1
|
Wang X, Ye T, Huang J, Hu F, Huang C, Gu B, Xu X, Yang J. Aberrant Chitinase 3-Like 1 Expression in Basal Cells Contributes to Systemic Sclerosis Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2310169. [PMID: 39686726 DOI: 10.1002/advs.202310169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 10/06/2024] [Indexed: 12/18/2024]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by extensive skin and internal organ fibrosis. However, the mechanism underlying fibrosis remains unclear, and effective treatments for halting or reversing fibrosis are lacking. In this study, single-cell RNA sequencing is used to obtain a comprehensive overview of skin cells from patients with SSc and healthy controls. A subset of basal cells with high chitinase 3-like 1 (Chi3L1) expression, which potentially plays an important role in fibroblast activation, is identified in SSc. Subsequently, patients with SSc are present with increased expression of Chi3L1 in the skin and serum, and elevated serum levels are associated with skin induration and pulmonary function. Furthermore, Chi3L1 promoted the differentiation of SSc dermal fibroblasts into myofibroblasts, and Chi3L1-deficient (Chi3L1-/-) mice showed amelioration of fibrosis in a bleomycin-induced SSc (BLM-SSc) model. Mechanistically, Chi3L1 mediates fibroblast activation primarily by interacting with interleukin-17 receptor A (IL-17RA), thereby initiating downstream nuclear factor kappa B and mitogen-activated protein kinases signaling pathways. Moreover, the anti-fibrotic effect of IL-17RA antagonists in BLM-SSc mice is demonstrated. In conclusion, Chi3L1 is a potential biomarker for the degree of fibrosis in SSc. Chi3L1 and its receptor, IL-17RA, are promising therapeutic targets for patients with SSc.
Collapse
Affiliation(s)
- Xiuyuan Wang
- Department of Dermatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Tianbao Ye
- Sixth People's Hospital affiliated to Shanghai Jiao Tong University, Shanghai, 200233, China
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361008, China
| | - Junxia Huang
- Department of Dermatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Feifei Hu
- Department of Dermatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Chengjie Huang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Bei Gu
- Shanghai Normal University, Shanghai, 200233, China
| | - Xinzhi Xu
- Department of Dermatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Ji Yang
- Department of Dermatology, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| |
Collapse
|
2
|
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
|
3
|
Sorkhdini P, Klubock-Shukla K, Sheth S, Yang D, Yang AX, Norbrun C, Introne WJ, Gochuico BR, Zhou Y. Type 2 innate immunity promotes the development of pulmonary fibrosis in Hermansky-Pudlak syndrome. JCI Insight 2024; 9:e178381. [PMID: 39405112 PMCID: PMC11601950 DOI: 10.1172/jci.insight.178381] [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: 12/08/2023] [Accepted: 10/08/2024] [Indexed: 10/20/2024] Open
Abstract
Hermansky-Pudlak syndrome (HPS), particularly types 1 and 4, is characterized by progressive pulmonary fibrosis, a major cause of morbidity and mortality. However, the precise mechanisms driving pulmonary fibrosis in HPS are not fully elucidated. Our previous studies suggested that CHI3L1-driven fibroproliferation may be a notable factor in HPS-associated fibrosis. This study aimed to explore the role of CHI3L1-CRTH2 interaction on type 2 innate lymphoid cells (ILC2s) and explored the potential contribution of ILC2-fibroblast crosstalk in the development of pulmonary fibrosis in HPS. We identified ILC2s in lung tissues from patients with idiopathic pulmonary fibrosis and HPS. Using bleomycin-challenged WT and Hps1-/- mice, we observed that ILC2s were recruited and appeared to contribute to fibrosis development in the Hps1-/- mice, with CRTH2 playing a notable role in ILC2 accumulation. We sorted ILC2s, profiled fibrosis-related genes and mediators, and conducted coculture experiments with primary lung ILC2s and fibroblasts. Our findings suggest that ILC2s may directly stimulate the proliferation and differentiation of primary lung fibroblasts partially through amphiregulin-EGFR-dependent mechanisms. Additionally, specific overexpression of CHI3L1 in the ILC2 population using the IL-7Rcre driver, which was associated with increased fibroproliferation, indicates that ILC2-mediated, CRTH2-dependent mechanisms might contribute to optimal CHI3L1-induced fibroproliferative repair in HPS-associated pulmonary fibrosis.
Collapse
Affiliation(s)
- Parand Sorkhdini
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Kiran Klubock-Shukla
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Selena Sheth
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Dongqin Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Alina Xiaoyu Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Carmelissa Norbrun
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Wendy J. Introne
- Medical Genetics Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Bernadette R. Gochuico
- Medical Genetics Branch, National Human Genome Research Institute (NHGRI), NIH, Bethesda, Maryland, USA
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
4
|
Ma B, Kamle S, Sadanaga T, Lee CM, Lee JH, Yee DC, Zhu Z, Silverman EK, DeMeo DL, Choi AMK, Lee CG, Elias JA. Chitinase 3-like-1 Inhibits Innate Antitumor and Tissue Remodeling Immune Responses by Regulating CD47-SIRPα- and CD24-Siglec10-Mediated Phagocytosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1279-1291. [PMID: 39291933 DOI: 10.4049/jimmunol.2400035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024]
Abstract
Innate immune responses such as phagocytosis are critically linked to the generation of adaptive immune responses against the neoantigens in cancer and the efferocytosis that is essential for homeostasis in diseases characterized by lung injury, inflammation, and remodeling as in chronic obstructive pulmonary disease (COPD). Chitinase 3-like-1 (CHI3L1) is induced in many cancers where it inhibits adaptive immune responses by stimulating immune checkpoint molecules (ICPs) and portends a poor prognosis. CHI3L1 is also induced in COPD where it regulates epithelial cell death. In this study, we demonstrate that pulmonary melanoma metastasis inhibits macrophage phagocytosis by stimulating the CD47-SIRPα and CD24-Siglec10 phagocytosis checkpoint pathways while inhibiting macrophage "eat me" signals from calreticulin and HMGB1. We also demonstrate that these effects on macrophage phagocytosis are associated with CHI3L1 stimulation of the SHP-1 and SHP-2 phosphatases and inhibition of the accumulation and phosphorylation of cytoskeleton-regulating nonmuscle myosin IIa. This inhibition of innate immune responses such as phagocytosis provides a mechanistic explanation for the ability of CHI3L1 to stimulate ICPs and inhibit adaptive immune responses in cancer and diseases such as COPD. The ability of CHI3L1 to simultaneously inhibit innate immune responses, stimulate ICPs, inhibit T cell costimulation, and regulate a number of other oncogenic and inflammation pathways suggests that CHI3L1-targeted therapeutics are promising interventions in cancer, COPD, and other disorders.
Collapse
Affiliation(s)
- Bing Ma
- Molecular Microbiology and Immunology, Brown University, Providence, RI
| | - Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, Providence, RI
| | - Takayuki Sadanaga
- Molecular Microbiology and Immunology, Brown University, Providence, RI
| | - Chang-Min Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI
| | - Joyce H Lee
- Johns Hopkins School of Medicine, Baltimore, MD
| | - Daniel C Yee
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY
| | - Zhou Zhu
- Department of Pediatrics, Brown University, Providence, RI
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Woman's Hospital, Harvard Medical School, Boston, MA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Woman's Hospital, Harvard Medical School, Boston, MA
| | | | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI
| | - Jack A Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI
- Department of Medicine, Brown University, Providence, RI
| |
Collapse
|
5
|
Li K, Wang XQ, Liao ZL, Liu JY, Feng BH, Ren YC, Dai NN, Yu K, Yu H, Chen HJ, Mei H, Qin S. Wedelolactone inhibits ferroptosis and alleviates hyperoxia-induced acute lung injury via the Nrf2/HO-1 signaling pathway. Toxicol Sci 2024; 202:25-35. [PMID: 39110510 DOI: 10.1093/toxsci/kfae099] [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] [Indexed: 10/29/2024] Open
Abstract
Hyperoxia-induced acute lung injury (HALI) is a complication of oxygen therapy. Ferroptosis is a vital factor in HALI. This paper was anticipated to investigate the underlying mechanism of wedelolactone (WED) on ferroptosis in HALI. The current study used hyperoxia to injure two models, one HALI mouse model and one MLE-12 cell injury model. We found that WED treatment attenuated HALI by decreasing the lung injury score and lung wet/dry (W/D) weight ratio and alleviating pathomorphological changes. Then, the inflammatory reaction and apoptosis in HALI mice and hyperoxia-mediated MLE-12 cells were inhibited by WED treatment. Moreover, WED alleviated ferroptosis with less iron accumulation and reversed expression alterations of ferroptosis markers, including MDA, GSH, GPX4, SLC7A11, FTH1, and TFR1 in hyperoxia-induced MLE-12 cells in vitro and in vivo. Nrf2-KO mice and Nrf2 inhibitor (ML385) decreased WED's ability to protect against apoptosis, inflammatory response, and ferroptosis in hyperoxia-induced MLE-12 cells. Collectively, our data highlighted the alleviatory role of WED in HALI by activating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Kang Li
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Xiao-Qin Wang
- Department of Pediatric, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Zhen-Liang Liao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Jun-Ya Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Bang-Hai Feng
- Department of Critical Care Medicine, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou 563000, P.R. China
| | - Ying-Cong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Ni-Nan Dai
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Kun Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hong Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hua-Jun Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Hong Mei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| | - Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
6
|
Kamle S, Ma B, Schor G, Bailey M, Pham B, Cho I, Khan H, Azzoli C, Hofstetter M, Sadanaga T, Herbst R, Politi K, Lee CG, Elias JA. Chitinase 3-like-1 (CHI3L1) in the pathogenesis of epidermal growth factor receptor mutant non-small cell lung cancer. Transl Oncol 2024; 49:102108. [PMID: 39178575 PMCID: PMC11388375 DOI: 10.1016/j.tranon.2024.102108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/26/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for 85 % of all lung cancers. In NSCLC, 10-20 % of Caucasian patients and 30-50 % of Asian patients have tumors with activating mutations in the Epidermal Growth Factor Receptor (EGFR). A high percentage of these patients exhibit favorable responses to treatment with tyrosine kinase inhibitors (TKI). Unfortunately, a majority of these patients develop therapeutic resistance with progression free survival lasting 9-18 months. The mechanisms that underlie the tumorigenic effects of EGFR and the ability of NSCLC to develop resistance to TKI therapies, however, are poorly understood. Here we demonstrate that CHI3L1 is produced by EGFR activation of normal epithelial cells, transformed epithelial cells with wild type EGFR and cells with cancer-associated, activating EGFR mutations. We also demonstrate that CHI3L1 auto-induces itself and feeds back to stimulate EGFR and its ligands via a STAT3-dependent mechanism(s). Highly specific antibodies against CHI3L1 (anti-CHI3L1/FRG) and TKI, individually and in combination, abrogated the effects of EGFR activation on CHI3L1 and the ability of CHI3L1 to stimulate the EGFR axis. Anti-CHI3L1 also interacted with osimertinib to reverse TKI therapeutic resistance and induce tumor cell death and inhibit pulmonary metastasis while stimulating tumor suppressor genes including KEAP1. CHI3L1 is a downstream target of EGFR that feeds back to stimulate and activate the EGFR axis. Anti-CHI3L1 is an exciting potential therapeutic for EGFR mutant NSCLC, alone and in combination with osimertinib or other TKIs.
Collapse
Affiliation(s)
- Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA; Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Bing Ma
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Gail Schor
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Madison Bailey
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Brianna Pham
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Inyoung Cho
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Hina Khan
- Medical Oncology, Department of Medicine, Warren Alpert Medical School Brown University, USA
| | - Christopher Azzoli
- Medical Oncology, Department of Medicine, Warren Alpert Medical School Brown University, USA
| | - Mara Hofstetter
- Department of Chemistry, Yale University, USA; University of Zurich, Switzerland
| | - Takayuki Sadanaga
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Roy Herbst
- Medical Oncology, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Katerina Politi
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA; Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Jack A Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA; Legorreta Cancer Center, Brown University, Providence, RI, USA; Departments of Medicine, Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
7
|
Johnston RA, Pilkington AW, Atkins CL, Boots TE, Brown PL, Jackson WT, Spencer CY, Siddiqui SR, Haque IU. Inconsequential role for chemerin-like receptor 1 in the manifestation of ozone-induced lung pathophysiology in male mice. Physiol Rep 2024; 12:e16008. [PMID: 38631890 PMCID: PMC11023814 DOI: 10.14814/phy2.16008] [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: 02/22/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/19/2024] Open
Abstract
We executed this study to determine if chemerin-like receptor 1 (CMKLR1), a Gi/o protein-coupled receptor expressed by leukocytes and non-leukocytes, contributes to the development of phenotypic features of non-atopic asthma, including airway hyperresponsiveness (AHR) to acetyl-β-methylcholine chloride, lung hyperpermeability, airway epithelial cell desquamation, and lung inflammation. Accordingly, we quantified sequelae of non-atopic asthma in wild-type mice and mice incapable of expressing CMKLR1 (CMKLR1-deficient mice) following cessation of acute inhalation exposure to either filtered room air (air) or ozone (O3), a criteria pollutant and non-atopic asthma stimulus. Following exposure to air, lung elastic recoil and airway responsiveness were greater while the quantity of adiponectin, a multi-functional adipocytokine, in bronchoalveolar lavage (BAL) fluid was lower in CMKLR1-deficient as compared to wild-type mice. Regardless of genotype, exposure to O3 caused AHR, lung hyperpermeability, airway epithelial cell desquamation, and lung inflammation. Nevertheless, except for minimal genotype-related effects on lung hyperpermeability and BAL adiponectin, we observed no other genotype-related differences following O3 exposure. In summary, we demonstrate that CMKLR1 limits the severity of innate airway responsiveness and lung elastic recoil but has a nominal effect on lung pathophysiology induced by acute exposure to O3.
Collapse
Affiliation(s)
- Richard A. Johnston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and PreventionUnited States Department of Health and Human ServicesMorgantownWest VirginiaUSA
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of MedicineWest Virginia UniversityMorgantownWest VirginiaUSA
- Division of Critical Care Medicine, Department of PediatricsMcGovern Medical School at the University of Texas Health Science Center at HoustonHoustonTexasUSA
- Department of Integrative Biology and PharmacologyMcGovern Medical School at the University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Albert W. Pilkington
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and PreventionUnited States Department of Health and Human ServicesMorgantownWest VirginiaUSA
| | - Constance L. Atkins
- Division of Pulmonary Medicine, Department of PediatricsMcGovern Medical School at the University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Theresa E. Boots
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and PreventionUnited States Department of Health and Human ServicesMorgantownWest VirginiaUSA
| | - Philip L. Brown
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and PreventionUnited States Department of Health and Human ServicesMorgantownWest VirginiaUSA
| | - William T. Jackson
- Division of Critical Care Medicine, Department of PediatricsMcGovern Medical School at the University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Chantal Y. Spencer
- Section of Pediatric Pulmonology, Department of PediatricsBaylor College of MedicineHoustonTexasUSA
| | - Saad R. Siddiqui
- Division of Critical Care Medicine, Department of PediatricsMcGovern Medical School at the University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Ikram U. Haque
- Division of Critical Care Medicine, Department of PediatricsMcGovern Medical School at the University of Texas Health Science Center at HoustonHoustonTexasUSA
- Division of Critical Care, Department of PediatricsSidra MedicineDohaQatar
| |
Collapse
|
8
|
Li L, Xu S, Li M, Yin X, Xi H, Yang P, Ma L, Zhang L, Li X. Combined gestational age and serum fucose for early prediction of risk for bronchopulmonary dysplasia in premature infants. BMC Pediatr 2024; 24:107. [PMID: 38347448 PMCID: PMC10860215 DOI: 10.1186/s12887-024-04556-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
OBJECTIVE As the predominant complication in preterm infants, Bronchopulmonary Dysplasia (BPD) necessitates accurate identification of infants at risk and expedited therapeutic interventions for an improved prognosis. This study evaluates the potential of Monosaccharide Composite (MC) enriched with environmental information from circulating glycans as a diagnostic biomarker for early-onset BPD, and, concurrently, appraises BPD risk in premature neonates. MATERIALS AND METHODS The study incorporated 234 neonates of ≤32 weeks gestational age. Clinical data and serum samples, collected one week post-birth, were meticulously assessed. The quantification of serum-free monosaccharides and their degraded counterparts was accomplished via High-performance Liquid Chromatography (HPLC). Logistic regression analysis facilitated the construction of models for early BPD diagnosis. The diagnostic potential of various monosaccharides for BPD was determined using Receiver Operating Characteristic (ROC) curves, integrating clinical data for enhanced diagnostic precision, and evaluated by the Area Under the Curve (AUC). RESULTS Among the 234 neonates deemed eligible, BPD development was noted in 68 (29.06%), with 70.59% mild (48/68) and 29.41% moderate-severe (20/68) cases. Multivariate analysis delineated several significant risk factors for BPD, including gestational age, birth weight, duration of both invasive mechanical and non-invasive ventilation, Patent Ductus Arteriosus (PDA), pregnancy-induced hypertension, and concentrations of two free monosaccharides (Glc-F and Man-F) and five degraded monosaccharides (Fuc-D, GalN-D, Glc-D, and Man-D). Notably, the concentrations of Glc-D and Fuc-D in the moderate-to-severe BPD group were significantly diminished relative to the mild BPD group. A potent predictive capability for BPD development was exhibited by the conjunction of gestational age and Fuc-D, with an AUC of 0.96. CONCLUSION A predictive model harnessing the power of gestational age and Fuc-D demonstrates promising efficacy in foretelling BPD development with high sensitivity (95.0%) and specificity (94.81%), potentially enabling timely intervention and improved neonatal outcomes.
Collapse
Affiliation(s)
- Liangliang Li
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Shimin Xu
- Division of Neonatology, Beijing jingdu Children's Hospital, Beijing, China
| | - Miaomiao Li
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Xiangyun Yin
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Hongmin Xi
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Ping Yang
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Lili Ma
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China
| | - Lijuan Zhang
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China.
| | - Xianghong Li
- Division of Neonatology, The Affiliated Hospital of Qingdao University, Shandong, China.
| |
Collapse
|
9
|
Kui L, Kim AD, Onyuru J, Hoffman HM, Feldstein AE. BRP39 Regulates Neutrophil Recruitment in NLRP3 Inflammasome-Induced Liver Inflammation. Cell Mol Gastroenterol Hepatol 2023; 17:481-497. [PMID: 38092312 PMCID: PMC10837621 DOI: 10.1016/j.jcmgh.2023.12.002] [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: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND & AIMS Breast regression protein 39 (BRP39) (Chi3L1) and its human homolog YKL-40, is an established biomarker of liver fibrosis in nonalcoholic steatohepatitis (NASH) patients, but its role in NASH pathogenesis remains unclear. We recently identified Chi3L1 as one of the top up-regulated genes in mice with inducible gain-of-function NOD-like receptor protein 3 (NLRP3) activation that mimics several liver features of NASH. This study aimed to investigate the effects of BRP39 deficiency on NLRP3-induced liver inflammation using tamoxifen-inducible Nlrp3 knockin mice sufficient (Nlrp3A350V CRT) and deficient for BRP39 (Nlrp3A350V/BRP-/- CRT). METHODS Using Nlrp3A350V CRT mice and Nlrp3A350V BRP-/- CRT, we investigated the consequences of BRP39 deficiency influencing NLRP3-induced liver inflammation. RESULTS Our results showed that BRP39 deficiency in NLRP3-induced inflammation improved body weight and liver weight. Moreover, liver inflammation, fibrosis, and hepatic stellate cell activation were reduced significantly, corresponding to significantly decreased Ly6C+ infiltrating macrophages, CD68+ osteopontin-positive hepatic lipid-associated macrophages, and activated Lymphocyte antigen 6 complex locus G6D positive (Ly6G+) and citrullinated histone H3 postivie (H3Cit+) neutrophil accumulation in the liver. Further investigation showed that circulatory neutrophils from NLRP3-induced BRP39-deficient mice have impaired chemotaxis and migration ability, and this was confirmed by RNA bulk sequencing showing reduced immune activation, migration, and signaling responses in neutrophils. CONCLUSIONS These data showcase the importance of BRP39 in regulating the NLRP3 inflammasome during liver inflammation and fibrotic NASH by altering cellular activation, recruitment, and infiltration during disease progression, and revealing BRP39 to be a potential therapeutic target for future treatment of inflammatory NASH and its associated diseases.
Collapse
Affiliation(s)
- Lin Kui
- Department of Pediatrics, University of California San Diego, San Diego, California
| | - Andrea D Kim
- Department of Pediatrics, University of California San Diego, San Diego, California; Department of Surgery, Johns Hopkins University, Baltimore, Maryland
| | - Janset Onyuru
- Department of Pediatrics, University of California San Diego, San Diego, California
| | - Hal M Hoffman
- Department of Pediatrics, University of California San Diego, San Diego, California
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, San Diego, California; Global Drug Discovery, Novo Nordisk, Denmark.
| |
Collapse
|
10
|
Sipeki N, Kovats PJ, Deutschmann C, Schierack P, Roggenbuck D, Papp M. Location-based prediction model for Crohn's disease regarding a novel serological marker, anti-chitinase 3-like 1 autoantibodies. World J Gastroenterol 2023; 29:5728-5750. [PMID: 38075846 PMCID: PMC10701337 DOI: 10.3748/wjg.v29.i42.5728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/03/2023] [Accepted: 11/02/2023] [Indexed: 11/13/2023] Open
Abstract
BACKGROUND Defective neutrophil regulation in inflammatory bowel disease (IBD) is thought to play an important role in the onset or manifestation of IBD, as it could lead to damage of the intestinal mucosal barrier by the infiltration of neutrophils in the inflamed mucosa and the accumulation of pathogens. Like neutrophils in the context of innate immune responses, immunoglobulin A (IgA) as an acquired immune response partakes in the defense of the intestinal epithelium. Under normal conditions, IgA contributes to the elimination of microbes, but in connection with the loss of tolerance to chitinase 3-like 1 (CHI3L1) in IBD, IgA could participate in CHI3L1-mediated improved adhesion and invasion of potentially pathogenic microorganisms. The tolerance brake to CHI3L1 and the occurrence of IgA autoantibodies to this particular target, the exact role and underlying mechanisms of CHI3L1 in the pathogenesis of IBD are still unclear. AIM To determine the predictive potential of Ig subtypes of a novel serological marker, anti-CHI3L1 autoantibodies (aCHI3L1) in determining the disease phenotype, therapeutic strategy and long-term disease course in a prospective referral cohort of adult IBD patients. METHODS Sera of 257 Crohn's disease (CD) and 180 ulcerative colitis (UC) patients from a tertiary IBD referral center of Hungary (Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen) were assayed for IgG, IgA, and secretory IgA (sIgA) type aCHI3L1 by enzyme-linked immunosorbent assay using recombinant CHI3L1, along with 86 healthy controls (HCONT). RESULTS The IgA type was more prevalent in CD than in UC (29.2% vs 11.1%) or HCONT (2.83%; P < 0.0001 for both). However, sIgA subtype aCHI3L1 positivity was higher in both CD and UC patients than in HCONT (39.3% and 32.8% vs 4.65%, respectively; P < 0.0001). The presence of both IgA and sIgA aCHI3L1 antibodies was associated with colonic involvement (P < 0.0001 and P = 0.038, respectively) in patients with CD. Complicated disease behavior at sample procurement was associated with aCHI3L1 sIgA positivity (57.1% vs 36.0%, P = 0.009). IgA type aCH3L1 was more prevalent in patients with frequent relapse during the disease course in the CD group (46.9% vs 25.7%, P = 0.005). In a group of patients with concomitant presence of pure inflammatory luminal disease and colon involvement at the time of diagnosis, positivity for IgA or sIgA type aCH3L1 predicted faster progression towards a complicated disease course in time-dependent models. This association disappeared after merging subgroups of different disease locations. CONCLUSION CHI3L1 is a novel neutrophil autoantigenic target in IBD. The consideration of antibody classes along with location-based prediction may transform the future of serology in IBD.
Collapse
Affiliation(s)
- Nora Sipeki
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
| | - Patricia Julianna Kovats
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
- Kálmán Laki Doctoral School, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
| | - Claudia Deutschmann
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
| | - Peter Schierack
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
| | - Dirk Roggenbuck
- Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg 01968, Germany
- Medipan GmbH & GA Generic Assays GmbH, Dahlewitz-Berlin 15827, Germany
| | - Maria Papp
- Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen H-4032, Hungary
| |
Collapse
|
11
|
NLRX1 knockdown attenuates pro-apoptotic signaling and cell death in pulmonary hyperoxic acute injury. Sci Rep 2023; 13:3441. [PMID: 36859435 PMCID: PMC9975446 DOI: 10.1038/s41598-023-28206-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/13/2023] [Indexed: 03/03/2023] Open
Abstract
Hyperoxia is frequently used for treating acute respiratory failure, but it can cause acute lung injury. Nucleotide-binding domain and leucine-rich-repeat-containing family member X1 (NLRX1) is localized in mitochondria and involved in production of reactive oxygen species, inflammation, and apoptosis, which are the features of hyperoxic acute lung injury (HALI). The contribution of NLRX1 to HALI has not previously been addressed. Thus, to investigate the role of NLRX1 in hyperoxia, we generated a murine model of HALI in wild-type (WT) and NLRX1-/- mice by exposure to > 95% oxygen for 72 h. As a result, NLRX1 expression was elevated in mice exposed to hyperoxia. In acute lung injury, levels of inflammatory cells, protein leakage, cell cytotoxicity, and pro-inflammatory cytokines were diminished in NLRX1-/- mice compared to WT mice. In a survival test, NLRX1-/- mice showed reduced mortality under hyperoxic conditions, and apoptotic cell death and caspase expression and activity were also lower in NLRX1-/- mice. Furthermore, levels of the MAPK signaling proteins ERK 1/2, JNK, and p38 were decreased in NLRX1-deficient mice than in WT mice exposed to hyperoxia. The study shows that a genetic deficit in NLRX1 can suppress hyperoxia-induced apoptosis, suggesting that NLRX1 acts as a pivotal regulator of HALI.
Collapse
|
12
|
Sun X, Nakajima E, Norbrun C, Sorkhdini P, Yang AX, Yang D, Ventetuolo CE, Braza J, Vang A, Aliotta J, Banerjee D, Pereira M, Baird G, Lu Q, Harrington EO, Rounds S, Lee CG, Yao H, Choudhary G, Klinger JR, Zhou Y. Chitinase 3-like-1 contributes to the development of pulmonary vascular remodeling in pulmonary hypertension. JCI Insight 2022; 7:159578. [PMID: 35951428 DOI: 10.1172/jci.insight.159578] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Chitinase 3-like 1 (CHI3L1) is the prototypic chitinase-like protein mediating inflammation, cell proliferation, and tissue remodeling. Limited data suggests CHI3L1 is elevated in human pulmonary arterial hypertension (PAH) and is associated with disease severity. Despite its importance as a regulator of injury/repair responses, the relationship between CHI3L1 and pulmonary vascular remodeling is not well understood. We hypothesize that CHI3L1 and its signaling pathways contribute to the vascular remodeling responses that occur in pulmonary hypertension (PH). We examined the relationship of plasma CHI3L1 levels and severity of PH in patients with various forms of PH, including Group 1 PAH and Group 3 PH, and found that circulating levels of serum CHI3L1 were associated with worse hemodynamics and correlated directly with mean pulmonary artery pressure and pulmonary vascular resistance. We also used transgenic mice with constitutive knockout and inducible overexpression of CHI3L1 to examine its role in hypoxia-, monocrotaline-, and bleomycin-induced models of pulmonary vascular disease. In all 3 mouse models of pulmonary vascular disease, pulmonary hypertensive responses were mitigated in CHI3L1 null mice and accentuated in transgenic mice that overexpress CHI3L1. Finally, CHI3L1 alone was sufficient to induce pulmonary arterial smooth muscle cell proliferation, inhibit pulmonary vascular endothelial cell apoptosis, induce the loss of endothelial barrier function, and induce endothelial-to-mesenchymal transition. These findings demonstrate that CHI3L1 and its receptors play an integral role in pulmonary vascular disease pathobiology and may offer a novel target for the treatment PAH and PH associated with fibrotic lung disease.
Collapse
Affiliation(s)
- Xiuna Sun
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Erika Nakajima
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Carmelissa Norbrun
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Parand Sorkhdini
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Alina Xiaoyu Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Dongqin Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Corey E Ventetuolo
- Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Julie Braza
- Providence VA Medical Center, Providence, United States of America
| | - Alexander Vang
- Research, Providence VA Medical Center, Providence, United States of America
| | - Jason Aliotta
- Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Debasree Banerjee
- Department of Internal Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Mandy Pereira
- Department of Hematology/Oncology, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Grayson Baird
- Department of DIagnostic Imaging, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Qing Lu
- Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | | | - Sharon Rounds
- Providence VA Medical Center, Providence, United States of America
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology, and Biochemistry,, Brown University, Providence, United States of America
| | - Gaurav Choudhary
- Providence VA Medical Center, Providence, United States of America
| | - James R Klinger
- Department of Pulmonary, Sleep, and Critical Care Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, United States of America
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, United States of America
| |
Collapse
|
13
|
Cao Y, Rudrakshala J, Williams R, Rodriguez S, Sorkhdini P, Yang AX, Mundy M, Yang D, Palmisciano A, Walsh T, Delcompare C, Caine T, Tomasi L, Shea BS, Zhou Y. CRTH2 Mediates Pro-fibrotic Macrophage Differentiation and Promotes Lung Fibrosis. Am J Respir Cell Mol Biol 2022; 67:201-214. [PMID: 35585756 DOI: 10.1165/rcmb.2021-0504oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a particularly deadly form of pulmonary fibrosis with unknown reason. In patients with IPF, high serum and lung levels of CHI3L1 can be detected and are associated with poor survival. However, the roles of CHI3L1 in these diseases have not been fully elucidated. We hypothesize that CHI3L1 interacts with CRTH2 to stimulate pro-fibrotic macrophage differentiation and the development of pulmonary fibrosis and that circulating blood monocytes from patients with IPF are hyperresponsive to CHI3L1-CRTH2 signaling. We used murine pulmonary fibrosis models to investigate the role of CRTH2 on pro-fibrotic macrophage differentiation and fibrosis development, and primary human PBMC cell culture to detect the difference of monocytes in the responses to CHI3L1 stimulation and CRTH2 inhibition between IPF patients and normal controls. Our results showed that null mutation or small molecule inhibition of CRTH2 prevents the development of pulmonary fibrosis in murine models. Furthermore, CHI3L1 stimulation induces a greater increase in CD206 expression in IPF monocytes than control monocytes. These results demonstrated that monocytes from IPF patients appear to be hyperresponsive to CHI3L1 stimulation. These studies support targeting CHI3L1-CRTH2 pathway as a promising therapeutic approach in IPF and that the sensitivity of blood monocytes to CHI3L1-induced pro-fibrotic differentiation may serve as a biomarker that predicts responsiveness to CHI3L1 or CRTH2 based interventions.
Collapse
Affiliation(s)
- Yueming Cao
- Brown University, 6752, Providence, Rhode Island, United States
| | | | - River Williams
- Brown University, 6752, Providence, Rhode Island, United States
| | - Shade Rodriguez
- Brown University, 6752, Providence, Rhode Island, United States
| | | | - Alina X Yang
- Brown University, 6752, Providence, Rhode Island, United States
| | - Miles Mundy
- Brown University, 6752, Providence, Rhode Island, United States
| | - Dongqin Yang
- Brown University, 6752, Providence, Rhode Island, United States
| | - Amy Palmisciano
- Rhode Island Hospital, Pulmonary, Critical Care and Sleep, Providence, Rhode Island, United States
| | - Thomas Walsh
- Rhode Island Hospital, 23325, Providence, Rhode Island, United States
| | - Cesar Delcompare
- Rhode Island Hospital, Pulmonary, Critical Care and Sleep, Providence, Rhode Island, United States
| | - Tanis Caine
- Rhode Island Hospital, Pulmonary, Critical Care and Sleep, Providence, Rhode Island, United States
| | - Luca Tomasi
- Rhode Island Hospital, Pulmonary, Critical Care and Sleep, Providence, Rhode Island, United States
| | - Barry S Shea
- Rhode Island Hospital, Pulmonary, Critical Care and Sleep, Providence, Rhode Island, United States
| | - Yang Zhou
- Brown University, Molecular Microbiology and Immunology, Providence, Rhode Island, United States;
| |
Collapse
|
14
|
Lee SY, Lee CM, Ma B, Kamle S, Elias JA, Zhou Y, Lee CG. Targeting Chitinase 1 and Chitinase 3-Like 1 as Novel Therapeutic Strategy of Pulmonary Fibrosis. Front Pharmacol 2022; 13:826471. [PMID: 35370755 PMCID: PMC8969576 DOI: 10.3389/fphar.2022.826471] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/18/2022] [Indexed: 11/21/2022] Open
Abstract
Chitinase 1 (CHIT1) and chitinase 3-like-1 (CHI3L1), two representative members of 18-Glycosyl hydrolases family, are significantly implicated in the pathogenesis of various human diseases characterized by inflammation and remodeling. Notably, dysregulated expression of CHIT1 and CHI3L1 was noted in the patients with pulmonary fibrosis and their levels were inversely correlated with clinical outcome of the patients. CHIT1 and CHI3L1, mainly expressed in alveolar macrophages, regulate profibrotic macrophage activation, fibroblast proliferation and myofibroblast transformation, and TGF-β signaling and effector function. Although the mechanism or the pathways that CHIT1 and CHI3L1 use to regulate pulmonary fibrosis have not been fully understood yet, these studies identify CHIT1 and CHI3L1 as significant modulators of fibroproliferative responses leading to persistent and progressive pulmonary fibrosis. These studies suggest a possibility that CHIT1 and CHI3L1 could be reasonable therapeutic targets to intervene or reverse established pulmonary fibrosis. In this review, we will discuss specific roles and regulatory mechanisms of CHIT1 and CHI3L1 in profibrotic cell and tissue responses as novel therapeutic targets of pulmonary fibrosis.
Collapse
Affiliation(s)
- Suh-Young Lee
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
- Devision of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Chang-Min Lee
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
| | - Bing Ma
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
| | - Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
| | - Jack A. Elias
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
| | - Yang Zhou
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, 185 Meeting St., Providence, RI, United States
| |
Collapse
|
15
|
Inhalation of MSC-EVs is a noninvasive strategy for ameliorating acute lung injury. J Control Release 2022; 345:214-230. [DOI: 10.1016/j.jconrel.2022.03.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/21/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
|
16
|
Ma B, Kamle S, Akosman B, Khan H, Lee CM, Lee CG, Elias JA. CHI3L1 enhances melanoma lung metastasis via regulation of T cell co-stimulators and CTLA-4/B7 axis. Front Immunol 2022; 13:1056397. [PMID: 36618349 PMCID: PMC9812560 DOI: 10.3389/fimmu.2022.1056397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
ICOS/ICOSL and CD28/B7-1/B7-2 are T cell co-stimulators and CTLA-4 is an immune checkpoint inhibitor that play critical roles in the pathogenesis of neoplasia. Chitinase 3-like-1 (CHI3L1) is induced in many cancers where it portends a poor prognosis and contributes to tumor metastasis. Here we demonstrate that CHI3L1 inhibits the expression of ICOS, ICOSL and CD28 while stimulating CTLA-4 and the B7 moieties in melanoma lung metastasis. We also demonstrate that RIG-like helicase innate immune activation augments T cell co-stimulation, inhibits CTLA-4 and suppresses pulmonary metastasis. At least additive antitumor responses were seen in melanoma lung metastasis treated with anti-CTLA-4 and anti-CHI3L1 antibodies in combination. Synergistic cytotoxic T cell-induced tumor cell death and the heightened induction of the tumor suppressor PTEN were seen in co-cultures of T and tumor cells treated with bispecific antibodies that target both CHI3L1 and CTLA-4. Thus, CHI3L1 contributes to pulmonary metastasis by inhibiting T cell co-stimulation and stimulating CTLA-4. The simultaneous targeting of CHI3L1 and the CTLA-4 axis with individual and, more powerfully with bispecific antibodies, represent promising therapeutic strategies for pulmonary metastasis.
Collapse
Affiliation(s)
- Bing Ma
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Suchitra Kamle
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Bedia Akosman
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Hina Khan
- Division of Hematology-Oncology, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Chang-Min Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Jack A. Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
- Department of Medicine, Brown University, Providence, RI, United States
- *Correspondence: Jack A. Elias,
| |
Collapse
|
17
|
Huan W, Yandong L, Chao W, Sili Z, Jun B, Mingfang L, Yu C, Lefeng Q. YKL-40 Aggravates Early-Stage Atherosclerosis by Inhibiting Macrophage Apoptosis in an Aven-dependent Way. Front Cell Dev Biol 2021; 9:752773. [PMID: 34950656 PMCID: PMC8688858 DOI: 10.3389/fcell.2021.752773] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: programmed cell removal in atherosclerotic plaques plays a crucial role in retarding lesion progression. Macrophage apoptosis has a critical role in PrCR, especially in early-stage lesions. YKL-40 has been shown to be elevated as lesions develop and is closely related to macrophages. This study aimed to determine the effect of YKL-40 on regulating macrophage apoptosis and early-stage atherosclerosis progression. Research design and Methods: The correlations among the expression level of YKL-40, the area of early-stage plaque, and the macrophage apoptosis rate in plaques have been shown in human carotid atherosclerotic plaques through pathological and molecular biological detection. These results were successively confirmed in vivo (Ldlr−/- mice treated by YKL-40 recombinant protein/neutralizing antibody) and in vitro (macrophages that Ykl40 up-/down-expressed) experiments. The downstream targets were predicted by iTRAQ analysis. Results: In early-stage human carotid plaques and murine plaques, the YKL-40 expression level had a significant positive correlation with the area of the lesion and a significant negative correlation with the macrophage apoptosis rate. In vivo, the plaque area of aortic roots was significantly larger in the recomb-YKL-40 group than that in IgG group (p = 0.0247) and was significantly smaller in the anti-YKL-40 group than in the IgG group (p = 0.0067); the macrophage apoptosis rate of the plaque in aortic roots was significantly lower in the recomb-YKL-40 group than that in IgG group (p = 0.0018) and was higher in anti-YKL-40 group than that in VC group. In vitro, the activation level of caspase-9 was significantly lower in RAW264.7 with Ykl40 overexpressed than that in controls (p = 0.0054), while the expression level of Aven was significantly higher than that in controls (p = 0.0031). The apoptosis rate of RAW264.7 treated by recomb-YKL40 was significantly higher in the Aven down-regulated group than that in the control group (p < 0.001). The apoptosis inhibitor Aven was confirmed as the target molecule of YKL-40. Mechanistically, YKL-40 could inhibit macrophage apoptosis by upregulating Aven to suppress the activation of caspase-9. Conclusion: YKL-40 inhibits macrophage apoptosis by upregulating the apoptosis inhibitor Aven to suppress the activation of caspase-9, which may impede normal PrCR and promote substantial accumulation in early-stage plaques, thereby leading to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Wei Huan
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Liu Yandong
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wang Chao
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zou Sili
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Bai Jun
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Liao Mingfang
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chen Yu
- Yueyang Hospital of Integrated Traditional Chinese Medicine & Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qu Lefeng
- Department of Vascular and Endovascular Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
18
|
Kamle S, Ma B, He CH, Akosman B, Zhou Y, Lee CM, El-Deiry WS, Huntington K, Liang O, Machan JT, Kang MJ, Shin HJ, Mizoguchi E, Lee CG, Elias JA. Chitinase 3-like-1 is a therapeutic target that mediates the effects of aging in COVID-19. JCI Insight 2021; 6:e148749. [PMID: 34747367 PMCID: PMC8663553 DOI: 10.1172/jci.insight.148749] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is caused by SARS-CoV-2 (SC2) and is more prevalent and severe in elderly and patients with comorbid diseases (CM). Because chitinase 3-like-1 (CHI3L1) is induced during aging and CM, the relationships between CHI3L1 and SC2 were investigated. Here, we demonstrate that CHI3L1 is a potent stimulator of the SC2 receptor angiotensin converting enzyme 2 (ACE2) and viral spike protein priming proteases (SPP), that ACE2 and SPP are induced during aging, and that anti-CHI3L1, kasugamycin, and inhibitors of phosphorylation abrogate these ACE2- and SPP-inductive events. Human studies also demonstrate that the levels of circulating CHI3L1 are increased in the elderly and patients with CM, where they correlate with COVID-19 severity. These studies demonstrate that CHI3L1 is a potent stimulator of ACE2 and SPP, that this induction is a major mechanism contributing to the effects of aging during SC2 infection, and that CHI3L1 co-opts the CHI3L1 axis to augment SC2 infection. CHI3L1 plays a critical role in the pathogenesis of and is an attractive therapeutic target in COVID-19.
Collapse
Affiliation(s)
| | - Bing Ma
- Molecular Microbiology and Immunology
| | | | | | - Yang Zhou
- Molecular Microbiology and Immunology
| | | | - Wafik S. El-Deiry
- Pathology and Laboratory Medicine
- Hematology-Oncology Division, Department of Medicine
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
| | - Kelsey Huntington
- Pathology and Laboratory Medicine
- Hematology-Oncology Division, Department of Medicine
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
| | - Olin Liang
- Hematology-Oncology Division, Department of Medicine
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
| | - Jason T. Machan
- Department of Biostatistics, Lifespan Health System, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hyeon Jun Shin
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emiko Mizoguchi
- Molecular Microbiology and Immunology
- Department of Immunology, Kurume University, School of Medicine, Kurume, Fukuoka, Japan
| | | | - Jack A. Elias
- Molecular Microbiology and Immunology
- The Joint Program in Cancer Biology
- Cancer Center at Brown University, and
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
19
|
Wang F, Li W, Liu Z, Yu R, Wang D. LPS-induced inflammatory response and apoptosis are mediated by Fra-1 upregulation and binding to YKL-40 in A549 cells. Exp Ther Med 2021; 22:1474. [PMID: 34737814 PMCID: PMC8561753 DOI: 10.3892/etm.2021.10909] [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: 04/14/2021] [Accepted: 07/16/2021] [Indexed: 12/05/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a multifactorial syndrome that leads to increased morbidity and mortality in infants and children. The identification of novel biomarkers is critical for the treatment of ARDS. The present study aimed to investigate the effects of chitinase-3-like-1 protein (CHI3L1 or YKL-40) in an in vitro model of ARDS and to explore the potential underlying mechanisms. The in vitro model of ARDS was established in A549 alveolar epithelial type II cells, which were treated by lipopolysaccharide (LPS) to induce inflammation. Transfection was performed to alter YKL-40 expression. The mRNA and protein expression of YKL-40 was determined using reverse transcription-quantitative PCR and western blotting, respectively. Cell Counting Kit-8 and TUNEL assays were used to evaluate the cell viability and apoptosis, respectively. The production of cytokines was evaluated using specific ELISA kits. The relationship between YKL-40 and Fos-related antigen 1 (Fra-1) was verified using luciferase reporter and chromatin immunoprecipitation assays. The expression of the apoptotic proteins was detected using western blotting. The expression levels of YKL-40 and Fra-1 were increased in LPS-treated A549 cells. Higher levels of pro-inflammatory cytokines and induction of cell apoptosis were observed in LPS-treated A549 cells compared with the control. YKL-40 knockdown in LPS-treated A549 cells significantly decreased the production of pro-inflammatory cytokines and reduced cell apoptosis, whereas it concomitantly caused upregulation of Bax and downregulation of Bcl-2, cleaved caspase-3 and cleaved caspase-9. In addition, Fra-1 could directly bind to YKL-40 promoter and regulate its expression level. Overexpression of YKL-40 partly decreased the inhibitory effects of Fra-1 knockdown on the inflammatory response and induction of apoptosis. In summary, the findings from the present study indicated that Fra-1 could bind to YKL-40 and regulate its expression, whereas YKL-40 knockdown could further suppress LPS-induced inflammatory response and apoptosis in A549 cells. These data may provide novel evidence on the diagnosis and therapy of ARDS.
Collapse
Affiliation(s)
- Fei Wang
- Department of Pediatrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Wenxuan Li
- Department of Pediatrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Zhen Liu
- Department of Pediatrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Ronghua Yu
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200062, P.R. China
| | - Dalian Wang
- Department of Pediatrics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
20
|
Ma B, Akosman B, Kamle S, Lee CM, He CH, Koo JS, Lee CG, Elias JA. CHI3L1 regulates PD-L1 and anti-CHI3L1-PD-1 antibody elicits synergistic antitumor responses. J Clin Invest 2021; 131:137750. [PMID: 34720089 DOI: 10.1172/jci137750] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Evasion of the immune response is a hallmark of cancer, and programmed cell death 1 (PD-1) and PD-1 ligand 1 (PD-L1) are major mediators of this immunosuppression. Chitinase 3-like 1 (CHI3L1) is induced in many cancers, where it portends a poor prognosis and contributes to tumor metastasis and spread. However, the mechanism(s) that CHI3L1 uses in metastasis have not been defined. Here we demonstrate that CHI3L1 regulates the expression of PD-L1, PD-L2, PD-1, LAG3, and TIM3 and plays a critical role in melanoma progression and lymphatic spread. CHI3L1 also contributed to IFN-γ-stimulated macrophage PD-L1 expression, and RIG-like helicase innate immunity suppressed CHI3L1, PD-L1, and melanoma progression. Individual antibodies against CHI3L1 or PD-1 had discrete antitumor effects and additive antitumor responses in metastasis models and T cell-tumor cell cocultures when administered simultaneously. Synergistic cytotoxic tumor cell death was seen in T cell-tumor cell cocultures, and significantly enhanced antitumor responses were seen in in vivo tumor models treated with bispecific antibodies that simultaneously target CHI3L1 and PD-1. CHI3L1 contributes to tumor progression by stimulating the PD-1/PD-L1 axis and other checkpoint molecules. The simultaneous targeting of CHI3L1 and the PD-1/PD-L1 axis with individual and, more powerfully, with bispecific antibodies represents a promising therapy for pulmonary metastasis and progression.
Collapse
Affiliation(s)
- Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Bedia Akosman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Suchitra Kamle
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chuan Hua He
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Ja Seok Koo
- Section of Medical Oncology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA.,Department of Medicine, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
21
|
Hong JY, Kim MN, Kim EG, Lee JW, Kim HR, Kim SY, Lee SM, Kim YH, Kim KW, Sohn MH. Clusterin Deficiency Exacerbates Hyperoxia-Induced Acute Lung Injury. Cells 2021; 10:944. [PMID: 33921872 PMCID: PMC8073575 DOI: 10.3390/cells10040944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/07/2021] [Accepted: 04/16/2021] [Indexed: 02/05/2023] Open
Abstract
Exposure to high oxygen concentrations leads to generation of excessive reactive oxygen species, causing cellular injury and multiple organ dysfunctions and is associated with a high mortality rate. Clusterin (CLU) is a heterodimeric glycoprotein that mediates several intracellular signaling pathways, including cell death and inflammation. However, the role of CLU in the pathogenesis of hyperoxic acute lung injury (HALI) is unknown. Wild-type (WT) and CLU-deficient mice and cultured human airway epithelial cells were used. Changes in cell death- and inflammation-related molecules with or without hyperoxia exposure in cells and animals were determined. Hyperoxia induced an increase in CLU expression in mouse lungs and human airway epithelial cells. Mice lacking CLU had increased HALI and mortality rate compared with WT mice. In vitro, CLU-disrupted cells showed enhanced release of cytochrome c, Bax translocation, cell death and inflammatory cytokine expression. However, treatment with recombinant CLU attenuated hyperoxia-induced apoptosis. Moreover, the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses revealed metabolic pathways, hematopoietic cell lineage, response to stress and localization and regulation of immune system that were differentially regulated between WT and CLU-/- mice. These results demonstrate that prolonged hyperoxia-induced lung injury is associated with CLU expression and that CLU replenishment may alleviate hyperoxia-induced cell death.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine,134 Sinchon-Dong, Seoul 03722, Korea; (J.Y.H.); (M.N.K.); (E.G.K.); (J.W.L.); (H.R.K.); (S.Y.K.); (S.M.L.); (Y.H.K.); (K.W.K.)
| |
Collapse
|
22
|
Mohanty AK, Choudhary S, Kaushik JK, Fisher AJ. Crystal structure of breast regression protein 39 (BRP39), a signaling glycoprotein expressed during mammary gland apoptosis, at 2.6 Å resolution. J Struct Biol 2021; 213:107737. [PMID: 33838225 DOI: 10.1016/j.jsb.2021.107737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 11/16/2022]
Abstract
Breast regression protein 39 (BRP39) is a 39 kDa protein that is a member of chitolectin class of glycosyl hydrolase family 18 (GH18). High expression levels of BRP39 have been detected in breast carcinoma. It helps in proliferation of cells during the progression of this disease and may act as a signaling factor. BRP39 may act as a potential candidate for rational structure-based drug design against breast carcinoma. In this study, we report the crystal structure of mouse recombinant BRP39 expressed in E. coli. The structure was solved by molecular replacement and refined to 2.6 Å resolution. The overall structure of BRP39 consisted of two globular domains: a large (β/α)8 triosephosphate isomerase (TIM) barrel domain and a small (α + β) domain. Three non-proline cis-peptides were detected in the sugar-binding cleft of BRP39, including Ser57-Phe58, Leu141-Tyr142, and Trp353-Ala354. The latter residues were conserved in other GH18 family members. It was notable that the conformation of critical Trp100 residue within the sugar-binding cleft was oriented away from the barrel. The side-chain conformation was found to be similar to that observed in chitinases, however, it was oriented into the barrel in other chitinase-like proteins (CLPs). The conformation of this critical residue may have significant implications in sugar binding. Further, two amino acid substitutions were observed in the sugar-binding groove of BRP39. The conserved Asn100 and Arg263 in Hcgp39 and other CLPs proteins (SPX-40 structures) were substituted by Lys101 and Lys264 in BRP39 which may have a significant impact on the sugar-binding properties.
Collapse
Affiliation(s)
- Ashok K Mohanty
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal 132001, Haryana, India.
| | - Suman Choudhary
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Jai K Kaushik
- Animal Biotechnology Centre, National Dairy Research Institute, Karnal 132001, Haryana, India
| | - Andrew J Fisher
- Department of Chemistry, University of California Davis, Davis, CA 95616, United States; Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, United States
| |
Collapse
|
23
|
Velázquez-Díaz P, Nakajima E, Sorkhdini P, Hernandez-Gutierrez A, Eberle A, Yang D, Zhou Y. Hermansky-Pudlak Syndrome and Lung Disease: Pathogenesis and Therapeutics. Front Pharmacol 2021; 12:644671. [PMID: 33841163 PMCID: PMC8028140 DOI: 10.3389/fphar.2021.644671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/11/2021] [Indexed: 12/19/2022] Open
Abstract
Hermansky-Pudlak Syndrome (HPS) is a rare, genetic, multisystem disorder characterized by oculocutaneous albinism (OCA), bleeding diathesis, immunodeficiency, granulomatous colitis, and pulmonary fibrosis. HPS pulmonary fibrosis (HPS-PF) occurs in 100% of patients with subtype HPS-1 and has a similar presentation to idiopathic pulmonary fibrosis. Upon onset, individuals with HPS-PF have approximately 3 years before experiencing signs of respiratory failure and eventual death. This review aims to summarize current research on HPS along with its associated pulmonary fibrosis and its implications for the development of novel treatments. We will discuss the genetic basis of the disease, its epidemiology, and current therapeutic and clinical management strategies. We continue to review the cellular processes leading to the development of HPS-PF in alveolar epithelial cells, lymphocytes, mast cells, and fibrocytes, along with the molecular mechanisms that contribute to its pathogenesis and may be targeted in the treatment of HPS-PF. Finally, we will discuss emerging new cellular and molecular approaches for studying HPS, including lentiviral-mediated gene transfer, induced pluripotent stem cells (iPSCs), organoid and 3D-modelling, and CRISPR/Cas9-based gene editing approaches.
Collapse
Affiliation(s)
| | - Erika Nakajima
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Parand Sorkhdini
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | | | - Adam Eberle
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Dongqin Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States
| |
Collapse
|
24
|
Amarelle L, Quintela L, Hurtado J, Malacrida L. Hyperoxia and Lungs: What We Have Learned From Animal Models. Front Med (Lausanne) 2021; 8:606678. [PMID: 33768102 PMCID: PMC7985075 DOI: 10.3389/fmed.2021.606678] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Although oxygen (O2) is essential for aerobic life, it can also be an important source of cellular damage. Supra-physiological levels of O2 determine toxicity due to exacerbated reactive oxygen species (ROS) production, impairing the homeostatic balance of several cellular processes. Furthermore, injured cells activate inflammation cascades, amplifying the tissue damage. The lung is the first (but not the only) organ affected by this condition. Critically ill patients are often exposed to several insults, such as mechanical ventilation, infections, hypo-perfusion, systemic inflammation, and drug toxicity. In this scenario, it is not easy to dissect the effect of oxygen toxicity. Translational investigations with animal models are essential to explore injuring stimuli in controlled experimental conditions, and are milestones in understanding pathological mechanisms and developing therapeutic strategies. Animal models can resemble what happens in critical care or anesthesia patients under mechanical ventilation and hyperoxia, but are also critical to explore the effect of O2 on lung development and the role of hyperoxic damage on bronchopulmonary dysplasia. Here, we set out to review the hyperoxia effects on lung pathology, contributing to the field by describing and analyzing animal experimentation's main aspects and its implications on human lung diseases.
Collapse
Affiliation(s)
- Luciano Amarelle
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Lucía Quintela
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Javier Hurtado
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Leonel Malacrida
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay.,Advanced Bioimaging Unit, Institut Pasteur Montevideo and Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
25
|
Kamle S, Ma B, He CH, Akosman B, Zhou Y, Lee CM, El-Deiry WS, Huntington K, Liang O, Machan JT, Kang MJ, Shin HJ, Mizoguchi E, Lee CG, Elias JA. Chitinase 3-like-1 is a Therapeutic Target That Mediates the Effects of Aging in COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.01.05.425478. [PMID: 33442679 PMCID: PMC7805436 DOI: 10.1101/2021.01.05.425478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
COVID-19 is caused by the SARS-CoV-2 (SC2) virus and is more prevalent and severe in the elderly and patients with comorbid diseases (CM). Because chitinase 3-like-1 (CHI3L1) is induced during aging and CM, the relationships between CHI3L1 and SC2 were investigated. Here we demonstrate that CHI3L1 is a potent stimulator of the SC2 receptor ACE2 and viral spike protein priming proteases (SPP), that ACE2 and SPP are induced during aging and that anti-CHI3L1, kasugamycin and inhibitors of phosphorylation, abrogate these ACE2- and SPP- inductive events. Human studies also demonstrated that the levels of circulating CHI3L1 are increased in the elderly and patients with CM where they correlate with COVID-19 severity. These studies demonstrate that CHI3L1 is a potent stimulator of ACE2 and SPP; that this induction is a major mechanism contributing to the effects of aging during SC2 infection and that CHI3L1 coopts the CHI3L1 axis to augment SC2 infection. CHI3L1 plays a critical role in the pathogenesis of and is an attractive therapeutic target in COVID-19.
Collapse
|
26
|
Abstract
Despite advancements in standardizing the criteria for acute kidney injury (AKI), its definition remains based on changes in serum creatinine and urinary output that do not specifically represent tubular function or injury and that have significant limitations in the acute hospital setting. Much effort in nephrology has centered on identifying biomarkers of AKI to address these limitations. This review summarizes recent advances in our knowledge of biomarkers involved in pathophysiological processes during AKI and describes their potential clinical implications. Blood and urine biomarkers are released via various mechanisms during renal tubular injury. Urinary kidney injury molecule-1 (KIM-1), liver-type fatty acid binding protein (L-FABP), insulin-like growth factor-binding protein-7 (IGFBP-7), and tissue inhibitor of metalloprotease-2 (TIMP-2) are released from the proximal tubule while uromodulin (UMOD) is secreted from the loop of Henle and neutrophil gelatinase-associated lipocalin (NGAL) is released from the distal tubule. These biomarkers could therefore be used to localize specific segments of injured tubules. Biomarkers also have diverse roles in pathophysiological processes in AKI, including inflammation, repair, and fibrosis. Current evidence suggests that these biomarkers could be used to predict the transition to chronic kidney disease (CKD), decrease discard of AKI kidneys, differentiate between kidney dysfunction and injury, guide AKI management, and improve diagnosis of diseases such as acute interstitial nephritis (AIN). They could differentiate between disease phenotypes, facilitate the inclusion of a homogenous patient population in future trials of AKI, and shed light on therapeutic pathways to prevent the transition from AKI to CKD. However, a major limitation of current biomarker research in AKI is the lack of tissue correlation. The Kidney Precision Medicine Project, a large-scale national effort, is currently underway to construct a kidney tissue atlas and expand the use of biomarkers to assess nephron health. Numerous biomarkers are involved in distinct pathophysiological processes after kidney injury and have demonstrated potential to improve diagnosis and risk stratification as well as provide a prognosis for patients with AKI. Some biomarkers are ready for use in clinical trials of AKI and could guide management in various clinical settings. Further investigation of these biomarkers will provide insight that can be applied to develop novel therapeutic agents for AKI.
Collapse
Affiliation(s)
- Yumeng Wen
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chirag R Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Mu G, Deng Y, Lu Z, Li X, Chen Y. miR-20b suppresses mitochondrial dysfunction-mediated apoptosis to alleviate hyperoxia-induced acute lung injury by directly targeting MFN1 and MFN2. Acta Biochim Biophys Sin (Shanghai) 2021; 53:220-228. [PMID: 33347533 DOI: 10.1093/abbs/gmaa161] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Indexed: 12/21/2022] Open
Abstract
Supplemental oxygen is commonly used to treat severe respiratory failure, while prolonged exposure to hyperoxia can induce acute lung injury characterized by the accumulation of reactive oxygen species (ROS) and pulmonary inflammation. Dysregulation of microRNAs contributes to multiple diseases, including hyperoxia-induced acute lung injury (HALI). In this study, we explored the roles of miR-20b in mediating the response of type II alveolar epithelial cells (ACE IIs) to hyperoxia and the potential underlying mechanisms. We found that miR-20b was significantly decreased in the lung tissues of HALI models and H2O2-treated ACE IIs. Hyperoxia induced the release of TNF-α, decreased the mitochondrial membrane potential, and led to excessive ROS production and cell apoptosis. Overexpression of miR-20b suppressed the hyperoxia-induced biological effects in ACE IIs. miR-20b negatively regulated the expression levels of Mitofusin 1 (MFN1) and MFN2, the two key proteins of mitochondrial fusion, via complementarily binding to the 3'-untranslated regions of mRNAs. Furthermore, both in vivo and in vitro, upregulation of MFN1 and MFN2 aggravated lung damage and cell apoptosis that were alleviated by miR-20b overexpression. These results provided new insights into the involvement of the miR-20b/MFN1/2 signaling pathway in HALI.
Collapse
Affiliation(s)
- Genhua Mu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
- Department of Intensive Care Unit, The First People’s Hospital of Yancheng, Yancheng 224005, China
| | - Yijun Deng
- Department of Intensive Care Unit, The First People’s Hospital of Yancheng, Yancheng 224005, China
| | - Zhongqian Lu
- Department of Intensive Care Unit, The First People’s Hospital of Yancheng, Yancheng 224005, China
| | - Xing Li
- Department of Intensive Care Unit, The First People’s Hospital of Yancheng, Yancheng 224005, China
| | - Yanbin Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215000, China
| |
Collapse
|
28
|
Mackel JJ, Garth JM, Jones M, Ellis DA, Blackburn JP, Yu Z, Matalon S, Curtiss M, Lund FE, Hastie AT, Meyers DA, Steele C. Chitinase 3-like-1 protects airway function despite promoting type 2 inflammation during fungal-associated allergic airway inflammation. Am J Physiol Lung Cell Mol Physiol 2021; 320:L615-L626. [PMID: 33533316 DOI: 10.1152/ajplung.00528.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Joseph J Mackel
- Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama
| | - Jaleesa M Garth
- Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama
| | - MaryJane Jones
- Department of Microbiology and Immunology, Tulane University, New Orleans, Louisiana
| | - Diandra A Ellis
- Department of Microbiology and Immunology, Tulane University, New Orleans, Louisiana
| | | | - Zhihong Yu
- Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama
| | - Sadis Matalon
- Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama
| | - Miranda Curtiss
- Department of Medicine, University of Alabama Birmingham, Birmingham, Alabama.,Department of Microbiology, University of Alabama Birmingham, Birmingham, Alabama
| | - Frances E Lund
- Department of Microbiology, University of Alabama Birmingham, Birmingham, Alabama
| | - Annette T Hastie
- Department of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | | | - Chad Steele
- Department of Microbiology and Immunology, Tulane University, New Orleans, Louisiana
| |
Collapse
|
29
|
A Novel Association between YKL-40, a Marker of Structural Lung Disease, and Short Telomere Length in 10-Year-Old Children with Bronchopulmonary Dysplasia. CHILDREN-BASEL 2021; 8:children8020080. [PMID: 33498968 PMCID: PMC7912154 DOI: 10.3390/children8020080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
Extremely preterm infants are born with immature lungs and are exposed to an inflammatory environment as a result of oxidative stress. This may lead to airway remodeling, cellular aging and the development of bronchopulmonary dysplasia (BPD). Reliable markers that predict the long-term consequences of BPD in infancy are still lacking. We analyzed two biomarkers of cellular aging and lung function, telomere length and YKL-40, respectively, at 10 years of age in children born preterm with a history of BPD (n = 29). For comparison, these markers were also evaluated in sex-and-age-matched children born at term with childhood asthma (n = 28). Relative telomere length (RTL) was measured in whole blood with qPCR and serum YKL-40 with ELISA, and both were studied in relation to gas exchange and the regional ventilation/perfusion ratio using three-dimensional V/Q-scintigraphy (single photon emission computer tomography, SPECT) in children with BPD. Higher levels of YKL-40 were associated with shorter leukocyte RTL (Pearson's correlation: -0.55, p = 0.002), but were not associated with a lower degree of matching between ventilation and perfusion within the lung. Serum YKL-40 levels were significantly higher in children with BPD compared to children with asthma (17.7 vs. 13.2 ng/mL, p < 0.01). High levels of YKL-40 and short RTLs were associated to the need for ventilatory support more than 1 month in the neonatal period (p < 0.01). The link between enhanced telomere shortening in childhood and structural remodeling of the lung, as observed in children with former BPD but not in children with asthma at the age of 10 years, suggests altered lung development related to prematurity and early life inflammatory exposure. In conclusion, relative telomere length and YKL-40 may serve as biomarkers of altered lung development as a result of early-life inflammation in children with a history of prematurity.
Collapse
|
30
|
Abdelwahab SH, Reidel B, Martin JR, Ghosh A, Keating JE, Haridass P, Carpenter J, Glish GL, Tarran R, Doerschuk CM, Kesimer M. Cigarillos Compromise the Mucosal Barrier and Protein Expression in Airway Epithelia. Am J Respir Cell Mol Biol 2020; 63:767-779. [PMID: 32877614 PMCID: PMC7790145 DOI: 10.1165/rcmb.2019-0085oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 09/02/2020] [Indexed: 12/22/2022] Open
Abstract
Smoking remains a leading cause of preventable morbidity and mortality worldwide. Despite a downward trend in cigarette use, less-regulated tobacco products, such as cigarillos, which are often flavored to appeal to specific demographics, such as younger people, are becoming increasingly popular. Cigar/cigarillo smoking has been considered a safer alternative to cigarettes; however, the health risks associated with cigar in comparison with cigarette smoking are not well understood. To address this knowledge gap, we characterized the effects of multiple brands of cigarillos on the airway epithelium using ex vivo and in vivo models. To analyze these effects, we assessed the cellular viability and integrity of smoke-exposed primary airway cell cultures. We also investigated the protein compositions of apical secretions from cigarillo-exposed airway epithelial cultures and BAL fluid of cigarillo-exposed mice through label-free quantitative proteomics and determined the chemical composition of smoke collected from the investigated cigarillo products. We found that cigarillo smoke exerts similar or greater effects than cigarette smoke in terms of reduced cell viability; altered protein levels, including those of innate immune proteins; induced oxidative-stress markers; and greater nicotine delivery to cells. The analysis of the chemical composition of the investigated cigarillo products revealed differences that might be linked to the differential effects of these products on cell viability and protein abundance profiles, which have been associated with a range of health risks in the context of airway biology. These findings contradict the assumption that cigarillos might be safer and less harmful than cigarettes. Instead, our results indicate that cigarillo smoke is associated with equal or greater health risks and the same or increased airway toxicity compared with cigarette smoke.
Collapse
Affiliation(s)
| | - Boris Reidel
- Department of Pathology and Laboratory Medicine
- Marsico Lung Institute
| | | | | | | | | | - Jerome Carpenter
- Department of Pathology and Laboratory Medicine
- Marsico Lung Institute
| | | | - Robert Tarran
- Marsico Lung Institute
- Department of Cell Biology and Physiology, and
| | - Claire M. Doerschuk
- Department of Pathology and Laboratory Medicine
- Marsico Lung Institute
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine
- Marsico Lung Institute
| |
Collapse
|
31
|
He CH, Lee CG, Ma B, Kamle S, Choi AMK, Elias JA. N-Glycosylation Regulates Chitinase 3-like-1 and IL-13 Ligand Binding to IL-13 Receptor α2. Am J Respir Cell Mol Biol 2020; 63:386-395. [PMID: 32402213 DOI: 10.1165/rcmb.2019-0446oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chitinase 3-like-1 (Chi3l1) and IL-13 are both ligands of IL-13 receptor α2 (IL-13Rα2). The binding of the former activates mitogen-activated protein kinase, AKT, and Wnt/β-catenin signaling, and plays important roles in innate and adaptive immunity, cellular apoptosis, oxidative injury, allergic inflammation, tumor metastasis and wound healing, fibrosis, and repair in the lung. In contrast, the latter binding is largely a decoy event that diminishes the effects of IL-13. Here, we demonstrate that IL-13Rα2 N-glycosylation is a critical determinant of which ligand binds. Structure-function evaluations demonstrated that Chi3l1-IL-13Rα2 binding was increased when sites of N-glycosylation are mutated, and studies with tunicamycin and Peptide:N-glycosidase F (PNGase F) demonstrated that Chi3l1-IL-13Rα2 binding and signaling were increased when N-glycosylation was diminished. In contrast, structure-function experiments demonstrated that IL-13 binding to IL-13Rα2 was dependent on each of the four sites of N-glycosylation in IL-13Rα2, and experiments with tunicamycin and PNGase F demonstrated that IL-13-IL-13Rα2 binding was decreased when IL-13Rα2 N-glycosylation was diminished. Studies with primary lung epithelial cells also demonstrated that Chi3l1 inhibited, whereas IL-13 stimulated, N-glycosylation as evidenced by the ability of Chi3l1 to inhibit and IL-13 to stimulate the subunits of the oligosaccharide complex A and B (STT3A and STT3B). These studies demonstrate that N-glycosylation is a critical determinant of Chi3l1 and IL-13 binding to IL-13Rα2, and highlight the ability of Chi3l1 and IL-13 to alter key elements of the N-glycosylation apparatus in a manner that would augment their respective binding.
Collapse
Affiliation(s)
- Chuan Hua He
- Department of Molecular Microbiology and Immunology and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology and
| | - Bing Ma
- Department of Molecular Microbiology and Immunology and
| | | | - Augustine M K Choi
- Department of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology and.,Department of Medicine, Brown University, Providence, Rhode Island; and
| |
Collapse
|
32
|
Chitinase-3 like-protein-1 function and its role in diseases. Signal Transduct Target Ther 2020; 5:201. [PMID: 32929074 PMCID: PMC7490424 DOI: 10.1038/s41392-020-00303-7] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/28/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
Non-enzymatic chitinase-3 like-protein-1 (CHI3L1) belongs to glycoside hydrolase family 18. It binds to chitin, heparin, and hyaluronic acid, and is regulated by extracellular matrix changes, cytokines, growth factors, drugs, and stress. CHI3L1 is synthesized and secreted by a multitude of cells including macrophages, neutrophils, synoviocytes, chondrocytes, fibroblast-like cells, smooth muscle cells, and tumor cells. It plays a major role in tissue injury, inflammation, tissue repair, and remodeling responses. CHI3L1 has been strongly associated with diseases including asthma, arthritis, sepsis, diabetes, liver fibrosis, and coronary artery disease. Moreover, following its initial identification in the culture supernatant of the MG63 osteosarcoma cell line, CHI3L1 has been shown to be overexpressed in a wealth of both human cancers and animal tumor models. To date, interleukin-13 receptor subunit alpha-2, transmembrane protein 219, galectin-3, chemo-attractant receptor-homologous 2, and CD44 have been identified as CHI3L1 receptors. CHI3L1 signaling plays a critical role in cancer cell growth, proliferation, invasion, metastasis, angiogenesis, activation of tumor-associated macrophages, and Th2 polarization of CD4+ T cells. Interestingly, CHI3L1-based targeted therapy has been increasingly applied to the treatment of tumors including glioma and colon cancer as well as rheumatoid arthritis. This review summarizes the potential roles and mechanisms of CHI3L1 in oncogenesis and disease pathogenesis, then posits investigational strategies for targeted therapies.
Collapse
|
33
|
Liu C, Mor MK, Palevsky PM, Kaufman JS, Thiessen Philbrook H, Weisbord SD, Parikh CR. Postangiography Increases in Serum Creatinine and Biomarkers of Injury and Repair. Clin J Am Soc Nephrol 2020; 15:1240-1250. [PMID: 32839195 PMCID: PMC7480551 DOI: 10.2215/cjn.15931219] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 07/02/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND OBJECTIVES It is unknown whether iodinated contrast causes kidney parenchymal damage. Biomarkers that are more specific to nephron injury than serum creatinine may provide insight into whether contrast-associated AKI reflects tubular damage. We assessed the association between biomarker changes after contrast angiography with contrast-associated AKI and 90-day major adverse kidney events and death. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We conducted a longitudinal analysis of participants from the biomarker substudy of the Prevention of Serious Adverse Events following Angiography trial. We measured injury (kidney injury molecule-1, neutrophil gelatinase-associated lipocalin, IL-18) and repair (monocyte chemoattractant protein-1, uromodulin, YKL-40) proteins from plasma and urine samples at baseline and 2-4 hours postangiography. We assessed the associations between absolute changes and relative ratios of biomarkers with contrast-associated AKI and 90-day major adverse kidney events and death. RESULTS Participants (n=922) were predominately men (97%) with diabetes (82%). Mean age was 70±8 years, and eGFR was 48±13 ml/min per 1.73 m2; 73 (8%) and 60 (7%) participants experienced contrast-associated AKI and 90-day major adverse kidney events and death, respectively. No postangiography urine biomarkers were associated with contrast-associated AKI. Postangiography plasma kidney injury molecule-1 and IL-18 were significantly higher in participants with contrast-associated AKI compared with those who did not develop contrast-associated AKI: 428 (248, 745) versus 306 (179, 567) mg/dl; P=0.04 and 325 (247, 422) versus 280 (212, 366) mg/dl; P=0.009, respectively. The majority of patients did not experience an increase in urine or plasma biomarkers. Absolute changes in plasma IL-18 were comparable in participants with contrast-associated AKI (-30 [-71, -9] mg/dl) and those without contrast-associated AKI (-27 [-53, -10] mg/dl; P=0.62). Relative ratios of plasma IL-18 were also comparable in participants with contrast-associated AKI (0.91; 0.86, 0.97) and those without contrast-associated AKI (0.91; 0.85, 0.96; P=0.54). CONCLUSIONS The lack of significant differences in the absolute changes and relative ratios of injury and repair biomarkers by contrast-associated AKI status suggests that the majority of mild contrast-associated AKI cases may be driven by hemodynamic changes at the kidney.
Collapse
Affiliation(s)
- Caroline Liu
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria K Mor
- Center for Health Equity Research and Promotion, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania .,Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Paul M Palevsky
- Renal Section, Medical Service, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James S Kaufman
- Division of Nephrology, Veterans Affairs New York Harbor Healthcare System and New York University School of Medicine, New York, New York
| | | | - Steven D Weisbord
- Renal Section, Medical Service, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.,Renal Section, Medical Service and Center for Health Equity Research and Promotion, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Chirag R Parikh
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
34
|
Abstract
The current unidimensional paradigm of kidney disease detection is incompatible with the complexity and heterogeneity of renal pathology. The diagnosis of kidney disease has largely focused on glomerular filtration, while assessment of kidney tubular health has notably been absent. Following insult, the kidney tubular cells undergo a cascade of cellular responses that result in the production and accumulation of low-molecular-weight proteins in the urine and systemic circulation. Modern advancements in molecular analysis and proteomics have allowed the identification and quantification of these proteins as biomarkers for assessing and characterizing kidney diseases. In this review, we highlight promising biomarkers of kidney tubular health that have strong underpinnings in the pathophysiology of kidney disease. These biomarkers have been applied to various specific clinical settings from the spectrum of acute to chronic kidney diseases, demonstrating the potential to improve patient care.
Collapse
Affiliation(s)
- William R Zhang
- Kidney Health Research Collaborative, University of California San Francisco School of Medicine, San Francisco, California 94121, USA
| | - Chirag R Parikh
- Division of Nephrology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, USA;
| |
Collapse
|
35
|
Zhou Y, Meng LJ, Wang J. [Changes in serum human cartilage glycoprotein-39 and high-mobility group box 1 in preterm infants with bronchopulmonary dysplasia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:334-338. [PMID: 32312371 PMCID: PMC7389695 DOI: 10.7499/j.issn.1008-8830.2001041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/03/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To study the association of the dynamic changes of peripheral blood human cartilage glycoprotein-39 (YKL-40) and high-mobility group box 1 (HMGB1) with bronchopulmonary dysplasia (BPD) in preterm infants. METHODS Preterm infants, with a gestational age of 28-32 weeks and a birth weight of <1 500 g, who were admitted to the neonatal intensive care unit from July 2017 to August 2019 were prospectively selected and divided into a BPD group with 35 infants and a non-BPD group with 51 infants. ELISA was used to measure the serum concentrations of YKL-40 and HMGB1 in preterm infants on days 3, 7, and 14 after birth. RESULTS The BPD group had a significantly lower serum YKL-40 concentration and a significantly higher serum HMGB1 concentration than the non-BPD group on days 3, 7, and 14 (P<0.001). The serum concentrations of YKL-40 and HMGB1 on days 7 and 14 were significantly higher than those on day 3 in both groups (P<0.017). In the BPD group, HMGB1 concentration on day 14 was significantly higher than that on day 7 (P<0.017), while there was no significant change in YKL-40 concentration from day 7 to day 14 (P>0.017). In the non-BPD group, YKL-40 concentration on day 14 was significantly higher than that on day 7 (P<0.017), while there was no significant change in HMGB1 concentration from day 7 to day 14 (P>0.017). CONCLUSIONS There are significant differences in the levels of YKL-40 and HMGB1 in peripheral blood between the preterm infants with BPD and those without BPD on days 3, 7, and 14 after birth, suggesting that YKL-40 and HMGB1 might be associated with the development of BPD.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Neonatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | | | | |
Collapse
|
36
|
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
|
37
|
Kwak EJ, Hong JY, Kim MN, Kim SY, Kim SH, Park CO, Kim KW, Lee CG, Elias JA, Jee HM, Sohn MH. Chitinase 3-like 1 drives allergic skin inflammation via Th2 immunity and M2 macrophage activation. Clin Exp Allergy 2019; 49:1464-1474. [PMID: 31397016 DOI: 10.1111/cea.13478] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/15/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterized by defective skin barrier and Th2 immune responses. Chitinase 3-like 1 (CHI3L1), also known as breast regression protein 39 (BRP-39) in mice and human homologue YKL-40, plays important roles in Th2 inflammation and allergen sensitization. CHI3L1 has been implicated in a variety of diseases including asthma characterized by inflammation, apoptosis and tissue remodelling, but its role in AD remains elusive. OBJECTIVE The aim of this study was to investigate the role of CHI3L1 in the development and progression of AD. RESULTS We investigated YKL-40 levels in the serum and skin of AD patients by ELISA and immunofluorescence, respectively. Using a murine model of AD induced by ovalbumin (OVA), we investigated Th2 immune responses, M2 macrophage activation and skin barrier gene expression using wild-type (WT) and BRP-39 null mutant (BRP-39-/- ) mice. YKL-40 level was significantly increased in serum of AD patients. In addition, both mRNA and protein expression levels of BRP-39 were higher in OVA-sensitized WT mice than in control mice. OVA-sensitized BRP-39-/- mice showed decreased epidermal thickness, lower total serum IgE, Th2 cytokine levels and CD4+ effector T cell populations than OVA-sensitized WT mice. Induction of BRP-39 was dominant in dermal macrophages. BRP-39 deficiency was found to be involved in M2 macrophage activation. Consistently, the YKL-40 level in the skin of AD patients was higher than in normal subjects and it was expressed in dermal macrophages. BRP-39 deficiency attenuated dysregulation of skin barrier and tight junction genes. CONCLUSIONS AND CLINICAL RELEVANCE These findings demonstrate that CHI3L1 mediates the development of AD induced by OVA, affecting Th2 inflammation, M2 macrophage activation and skin barrier function.
Collapse
Affiliation(s)
- Eun Ji Kwak
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Yeon Hong
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Na Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Yeon Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seo Hyeong Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Ook Park
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Chun Geun Lee
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA.,Department of Internal Medicine, Hanyang University, Seoul, Korea
| | - Jack A Elias
- Molecular Microbiology and Immunology, Brown University, Providence, RI, USA
| | - Hye Mi Jee
- Department of Pediatrics, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Korea
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
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
|
39
|
Kim MJ, Shim DH, Cha H, Moon K, Yang CM, Hwang SJ, Kim KW, Park JH, Lee C, Elias JA, Sohn MH, Lee JM. Chitinase 3-like 1 protein plays a critical role in respiratory syncytial virus-induced airway inflammation. Allergy 2019; 74:685-697. [PMID: 30402955 PMCID: PMC7159489 DOI: 10.1111/all.13661] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 09/09/2018] [Accepted: 09/19/2018] [Indexed: 12/28/2022]
Abstract
Background Chitinase 3‐like 1 protein (CHI3L1) (YKL‐40 in humans and breast regression protein [BRP]‐39 in mice) is required for optimal allergen sensitization and Th2 inflammation in various chronic inflammatory diseases including asthma. However, the role of CHI3L1 in airway inflammation induced by respiratory viruses has not been investigated. The aim of this study was to investigate the relationship between CHI3L1 and airway inflammation caused by respiratory syncytial virus (RSV) infection. Methods We measured YKL‐40 levels in human nasopharyngeal aspirate (NPA) from hospitalized children presenting with acute respiratory symptoms. Wild‐type (WT) and BRP‐39 knockout (KO) C57BL/6 mice were inoculated with live RSV (A2 strain). Bronchoalveolar lavage fluid and lung tissue samples were obtained on day 7 after inoculation to assess lung inflammation, airway reactivity, and expression of cytokines and BRP‐39. Results In human subjects, YKL‐40 and IL‐13 levels in NPA were higher in children with RSV infection than in control subjects. Expression of BRP‐39 and Th2 cytokines, IL‐13 in particular, was increased following RSV infection in mice. Airway inflammation caused by RSV infection was reduced in BRP‐39 KO mice as compared to WT mice. Th2 cytokine levels were not increased in the lungs of RSV‐infected BRP‐39 KO mice. BRP‐39 regulated M2 macrophage activation in RSV‐infected mice. Additionally, treatment with anti‐CHI3L1 antibody attenuated airway inflammation and Th2 cytokine production in RSV‐infected WT mice. Conclusion These findings suggest that CHI3L1 could contribute to airway inflammation induced by RSV infection. CHI3L1 could be a potential therapeutic candidate for attenuating Th2‐associated immunopathology during RSV infection.
Collapse
Affiliation(s)
- Min Jung Kim
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
- Department of Pediatrics Severance Hospital Institute of Allergy Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Doo Hee Shim
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Hye‐Ran Cha
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Kuk‐Young Moon
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Chang Mo Yang
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Su Jin Hwang
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Kyung Won Kim
- Department of Pediatrics Severance Hospital Institute of Allergy Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Jeon Han Park
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology Brown University Providence Rhode Island USA
| | - Jack A. Elias
- Department of Molecular Microbiology and Immunology Brown University Providence Rhode Island USA
| | - Myung Hyun Sohn
- Department of Pediatrics Severance Hospital Institute of Allergy Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| | - Jae Myun Lee
- Department of Microbiology and Immunology Brain Korea 21 PLUS Project for Medical Science Yonsei University College of Medicine Seoul Korea
| |
Collapse
|
40
|
Can U, Uysal S, Ruveyda Ugur A, Toker A, Aslan U, Taha Hidayetoglu B. Can YKL-40 be an Inflammatory Biomarker in Vitamin D Deficiency? INT J VITAM NUTR RES 2019; 89:309-313. [PMID: 30806610 DOI: 10.1024/0300-9831/a000545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Vitamin D deficiency is associated with several non-homeostatic conditions and/or diseases like inflammation, atherosclerosis, cardiovascular disease and mortality. YKL-40 is a glycoprotein, secreted by macrophages, neutrophils and different cell types and it is also associated with inflammation and pathological tissue remodeling. In this study, we aimed to evaluate relationship between the proinflammatory biomarkers YKL-40 and hs-CRP levels and vitamin D deficiency. Our study group includes 45 subjects with vitamin D deficiency (Group 1) (20 M, 25 F; mean age 37.72 ± 7.70 years) and 40 age and sex-matched healthy subjects with normal serum levels of vitamin D (Group 2) (19 M, 21 F; mean age 39.26 ± 7.41 years). Plasma 25 (OH) vitamin D levels were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. Plasma YKL-40 analysis was performed by ELISA. Serum hs-CRP levels were measured by nephelometric method. Plasma vitamin D levels below 20 ng/mL were accepted as vitamin D deficiency. Although we could not find any significant differences by means of serum hs-CRP levels between Group 1 and Group 2 (2.21 (0.27-11.70); 1.79 (0.16-9.85) mg/L, p = 0.247), plasma YKL-40 levels were significantly higher in group 1 than group2 (70.47 (17.84-198.50); 47.14 (4.80-135.48) ng/mL, p = 0.047). In literature, vitamin D deficiency is associated with inflammation. In our study, we found similar hs-CRP levels between groups and higher YKL-40 levels in group 1. Vitamin D deficiency may be related to high YKL-40 levels in terms of causing chronic inflammation.
Collapse
Affiliation(s)
- Ummugulsum Can
- Department of Biochemistry, Konya Education and Research Hospital, Konya, Turkey
| | - Saliha Uysal
- Department of Biochemistry, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Ayse Ruveyda Ugur
- Department of Microbiology, Konya Education and Research Hospital, Konya, Turkey
| | - Aysun Toker
- Department of Biochemistry, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Uysaler Aslan
- Department of Internal Medicine, Konya Education and Research Hospital, Konya, Turkey
| | | |
Collapse
|
41
|
Wu X, Zahari MS, Renuse S, Kelkar DS, Barbhuiya MA, Rojas PL, Stearns V, Gabrielson E, Malla P, Sukumar S, Mahajan NP, Pandey A. The non-receptor tyrosine kinase TNK2/ACK1 is a novel therapeutic target in triple negative breast cancer. Oncotarget 2018; 8:2971-2983. [PMID: 27902967 PMCID: PMC5356856 DOI: 10.18632/oncotarget.13579] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 10/10/2016] [Indexed: 12/04/2022] Open
Abstract
Breast cancer is the most prevalent cancer in women worldwide. About 15-20% of all breast cancers do not express estrogen receptor, progesterone receptor or HER2 receptor and hence are collectively classified as triple negative breast cancer (TNBC). These tumors are often relatively aggressive when compared to other types of breast cancer, and this issue is compounded by the lack of effective targeted therapy. In our previous phosphoproteomic profiling effort, we identified the non-receptor tyrosine kinase TNK2 as activated in a majority of aggressive TNBC cell lines. In the current study, we show that high expression of TNK2 in breast cancer cell lines correlates with high proliferation, invasion and colony forming ability. We demonstrate that knockdown of TNK2 expression can substantially suppress the invasiveness and proliferation advantage of TNBC cells in vitro and tumor formation in xenograft mouse models. Moreover, inhibition of TNK2 with small molecule inhibitor (R)-9bMS significantly compromised TNBC proliferation. Finally, we find that high levels of TNK2 expression in high-grade basal-like breast cancers correlates significantly with poorer patient outcome. Taken together, our study suggests that TNK2 is a novel potential therapeutic target for the treatment of TNBC.
Collapse
Affiliation(s)
- Xinyan Wu
- Department of Biological Chemistry, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Muhammad Saddiq Zahari
- Department of Biological Chemistry, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Santosh Renuse
- Department of Biological Chemistry, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
| | - Dhanashree S Kelkar
- Department of Biological Chemistry, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Mustafa A Barbhuiya
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Pamela L Rojas
- Department of Biological Chemistry, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Vered Stearns
- Department of Oncology, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Edward Gabrielson
- Department of Oncology, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,Department of Pathology, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Pavani Malla
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, U.S.A
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| | - Nupam P Mahajan
- Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, U.S.A.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, U.S.A
| | - Akhilesh Pandey
- Department of Biological Chemistry, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,Department of Oncology, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A.,Department of Pathology, Johns Hopkins University School of Medicine Baltimore, MD 21205, U.S.A
| |
Collapse
|
42
|
Conroy AL, Hawkes MT, Elphinstone R, Opoka RO, Namasopo S, Miller C, John CC, Kain KC. Chitinase-3-like 1 is a biomarker of acute kidney injury and mortality in paediatric severe malaria. Malar J 2018; 17:82. [PMID: 29448936 PMCID: PMC5815237 DOI: 10.1186/s12936-018-2225-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022] Open
Abstract
Background Chitinase-3-like 1 (CHI3L1) is a glycoprotein elevated in paediatric severe malaria, and an emerging urinary biomarker of acute kidney injury (AKI). Based on the hypothesis that elevated CHI3L1 levels in malaria are associated with disease severity, the relationship between plasma CHI3L1 levels, AKI and mortality was investigated in Ugandan children enrolled in a clinical trial evaluating inhaled nitric oxide (iNO) as an adjunctive therapy for severe malaria. Methods Plasma CHI3L1 levels were measured daily for 4 days in children admitted to hospital with severe malaria and at day 14 follow up. AKI was defined using the Kidney Disease: Improving Global Outcomes consensus criteria. This is a secondary analysis of a randomized double-blind placebo-controlled trial of iNO versus placebo as an adjunctive therapy for severe malaria. Inclusion criteria were: age 1–10 years, and selected criteria for severe malaria. Exclusion criteria included suspected bacterial meningitis, known chronic illness including renal disease, haemoglobinopathy, or severe malnutrition. iNO was administered by non-rebreather mask for up to 72 h at 80 ppm. Results CHI3L1 was elevated in patients with AKI and remained higher over hospitalization (p < 0.0001). Admission CHI3L1 levels were elevated in children who died. By multivariable analysis logCHI3L1 levels were associated with increased risk of in-hospital death (relative risk, 95% CI 4.10, 1.32–12.75, p = 0.015) and all-cause 6 month mortality (3.21, 1.47–6.98, p = 0.003) following correction for iNO and AKI. Treatment with iNO was associated with delayed CHI3L1 recovery with a daily decline of 34% in the placebo group versus 29% in the iNO group (p = 0.012). CHI3L1 levels correlated with markers of inflammation (CRP, sTREM-1, CXCL10), endothelial activation (Ang-2, sICAM-1) and intravascular haemolysis (LDH, haem, haemopexin). Conclusions CHI3L1 is a novel biomarker of malaria-associated AKI and an independent risk factor for mortality that is associated with well-established pathways of severe malaria pathogenesis including inflammation, endothelial activation, and haemolysis. Trial registration Clinicaltrials.gov, NCT01255215. Registered December 7th 2010 Electronic supplementary material The online version of this article (10.1186/s12936-018-2225-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea L Conroy
- Department of Pediatrics, Indiana University School of Medicine, 1044 West Walnut St., Building 4, Indianapolis, IN, 46202, USA. .,Sandra Rotman Centre for Global Health, Toronto General Hospital, University Health Network, MaRS Centre, 101 College St. TMDT 10-360A, Toronto, ON, M5G 1L7, Canada. .,Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Michael T Hawkes
- Division of Pediatric Infectious Diseases, 3-593 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, T6G1C9, Canada
| | - Robyn Elphinstone
- Sandra Rotman Centre for Global Health, Toronto General Hospital, University Health Network, MaRS Centre, 101 College St. TMDT 10-360A, Toronto, ON, M5G 1L7, Canada
| | - Robert O Opoka
- Department of Pediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Sophie Namasopo
- Department of Pediatrics, Jinja Regional Referral Hospital, P.O. Box 43, Jinja, Uganda
| | | | - Chandy C John
- Department of Pediatrics, Indiana University School of Medicine, 1044 West Walnut St., Building 4, Indianapolis, IN, 46202, USA
| | - Kevin C Kain
- Sandra Rotman Centre for Global Health, Toronto General Hospital, University Health Network, MaRS Centre, 101 College St. TMDT 10-360A, Toronto, ON, M5G 1L7, Canada.,Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
43
|
Zhou Y, He CH, Yang DS, Nguyen T, Cao Y, Kamle S, Lee CM, Gochuico BR, Gahl WA, Shea BS, Lee CG, Elias JA. Galectin-3 Interacts with the CHI3L1 Axis and Contributes to Hermansky-Pudlak Syndrome Lung Disease. THE JOURNAL OF IMMUNOLOGY 2018; 200:2140-2153. [PMID: 29427412 DOI: 10.4049/jimmunol.1701442] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/04/2018] [Indexed: 01/08/2023]
Abstract
Hermansky-Pudlak syndrome (HPS) comprises a group of inherited disorders caused by mutations that alter the function of lysosome-related organelles. Pulmonary fibrosis is the major cause of morbidity and mortality in HPS-1 and HPS-4 patients. However, the mechanisms that underlie the exaggerated injury and fibroproliferative repair responses in HPS have not been adequately defined. In particular, although Galectin-3 (Gal-3) is dysregulated in HPS, its roles in the pathogenesis of HPS have not been adequately defined. In addition, although chitinase 3-like 1 (CHI3L1) and its receptors play major roles in the injury and repair responses in HPS, the ability of Gal-3 to interact with or alter the function of these moieties has not been evaluated. In this article, we demonstrate that Gal-3 accumulates in exaggerated quantities in bronchoalveolar lavage fluids, and traffics abnormally and accumulates intracellularly in lung fibroblasts and macrophages from bleomycin-treated pale ear, HPS-1-deficient mice. We also demonstrate that Gal-3 drives epithelial apoptosis when in the extracellular space, and stimulates cell proliferation and myofibroblast differentiation when accumulated in fibroblasts and M2-like differentiation when accumulated in macrophages. Biophysical and signaling evaluations also demonstrated that Gal-3 physically interacts with IL-13Rα2 and CHI3L1, and competes with TMEM219 for IL-13Rα2 binding. By doing so, Gal-3 diminishes the antiapoptotic effects of and the antiapoptotic signaling induced by CHI3L1 in epithelial cells while augmenting macrophage Wnt/β-catenin signaling. Thus, Gal-3 contributes to the exaggerated injury and fibroproliferative repair responses in HPS by altering the antiapoptotic and fibroproliferative effects of CHI3L1 and its receptor complex in a tissue compartment-specific manner.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912;
| | - Chuan Hua He
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Daniel S Yang
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Tung Nguyen
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Yueming Cao
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Suchitra Kamle
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Bernadette R Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - William A Gahl
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Barry S Shea
- Division of Pulmonary, Critical Care and Sleep Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903; and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912; .,Department of Internal Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02903
| |
Collapse
|
44
|
Kim MH, Wu WH, Choi JH, Kim J, Jun JH, Ko Y, Lee JH. Galectin-1 from conditioned medium of three-dimensional culture of adipose-derived stem cells accelerates migration and proliferation of human keratinocytes and fibroblasts. Wound Repair Regen 2017; 26 Suppl 1:S9-S18. [PMID: 28857355 DOI: 10.1111/wrr.12579] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/16/2017] [Indexed: 12/19/2022]
Abstract
Keratinocytes and fibroblasts cells play important roles in the skin-wound healing process and are the cell types activated by trauma. Activated cells participate in epithelialization, granulation, scar tissue formation, wound remodeling, and angiogenesis via a series of cellular activities including migration and proliferation. Previous studies reported that the conditioned medium (CM) of adipose-derived stem cells (ADSCs) stimulated the migration and proliferation of cell types involved in the skin wound healing process; however, these studies only show ADSC-CM effects that were obtained using 2-dimensional (2D) culture. Recently, 3-dimensional (3D) culture has been considered as a more physiologically appropriate system than 2D culture for ADSC cultures; therefore, ADSC-CM was collected from 3D culture (ADSC-CM-3D) and compared with ADSC-CM from 2D culture (ADSC-CM-2D) to investigate the effects on the migration and proliferation of human keratinocytes (HaCaTs) and fibroblasts. The migrations of the HaCaT cells and fibroblasts were significantly higher with ADSC-CM-3D compared with the 2D culture; similarly, the proliferation of HaCaT cells was also highly stimulated by ADSC-CM-3D. Proteomic analyses of the ADSC-CM revealed that collagens and actins were highly expressed in the 3D-culture system. Chitinase 3-like 1 (CHI3L1), tissue inhibitor of metalloproteinases (TIMP), and galectin-1 were specifically expressed only in ADSC-CM-3D. Especially, through antibody neutralization, galectin-1 in ADSC-CM-3D was found to be an important factor for the migration of human keratinocytes. Therefore, these results suggest that ADSC-CM-3D was more effective in the wound healing than ADSC-CM-2D, and galectin-1 in ADSC-CM-3D was could be a promising option for skin-wound healing. Furthermore, the differential expressions of several ADSC-CM proteins between the 2D- and 3D-culture systems may be used as basic information for the development of efficient wound-healing strategies.
Collapse
Affiliation(s)
- Min Ho Kim
- Eulji Medi-Bio Research Institute, Eulji University, Seoul, Republic of Korea.,Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Wen Hao Wu
- Eulji Medi-Bio Research Institute, Eulji University, Seoul, Republic of Korea
| | - Jee Hyun Choi
- Eulji Medi-Bio Research Institute, Eulji University, Seoul, Republic of Korea
| | - Jihyun Kim
- Department of Senior Healthcare, BK21 plus Program, Graduated School, Eulji University, Seongnam, Republic of Korea
| | - Jin Hyun Jun
- Eulji Medi-Bio Research Institute, Eulji University, Seoul, Republic of Korea.,Department of Senior Healthcare, BK21 plus Program, Graduated School, Eulji University, Seongnam, Republic of Korea.,Department of Biomedical Laboratory Science, Eulji University, Seongnam, Republic of Korea
| | - Yong Ko
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jong Hun Lee
- Eulji Medi-Bio Research Institute, Eulji University, Seoul, Republic of Korea.,Department of Plastic and Reconstructive Surgery, Eulji General Hospital, College of Medicine, Eulji University, Seoul, Republic of Korea
| |
Collapse
|
45
|
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
|
46
|
Li Z, Gu J, Liu J, Zhu Q, Lu H, Lu Y, Rao J, Lu L, Wang X. Chitinase 3-like-1 deficient donor splenocytes accentuated the pathogenesis of acute graft-versus-host diseases through regulating T cell expansion and type I inflammation. Int Immunopharmacol 2017; 46:201-209. [PMID: 28324830 DOI: 10.1016/j.intimp.2017.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/20/2017] [Accepted: 03/08/2017] [Indexed: 01/05/2023]
Abstract
Acute graft-versus-host disease (aGVHD) is a major complication following transplantation, limiting the success of this therapy. Chitinase 3-like-1 (CHI3L1), a member of the glycosyl hydrolase 18 family, plays a critical role in bacterial infections, allergic disease and a variety of malignancies. Here, we investigated whether CHI3L1 could affect the pathogenesis of aGVHD in a mouse allo-HCT model. In this study, we show that CHI3L1 deficiency in donor T cells increased the severity of aGVHD through enhancing systemic and local inflammation. In addition, we found that aGVHD induced by CHI3L1-knockout (CHI3L1-KO) donors resulted in massive expansion of donor CD3+ T cells, release of Th1-related cytokines and chemokines, and significant inhibition of CD4+CD25+Foxp3+ regulatory T cells (Tregs) without changing the suppressive ability of donor Tregs remarkably. Expression of PERK1/2 and PAkt increased both in the skin and intestine from CHI3L1-KO splenocytes-treated aGVHD mice. Moreover, at mRNA and protein levels, we defined several molecules that may account for the enhanced ability of CHI3L1-KO splenocytes to migrate into target organs and produce Th1-related cytokines and chemokines, such as CXCL9, CXCL11, IFN-γ and TNF-α. Therefore, these results imply that CHI3L1 levels in donor cells may be related to the risk of aGVHD and targeting CHI3L1 may be a promising clinical strategy to control aGVHD.
Collapse
Affiliation(s)
- Zengyao Li
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Jian Gu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Jing Liu
- Department of Radiotherapy, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Qin Zhu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Hao Lu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Yunjie Lu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Jianhua Rao
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Ling Lu
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Xuehao Wang
- Liver Transplantation Center, First Affiliated Hospital, Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China.
| |
Collapse
|
47
|
König K, Guy KJ, Nold-Petry CA, Barfield CP, Walsh G, Drew SM, Veldman A, Nold MF, Casalaz DM. BNP, troponin I, and YKL-40 as screening markers in extremely preterm infants at risk for pulmonary hypertension associated with bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1076-L1081. [PMID: 27760764 DOI: 10.1152/ajplung.00344.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/13/2016] [Indexed: 11/22/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is often complicated by pulmonary hypertension (PH). We investigated three biomarkers potentially suitable as screening markers for extremely preterm infants at risk of BPD-associated PH. In this prospective observational cohort study conducted in a tertiary neonatal intensive care unit, 83 preterm infants with BPD born <28-wk gestation and still inpatients at 36-wk corrected age received an echocardiogram and blood tests of B-type natriuretic peptide (BNP), troponin I, and YKL-40. Infants were analyzed according to echocardiographic evidence of tricuspid regurgitation (TR). Thirty infants had evidence of TR on echocardiogram at 36-wk corrected age. Infants with or without TR had similar baseline demographics: mean ± SD gestational age 261 ± 12 vs. 261 ± 11 wk and birth weight 830 ± 206 vs. 815 ± 187 g, respectively. There was no difference in duration of respiratory support. The right ventricular systolic pressure of infants with evidence of TR was 40 ± 16 mmHg. BNP was the only biomarker that proved to be significantly higher in infants with evidence of TR: median (interquartile range) serum level 54.5 (35-105) vs. 41.5 (30-59) pg/ml, P = 0.043. Subgroup analysis of infants with severe BPD requiring discharge on home oxygen or BPD-related mortality revealed similar results. There was no difference between groups for troponin I and YKL-40. In conclusion, increased serum levels of BNP were associated with evidence of TR at 36-wk corrected gestational age in extremely preterm infants, suggesting a potential role as a screening biomarker for BPD-associated PH.
Collapse
Affiliation(s)
- Kai König
- Mercy Hospital for Women, Department of Paediatrics, Melbourne, Victoria, Australia;
| | - Katelyn J Guy
- Mercy Hospital for Women, Department of Paediatrics, Melbourne, Victoria, Australia
| | - Claudia A Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; and.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Charles P Barfield
- Mercy Hospital for Women, Department of Paediatrics, Melbourne, Victoria, Australia
| | - Geraldine Walsh
- Mercy Hospital for Women, Department of Paediatrics, Melbourne, Victoria, Australia
| | - Sandra M Drew
- Mercy Hospital for Women, Department of Paediatrics, Melbourne, Victoria, Australia
| | - Alex Veldman
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; and.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Marcel F Nold
- Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria, Australia; and.,Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Dan M Casalaz
- Mercy Hospital for Women, Department of Paediatrics, Melbourne, Victoria, Australia
| |
Collapse
|
48
|
Lee CM, He CH, Nour AM, Zhou Y, Ma B, Park JW, Kim KH, Cruz CD, Sharma L, Nasr ML, Modis Y, Lee CG, Elias JA. IL-13Rα2 uses TMEM219 in chitinase 3-like-1-induced signalling and effector responses. Nat Commun 2016; 7:12752. [PMID: 27629921 PMCID: PMC5027616 DOI: 10.1038/ncomms12752] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/28/2016] [Indexed: 12/27/2022] Open
Abstract
Recent studies demonstrated that chitinase 3-like-1 (Chi3l1) binds to and signals via IL-13Rα2. However, the mechanism that IL-13Rα2 uses to mediate the effects of Chi3l1 has not been defined. Here, we demonstrate that the membrane protein, TMEM219, is a binding partner of IL-13Rα2 using yeast two-hybrid, co-immunoprecipitation, co-localization and bimolecular fluorescence complementation assays. Furthermore, fluorescence anisotropy nanodisc assays revealed a direct physical interaction between TMEM219 and IL-13Rα2-Chi3l1 complexes. Null mutations or siRNA silencing of TMEM219 or IL-13Rα2 similarly decreased Chi3l1-stimulated epithelial cell HB-EGF production and macrophage MAPK/Erk and PKB/Akt activation. Null mutations of TMEM219 or IL-13Rα2 also phenocopied one another as regards the ability of Chi3l1 to inhibit oxidant-induced apoptosis and lung injury, promote melanoma metastasis and stimulate TGF-β1. TMEM219 also contributed to the decoy function of IL-13Rα2. These studies demonstrate that TMEM219 plays a critical role in Chi3l1-induced IL-13Rα2 mediated signalling and tissue responses.
Collapse
Affiliation(s)
- Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Chuan Hua He
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Adel M. Nour
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Jin Wook Park
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Kyung Hee Kim
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Charles Dela Cruz
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, 300 Cedar Street, New Haven, Connecticut 06520, USA
| | - Lokesh Sharma
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, 300 Cedar Street, New Haven, Connecticut 06520, USA
| | - Mahmoud L. Nasr
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Yorgo Modis
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Jack A. Elias
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
- Division of Medicine and Biological Sciences, Warren Alpert School of Medicine, Brown University, Box G-A1, 97 Waterman Street, Providence, Rhode Island 02912, USA
| |
Collapse
|
49
|
Kozan A, Kilic N, Alacam H, Guzel A, Guvenc T, Acikgoz M. The Effects of Dexamethasone and L-NAME on Acute Lung Injury in Rats with Lung Contusion. Inflammation 2016; 39:1747-56. [DOI: 10.1007/s10753-016-0409-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
50
|
Puthumana J, Hall IE, Reese PP, Schröppel B, Weng FL, Thiessen-Philbrook H, Doshi MD, Rao V, Lee CG, Elias JA, Cantley LG, Parikh CR. YKL-40 Associates with Renal Recovery in Deceased Donor Kidney Transplantation. J Am Soc Nephrol 2016; 28:661-670. [PMID: 27451287 DOI: 10.1681/asn.2016010091] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/28/2016] [Indexed: 12/16/2022] Open
Abstract
Deceased donor kidneys with AKI are often discarded for fear of poor transplant outcomes. Donor biomarkers that predict post-transplant renal recovery could improve organ selection and reduce discard. We tested whether higher levels of donor urinary YKL-40, a repair phase protein, associate with improved recipient outcomes in a prospective cohort study involving deceased kidney donors from five organ procurement organizations. We measured urinary YKL-40 concentration in 1301 donors (111 had AKI, defined as doubling of serum creatinine) and ascertained outcomes in the corresponding 2435 recipients, 756 of whom experienced delayed graft function (DGF). Donors with AKI had higher urinary YKL-40 concentration (P<0.001) and acute tubular necrosis on procurement biopsies (P=0.05). In fully adjusted analyses, elevated donor urinary YKL-40 concentration associated with reduced risk of DGF in both recipients of AKI donor kidneys (adjusted relative risk, 0.51 [95% confidence interval (95% CI), 0.32 to 0.80] for highest versus lowest YKL-40 tertile) and recipients of non-AKI donor kidneys (adjusted relative risk, 0.79 [95% CI, 0.65 to 0.97]). Furthermore, in the event of DGF, elevated donor urinary YKL-40 concentration associated with higher 6-month eGFR (6.75 [95% CI, 1.49 to 12.02] ml/min per 1.73 m2) and lower risk of graft failure (adjusted hazard ratio, 0.50 [95% CI, 0.27 to 0.94]). These findings suggest that YKL-40 is produced in response to tubular injury and is independently associated with recovery from AKI and DGF. If ultimately validated as a prognostic biomarker, urinary YKL-40 should be considered in determining the suitability of donor kidneys for transplant.
Collapse
Affiliation(s)
- Jeremy Puthumana
- Program of Applied Translational Research, Department of Medicine and
| | - Isaac E Hall
- Program of Applied Translational Research, Department of Medicine and.,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Peter P Reese
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Francis L Weng
- Renal and Pancreas Transplant Division, Saint Barnabas Medical Center, Livingston, New Jersey
| | | | - Mona D Doshi
- Department of Medicine, Division of Nephrology, Wayne State University School of Medicine, Detroit, Michigan
| | - Veena Rao
- Program of Applied Translational Research, Department of Medicine and
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island; and
| | - Jack A Elias
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island; and
| | - Lloyd G Cantley
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Chirag R Parikh
- Program of Applied Translational Research, Department of Medicine and .,Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut.,Section of Nephrology, Veterans Affairs Medical Center, West Haven, Connecticut
| |
Collapse
|