a screening center, local hospitals, and two regional university hospitals. The cases were evaluated according to clinical and biological markers of severity and multi-organ manifestations and compared to 35 healthy controls. RESULTS Blood neutrophil elastase, histone-DNA, myeloperoxidase-DNA, and free dsDNA were dramatically increased, and DNase activity was decreased by 10-fold, compared with controls. Neutrophil elastase and histone-DNA were associated with intensive care admission, body temperature, lung damage, and markers of cardiovascular outcomes, renal failure, and increased interleukin-6 (IL-6), IL-8, and CXCR2. Neutrophil elastase was an independent predictor of the computed tomography score of COVID-19 lung damage and the number of affected organs, in multivariate analyses. The increased blood concentrations of NE and neutrophil extracellular traps were related to exacerbation of neutrophil stimulation through IL-8 and CXCR2 increased concentrations and increased serum DAMPs, and to impaired degradation of NETs as a consequence of the dramatic decrease in blood DNase activity. CONCLUSION Our results point out the key role of neutrophil innate immunity exacerbation in COVID-19. Neutrophil elastase and DNase could be potential biomarkers and therapeutic targets of severe systemic manifestations of COVID-19.
Collapse
Affiliation(s)
- Jean‐Louis Guéant
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | - Rosa‐Maria Guéant‐Rodriguez
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | - Julien Fromonot
- Center for CardioVascular and Nutrition Research (RG, JF)INSERM, INRA and Aix‐Marseille UniversityMarseilleFrance
| | - Abderrahim Oussalah
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | - Huguette Louis
- Department Inserm UMRS_1116 DCACUniversité de LorraineNancyFrance
| | - Celine Chery
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | - Mickael Gette
- Center for CardioVascular and Nutrition Research (RG, JF)INSERM, INRA and Aix‐Marseille UniversityMarseilleFrance
| | - Stanislas Gleye
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | - Jonas Callet
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | - Jeremie Raso
- Department InsermUMR_S 1256 Nutrition‐Genetics‐Environmental Risk ExposureUniversité de Lorraine and University Regional hospital of NancyNancyFrance
| | | | - Patrick Lacolley
- Department Inserm UMRS_1116 DCACUniversité de LorraineNancyFrance
| | - Régis Guieu
- Center for CardioVascular and Nutrition Research (RG, JF)INSERM, INRA and Aix‐Marseille UniversityMarseilleFrance
| | | |
Collapse
|
17
|
Al-Horani RA, Aliter KF, Kar S, Mottamal M. Sulfonated Nonsaccharide Heparin Mimetics Are Potent and Noncompetitive Inhibitors of Human Neutrophil Elastase. ACS OMEGA 2021; 6:12699-12710. [PMID: 34056422 PMCID: PMC8154244 DOI: 10.1021/acsomega.1c00935] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/13/2021] [Indexed: 05/03/2023]
Abstract
Human neutrophil elastase (HNE) is a serine protease that plays vital roles in inflammation, innate immune response, and tissue remodeling processes. HNE has been actively pursued as a drug target, particularly for the treatment of cardiopulmonary diseases. Although thousands of molecules have been reported to inhibit HNE, yet very few are being evaluated in early clinical trials, with sivelestat as the only approved HNE inhibitor. We report here a novel chemotype of sulfonated nonsaccharide heparin mimetics as potent and noncompetitive inhibitors of HNE. Using a chromogenic substrate hydrolysis assay, 14 sulfonated nonsaccharide heparin mimetics were tested for their inhibitory activity against HNE. Only 12 molecules inhibited HNE with IC50 values of 0.22-88.3 μM. The inhibition of HNE by these molecules was salt-dependent. Interestingly, a specific hexa-sulfonated molecule inhibited HNE with an IC50 value of 0.22 μM via noncompetitive mechanism, as demonstrated by Michaelis-Menten kinetics. The hexa-sulfonated derivative demonstrated at least 455-, 221-, 1590-, 21-, and 381-fold selectivity indices over other heparin-binding coagulation proteins including factors IIa, Xa, IXa, XIa, and FXIIIa, respectively. At the highest concentrations tested, the molecule also did not significantly inhibit other serine proteases of plasmin, trypsin, and chymotrypsin. Further supporting its selectivity, the molecule did not show heparin-like effects on clotting times of human plasma. The molecule also did not affect the proliferation of three cell lines at a concentration as high as 10 μM. Interestingly, the hexa-sulfonated molecule also inhibited cathepsin G with an IC50 value of 0.57 μM alluding to a dual anti-inflammatory action. A computational approach was exploited to identify putative binding site(s) for this novel class of HNE inhibitors. Overall, the reported hexa-sulfonated nonsaccharide heparin mimetic serves as a new platform to develop potent, selective, and noncompetitive inhibitors of HNE for therapeutic purposes.
Collapse
Affiliation(s)
- Rami A. Al-Horani
- Division
of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125, United States
- . Tel: (504) 520-7603. Fax: (504) 520-7954
| | - Kholoud F. Aliter
- Department
of Chemistry, School of STEM, Dillard University, New Orleans, Louisiana 70122, United States
| | - Srabani Kar
- Division
of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, Louisiana 70125, United States
| | - Madhusoodanan Mottamal
- Department
of Chemistry, Xavier University of Louisiana, New Orleans, Louisiana 70125, United States
| |
Collapse
|
18
|
Allaeys I, Ribeiro de Vargas F, Bourgoin SG, Poubelle PE. Human Inflammatory Neutrophils Express Genes Encoding Peptidase Inhibitors: Production of Elafin Mediated by NF-κB and CCAAT/Enhancer-Binding Protein β. THE JOURNAL OF IMMUNOLOGY 2021; 206:1943-1956. [PMID: 33762327 DOI: 10.4049/jimmunol.2000852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/11/2021] [Indexed: 11/19/2022]
Abstract
The concept of plasticity of neutrophils is highlighted by studies showing their ability to transdifferentiate into APCs. In this regard, transdifferentiated neutrophils were found at inflammatory sites of autoimmune arthritis (AIA). Exposure of neutrophils to inflammatory stimuli prolongs their survival, thereby favoring the acquisition of pathophysiologically relevant phenotypes and functions. By using microarrays, quantitative RT-PCR, and ELISAs, we showed that long-lived (LL) neutrophils obtained after 48 h of culture in the presence of GM-CSF, TNF, and IL-4 differentially expressed genes related to apoptosis, MHC class II, immune response, and inflammation. The expression of anti-inflammatory genes mainly of peptidase inhibitor families is upregulated in LL neutrophils. Among these, the PI3 gene encoding elafin was the most highly expressed. The de novo production of elafin by LL neutrophils depended on a synergism between GM-CSF and TNF via the activation and cooperativity of C/EBPβ and NF-κB pathways, respectively. Elafin concentrations were higher in synovial fluids (SF) of patients with AIA than in SF of osteoarthritis. SF neutrophils produced more elafin than blood counterparts. These results are discussed with respect to implications of neutrophils in chronic inflammation and the potential influence of elafin in AIA.
Collapse
Affiliation(s)
- Isabelle Allaeys
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| | - Flavia Ribeiro de Vargas
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| | - Sylvain G Bourgoin
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| | - Patrice E Poubelle
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| |
Collapse
|
19
|
Tanaka T, Basisty N, Fantoni G, Candia J, Moore AZ, Biancotto A, Schilling B, Bandinelli S, Ferrucci L. Plasma proteomic biomarker signature of age predicts health and life span. eLife 2020; 9:61073. [PMID: 33210602 PMCID: PMC7723412 DOI: 10.7554/elife.61073] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
Older age is a strong shared risk factor for many chronic diseases, and there is increasing interest in identifying aging biomarkers. Here, a proteomic analysis of 1301 plasma proteins was conducted in 997 individuals between 21 and 102 years of age. We identified 651 proteins associated with age (506 over-represented, 145 underrepresented with age). Mediation analysis suggested a role for partial cis-epigenetic control of protein expression with age. Of the age-associated proteins, 33.5% and 45.3%, were associated with mortality and multimorbidity, respectively. There was enrichment of proteins associated with inflammation and extracellular matrix as well as senescence-associated secretory proteins. A 76-protein proteomic age signature predicted accumulation of chronic diseases and all-cause mortality. These data support the use of proteomic biomarkers to monitor aging trajectories and to identify individuals at higher risk of disease to be targeted for in depth diagnostic procedures and early interventions.
Collapse
Affiliation(s)
- Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States
| | - Nathan Basisty
- The Buck Institute for Research on Aging, Novato, United States
| | - Giovanna Fantoni
- National Institute on Aging, Intramural Research Program, Clinical Research Core, NIH, Baltimore, United States
| | - Julián Candia
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, United States
| | - Ann Z Moore
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States
| | - Angelique Biancotto
- Precision Immunology, Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, United States
| | | | | | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, United States
| |
Collapse
|
20
|
Pacharra S, McMahon S, Duffy P, Basnett P, Yu W, Seisel S, Stervbo U, Babel N, Roy I, Viebahn R, Wang W, Salber J. Cytocompatibility Evaluation of a Novel Series of PEG-Functionalized Lactide-Caprolactone Copolymer Biomaterials for Cardiovascular Applications. Front Bioeng Biotechnol 2020; 8:991. [PMID: 32903548 PMCID: PMC7438451 DOI: 10.3389/fbioe.2020.00991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/29/2020] [Indexed: 11/24/2022] Open
Abstract
Although the use of bioresorbable materials in stent production is thought to improve long-term safety compared to their durable counterparts, a recent FDA report on the 2-year follow-up of the first FDA-approved bioresorbable vascular stent showed an increased occurrence of major adverse cardiac events and thrombosis in comparison to the metallic control. In order to overcome the issues of first generation bioresorbable polymers, a series of polyethylene glycol-functionalized poly-L-lactide-co-ε-caprolactone copolymers with varying lactide-to-caprolactone content is developed using a novel one-step PEG-functionalization and copolymerization strategy. This approach represents a new facile way toward surface enhancement for cellular interaction, which is shown by screening these materials regarding their cyto- and hemocompatibility in terms of cytotoxicity, hemolysis, platelet adhesion, leucocyte activation and endothelial cell adhesion. By varying the lactide-to-caprolactone polymer composition, it is possible to gradually affect endothelial and platelet adhesion which allows fine-tuning of the biological response based on polymer chemistry. All polymers developed were non-cytotoxic, had acceptable leucocyte activation levels and presented non-hemolytic (<2% hemolysis rate) behavior except for PLCL-PEG 55:45 which presented hemolysis rate of 2.5% ± 0.5. Water contact angles were reduced in the polymers containing PEG functionalization (PLLA-PEG: 69.8° ± 2.3, PCL-PEG: 61.2° ± 7.5) versus those without (PLLA: 79.5° ± 3.2, PCL: 76.4° ± 10.2) while the materials PCL-PEG550, PLCL-PEG550 90:10 and PLCL-PEG550 70:30 demonstrated best endothelial cell adhesion. PLLA-PEG550 and PLCL-PEG550 70:30 presented as best candidates for cardiovascular implant use from a cytocompatibility perspective across the spectrum of testing completed. Altogether, these polymers are excellent innovative materials suited for an application in stent manufacture due to the ease in translation of this one-step synthesis strategy to device production and their excellent in vitro cyto- and hemocompatibility.
Collapse
Affiliation(s)
- Sandra Pacharra
- Salber Laboratory, Centre for Clinical Research, Department of Experimental Surgery, Ruhr-Universität Bochum, Bochum, Germany
| | - Seán McMahon
- Laboratory A, Synergy Centre, Ashland Specialties Ireland Ltd., Dublin, Ireland
| | - Patrick Duffy
- Laboratory A, Synergy Centre, Ashland Specialties Ireland Ltd., Dublin, Ireland
| | - Pooja Basnett
- School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Wenfa Yu
- Rosenhahn Group, Faculty of Chemistry and Biochemistry, Analytical Chemistry - Biointerfaces, Ruhr-Universität Bochum, Bochum, Germany
| | - Sabine Seisel
- Faculty of Chemistry and Biochemistry, Analytical Chemistry - Center for Electrochemical Sciences, Ruhr-Universität Bochum, Bochum, Germany
| | - Ulrik Stervbo
- Centre for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- Centre for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Ipsita Roy
- Roy Group, Kroto Innovation Centre, Department of Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Richard Viebahn
- Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum GmbH, Bochum, Germany
| | - Wenxin Wang
- The Charles Institute of Dermatology, School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Jochen Salber
- Salber Laboratory, Centre for Clinical Research, Department of Experimental Surgery, Ruhr-Universität Bochum, Bochum, Germany.,Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum GmbH, Bochum, Germany
| |
Collapse
|
21
|
Li K, Zhang F, Wei L, Han Z, Liu X, Pan Y, Guo C, Han W. Recombinant Human Elafin Ameliorates Chronic Hyperoxia-Induced Lung Injury by Inhibiting Nuclear Factor-Kappa B Signaling in Neonatal Mice. J Interferon Cytokine Res 2020; 40:320-330. [PMID: 32460595 DOI: 10.1089/jir.2019.0241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The study aimed to investigate whether recombinant human elafin can prevent hyperoxia-induced pulmonary inflammation in newborn mice, and to explore the mechanism underlying the inhibitory effects of elafin on nuclear factor-kappa B (NF-κB) signaling pathway. Neonatal C57BL/6J mice were exposed to 85% O2 for 1, 3, 7, 14, or 21 days. Then, elafin was administered daily for 20 days through intraperitoneal injection. After treatment, morphometric analysis, quantitative real-time polymerase chain reaction, terminal deoxynucleotidyl transferase dUTP nick end labeling staining, and Western blotting were carried out to determine the key markers involved in inflammatory process and the potential signaling pathways in hyperoxia-exposed newborn mice treated with elafin. In neonatal bronchopulmonary dysplasia (BPD) mice, hyperoxia induced apoptosis by increasing Bcl-2-associated X protein expression, and triggered inflammation by upregulating the expression levels of interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor-α. Moreover, hyperoxia activated NF-κB signaling pathway by promoting the nuclear translocation of p65 in lung tissue. However, all these changes could be inhibited or reversed by elafin at least partially. Elafin reduced apoptosis, suppressed inflammation cytokines, and improved NF-κB p65 nuclear accumulation in hyperoxia-exposed neonatal mice, indicating that this recombinant protein can serve as a novel target for the treatment of BPD.
Collapse
Affiliation(s)
- Kexin Li
- Laboratory Animal Center, Chongqing Medical University, Chongqing, P.R. China
| | - Fengmei Zhang
- Laboratory Animal Center, Chongqing Medical University, Chongqing, P.R. China
| | - Li Wei
- Centre for Lipid Research and Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, P.R. China
| | - Zhigang Han
- Laboratory Animal Center, Chongqing Medical University, Chongqing, P.R. China
| | - Xuwei Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Yongquan Pan
- Laboratory Animal Center, Chongqing Medical University, Chongqing, P.R. China
| | - Chunbao Guo
- Ministry of Education Key Laboratory of Child Development and Disorders, Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China.,Department of Hepatology and Liver Transplantation Center, Children's Hospital, Chongqing Medical University, Chongqing, P.R. China
| | - Wenli Han
- Laboratory Animal Center, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
22
|
Ghule BV, Kotagale NR, Patil KS. Inhibition of the pro-inflammatory mediators in rat neutrophils by shanzhiside methyl ester and its acetyl derivative isolated from Barleria prionitis. JOURNAL OF ETHNOPHARMACOLOGY 2020; 249:112374. [PMID: 31704416 DOI: 10.1016/j.jep.2019.112374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 09/21/2019] [Accepted: 10/31/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The aerial parts of Barleria prionitis Linn. (BP) (Acanthaceae) plant has long been used to treat inflammatory disorders such as toothache, swellings, arthritis and gout. AIM OF THE STUDY The purpose of this study was to evaluate the effects of shanzhiside methyl ester (SME), 8-O-acetyl shanzhiside methyl ester (ASME) and iridoid glycosides rich monoterpenoidal fraction (IFBp), isolated from the aerial part of BP, on the pro-inflammatory mediators in stimulated rat neutrophils. MATERIALS AND METHODS Rat neutrophils were incubated with or without test drugs. The influence of laboratory isolated and identified SME, ASME and IFBp on the production and release of pro-inflammatory mediators i.e. myeloperoxidase (MPO), elastase, matrix metalloproteinase-9 (MMP-9), interleukin 8 (IL-8), tumor necrosis factor alpha (TNF-α) and leukotriene B4 (LTB4) was evaluated in the formyl-met-leu-phenylalanine (f-MLP) and lipopolysaccharide (LPS) stimulated rat neutrophils using enzyme-linked immunosorbent assay (ELISA) methods. IFBp was also standardized with the high performance thin layer chromatography by simultaneous determination of SME and ASME marker compounds. RESULTS SME, ASME and IFBp displayed concentration-dependent inhibitory effects on the MPO, elastase and MMP-9 enzymes release, and IL-8, TNF-α and LTB4 cytokines production in the f-MLP and LPS stimulated rat neutrophils. The content of SME and ASME was found to be 17.32 ± 1.98 and 11.30 ± 1.06% w/w, respectively, in IFBp by HPTLC method. CONCLUSION Altogether, the present results suggest that the iridoidal glycosides of BP may be considered as therapeutic strategy against neutrophil-mediated inflammatory diseases. Developed and validated HPTLC method for the standardization of IFBp of BP can be used as a quality control tool for the routine qualitative and quantitative analysis of Barleria species containing SME and/or ASME.
Collapse
Affiliation(s)
- B V Ghule
- Government College of Pharmacy, Kathora Naka, Amravati, 444 604, Maharashtra State, India; Institute of Pharmaceutical Education and Research, Wardha, 442 001, Maharashtra State, India.
| | - N R Kotagale
- Government College of Pharmacy, Kathora Naka, Amravati, 444 604, Maharashtra State, India.
| | - K S Patil
- Government College of Pharmacy, Kathora Naka, Amravati, 444 604, Maharashtra State, India; Institute of Pharmaceutical Education and Research, Wardha, 442 001, Maharashtra State, India.
| |
Collapse
|
23
|
Shavadia JS, Granger CB, Alemayehu W, Westerhout CM, Povsic TJ, Brener SJ, van Diepen S, Defilippi C, Armstrong PW. High-throughput targeted proteomics discovery approach and spontaneous reperfusion in ST-segment elevation myocardial infarction. Am Heart J 2020; 220:137-144. [PMID: 31812755 DOI: 10.1016/j.ahj.2019.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 09/20/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Although spontaneous reperfusion (SR) prior to primary percutaneous coronary intervention (pPCI) is associated with improved outcomes, its pathophysiology remains unclear. The objective of the study was to explore associations between SR in ST-segment elevation myocardial infarction (STEMI) using a multimarker cardiovascular proteins strategy METHODS: We evaluated STEMI patients from the Assessment of Pexelizumab in Acute Myocardial Infarction trial treated with pPCI within 6 hours from symptom onset. SR was core laboratory-defined as pre-PCI Thrombolysis in Myocardial Infarction flow 2 or 3. Ninety-one cardiovascular disease-related serum biomarkers drawn prior to PCI were analyzed using a high-throughput "targeted discovery" panel. Expression levels for individual biomarkers were compared between patients with/without SR. A hierarchical clustering method of biomarkers identified clusters of biomarkers that differentiated the 2 groups. Associations between individual biomarkers and clusters with SR were further evaluated by multivariable logistic regression. RESULTS Of 683 patients studied, 290 had spontaneous reperfusion; those with compared to without SR were more likely noninferior STEMI and had lower clinical acuity and lower baseline levels of troponin and creatine kinase. SR was associated with a lower occurrence of 90-day composite of death, heart failure, or cardiogenic shock. Fifty-two of 91 individual biomarkers were significantly univariably associated with SR. Forty-five remained significant with adjustment for false discovery rate. Using cluster analysis, 26 biomarkers clusters were identified, explaining 72% of total covariance, and 13 biomarker clusters were significantly associated with SR after multivariable adjustment. SR was associated with higher mean expression levels of proteins in all 13 clusters. The cluster most strongly associated with SR consisted of novel proteins across various distinct, yet interlinked, pathobiological processes (kallikrein-6, matrix extracellular phosphoglycoprotein, matrix mettaloproteinaise-3, and elafin). CONCLUSIONS Spontaneous reperfusion prior to pPCI in STEMI was associated with a lower risk of adverse clinical events. These exploratory data from a targeted discovery proteomics platform identifies novel proteins across diverse, yet complementary, pathobiological axes that show promise in providing mechanistic insights into spontaneous reperfusion in STEMI. CONDENSED ABSTRACT Spontaneous reperfusion has been established with improved STEMI outcomes, yet its pathobiology is unclear and appears to involve diverse physiological processes. Using a 91-biomarker high-throughput proteomics platform, we studied 683 STEMI patients in the APEX AMI trial (290 had core laboratory-adjudicated pre-PCI TIMI 2/3 flow) and identified 52 proteins that univariably associate with spontaneous reperfusion. Cluster analysis identified 26 biomarker clusters (explaining 72% of total variance), 13 of which, after multivariable adjustment, were significantly associated with spontaneous reperfusion. Four proteins (kallikrein-6, matrix extracellular phosphoglycoprotein, matrix mettaloproteinaise-3, and elafin) across diverse, yet complementary, pathways appear to be associated most strongly with spontaneous reperfusion.
Collapse
Affiliation(s)
- Jay S Shavadia
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; Duke Clinical Research Institute, Durham, NC, USA.
| | | | | | | | | | - Sorin J Brener
- Department of Medicine, Cardiac Catheterization Laboratory, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY, USA
| | - Sean van Diepen
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | - Paul W Armstrong
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Extracellular matrix-cell interactions: Focus on therapeutic applications. Cell Signal 2019; 66:109487. [PMID: 31778739 DOI: 10.1016/j.cellsig.2019.109487] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Extracellular matrix (ECM) macromolecules together with a multitude of different molecules residing in the extracellular space play a vital role in the regulation of cellular phenotype and behavior. This is achieved via constant reciprocal interactions between the molecules of the ECM and the cells. The ECM-cell interactions are mediated via cell surface receptors either directly or indirectly with co-operative molecules. The ECM is also under perpetual remodeling process influencing cell-signaling pathways on its part. The fragmentation of ECM macromolecules provides even further complexity for the intricate environment of the cells. However, as long as the interactions between the ECM and the cells are in balance, the health of the body is retained. Alternatively, any dysregulation in these interactions can lead to pathological processes and finally to various diseases. Thus, therapeutic applications that are based on retaining normal ECM-cell interactions are highly rationale. Moreover, in the light of the current knowledge, also concurrent multi-targeting of the complex ECM-cell interactions is required for potent pharmacotherapies to be developed in the future.
Collapse
|
25
|
Bahadır Acıkara Ö, Ilhan M, Kurtul E, Šmejkal K, Küpeli Akkol E. Inhibitory activity of Podospermum canum and its active components on collagenase, elastase and hyaluronidase enzymes. Bioorg Chem 2019; 93:103330. [PMID: 31614286 DOI: 10.1016/j.bioorg.2019.103330] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 08/21/2019] [Accepted: 09/28/2019] [Indexed: 10/25/2022]
Abstract
Present study is aimed to investigate in vitro inhibitory effects of the extract prepared from the aerial parts of Podospermum canum (syn: Scorzonera cana var. jacquiniana) (Asteraceae) on hyaluronidase, collagenase, and elastase enzymes using a bioassay-guided fractionation. Inhibitory effects of the extract, sub-extracts, fractions obtained by column chromatography, and isolated compounds on collagenase, elastase, and hyaluronidase were performed by using in vitro enzyme inhibitory assays based on spectrophotometric evaluation. The methanolic extract obtained from P. canum exhibited strong inhibitory activities on elastase and collagenase while the insignificant activity was observed on hyaluronidase. Through bioactivity-guided fractionation, the ethyl acetate and remaining water sub-extracts obtained from the methanolic extract displayed significant inhibitory activities on collagenase and elastase, while petroleum ether and chloroform extracts did not show any inhibitory activity. Eleven known compounds: arbutin, 6́-O-caffeoylarbutin, cichoriin, 3,5-dicaffeoylquinic acid methyl ester, apigenin 7-O-β-glucoside, luteolin 7-O-β-glucoside, apigenin 7-O-β-rutinoside, isoorientin, orientin, vitexin, procatechuic acid, and new compound 4-hydroxy-benzoic acid 4-(6-O-α-rhamnopyranosyl-β-glucopyranosyl) benzyl ester have been obtained from ethyl acetate sub-extract. Results of the present study have revealed that apigenin 7-O-β-glucoside, luteolin 7-O-β-glucoside, apigenin 7-O-β-rutinoside, and isoorientin showed potent enzyme inhibitory activities. However, methanolic extract of P. canum displayed a greater inhibitory activity than fractions and isolated compounds both on collagenase and elastase.
Collapse
Affiliation(s)
- Özlem Bahadır Acıkara
- Department of Pharmacognosy, Faculty of Pharmacy, Ankara University, Tandoğan, 06100 Ankara, Turkey
| | - Mert Ilhan
- Department of Pharmacognosy, Faculty of Pharmacy, Van Yüzüncü Yıl University, Tuşba 65080, Van, Turkey
| | - Ekin Kurtul
- Department of Pharmacognosy, Faculty of Pharmacy, Ankara University, Tandoğan, 06100 Ankara, Turkey
| | - Karel Šmejkal
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 61242 Brno, Czech Republic
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Etiler, 06330 Ankara, Turkey.
| |
Collapse
|
26
|
Crocetti L, Quinn MT, Schepetkin IA, Giovannoni MP. A patenting perspective on human neutrophil elastase (HNE) inhibitors (2014-2018) and their therapeutic applications. Expert Opin Ther Pat 2019; 29:555-578. [PMID: 31204543 PMCID: PMC9642779 DOI: 10.1080/13543776.2019.1630379] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/07/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Human neutrophil elastase (HNE) is involved in a variety of serious chronic diseases, especially cardiopulmonary pathologies. For this reason, the regulation of HNE activity represents a promising therapeutic approach, which is evident by the development of a number of new and selective HNE inhibitors, both in the academic and pharmaceutical environments. AREAS COVERED The present review analyzes and summarizes the patent literature regarding human neutrophil elastase inhibitors for the treatment of cardiopulmonary diseases over 2014-2018. EXPERT OPINION HNE is an interesting and defined target to treat various inflammatory diseases, including a number of cardiopulmonary pathologies. The research in this field is quite active, and a number of HNE inhibitors are currently in various stages of clinical development. In addition, new opportunities for HNE inhibitor development stem from recent studies demonstrating the involvement of HNE in many other inflammatory pathologies, including rheumatoid arthritis, inflammatory bowel disease, skin diseases, and cancer. Furthermore, the development of dual HNE/proteinase 3 inhibitors is being pursued as an innovative approach for the treatment of neutrophilic inflammatory diseases. Thus, these new developments will likely stimulate new and increased interest in this important therapeutic target and for the development of novel and selective HNE inhibitors.
Collapse
Affiliation(s)
- L Crocetti
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - MT Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - IA Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - MP Giovannoni
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| |
Collapse
|
27
|
Intracardiac administration of neutrophil protease cathepsin G activates noncanonical inflammasome pathway and promotes inflammation and pathological remodeling in non-injured heart. J Mol Cell Cardiol 2019; 134:29-39. [PMID: 31252040 DOI: 10.1016/j.yjmcc.2019.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Inflammatory serine proteases (ISPs) play an important role in cardiac repair after injury through hydrolysis of dead cells and extracellular matrix (ECM) debris. Evidence also suggests an important role of ISPs in the coordination of the inflammatory response. However, the effect of ISPs on inflammation is obfuscated by the confounding factors associated with cell death and inflammatory cell infiltration induced after cardiac injury. This study investigated whether neutrophil-derived cathepsin G (Cat.G) influences inflammation and remodeling in the absence of prior cardiac injury and cell death. METHODS AND RESULTS Intracardiac catheter delivery of Cat.G (1 mg/kg) in rats induced significant left ventricular (LV) dilatation and cardiac contractile dysfunction at day 5, but not at day 2, post-delivery compared to vehicle-treated animals. Cat.G delivery also significantly increased matrix metalloprotease activity and collagen and fibronectin degradation at day 5 compared to vehicle-treated rats and these changes were associated with increased death signaling pathways and myocyte apoptosis. Mechanistic analysis shows that Cat.G-treatment induced potent chemotactic activity in hearts at day 2 and 5 post-delivery, characterized by processing and activation of interleukin (IL)-1β and IL-18, stimulation of inflammatory signaling pathways and accumulation of myeloid cells when compared to vehicle-treated rats. Cat.G-induced processing of IL-1β and IL-18 was independent of the canonical NLRP-3 inflammasome pathway and treatment of isolated cardiomyocytes with inhibitors of NLRP-3 or caspase-1 failed to reduce Cat.G-induced cardiomyocyte death. Notably, rats treated with IL-1 receptor antagonist (IL-1Ra) show reduced inflammation and improved cardiac remodeling and function following Cat.G delivery. CONCLUSIONS Cat.G exerts potent chemoattractant and pro-inflammatory effects in non-stressed or injured heart in part through processing and activation of IL-1 family cytokines, subsequently leading to adverse cardiac remodeling and function. Thus, targeting ISPs could be a novel therapeutic strategy to reduce cardiac inflammation and improve cardiac remodeling and function after injury or stress.
Collapse
|
28
|
Kulasingam A, Hvas AM, Grove EL, Funck KL, Kristensen SD. Detection of biomarkers using a novel proximity extension assay in patients with ST-elevation myocardial infarction. Thromb Res 2018; 172:21-28. [DOI: 10.1016/j.thromres.2018.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/03/2018] [Accepted: 10/11/2018] [Indexed: 11/15/2022]
|
29
|
Bronze-da-Rocha E, Santos-Silva A. Neutrophil Elastase Inhibitors and Chronic Kidney Disease. Int J Biol Sci 2018; 14:1343-1360. [PMID: 30123081 PMCID: PMC6097478 DOI: 10.7150/ijbs.26111] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/18/2018] [Indexed: 12/11/2022] Open
Abstract
End-stage renal disease (ESRD), the last stage of chronic kidney disease (CKD), is characterized by chronic inflammation and oxidative stress. Neutrophils are the front line cells that mediate an inflammatory response against microorganisms as they can migrate, produce reactive oxygen species (ROS), secrete neutrophil serine proteases (NSPs), and release neutrophil extracellular traps (NETs). Serine proteases inhibitors regulate the activity of serine proteases and reduce neutrophil accumulation at inflammatory sites. This review intends to relate the role of neutrophil elastase in CKD and the effects of neutrophil elastase inhibitors in predicting or preventing inflammation.
Collapse
Affiliation(s)
- Elsa Bronze-da-Rocha
- UCIBIO/REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | | |
Collapse
|
30
|
Serine protease inhibitors containing a Kunitz domain: their role in modulation of host inflammatory responses and parasite survival. Microbes Infect 2018; 20:606-609. [PMID: 29355617 DOI: 10.1016/j.micinf.2018.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/09/2018] [Indexed: 11/21/2022]
Abstract
Proteins containing a Kunitz domain have the typical serine protease inhibition function ranging from sea anemone to man. Protease inhibitors play major roles in infection, inflammation disorders and cancer. This review discusses the role of serine proteases containing a Kunitz domain in immunomodulation induced by helminth parasites. Helminth parasites are associated with protection from inflammatory conditions. Therefore, interest has raised whether worm parasites or their products hold potential as drugs for treatment of immunological disorders. Finally, we also propose the use of recombinant SmKI-1 from Schistosoma mansoni as a potential therapeutic molecule to treat inflammatory diseases.
Collapse
|
31
|
Morais SB, Figueiredo BC, Assis NRG, Alvarenga DM, de Magalhães MTQ, Ferreira RS, Vieira AT, Menezes GB, Oliveira SC. Schistosoma mansoni SmKI-1 serine protease inhibitor binds to elastase and impairs neutrophil function and inflammation. PLoS Pathog 2018; 14:e1006870. [PMID: 29425229 PMCID: PMC5823468 DOI: 10.1371/journal.ppat.1006870] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 02/22/2018] [Accepted: 01/09/2018] [Indexed: 12/27/2022] Open
Abstract
Protease inhibitors have important function during homeostasis, inflammation and tissue injury. In this study, we described the role of Schistosoma mansoni SmKI-1 serine protease inhibitor in parasite development and as a molecule capable of regulating different models of inflammatory diseases. First, we determine that recombinant (r) SmKI-1 and its Kunitz domain but not the C-terminal region possess inhibitory activity against trypsin and neutrophil elastase (NE). To better understand the molecular basis of NE inhibition by SmKI-1, molecular docking studies were also conducted. Docking results suggest a complete blockage of NE active site by SmKI-1 Kunitz domain. Additionally, rSmKI-1 markedly inhibited the capacity of NE to kill schistosomes. In order to further investigate the role of SmKI-1 in the parasite, we designed specific siRNA to knockdown SmKI-1 in S. mansoni. SmKI-1 gene suppression in larval stage of S. mansoni robustly impact in parasite development in vitro and in vivo. To determine the ability of SmKI-1 to interfere with neutrophil migration and function, we tested SmKI-1 anti-inflammatory potential in different murine models of inflammatory diseases. Treatment with SmKI-1 rescued acetaminophen (APAP)-mediated liver damage, with a significant reduction in both neutrophil recruitment and elastase activity. In the model of gout arthritis, this protein reduced neutrophil accumulation, IL-1β secretion, hypernociception, and overall pathological score. Finally, we demonstrated the ability of SmKI-1 to inhibit early events that trigger neutrophil recruitment in pleural cavities of mice in response to carrageenan. In conclusion, SmKI-1 is a key protein in S. mansoni survival and it has the ability to inhibit neutrophil function as a promising therapeutic molecule against inflammatory diseases. Schistosoma mansoni is one of the main agents of schistosomiasis, which is the most important human helminthic infection in terms of global morbidity and mortality. Although schistosomiasis represents a major public health problem in endemic countries, evidences show that S. mansoni downregulates inflammatory responses in many diseases. Fortunately, the control of inflammatory responses is extended to pathogen-derived antigens, leading us to study one S. mansoni Kunitz type protease inhibitor (SmKI-1), found in larval and adult phases of the parasite. We demonstrate that SmKI-1 inhibits trypsin and neutrophil elastase (NE). Additionally, live parasites that SmKI-1 gene has been suppressed using siRNA displayed an impaired schistosome development both in vitro and in vivo. Further, we demonstrate that SmKI-1 possesses an anti-inflammatory potential in three different murine models of inflammatory diseases: acetaminophen (APAP)-mediated liver damage, gout arthritis, and pleural inflammation in response to carrageenan. In these inflammatory disease models, we evaluated SmKI-1 effect on neutrophil and our results demonstrate this molecule is able to inhibit neutrophil migration and function, regulating inflammation. Thus, our data suggest that SmKI-1 is a promising therapeutic molecule against inflammatory diseases.
Collapse
Affiliation(s)
- Suellen B. Morais
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Barbara C. Figueiredo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Natan R. G. Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
| | - Debora M. Alvarenga
- Centro de Biologia Gastrointestinal, Departamento de Morfologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariana T. Q. de Magalhães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rafaela S. Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Angélica T. Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo B. Menezes
- Centro de Biologia Gastrointestinal, Departamento de Morfologia do Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C. Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação Salvador, Bahia, Brazil
- * E-mail:
| |
Collapse
|
32
|
Dong W, Lv H, Guo K, Wang T, Ouyang Y, Jin M, Zhang Y. Classical Swine Fever Virus Infection and Its NS4A Protein Expression Induce IL-8 Production through MAVS Signaling Pathway in Swine Umbilical Vein Endothelial Cells. Front Microbiol 2018; 8:2687. [PMID: 29375538 PMCID: PMC5770398 DOI: 10.3389/fmicb.2017.02687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/26/2017] [Indexed: 12/16/2022] Open
Abstract
Classical swine fever virus (CSFV) infection causes a severe disease of pigs, which is characterized by hemorrhage, disseminated intravascular coagulation, and leucopenia. IL-8, a main chemokine and activator of neutrophils, regulates the permeability of endothelium, which may be related to the hemorrhage upon CSFV infection. Until now, the molecular mechanisms of IL-8 regulation during CSFV infection are poorly defined. Here, we showed that CSFV infection induced IL-8 production and the upregulation of IL-8 required virus replication in swine umbilical vein endothelial cells (SUVECs). Additionally, MAVS expression was increased and was required for IL-8 production upon CSFV infection. Moreover, ROS was involved in CSFV-induced IL-8 production. Subsequent studies demonstrated that ROS was involved in MAVS-induced IL-8 production and CSFV induced ROS production through MAVS pathway. These results indicate that CSFV induces IL-8 production through MAVS pathway and production of ROS. The role of NS4A in the pathogenesis of CSFV is not well-understood. In this study, we further demonstrated that CSFV NS4A induced IL-8 production through enhancing MAVS pathway and promoted CSFV replication. In addition, we discovered that CSFV NS4A was localized in the cell nucleus and cytoplasm, including endoplasmic reticulum (ER) and mitochondria. Taken together, these results provide insights into the mechanisms of IL-8 regulation and NS4A functions during CSFV infection.
Collapse
Affiliation(s)
- Wang Dong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Huifang Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Tao Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yueling Ouyang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Mingxing Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
33
|
Liu R, Chen L, Wu W, Chen H, Zhang S. Neutrophil serine proteases and their endogenous inhibitors in coronary artery ectasia patients. Anatol J Cardiol 2016; 16:23-8. [PMID: 26467359 PMCID: PMC5336701 DOI: 10.5152/akd.2015.6072] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Objective: Proteolytic enzymes possibly contribute to coronary artery ectasia (CAE). This study aimed to determine whether neutrophils, neutrophil serine proteases (NSPs), and their endogenous inhibitors participated in the pathological process of CAE. Methods: The study consisted of 30 patients with CAE, 30 patients with coronary artery disease (CAD), and 29 subjects with normal coronary arteries (Control). The following circulating items were measured: the main NSPs, including human neutrophil elastase (HNE), cathepsin G (CG), and proteinase 3 (PR3); soluble elastin (sElastin), which was a degradation product of elastin fibres; NSP inhibitors such as α1-protease inhibitor (α1-PI), α2-macroglobulin (α2-MG), secretory leucoprotease inhibitor (SLPI), and elafin; as well as two neutrophil activation markers (myeloperoxidase and lactoferrin) and three classic neutrophil activators [tumor necrosis factor-α (TNF-α), interleukin-8 (IL-8), and bacterial endotoxin]. Results: The levels of HNE, CG, and sElastin were elevated in the CAE group. The levels of α1-PI and α2-MG were also significantly increased in the CAE group. The levels of myeloperoxidase and lactoferrin were higher in the CAE group. The levels of TNF-α, IL-8, and endotoxin were unchanged in the CAE group compared with those in the CAD group. Conclusion: Neutrophils may participate in the process of vessel extracellular matrix destruction and coronary ectasia by releasing NSPs in a non-classical manner.
Collapse
Affiliation(s)
- Ruifeng Liu
- Cardiac Department, Peking Union Medical College Hospital (PUMCH), Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing-China.
| | | | | | | | | |
Collapse
|
34
|
Lee KS, Kim BY, Yoon HJ, Choi YS, Jin BR. Secapin, a bee venom peptide, exhibits anti-fibrinolytic, anti-elastolytic, and anti-microbial activities. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 63:27-35. [PMID: 27208884 DOI: 10.1016/j.dci.2016.05.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/15/2016] [Accepted: 05/16/2016] [Indexed: 05/27/2023]
Abstract
Bee venom contains a variety of peptide constituents that have various biological, toxicological, and pharmacological actions. However, the biological actions of secapin, a venom peptide in bee venom, remain largely unknown. Here, we provide the evidence that Asiatic honeybee (Apis cerana) secapin (AcSecapin-1) exhibits anti-fibrinolytic, anti-elastolytic, and anti-microbial activities. The recombinant mature AcSecapin-1 peptide was expressed in baculovirus-infected insect cells. AcSecapin-1 functions as a serine protease inhibitor-like peptide that has inhibitory effects against plasmin, elastases, microbial serine proteases, trypsin, and chymotrypsin. Consistent with these functions, AcSecapin-1 inhibited the plasmin-mediated degradation of fibrin to fibrin degradation products, thus indicating the role of AcSecapin-1 as an anti-fibrinolytic agent. AcSecapin-1 also inhibited both human neutrophil and porcine pancreatic elastases. Furthermore, AcSecapin-1 bound to bacterial and fungal surfaces and exhibited anti-microbial activity against fungi and gram-positive and gram-negative bacteria. Taken together, our data demonstrated that the bee venom peptide secapin has multifunctional roles as an anti-fibrinolytic agent during fibrinolysis and an anti-microbial agent in the innate immune response.
Collapse
Affiliation(s)
- Kwang Sik Lee
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea
| | - Bo Yeon Kim
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea
| | - Hyung Joo Yoon
- Department of Agricultural Biology, National Academy of Agricultural Science, Wanju 55365, Republic of Korea
| | - Yong Soo Choi
- Department of Agricultural Biology, National Academy of Agricultural Science, Wanju 55365, Republic of Korea
| | - Byung Rae Jin
- College of Natural Resources and Life Science, Dong-A University, Busan 604-714, Republic of Korea.
| |
Collapse
|
35
|
Regn M, Laggerbauer B, Jentzsch C, Ramanujam D, Ahles A, Sichler S, Calzada-Wack J, Koenen RR, Braun A, Nieswandt B, Engelhardt S. Peptidase inhibitor 16 is a membrane-tethered regulator of chemerin processing in the myocardium. J Mol Cell Cardiol 2016; 99:57-64. [PMID: 27539859 DOI: 10.1016/j.yjmcc.2016.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 08/09/2016] [Accepted: 08/15/2016] [Indexed: 10/21/2022]
Abstract
A key response of the myocardium to stress is the secretion of factors with paracrine or endocrine function. Intriguing in this respect is peptidase inhibitor 16 (PI16), a member of the CAP family of proteins which we found to be highly upregulated in cardiac disease. Up to this point, the mechanism of action and physiological function of PI16 remained elusive. Here, we show that PI16 is predominantly expressed by cardiac fibroblasts, which expose PI16 to the interstitium via a glycophosphatidylinositol (-GPI) membrane anchor. Based on a reported genetic association of PI16 and plasma levels of the chemokine chemerin, we investigated whether PI16 regulates post-translational processing of its precursor pro-chemerin. PI16-deficient mice were engineered and found to generate higher levels of processed chemerin than wildtype mice. Purified recombinant PI16 efficiently inhibited cathepsin K, a chemerin-activating protease, in vitro. Moreover, we show that conditioned medium from PI16-overexpressing cells impaired the activation of pro-chemerin. Together, our data indicate that PI16 suppresses chemerin activation in the myocardium and suggest that this circuit may be part of the cardiac stress response.
Collapse
Affiliation(s)
- Michael Regn
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Bernhard Laggerbauer
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Claudia Jentzsch
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Andrea Ahles
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Sonja Sichler
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Julia Calzada-Wack
- Institute of Pathology, Helmholtz Center Munich, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Attila Braun
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider Straße 2, 97080 Würzburg, Germany
| | - Bernhard Nieswandt
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider Straße 2, 97080 Würzburg, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Straße 29, 80802 Munich, Germany; DZHK (German Center for Cardiovascular Research) partner site Munich Heart Alliance, 80802 Munich, Germany.
| |
Collapse
|
36
|
Muto J, Fujimoto N, Ono K, Kobayashi T, Chen KR, Suzuki S, Wachi H, Tajima S. Deposition of elafin in the involved vascular wall of neutrophil-mediated cutaneous vasculitis. J Eur Acad Dermatol Venereol 2016; 30:1544-9. [PMID: 27060697 DOI: 10.1111/jdv.13650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/15/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Neutrophil elastase plays an important role in skin inflammation induced by neutrophil infiltration. Elafin is an inducible elastase inhibitor expressed by keratinocytes, and is known to be involved in pathogenesis of neutrophilic skin disorders such as psoriasis. METHODS Immunohistochemical studies of elafin expression in the cases of vasculitis were performed. Induction of elafin expression in cultured vascular cells and its effect on neutrophil migration were studied in vitro. RESULTS A positive immunoreactivity was detected in polyarteritis nodosa, giant cell arteritis and Schönlein-Henoch purpura, but no immunoreactivity was found in Churg-Strauss syndrome. Elafin expression in cultured venous endothelial cells and arterial smooth muscle cells was undetectable, but induced by interleukin-1β (IL-1β) and IL-8. Elafin inhibited the elastin peptide-induced neutrophil chemotaxis at the concentration of 10(-8) -10(-5) mol/L. CONCLUSION Elafin deposition induced by cytokines (IL-1β or IL-8) will be an important regulator for the progress of leucocytoclastic vasculitis by functioning as an inhibitor for neutrophil chemotaxis as well as for vascular elastin degradation.
Collapse
Affiliation(s)
- J Muto
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| | - N Fujimoto
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| | - K Ono
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| | - T Kobayashi
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| | - K R Chen
- Department of Dermatology, Saiseikai Central Hospital, Tokyo, Japan
| | - S Suzuki
- Department of Medicine, National Defense Medical College, Saitama, Japan
| | - H Wachi
- Department of Clinical Chemistry, Hoshi College of Pharmacy, Tokyo, Japan
| | - S Tajima
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| |
Collapse
|
37
|
Duca L, Blaise S, Romier B, Laffargue M, Gayral S, El Btaouri H, Kawecki C, Guillot A, Martiny L, Debelle L, Maurice P. Matrix ageing and vascular impacts: focus on elastin fragmentation. Cardiovasc Res 2016; 110:298-308. [DOI: 10.1093/cvr/cvw061] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/07/2016] [Indexed: 12/17/2022] Open
|
38
|
Wu W, Liu R, Chen L, Chen H, Zhang S. Disequilibrium of Blood Coagulation and Fibrinolytic System in Patients With Coronary Artery Ectasia. Medicine (Baltimore) 2016; 95:e2779. [PMID: 26937905 PMCID: PMC4779002 DOI: 10.1097/md.0000000000002779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Thrombus formation and myocardial infarction are not uncommon in patients with coronary artery ectasia (CAE). In light of this, the present study aims to systemically evaluate the blood coagulation and fibrinolytic systems in CAE patients. In this study, we enrolled 30 patients with CAE, 30 patients with coronary atherosclerosis disease (CAD), and 29 subjects with normal coronary arteries (control). The coagulation system was evaluated using a routine coagulation function test performed in the hospital laboratory before coronary angiography, and measurements included prothrombin time, international normalized ratio, activated partial thromboplastin time, fibrinogen time, and thrombin time. The evaluation of the fibrinolytic system included measurements of D-dimer, euglobulin lysis time, plasminogen activator inhibitor 1, plasminogen, plasminogen activity assay, α1-antitrypsin (α1-AT), α2 plasmin inhibitor (α2-PI), and α2-macroglobulin (α2-MG). Alpha1-AT, α2-PI, and α2-MG also inhibit activities of 3 neutrophil serine proteases, namely human neutrophil elastase (HNE), cathepsin G (CG), and proteinase 3 (PR3); therefore, the plasma levels of these 3 proteinases were also evaluated.In CAE patients, the circulating coagulation system was normal. For the fibrinolytic system, a decrease of plasminogen activity was observed (P = 0.029) when compared with CAD patients, and the concentrations of α1-AT (both P < 0.001), α2-PI (P = 0.002 and P = 0.025), and α2-MG (P = 0.034 and P < 0.001) were significantly elevated when compared with CAD patients and normal controls. Moreover, the plasma levels of HNE (both P < 0.001) and CG (P = 0.027 and 0.016) in CAE patients were also significantly higher than those of the CAD and control groups. There was no difference in plasma PR3 concentration among these 3 groups.Disequilibrium of the coagulation/fibrinolytic system may contribute to thrombus formation and clinical coronary events in patients with CAE. The increased plasma concentrations of α1-AT, α2-PI, and α2-MG might provide beneficial effects by inhibiting the proteinases and restraining the ectatic process; on other hand, they led to unfavorable results by inhibiting plasmin and decreasing thrombus degradation in CAE patients.
Collapse
Affiliation(s)
- Wei Wu
- From the Department of Cardiology, Chinese Academy of Medical Sciences and Peking Union Medical College, Peking Union Medical College Hospital (WW, RL, LC, SZ); and National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College (HC), Beijing, China
| | | | | | | | | |
Collapse
|
39
|
Alam SR, Lewis SC, Zamvar V, Pessotto R, Dweck MR, Krishan A, Goodman K, Oatey K, Harkess R, Milne L, Thomas S, Mills NM, Moore C, Semple S, Wiedow O, Stirrat C, Mirsadraee S, Newby DE, Henriksen PA. Perioperative elafin for ischaemia-reperfusion injury during coronary artery bypass graft surgery: a randomised-controlled trial. Heart 2015; 101:1639-45. [PMID: 26310261 PMCID: PMC4621368 DOI: 10.1136/heartjnl-2015-307745] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/22/2015] [Indexed: 11/16/2022] Open
Abstract
Background Elafin is a potent endogenous neutrophil elastase inhibitor that protects against myocardial inflammation and injury in preclinical models of ischaemic-reperfusion injury. We investigated whether elafin could inhibit myocardial ischaemia-reperfusion injury induced during coronary artery bypass graft (CABG) surgery. Methods and results In a randomised double-blind placebo-controlled parallel group clinical trial, 87 patients undergoing CABG surgery were randomised 1:1 to intravenous elafin 200 mg or saline placebo administered after induction of anaesthesia and prior to sternotomy. Myocardial injury was measured as cardiac troponin I release over 48 h (area under the curve (AUC)) and myocardial infarction identified with MRI. Postischaemic inflammation was measured by plasma markers including AUC high-sensitive C reactive protein (hs-CRP) and myeloperoxidase (MPO). Elafin infusion was safe and resulted in >3000-fold increase in plasma elafin concentrations and >50% inhibition of elastase activity in the first 24 h. This did not reduce myocardial injury over 48 h (ratio of geometric means (elafin/placebo) of AUC troponin I 0.74 (95% CI 0.47 to 1.15, p=0.18)) although post hoc analysis of the high-sensitive assay revealed lower troponin I concentrations at 6 h in elafin-treated patients (median 2.4 vs 4.1 μg/L, p=0.035). Elafin had no effect on myocardial infarction (elafin, 7/34 vs placebo, 5/35 patients) or on markers of inflammation: mean differences for AUC hs-CRP of 499 mg/L/48 h (95% CI −207 to 1205, p=0.16), and AUC MPO of 238 ng/mL/48 h (95% CI −235 to 711, p=0.320). Conclusions There was no strong evidence that neutrophil elastase inhibition with a single-dose elafin treatment reduced myocardial injury and inflammation following CABG-induced ischaemia-reperfusion injury. Trial registration number (EudraCT 2010-019527-58, ISRCTN82061264).
Collapse
Affiliation(s)
- S R Alam
- British Heart Foundation/Centre for Population Health Sciences, University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - S C Lewis
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - V Zamvar
- Department of Cardio-thoracic Surgery, Edinburgh Royal Infirmary, Edinburgh, UK
| | - R Pessotto
- Department of Cardio-thoracic Surgery, Edinburgh Royal Infirmary, Edinburgh, UK
| | - M R Dweck
- British Heart Foundation/Centre for Population Health Sciences, University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - A Krishan
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - K Goodman
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - K Oatey
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - R Harkess
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - L Milne
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - S Thomas
- Edinburgh Clinical Trials Unit, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - N M Mills
- British Heart Foundation/Centre for Population Health Sciences, University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - C Moore
- Department of Cardio-thoracic Surgery, Edinburgh Royal Infirmary, Edinburgh, UK
| | - S Semple
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, UK
| | - O Wiedow
- Department of Dermatology, University of Kiel, Kiel, Germany
| | - C Stirrat
- British Heart Foundation/Centre for Population Health Sciences, University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| | - S Mirsadraee
- Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, UK
| | - D E Newby
- British Heart Foundation/Centre for Population Health Sciences, University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK Clinical Research Imaging Centre, University of Edinburgh, Edinburgh, UK
| | - P A Henriksen
- British Heart Foundation/Centre for Population Health Sciences, University of Edinburgh Centre for Cardiovascular Science, Edinburgh, UK
| |
Collapse
|
40
|
Vicuña L, Strochlic DE, Latremoliere A, Bali KK, Simonetti M, Husainie D, Prokosch S, Riva P, Griffin RS, Njoo C, Gehrig S, Mall MA, Arnold B, Devor M, Woolf CJ, Liberles SD, Costigan M, Kuner R. The serine protease inhibitor SerpinA3N attenuates neuropathic pain by inhibiting T cell-derived leukocyte elastase. Nat Med 2015; 21:518-23. [PMID: 25915831 PMCID: PMC4450999 DOI: 10.1038/nm.3852] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 03/30/2015] [Indexed: 12/13/2022]
Abstract
Neuropathic pain is a major, intractable clinical problem and its pathophysiology is not well understood. Although recent gene expression profiling studies have enabled the identification of novel targets for pain therapy, classical study designs provide unclear results owing to the differential expression of hundreds of genes across sham and nerve-injured groups, which can be difficult to validate, particularly with respect to the specificity of pain modulation. To circumvent this, we used two outbred lines of rats, which are genetically similar except for being genetically segregated as a result of selective breeding for differences in neuropathic pain hypersensitivity. SerpinA3N, a serine protease inhibitor, was upregulated in the dorsal root ganglia (DRG) after nerve injury, which was further validated for its mouse homolog. Mice lacking SerpinA3N developed more neuropathic mechanical allodynia than wild-type (WT) mice, and exogenous delivery of SerpinA3N attenuated mechanical allodynia in WT mice. T lymphocytes infiltrate the DRG after nerve injury and release leukocyte elastase (LE), which was inhibited by SerpinA3N derived from DRG neurons. Genetic loss of LE or exogenous application of a LE inhibitor (Sivelastat) in WT mice attenuated neuropathic mechanical allodynia. Overall, we reveal a novel and clinically relevant role for a member of the serpin superfamily and a leukocyte elastase and crosstalk between neurons and T cells in the modulation of neuropathic pain.
Collapse
Affiliation(s)
- Lucas Vicuña
- Pharmacology Institute, University of Heidelberg, Heidelberg, Germany
| | - David E Strochlic
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Alban Latremoliere
- 1] F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kiran Kumar Bali
- Pharmacology Institute, University of Heidelberg, Heidelberg, Germany
| | - Manuela Simonetti
- Pharmacology Institute, University of Heidelberg, Heidelberg, Germany
| | - Dewi Husainie
- Pharmacology Institute, University of Heidelberg, Heidelberg, Germany
| | - Sandra Prokosch
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Priscilla Riva
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Robert S Griffin
- Department of Anesthesiology (Pain Management), Hospital for Special Surgery, New York, New York, USA
| | - Christian Njoo
- Pharmacology Institute, University of Heidelberg, Heidelberg, Germany
| | - Stefanie Gehrig
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Bernd Arnold
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Marshall Devor
- Institute of Life Sciences and Center for Research on Pain, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Clifford J Woolf
- 1] F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen D Liberles
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Costigan
- 1] F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA. [3] Department of Anesthesia, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rohini Kuner
- Pharmacology Institute, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
41
|
Expression of Elafin in Fallopian Tubes of Ectopic Pregnancies Is Reduced. Appl Immunohistochem Mol Morphol 2015; 23:349-54. [DOI: 10.1097/pai.0000000000000091] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Sulic AM, Kurppa K, Rauhavirta T, Kaukinen K, Lindfors K. Transglutaminase as a therapeutic target for celiac disease. Expert Opin Ther Targets 2014; 19:335-48. [PMID: 25410283 DOI: 10.1517/14728222.2014.985207] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The only current treatment for celiac disease is a strict gluten-free diet. The ubiquitous presence of gluten in groceries, however, makes the diet burdensome and difficult to maintain, and alternative treatment options are thus needed. Here, the important role of transglutaminase 2 (TG2) in the pathogenesis of celiac disease makes it an attractive target for drug development. AREAS COVERED The present paper gives an overview of TG2 and addresses its significance in the pathogenesis of celiac disease. Moreover, the article summarizes preclinical studies performed with TG2 inhibitors and scrutinizes issues related to this therapeutic approach. EXPERT OPINION Activation of TG2 in the intestinal mucosa is central in celiac disease pathogenesis and researchers have therefore suggested TG2 inhibitors as a potential therapeutic approach. However, a prerequisite for such a drug is that it should be specific for TG2 and not affect the activity of other members of the transglutaminase family. Such compounds have already been introduced and tested in vitro, but a major obstacle to further development is the lack of a well-defined animal model for celiac disease. Nonetheless, with encouraging results in preclinical studies clinical trials with TG2 inhibitors are eagerly awaited.
Collapse
Affiliation(s)
- Ana-Marija Sulic
- Tampere Center for Child Health Research, University of Tampere and Tampere University Hospital , Tampere , Finland +358 50 3186306; +358 3 3641369 ;
| | | | | | | | | |
Collapse
|
43
|
Hingorani S, Finn LS, Pao E, Lawler R, Schoch G, McDonald GB, Najafian B, Sandmaier B, Gooley T. Urinary elafin and kidney injury in hematopoietic cell transplant recipients. Clin J Am Soc Nephrol 2014; 10:12-20. [PMID: 25388519 DOI: 10.2215/cjn.01840214] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Graft-versus-host disease (GVHD) is associated with kidney injury after hematopoietic cell transplantation (HCT). Because plasma elafin levels correlate with skin GVHD, this study examined urinary elafin as a potential marker of renal inflammation and injury. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Urine was collected prospectively on 205 patients undergoing their first HCT from 2003 to 2010. Collections were done at baseline, weekly through day 100, and monthly through year 1 to measure elafin and urine albumin-to-creatinine ratio (ACR). Associations between urinary elafin levels and microalbuminuria, macroalbuminuria, AKI and CKD, and mortality were examined using Cox proportional hazards or linear regression models. Available kidney biopsy specimens were processed for immunohistochemistry. RESULTS Mean urinary elafin levels to day 100 were higher in patients with micro- or macroalbuminuria (adjusted mean difference, 529 pg/ml; P=0.03) at day 100 than in those with a normal ACR (adjusted mean difference, 1295 pg/ml; P<0.001). Mean urinary elafin levels were higher in patients with AKI compared with patients without AKI (adjusted mean difference, 558 pg/ml; P<0.01). The average urinary elafin levels within the first 100 days after HCT were higher in patients who developed CKD at 1 year than in patients without CKD (adjusted mean difference, 894 pg/ml; P=0.002). Among allogeneic recipients, a higher proportion of patients with micro- or macroalbuminuria at day 100 also had grade II-IV acute GVHD (80% and 86%, respectively) compared with patients with a normal ACR (58%; global P<0.01). Each increase in elafin of 500 pg/ml resulted in a 10% increase in risk of persistent macroalbuminuria (hazard ratio, 1.10; 95% confidence interval [95% CI], 1.06 to 1.13; P<0.001) and a 7% increase in the risk of overall mortality (95% CI, 1.02 to 1.13, P<0.01). Renal biopsy specimens from a separate cohort of HCT survivors demonstrated elafin staining in distal and collecting duct tubules. CONCLUSION Higher urinary elafin levels are associated with an increased risk of micro- and macroalbuminuria, AKI and CKD, and death after HCT.
Collapse
Affiliation(s)
- Sangeeta Hingorani
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington; Seattle Children's Hospital, Seattle, Washington; and Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Laura S Finn
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington; Seattle Children's Hospital, Seattle, Washington; and
| | - Emily Pao
- Seattle Children's Hospital, Seattle, Washington; and
| | - Rick Lawler
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Gary Schoch
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - George B McDonald
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Behzad Najafian
- Departments of Pediatrics, Gastroenterology, and Pathology, University of Washington, Seattle, Washington
| | - Brenda Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ted Gooley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
44
|
Plasmodium falciparum infection induces expression of a mosquito salivary protein (Agaphelin) that targets neutrophil function and inhibits thrombosis without impairing hemostasis. PLoS Pathog 2014; 10:e1004338. [PMID: 25211214 PMCID: PMC4161438 DOI: 10.1371/journal.ppat.1004338] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/02/2014] [Indexed: 01/05/2023] Open
Abstract
Background Invasion of mosquito salivary glands (SGs) by Plasmodium falciparum sporozoites is an essential step in the malaria life cycle. How infection modulates gene expression, and affects hematophagy remains unclear. Principal Findings Using Affimetrix chip microarray, we found that at least 43 genes are differentially expressed in the glands of Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Among the upregulated genes, one codes for Agaphelin, a 58-amino acid protein containing a single Kazal domain with a Leu in the P1 position. Agaphelin displays high homology to orthologs present in Aedes sp and Culex sp salivary glands, indicating an evolutionarily expanded family. Kinetics and surface plasmon resonance experiments determined that chemically synthesized Agaphelin behaves as a slow and tight inhibitor of neutrophil elastase (KD∼10 nM), but does not affect other enzymes, nor promotes vasodilation, or exhibit antimicrobial activity. TAXIscan chamber assay revealed that Agaphelin inhibits neutrophil chemotaxis toward fMLP, affecting several parameter associated with cell migration. In addition, Agaphelin reduces paw edema formation and accumulation of tissue myeloperoxidase triggered by injection of carrageenan in mice. Agaphelin also blocks elastase/cathepsin-mediated platelet aggregation, abrogates elastase-mediated cleavage of tissue factor pathway inhibitor, and attenuates neutrophil-induced coagulation. Notably, Agaphelin inhibits neutrophil extracellular traps (NETs) formation and prevents FeCl3-induced arterial thrombosis, without impairing hemostasis. Conclusions Blockade of neutrophil elastase emerges as a novel antihemostatic mechanism in hematophagy; it also supports the notion that neutrophils and the innate immune response are targets for antithrombotic therapy. In addition, Agaphelin is the first antihemostatic whose expression is induced by Plasmodium sp infection. These results suggest that an important interplay takes place in parasite-vector-host interactions. Malaria is transmitted by Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Salivary gland contributes to the development of the parasite by creating a favorable environment for the infection and facilitating blood feeding and reproduction of the vector. However, the molecular mechanism by which the vector salivary gland modulates parasite/host interactions is not understood. We discovered that infection of the mosquito salivary gland upregulates several genes; among them, one codes for a protease inhibitor named Agaphelin. Notably, Agaphelin was found to exhibit multiple antihemostatic functions by targeting elastase. As a result, it inhibits platelet function which is required for blood to clot, and it prevents cleavage of TFPI, an anticoagulant that has recently been found to play a crucial role in thrombus formation in vivo. Agaphelin also attenuates neutrophils chemotaxis and the release of Neutrophil Extracellular Traps. These results provide evidence that neutrophils serve as a link between coagulation and the innate immune response. Agaphelin also exhibits anti-inflammatory and antithrombotic effects in vivo. Furthermore, Agaphelin did not promote bleeding, suggesting that targeting neutrophil exhibits potential therapeutic value. Altogether, these results highlight that the interplay between parasite, vector and host is a dynamic process that contributes and sustains the interface among Plasmodium, Anopheles and humans.
Collapse
|
45
|
Abbott DS, Chin-Smith EC, Seed PT, Chandiramani M, Shennan AH, Tribe RM. Raised trappin2/elafin protein in cervico-vaginal fluid is a potential predictor of cervical shortening and spontaneous preterm birth. PLoS One 2014; 9:e100771. [PMID: 25075964 PMCID: PMC4116119 DOI: 10.1371/journal.pone.0100771] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/22/2014] [Indexed: 12/22/2022] Open
Abstract
Early spontaneous preterm birth is associated with inflammation/infection and shortening of the cervix. We hypothesised that cervico-vaginal production of trappin2/elafin (peptidase inhibitor 3) and cathelicidin antimicrobial peptide (cathelicidin), key components of the innate immune system, are altered in women who have a spontaneous preterm birth. The aim was to determine the relationship between cervico-vaginal fluid (CVF) trappin2/elafin and cathelicidin protein concentrations with cervical length in woman at risk of spontaneous preterm birth. Trappin2/elafin and cathelicidin were measured using ELISA in longitudinal CVF samples (taken between 13 to 30 weeks' gestation) from 74 asymptomatic high risk women (based on obstetric history) recruited prospectively. Thirty six women developed a short cervix (<25 mm) by 24 weeks' and 38 women did not. Women who developed a short cervix had 2.71 times higher concentrations of CVF trappin2/elafin from 14 weeks' versus those who did not (CI 1.94–3.79, p<0.0005). CVF trappin2/elafin before 24 weeks' was 1.79 times higher in women who had a spontaneous preterm birth <37 weeks' (CI: 1.05–3.05, p = 0.034). Trappin2/elafin (>200 ng/ml) measured between 14+0–14+6 weeks' of pregnancy predicted women who subsequently developed a short cervix (n = 11, ROC area = 1.00, p = 0.008) within 8 weeks. Cathelicidin was not predictive of spontaneous delivery. Vitamin D status did not correlate with CVF antimicrobial peptide concentrations. Raised CVF trappin2/elafin has potential as an early pregnancy test for prediction of cervical shortening and spontaneous preterm birth. This justifies validation in a larger cohort.
Collapse
Affiliation(s)
- Danielle S. Abbott
- Division of Women's Health, King's College London, Women's Health Academic Centre King's Health Partners, London, United Kingdom
| | - Evonne C. Chin-Smith
- Division of Women's Health, King's College London, Women's Health Academic Centre King's Health Partners, London, United Kingdom
| | - Paul T. Seed
- Division of Women's Health, King's College London, Women's Health Academic Centre King's Health Partners, London, United Kingdom
| | - Manju Chandiramani
- Parturition Research Group, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
| | - Andrew H. Shennan
- Division of Women's Health, King's College London, Women's Health Academic Centre King's Health Partners, London, United Kingdom
| | - Rachel M. Tribe
- Division of Women's Health, King's College London, Women's Health Academic Centre King's Health Partners, London, United Kingdom
- Division of Women's Health, King's College London Women's Health Academic Centre KHP, St. Thomas' Hospital Campus, London, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Norman P. Pulmonary arterial hypertension: a rare disease that encourages the development of multiple treatments. Expert Opin Orphan Drugs 2014. [DOI: 10.1517/21678707.2014.924851] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
47
|
Novel role of the serine protease inhibitor elafin in gluten-related disorders. Am J Gastroenterol 2014; 109:748-56. [PMID: 24710505 PMCID: PMC4219532 DOI: 10.1038/ajg.2014.48] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/02/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Elafin, an endogenous serine protease inhibitor, modulates colonic inflammation. We investigated the role of elafin in celiac disease (CD) using human small intestinal tissues and in vitro assays of gliadin deamidation. We also investigated the potential beneficial effects of elafin in a mouse model of gluten sensitivity. METHODS Epithelial elafin expression in the small intestine of patients with active CD, treated CD, and controls without CD was determined by immunofluorescence. Interaction of elafin with human tissue transglutaminase-2 (TG-2) was investigated in vitro. The 33-mer peptide, a highly immunogenic gliadin peptide, was incubated with TG-2 and elafin at different concentrations. The degree of deamidation of the 33-mer peptide was analyzed by liquid chromatography-mass spectrometry. Elafin was delivered to the intestine of gluten-sensitive mice using a recombinant Lactococcus lactis vector. Small intestinal barrier function, inflammation, proteolytic activity, and zonula occludens-1 (ZO-1) expression were assessed. RESULTS Elafin expression in the small intestinal epithelium was lower in patients with active CD compared with control patients. In vitro, elafin significantly slowed the kinetics of the deamidation of the 33-mer peptide to its more immunogenic form. Treatment of gluten-sensitive mice with elafin delivered by the L. lactis vector normalized inflammation, improved permeability, and maintained ZO-1 expression. CONCLUSIONS The decreased elafin expression in the small intestine of patients with active CD, the reduction of 33-mer peptide deamidation by elafin, coupled to the barrier enhancing and anti-inflammatory effects observed in gluten-sensitive mice, suggest that this molecule may have pathophysiological and therapeutic importance in gluten-related disorders.
Collapse
|
48
|
Kim JM. Antimicrobial proteins in intestine and inflammatory bowel diseases. Intest Res 2014; 12:20-33. [PMID: 25349560 PMCID: PMC4204685 DOI: 10.5217/ir.2014.12.1.20] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 12/21/2022] Open
Abstract
Mucosal surface of the intestinal tract is continuously exposed to a large number of microorganisms. To manage the substantial microbial exposure, epithelial surfaces produce a diverse arsenal of antimicrobial proteins (AMPs) that directly kill or inhibit the growth of microorganisms. Thus, AMPs are important components of innate immunity in the gut mucosa. They are frequently expressed in response to colonic inflammation and infection. Expression of many AMPs, including human β-defensin 2-4 and cathelicidin, is induced in response to invasion of pathogens or enteric microbiota into the mucosal barrier. In contrast, some AMPs, including human α-defensin 5-6 and human β-defensin 1, are constitutively expressed without microbial contact or invasion. In addition, specific AMPs are reported to be associated with inflammatory bowel disease (IBD) due to altered expression of AMPs or development of autoantibodies against AMPs. The advanced knowledge for AMPs expression in IBD can lead to its potential use as biomarkers for disease activity. Although the administration of exogenous AMPs as therapeutic strategies against IBD is still at an early stage of development, augmented induction of endogenous AMPs may be another interesting future research direction for the protective and therapeutic purposes. This review discusses new advances in our understanding of how intestinal AMPs protect against pathogens and contribute to pathophysiology of IBD.
Collapse
Affiliation(s)
- Jung Mogg Kim
- Department of Microbiology, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
|
50
|
Laquer VT, Hevezi PA, Albrecht H, Chen TS, Zlotnik A, Kelly KM. Microarray analysis of port wine stains before and after pulsed dye laser treatment. Lasers Surg Med 2013; 45:67-75. [PMID: 23440713 DOI: 10.1002/lsm.22087] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Neither the pathogenesis of port wine stain (PWS) birthmarks nor tissue effects of pulsed dye laser (PDL) treatment of these lesions is fully understood. There are few published reports utilizing gene expression analysis in human PWS skin. We aim to compare gene expression in PWS before and after PDL, using DNA microarrays that represent most, if not all, human genes to obtain comprehensive molecular profiles of PWS lesions and PDL-associated tissue effects. MATERIALS AND METHODS Five human subjects had PDL treatment of their PWS. One week later, three biopsies were taken from each subject: normal skin (N); untreated PWS (PWS); PWS post-PDL (PWS + PDL). Samples included two lower extremity lesions, two facial lesions, and one facial nodule. High-quality total RNA isolated from skin biopsies was processed and applied to Affymetrix Human gene 1.0ST microarrays for gene expression analysis. We performed a 16 pair-wise comparison identifying either up- or down-regulated genes between N versus PWS and PWS versus PWS + PDL for four of the donor samples. The PWS nodule (nPWS) was analyzed separately. RESULTS There was significant variation in gene expression profiles between individuals. By doing pair-wise comparisons between samples taken from the same donor, we were able to identify genes that may participate in the formation of PWS lesions and PDL tissue effects. Genes associated with immune, epidermal, and lipid metabolism were up-regulated in PWS skin. The nPWS exhibited more profound differences in gene expression than the rest of the samples, with significant differential expression of genes associated with angiogenesis, tumorigenesis, and inflammation. CONCLUSION In summary, gene expression profiles from N, PWS, and PWS + PDL demonstrated significant variation within samples from the same donor and between donors. By doing pair-wise comparisons between samples taken from the same donor and comparing these results between donors, we were able to identify genes that may participate in formation of PWS and PDL effects. Our preliminary results indicate changes in gene expression of angiogenesis-related genes, suggesting that dysregulation of angiogenic signals and/or components may contribute to PWS pathology.
Collapse
Affiliation(s)
- Vivian T Laquer
- Department of Dermatology, University of California, Irvine, Irvine, California, USA.
| | | | | | | | | | | |
Collapse
|