1
|
Liu D, Hu X, Ding X, Li M, Ding L. Inflammatory Effects and Regulatory Mechanisms of Chitinase-3-like-1 in Multiple Human Body Systems: A Comprehensive Review. Int J Mol Sci 2024; 25:13437. [PMCID: PMC11678640 DOI: 10.3390/ijms252413437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/29/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
Chitinase-3-like-1 (Chi3l1), also known as YKL-40 or BRP-39, is a highly conserved mammalian chitinase with a chitin-binding ability but no chitinase enzymatic activity. Chi3l1 is secreted by various cell types and induced by several inflammatory cytokines. It can mediate a series of cell biological processes, such as proliferation, apoptosis, migration, differentiation, and polarization. Accumulating evidence has verified that Chi3l1 is involved in diverse inflammatory conditions; however, a systematic and comprehensive understanding of the roles and mechanisms of Chi3l1 in almost all human body system-related inflammatory diseases is still lacking. The human body consists of ten organ systems, which are combinations of multiple organs that perform one or more physiological functions. Abnormalities in these human systems can trigger a series of inflammatory environments, posing serious threats to the quality of life and lifespan of humans. Therefore, exploring novel and reliable biomarkers for these diseases is highly important, with Chi3l1 being one such parameter because of its physiological and pathophysiological roles in the development of multiple inflammatory diseases. Reportedly, Chi3l1 plays an important role in diagnosing and determining disease activity/severity/prognosis related to multiple human body system inflammation disorders. Additionally, many studies have revealed the influencing factors and regulatory mechanisms (e.g., the ERK and MAPK pathways) of Chi3l1 in these inflammatory conditions, identifying potential novel therapeutic targets for these diseases. In this review, we comprehensively summarize the potential roles and underlying mechanisms of Chi3l1 in inflammatory disorders of the respiratory, digestive, circulatory, nervous, urinary, endocrine, skeletal, muscular, and reproductive systems, which provides a more systematic understanding of Chi3l1 in multiple human body system-related inflammatory diseases. Moreover, this article summarizes potential therapeutic strategies for inflammatory diseases in these systems on the basis of the revealed roles and mechanisms mediated by Chi3l1.
Collapse
Affiliation(s)
- Dong Liu
- School of Life Sciences, Yunnan University, Kunming 650500, China;
| | - Xin Hu
- Yunnan Key Laboratory of Soil Erosion Prevention and Green Development, Institute of International Rivers and Ecosecurity, Yunnan University, Kunming 650500, China;
| | - Xiao Ding
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China;
| | - Ming Li
- School of Life Sciences, Yunnan University, Kunming 650500, China;
| | - Lei Ding
- School of Life Sciences, Yunnan University, Kunming 650500, China;
| |
Collapse
|
2
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
3
|
缪 治, 冉 晶, 牟 大, 吴 沙, 陈 艳, 李 聪, 陈 月, 杨 闵, 谢 其. [YKL-40 Promotes the Expression of Inflammatory Factors in Type Ⅱ Alveolar Epithelial Cell Model of A549 Cell Line]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:954-958. [PMID: 37866952 PMCID: PMC10579078 DOI: 10.12182/20230960201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Indexed: 10/24/2023]
Abstract
Objective YKL-40, also known as chitinase-3-like-1 (CHI3L1), is a human cartilage glycoprotein-39, with its N-terminus consisting of tyrosine (Y), lysine (K), and leucine (L), hence the name YKL-40. In this study, we explored whether YKL-40 could promote the expression of inflammatory factors in type Ⅱ alveolar epithelial cells. Methods A549 cells were cultured in vitro with interleukin (IL)-1β (20 ng/mL), IL-6 (20 ng/mL), tumor necrosis factor-alpha (TNF-α) (20 ng/mL), and interferon-gamma (IFN-γ) (20 ng/mL). The expression of YKL-40 transcription was determined by RT-qPCR. A549 cells were cultured with IL-1β at 5, 10, and 20 ng/mL and the expression of YKL-40 protein was determined by Western blot. A549 cells were cultured with recombinant YKL-40 protein at 0, 100, 500, and 1 000 ng/mL and the expression levels of IL-6 and IL-8 were measured by RT-qPCR. Three pairs of small interfering RNAs targeting YKL-40 (si- YKL-40-1/2/3) and the negative control (NC) were designed and used to transfect A549 cells, respectively, and the expression of YKL-40 was determined by RT-qPCR and Western blot. si- YKL-40-3 was screened out for subsequent experiments. In A549 cells, si- YKL-40-3 and si-NC were transfected and, then, IL-1β (20 ng/mL) was added in for culturing. The expression of YKL-40, IL-6, and IL-8 was determined by RT-qPCR and the expression of multiple factors in the supernatant was measured with the QAH-INF-1 kit. Results RT-qPCR results showed that IL-1β could up-regulate YKL-40 protein transcription level compared with that of the control group and the difference was statistically significant ( P<0.01), but IL-6, TNF-α, and IFN-γ could not up-regulate YKL-40 protein transcription level. Western blot results showed that IL-1β (20 ng/mL) could significantly promote the expression of YKL-40 and, compared with that of the control group, the differences showed by groups treated with different concentrations of IL-1β were all statistical significant ( P<0.01). After adding human recombinant YKL-40 protein to A549 cells, the results showed that the expression of inflammatory factors IL-6 and IL-8 was significantly increased and the difference was statistically significant compared with that of the control group ( P<0.05). After the expression of YKL-40 was decreased by si- YKL-40-3 transfection, the expression of IL-6 ( P<0.05), IL-8 ( P<0.05), and other inflammatory factors was inhibited compared with that of the control group. Conclusion YKL-40 can promote the expression and secretion of IL-6, IL-8, and other acute inflammatory factors in A549 cell line, a type Ⅱ alveolar epithelial cell model, thus aggravating the inflammatory response. Targeted inhibition of YKL-40 expression may effectively inhibit inflammatory response.
Collapse
Affiliation(s)
- 治永 缪
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 晶晶 冉
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 大超 牟
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 沙沙 吴
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 艳琼 陈
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 聪 李
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 月红 陈
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 闵 杨
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 其冰 谢
- 四川大学华西医院 人类疾病与免疫治疗研究室 (成都 610041)Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Kuret T, Frank-Bertoncelj M, Lakota K, Žigon P, Thallinger GG, Kopitar AN, Čučnik S, Tomšič M, Hočevar A, Sodin-Šemrl S. From Active to Non-active Giant Cell Arteritis: Longitudinal Monitoring of Patients on Glucocorticoid Therapy in Combination With Leflunomide. Front Med (Lausanne) 2022; 8:827095. [PMID: 35127774 PMCID: PMC8811148 DOI: 10.3389/fmed.2021.827095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
In the present study, we longitudinally monitored leukocyte subsets, expression of neutrophil surface adhesion molecules (CD62L and CD11b) and serum analytes in therapy-naïve patients with active giant cell arteritis (GCA). We collected blood samples at the baseline, and at weeks 1, 4, 12, 24, and 48 of follow-up, and evaluated short- and long-term effects of glucocorticoids (GC) vs. GC and leflunomide. Our aim was to identify candidate biomarkers that could be used to monitor disease activity and predict an increased risk of a relapse. Following high doses of GC, the numbers of CD4+ T-lymphocytes and B-lymphocytes transiently increased and then subsided when GC dose tapering started at week 4. In contrast, the numbers of neutrophils significantly increased during the follow-up time of 12 weeks compared to pre-treatment time. Neutrophil CD62L rapidly diminished after initiation of GC therapy, however its expression remained low at week 48, only in patients under combinatorial therapy with leflunomide. Levels of acute phase reactant SAA and IL-6 decreased significantly after treatment with GC and leflunomide, while levels of IL-8, IL-18, and CHI3L1 did not change significantly during the follow-up period. CHI3L1 was associated with signs of transmural inflammation and vessel occlusion and might therefore serve as a marker of fully developed active GCA, and a promising therapeutic target. Patients with relapses had higher levels of IL-23 at presentation than patients without relapses (p = 0.021). Additionally, the levels of IL-23 were higher at the time of relapse compared to the last follow-up point before relapse. IL-23 might present a promising biomarker of uncontrolled and active disease and could give early indication of upcoming relapses.
Collapse
Affiliation(s)
- Tadeja Kuret
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Science and Information Technologies, University of Primorska, Koper, Slovenia
| | | | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Science and Information Technologies, University of Primorska, Koper, Slovenia
| | - Polona Žigon
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Science and Information Technologies, University of Primorska, Koper, Slovenia
| | - Gerhard G. Thallinger
- Institute for Biomedical Informatics, Graz University of Technology, Graz, Austria
- OMICS Center Graz, BioTechMed Graz, Graz, Austria
| | - Andreja N. Kopitar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Matija Tomšič
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Alojzija Hočevar
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Snežna Sodin-Šemrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Mathematics, Natural Science and Information Technologies, University of Primorska, Koper, Slovenia
- *Correspondence: Snežna Sodin-Šemrl
| |
Collapse
|
5
|
Lee JW, Kim MN, Kim EG, Leem JS, Baek SM, Kim MJ, Kim KW, Sohn MH. Chitinase 3-like 1 is involved in the induction of IL-8 expression by double-stranded RNA in airway epithelial cells. Biochem Biophys Res Commun 2022; 592:106-112. [PMID: 35033868 DOI: 10.1016/j.bbrc.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
Viral respiratory infection causes inflammatory lung disease. Chitinase 3-like 1 (CHI3L1) contributes to airway inflammation, but its role in human airway epithelial cells following viral infection is unclear. Thus, we investigated whether CHI3L1 regulates inflammatory responses caused by viral infections in airway epithelial cells. Human bronchial epithelial cells, BEAS-2B, were stimulated with a synthetic analog of viral double-stranded RNA, polyinosinic:polycytidylic acid (poly(I:C)). To confirm the specific role of CHI3L1, CHI3L1 was knocked down in BEAS-2B cells using shRNA lentivirus. The expression of CHI3L1 and proinflammatory cytokines such as IL-8 and phosphorylation of mitogen-activated protein kinase (MAPK) pathways were analyzed. In addition to poly(I:C), BEAS-2B cells were infected with the human respiratory syncytial virus (RSV) A2 strain, and CHI3L1 and IL-8 expression was analyzed. Stimulating the cells with poly(I:C) increased CHI3L1 and IL-8 expression, whereas IL-8 expression was abrogated in CHI3L1 knockdown BEAS-2B cells. Poly(I:C) stimulation of BEAS-2B cells resulted in phosphorylation of MAPK pathways, and inhibition of MAPK pathways significantly abolished IL-8 secretion. Phosphorylation of MAPK pathways was diminished in CHI3L1 knockdown BEAS-2B cells. Infection with RSV increased CHI3L1 and IL-8 expression. IL-8 expression induced by RSV infection was abrogated in CHI3L1 knockdown cells. In conclusion, CHI3L1 may be involved in IL-8 secretion by regulating MAPK pathways during respiratory viral infections in airway epithelial cells.
Collapse
Affiliation(s)
- Jae Woo Lee
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Mi Na Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Eun Gyul Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Ji Su Leem
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Seung Min Baek
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Min Jung Kim
- Department of Pediatrics, Yonsei University Yongin Severance Hospital, 363 Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 16995, South Korea.
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project, Yonsei University College of Medicine, 134 Sinchon-Dong, Seoul, 03722, South Korea.
| |
Collapse
|
6
|
Conceptual advances and evolving terminology in acute kidney disease. Nat Rev Nephrol 2021; 17:493-502. [PMID: 33712812 DOI: 10.1038/s41581-021-00410-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 01/31/2023]
Abstract
Over the past decade, new insights into epidemiology, pathophysiology and biomarkers have modified our understanding of acute kidney dysfunction and damage, and their association with subsequent chronic kidney disease. The concept of acute kidney injury (AKI), which has relied on established but nonetheless flawed biomarkers of solute clearance (serum creatinine levels and urinary output), has been challenged by the identification of novel biomarkers of tubular stress and/or damage. The expression of some of these novel biomarkers precedes changes in conventional biomarkers or can increase their predictive power, and might therefore enhance the clinical accuracy of the definition of AKI. In addition, the need to consider AKI recurrence, duration and progression to chronic kidney disease within the clinical and epidemiological framework of AKI led to the emergence of the concept of acute kidney disease. New definitions of acute syndromes of kidney impairment and injury are needed.
Collapse
|
7
|
van Sleen Y, Jiemy WF, Pringle S, van der Geest KSM, Abdulahad WH, Sandovici M, Brouwer E, Heeringa P, Boots AMH. A Distinct Macrophage Subset Mediating Tissue Destruction and Neovascularization in Giant Cell Arteritis: Implication of the YKL-40 - IL-13 Receptor α2 Axis. Arthritis Rheumatol 2021; 73:2327-2337. [PMID: 34105308 PMCID: PMC9298326 DOI: 10.1002/art.41887] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/27/2021] [Indexed: 11/17/2022]
Abstract
Objective Macrophages mediate inflammation, angiogenesis, and tissue destruction in giant cell arteritis (GCA). Serum levels of the macrophage‐associated protein YKL‐40 (chitinase 3–like protein 1), previously linked to angiogenesis and tissue remodeling, remain elevated in GCA despite glucocorticoid treatment. This study was undertaken to investigate the contribution of YKL‐40 to vasculopathy in GCA. Methods Immunohistochemistry was performed on GCA temporal artery biopsy specimens (n = 12) and aortas (n = 10) for detection of YKL‐40, its receptor interleukin‐13 receptor α2 (IL‐13Rα2), macrophage markers PU.1 and CD206, and the tissue‐destructive protein matrix metalloproteinase 9 (MMP‐9). Ten noninflamed temporal artery biopsy specimens served as controls. In vitro experiments with granulocyte–macrophage colony‐stimulating factor (GM‐CSF)– or macrophage colony‐stimulating factor (M‐CSF)–skewed monocyte‐derived macrophages were conducted to study the dynamics of YKL‐40 production. Next, small interfering RNA–mediated knockdown of YKL‐40 in GM‐CSF–skewed macrophages was performed to study its effect on MMP‐9 production. Finally, the angiogenic potential of YKL‐40 was investigated by tube formation experiments using human microvascular endothelial cells (HMVECs). Results YKL‐40 was abundantly expressed by a CD206+MMP‐9+ macrophage subset in inflamed temporal arteries and aortas. GM‐CSF–skewed macrophages from GCA patients, but not healthy controls, released significantly higher levels of YKL‐40 compared to M‐CSF–skewed macrophages (P = 0.039). In inflamed temporal arteries, IL‐13Rα2 was expressed by macrophages and endothelial cells. Functionally, knockdown of YKL‐40 led to a 10–50% reduction in MMP‐9 production by macrophages, whereas exposure of HMVECS to YKL‐40 led to significantly increased tube formation. Conclusion In GCA, a GM‐CSF–skewed, CD206+MMP‐9+ macrophage subset expresses high levels of YKL‐40 which may stimulate tissue destruction and angiogenesis through IL‐13Rα2 signaling. Targeting YKL‐40 or GM‐CSF may inhibit macrophages that are currently insufficiently suppressed by glucocorticoids.
Collapse
Affiliation(s)
- Yannick van Sleen
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - William F Jiemy
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Faculty of Applied Science, UCSI University, UCSI Heights, Cheras Kuala Lumpur, Malaysia
| | - Sarah Pringle
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kornelis S M van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wayel H Abdulahad
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria Sandovici
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annemieke M H Boots
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
8
|
Gaur N, Huss E, Prell T, Steinbach R, Guerra J, Srivastava A, Witte OW, Grosskreutz J. Monocyte-Derived Macrophages Contribute to Chitinase Dysregulation in Amyotrophic Lateral Sclerosis: A Pilot Study. Front Neurol 2021; 12:629332. [PMID: 34054686 PMCID: PMC8160083 DOI: 10.3389/fneur.2021.629332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/21/2021] [Indexed: 12/01/2022] Open
Abstract
Neuroinflammation significantly contributes to Amyotrophic Lateral Sclerosis (ALS) pathology. In lieu of this, reports of elevated chitinase levels in ALS are interesting, as they are established surrogate markers of a chronic inflammatory response. While post-mortem studies have indicated glial expression, the cellular sources for these moieties remain to be fully understood. Therefore, the objective of this pilot study was to examine whether the peripheral immune system also contributes to chitinase dysregulation in ALS. The temporal expression of CHIT1, CHI3L1, and CHI3L2 in non-polarized monocyte-derived macrophages (MoMas) from ALS patients and healthy controls (HCs) was examined. We demonstrate that while CHIT1 and CHI3L1 display similar temporal expression dynamics in both groups, profound between-group differences were noted for these targets at later time-points i.e., when cells were fully differentiated. CHIT1 and CHI3L1 expression were significantly higher in MoMas from ALS patients at both the transcriptomic and protein level, with CHI3L1 levels also being influenced by age. Conversely, CHI3L2 expression was not influenced by disease state, culture duration, or age. Here, we demonstrate for the first time, that in ALS, circulating immune cells have an intrinsically augmented potential for chitinase production that may propagate chronic neuroinflammation, and how the ageing immune system itself contributes to neurodegeneration.
Collapse
Affiliation(s)
- Nayana Gaur
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Elena Huss
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Tino Prell
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Centre for Healthy Ageing, Jena University Hospital, Jena, Germany
| | - Robert Steinbach
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Joel Guerra
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Akash Srivastava
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Centre for Healthy Ageing, Jena University Hospital, Jena, Germany
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.,Jena Centre for Healthy Ageing, Jena University Hospital, Jena, Germany
| |
Collapse
|
9
|
Tombetti E, Hysa E, Mason JC, Cimmino MA, Camellino D. Blood Biomarkers for Monitoring and Prognosis of Large Vessel Vasculitides. Curr Rheumatol Rep 2021; 23:17. [PMID: 33569633 PMCID: PMC7875948 DOI: 10.1007/s11926-021-00980-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Large vessel vasculitides (LVVs) are inflammatory conditions of the wall of large-sized arteries, mainly represented by giant cell arteritis (GCA) and Takayasu arteritis (TA). The inflammatory process within the vessel wall can lead to serious consequences such as development of aneurysms, strokes and blindness; therefore, early diagnosis and follow-up of LVV are fundamental. However, the arterial wall is poorly accessible and blood biomarkers are intended to help physicians not only in disease diagnosis but also in monitoring and defining the prognosis of these conditions, thus assisting therapeutic decisions and favouring personalised management. The field is the object of intense research as the identification of reliable biomarkers is likely to shed light on the mechanisms of disease progression and arterial remodelling. In this review, we will discuss the role of blood biomarkers in LVVs in the light of the latest evidence. RECENT FINDINGS In clinical practice, the most widely performed laboratory investigations are the erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP). However, these indices may be within normal limits during disease relapse and they are not reliable in patients receiving interleukin-6 (IL-6) receptor inhibitors. New biomarkers struggle to gain traction in clinical practice and no molecule with good accuracy has been identified to date. IL-6, a pro-inflammatory cytokine that drives CRP synthesis and increases the ESR, is one of the most promising biomarkers in the field. IL-6 analysis is increasingly performed, and serum levels are more sensitive than ESR for active GCA and might reflect persistent inflammation with high risk of relapse in patients on IL-6 receptor inhibitors. A future with biomarkers that reflect different disease features is an important aspiration. Accordingly, intense effort is being made to identify IL-6-independent inflammatory biomarkers, such as S100 proteins, pentraxin-3 and osteopontin. Moreover, metalloproteinases such as MMP2/9 and angiogenic modulators such as VEGF, YLK-40 and angiopoietins are being studied as markers of arterial remodelling. Lastly, biomarkers indicating organ damage may guide prognostic stratification as well as emergency therapeutic decisions: the most promising biomarkers so far identified are NT-proBNP, which reflects myocardial strain; pentraxin-3, which has been associated with recent optic nerve ischemia; and endothelin-1, which is associated with ischaemic complications. Currently, the use of these molecules in clinical practice is limited because of their restricted availability, lack of sufficient studies supporting their validity and associated costs. Further evidence is required to better interpret their biological and clinical value.
Collapse
Affiliation(s)
- Enrico Tombetti
- Internal Medicine, Department of Biomedical and Clinical Sciences "Luigi Sacco", Milan, Italy
- Internal Medicine and Rheumatology, Sacco and Fatebenefratelli Hospitals, Milan, Italy
| | - Elvis Hysa
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Justin C Mason
- National Heart and Lung Institute, Imperial College London, London, UK
- Rheumatology, Hammersmith Hospital, Imperial College NHS Trust, London, UK
| | - Marco A Cimmino
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Dario Camellino
- Division of Rheumatology, Musculoskeletal System Department, La Colletta Hospital, Local Health Trust 3 Genoa, Via del Giappone 3, 16011, Arenzano, Italy.
- Autoimmunology Laboratory, Department of Internal Medicine, University of Genoa, Genoa, Italy.
| |
Collapse
|
10
|
Przysucha N, Górska K, Krenke R. Chitinases and Chitinase-Like Proteins in Obstructive Lung Diseases - Current Concepts and Potential Applications. Int J Chron Obstruct Pulmon Dis 2020; 15:885-899. [PMID: 32368034 PMCID: PMC7185641 DOI: 10.2147/copd.s236640] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/10/2020] [Indexed: 01/14/2023] Open
Abstract
Chitinases, enzymes that cleave chitin’s chain to low molecular weight chitooligomers, are widely distributed in nature. Mammalian chitinases belong to the 18-glycosyl-hydrolase family and can be divided into two groups: true chitinases with enzymatic activity (AMCase and chitotriosidase) and chitinase-like proteins (CLPs) molecules which can bind to chitin or chitooligosaccharides but lack enzymatic activity (eg, YKL-40). Chitinases are thought to be part of an innate immunity against chitin-containing parasites and fungal infections. Both groups of these hydrolases are lately evaluated also as chemical mediators or biomarkers involved in airway inflammation and fibrosis. The aim of this article is to present the current knowledge on the potential role of human chitinases and CLPs in the pathogenesis, diagnosis, and course of obstructive lung diseases. We also assessed the potential role of chitinase and CLPs inhibitors as therapeutic targets in chronic obstructive pulmonary disease and asthma.
Collapse
Affiliation(s)
- Natalia Przysucha
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Górska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Rafal Krenke
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
11
|
Kzhyshkowska J, Larionova I, Liu T. YKL-39 as a Potential New Target for Anti-Angiogenic Therapy in Cancer. Front Immunol 2020; 10:2930. [PMID: 32038607 PMCID: PMC6988383 DOI: 10.3389/fimmu.2019.02930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/28/2019] [Indexed: 12/20/2022] Open
Abstract
YKL-39 belongs to the evolutionarily conserved family of Glyco_18-containing proteins composed of chitinases and chitinase-like proteins. Chitinase-like proteins (CLPs) are secreted lectins that lack hydrolytic activity due to the amino acid substitutions in their catalytic domain and combine the functions of cytokines and growth factors. One of the major cellular sources that produce CLPs in various pathologies, including cancer, are macrophages. Monocytes recruited to the tumor site and programmed by tumor cells differentiate into tumor-associated macrophages (TAMs), which are the primary source of pro-angiogenic factors. Tumor angiogenesis is a crucial process for supplying rapidly growing tumors with essential nutrients and oxygen. We recently determined that YKL-39 is produced by tumor-associated macrophages in breast cancer. YKL-39 acts as a strong chemotactic factor for monocytes and stimulates angiogenesis. Chemotherapy is a common strategy to reduce tumor size and aggressiveness before surgical intervention, but chemoresistance, resulting in the relapse of tumors, is a common clinical problem that is critical for survival in cancer patients. Accumulating evidence indicates that TAMs are essential regulators of chemoresistance. We have recently found that elevated levels of YKL-39 expression are indicative of the efficiency of the metastatic process in patients who undergo neoadjuvant chemotherapy. We suggest YKL-39 as a new target for anti-angiogenic therapy that can be combined with neoadjuvant chemotherapy to reduce chemoresistance and inhibit metastasis in breast cancer patients.
Collapse
Affiliation(s)
- Julia Kzhyshkowska
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - Tengfei Liu
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
12
|
Yeo IJ, Lee CK, Han SB, Yun J, Hong JT. Roles of chitinase 3-like 1 in the development of cancer, neurodegenerative diseases, and inflammatory diseases. Pharmacol Ther 2019; 203:107394. [PMID: 31356910 DOI: 10.1016/j.pharmthera.2019.107394] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2019] [Indexed: 02/07/2023]
Abstract
Chitinase 3-like 1 (CHI3L1) is a secreted glycoprotein that mediates inflammation, macrophage polarization, apoptosis, and carcinogenesis. The expression of CHI3L1 is strongly increased by various inflammatory and immunological conditions, including rheumatoid arthritis, multiple sclerosis, Alzheimer's disease, and several cancers. However, its physiological and pathophysiological roles in the development of cancer and neurodegenerative and inflammatory diseases remain unclear. Several studies have reported that CHI3L1 promotes cancer proliferation, inflammatory cytokine production, and microglial activation, and that multiple receptors, such as advanced glycation end product, syndecan-1/αVβ3, and IL-13Rα2, are involved. In addition, the pro-inflammatory action of CHI3L1 may be mediated via the protein kinase B and phosphoinositide-3 signaling pathways and responses to various pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interferon-γ. Therefore, CHI3L1 could contribute to a vast array of inflammatory diseases. In this article, we review recent findings regarding the roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of cancers, neurodegenerative diseases, and inflammatory diseases.
Collapse
Affiliation(s)
- In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Chong-Kil Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk 28160, Republic of Korea.
| |
Collapse
|
13
|
Plasm YKL-40 Levels Are Associated with Hypertension in Patients with Obstructive Sleep Apnea. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5193597. [PMID: 31001555 PMCID: PMC6436335 DOI: 10.1155/2019/5193597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/27/2018] [Accepted: 01/24/2019] [Indexed: 01/06/2023]
Abstract
Background Obstructive sleep apnea (OSA) is a common disease. It can cause many serious complications. OSA may increase the risk of hypertension. However, the exact mechanism of OSA causing hypertension is not fully understood. YKL-40/chitinase-3-like protein-1 plays an important role in vascular injury, repair, and generation. We sought to explore the role of YKL-40 in endothelial dysfunction and hypertension in OSA patients. Methods All subjects were examined by polysomnography (PSG) and the expression of YKL-40 in the plasm of the subjects was measured by luminex. Carotid intima-media thickness (CIMT) was measured by B-mode ultrasound. Results According to the conditions of OSA and hypertension, we studied four groups of 157 subjects, including OSA group (OSA, N=77), OSA with hypertension group (OSA+HT, N=37), hypertension group (HT, N=20), and healthly group (Con, N=23). YKL-40 levels were significantly elevated in OSA, OSA+HT, and HT group compared to Con groups. We used the ROC to predict the sensitivity and specificity of YKL-40 in all OSA patients or all hyperpietic patients. For OSA patients, the AUC of YKL-40 is 0.807 (95% confidence interval 0.725–0.888; p<0.01). For hyperpietic patients, the AUC of YKL-40 is 0.656 (95% confidence interval 0.570–0.742, p=0.01). There was a significant correlation between the parameter of OSA and hypertension and YKL-40 (P<0.05) and a significant correlation between Max-CIMT and YKL-40 (P<0.05). Conclusion Elevated circulating levels of YKL-40 are associated with hypertension in OSA patients. The specificity of YKL-40 suggests that it could be a potential biomarker for OSA and hypertension.
Collapse
|
14
|
van Sleen Y, Sandovici M, Abdulahad WH, Bijzet J, van der Geest KSM, Boots AMH, Brouwer E. Markers of angiogenesis and macrophage products for predicting disease course and monitoring vascular inflammation in giant cell arteritis. Rheumatology (Oxford) 2019; 58:kez034. [PMID: 30805622 PMCID: PMC6649803 DOI: 10.1093/rheumatology/kez034] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/21/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE GCA, a systemic vasculitis, is characterized by an IL-6-dependent acute-phase response. This response is typically suppressed by treatment rendering CRP/ESR unreliable for monitoring vascular inflammation. Also, there are no accurate biomarkers predicting a non-favourable disease course. Here we investigated macrophage products and markers of angiogenesis as biomarkers for prognosis and monitoring of vascular inflammation. METHODS Forty-one newly diagnosed, glucocorticoid-naive GCA patients were prospectively followed for relapses and glucocorticoid requirement for a median of 30 months (range 0-71). Serum markers at baseline and during follow-up were compared with 33 age-matched healthy controls and 13 infection controls. Concentrations of IL-6, serum amyloid A, soluble CD163, calprotectin, YKL-40, VEGF, angiopoietin-1 and -2 and sTie2 were determined by ELISA/Luminex assay. RESULTS Serum concentrations of all markers, but not angiopoietin-1, were elevated in GCA patients at baseline when compared with healthy controls. High VEGF (P = 0.0025) and angiopoietin-1 (P = 0.0174) and low YKL-40 (P = 0.0369) levels at baseline were predictive of a short time to glucocorticoid-free remission. Elevated angiopoietin-2 levels were associated with an imminent relapse during treatment (P < 0.05). IL-6 correlated strongly with acute-phase markers and soluble CD163 but not with markers of angiogenesis, YKL-40 or calprotectin. Glucocorticoid treatment down-modulated all markers except for calprotectin and YKL-40. Tissue expression of markers in temporal arteries was confirmed. CONCLUSION Markers of angiogenesis at baseline and during treatment predict GCA disease course, suggesting utility in patient stratification for glucocorticoid-sparing therapy. Calprotectin and YKL-40 are candidate markers for monitoring vessel wall inflammation.
Collapse
Affiliation(s)
- Yannick van Sleen
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria Sandovici
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Wayel H Abdulahad
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan Bijzet
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kornelis S M van der Geest
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annemieke M H Boots
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth Brouwer
- Vasculitis Expertise Center Groningen, Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
15
|
YKL-40 is a local marker for inflammation in patients with pseudoexfoliation syndrome. Eye (Lond) 2018; 33:772-776. [PMID: 30560917 DOI: 10.1038/s41433-018-0308-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 09/07/2018] [Accepted: 10/17/2018] [Indexed: 01/05/2023] Open
Abstract
PURPOSE To investigate the YKL-40, as a marker of inflammation, in aqueous humor and serum of cataract patients with and without pseudoexfoliation syndrome (PEX). METHODS Aqueous humor and serum samples were obtained from 44 patients who underwent phacoemulsification surgery. All patients were divided into two groups: PEX (n = 24) and control (n = 20). YKL-40 levels were measured with enzyme-linked immunosorbent assay (ELISA). The differences between the groups were assessed by using Chi-square and independent sample t-tests. The Pearson correlation coefficient was used to evaluate the correlation between variables. RESULTS There was a significant difference between the mean YKL-40 levels in the aqueous humor of PEX group (112.0 ± 35.8 ng/mL) and control subjects (88.2 ± 30.6 ng/mL) (P = 0.025). However, the difference between the mean YKL-40 levels in the serum of PEX group (53.5 ± 29.1 ng/mL) and control subjects (44.6 ± 30.2 ng/mL) was non-significant (P = 0.326). The correlation between aqueous humor and serum YKL-40 concentrations was significant in both the groups (r = 0.833, P < 0.001; r = 0.840, P < 0.001, respectively). CONCLUSIONS Increased aqueous humor levels of YKL-40 demonstrate that it is local, but not a systemic marker for inflammation in patients with PEX.
Collapse
|
16
|
Crotty Alexander LE, Drummond CA, Hepokoski M, Mathew D, Moshensky A, Willeford A, Das S, Singh P, Yong Z, Lee JH, Vega K, Du A, Shin J, Javier C, Tian J, Brown JH, Breen EC. Chronic inhalation of e-cigarette vapor containing nicotine disrupts airway barrier function and induces systemic inflammation and multiorgan fibrosis in mice. Am J Physiol Regul Integr Comp Physiol 2018; 314:R834-R847. [PMID: 29384700 DOI: 10.1152/ajpregu.00270.2017] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Electronic (e)-cigarettes theoretically may be safer than conventional tobacco. However, our prior studies demonstrated direct adverse effects of e-cigarette vapor (EV) on airway cells, including decreased viability and function. We hypothesize that repetitive, chronic inhalation of EV will diminish airway barrier function, leading to inflammatory protein release into circulation, creating a systemic inflammatory state, ultimately leading to distant organ injury and dysfunction. C57BL/6 and CD-1 mice underwent nose only EV exposure daily for 3-6 mo, followed by cardiorenal physiological testing. Primary human bronchial epithelial cells were grown at an air-liquid interface and exposed to EV for 15 min daily for 3-5 days before functional testing. Daily inhalation of EV increased circulating proinflammatory and profibrotic proteins in both C57BL/6 and CD-1 mice: the greatest increases observed were in angiopoietin-1 (31-fold) and EGF (25-fold). Proinflammatory responses were recapitulated by daily EV exposures in vitro of human airway epithelium, with EV epithelium secreting higher IL-8 in response to infection (227 vs. 37 pg/ml, respectively; P < 0.05). Chronic EV inhalation in vivo reduced renal filtration by 20% ( P = 0.017). Fibrosis, assessed by Masson's trichrome and Picrosirius red staining, was increased in EV kidneys (1.86-fold, C57BL/6; 3.2-fold, CD-1; P < 0.05), heart (2.75-fold, C57BL/6 mice; P < 0.05), and liver (1.77-fold in CD-1; P < 0.0001). Gene expression changes demonstrated profibrotic pathway activation. EV inhalation altered cardiovascular function, with decreased heart rate ( P < 0.01), and elevated blood pressure ( P = 0.016). These data demonstrate that chronic inhalation of EV may lead to increased inflammation, organ damage, and cardiorenal and hepatic disease.
Collapse
Affiliation(s)
- Laura E Crotty Alexander
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | | | - Mark Hepokoski
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Denzil Mathew
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Alex Moshensky
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Andrew Willeford
- Department of Pharmacology, University of California , San Diego, California
| | - Soumita Das
- Department of Pathology, University of California , San Diego, California
| | - Prabhleen Singh
- Division of Nephrology and Hypertension, Department of Medicine, University of California , San Diego, California.,Nephrology Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Zach Yong
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Jasmine H Lee
- Division of Physiology, Department of Medicine, University of California , San Diego, California
| | - Kevin Vega
- Department of Pathology, University of California , San Diego, California
| | - Ashley Du
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - John Shin
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Christian Javier
- Pulmonary Critical Care Section, Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California.,Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, University of California , San Diego, California
| | - Jiang Tian
- Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo , Toledo, Ohio.,Department of Medicine, College of Medicine and Life Sciences, University of Toledo , Toledo, Ohio
| | - Joan Heller Brown
- Department of Pharmacology, University of California , San Diego, California
| | - Ellen C Breen
- Division of Physiology, Department of Medicine, University of California , San Diego, California
| |
Collapse
|
17
|
Lorenz G, Schmalenberg M, Kemmner S, Haller B, Steubl D, Pham D, Schreiegg A, Bachmann Q, Schmidt A, Haderer S, Huber M, Angermann S, Günthner R, Braunisch M, Hauser C, Reichelt AL, Matschkal J, Suttmann Y, Moog P, Stock K, Küchle C, Thürmel K, Renders L, Bauer A, Baumann M, Heemann U, Luppa PB, Schmaderer C. Mortality prediction in stable hemodialysis patients is refined by YKL-40, a 40-kDa glycoprotein associated with inflammation. Kidney Int 2017; 93:221-230. [PMID: 28941940 DOI: 10.1016/j.kint.2017.07.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/01/2017] [Accepted: 07/13/2017] [Indexed: 12/21/2022]
Abstract
Chronic inflammation contributes to increased mortality in hemodialysis (HD) patients. YKL-40 is a novel marker of inflammation, tissue remodeling, and highly expressed in macrophages inside vascular lesions. Elevated levels of YKL-40 have been reported for HD patients but how it integrates into the proinflammatory mediator network as a predictor of mortality remains elusive. We studied serum YKL-40, Interleukin-6 (IL-6), high-sensitivity C-reactive protein, monocyte chemotactic protein-1 (MCP-1), and interferon-gamma induced protein-10 (IP-10) in 475 chronic hemodialysis patients. Patients were followed for mortality for a median of 37 [interquartile range: 25-49] months and checked for interrelation of the measured mediators. To plot cumulative incidence functions, patients were stratified into terciles per YKL-40, IL-6, MCP-1, and IP-10 levels. Multivariable Cox regression models were built to examine associations of YKL-40, IP-10, and MCP-1 with all-cause and cause-specific mortality. Net reclassification improvement was calculated for the final models containing YKL-40 and IL-6. Increased YKL-40 was independently associated with age, IP-10, and IL-6 serum levels. After adjustment for demographic and laboratory parameters, comorbidities, and IL-6, only YKL-40 significantly improved risk prediction for all-cause (hazard ratio 1.4; 95% confidence interval 1.1-1.8) and cardiovascular mortality (hazard ratio 1.5; 95% confidence interval 1.03-2.2). Thus, in contrast to other biomarkers of aberrant macrophage activation, YKL-40 reflects inflammatory activity, which is not covered by IL-6. Mechanistic and prospective studies are needed to test for causal involvement of YKL-40 and whether it might qualify as a therapeutic target.
Collapse
Affiliation(s)
- Georg Lorenz
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
| | - Michael Schmalenberg
- Department of Clinical Chemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Stephan Kemmner
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Bernhard Haller
- Department of Medical Statistics and Epidemiology, Technical University Munich, Munich, Germany
| | - Dominik Steubl
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Dang Pham
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Anita Schreiegg
- Department of Clinical Chemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Quirin Bachmann
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Alina Schmidt
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sandra Haderer
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Monika Huber
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Susanne Angermann
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Roman Günthner
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Matthias Braunisch
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Christine Hauser
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Anna-Lena Reichelt
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Julia Matschkal
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Yana Suttmann
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Philipp Moog
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Konrad Stock
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Claudius Küchle
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Klaus Thürmel
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Lutz Renders
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Axel Bauer
- Department of Cardiology, Ludwig-Maximilian University, Munich, Germany
| | - Marcus Baumann
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Peter B Luppa
- Department of Clinical Chemistry, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
| |
Collapse
|