1
|
Panigrahi T, Khamar P, Shetty R, Kannan R, Ashok N, Nishtala K, Ghosh A, Deshpande V. Longitudinal analysis of wound healing response post SMILE and LASIK surgery using proteomic profiling of tears. Exp Eye Res 2024; 246:109987. [PMID: 38964497 DOI: 10.1016/j.exer.2024.109987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Different types of refractive surgeries often exhibit differences in wound healing responses. The current study investigated post-operative tear protein profiles in subjects who underwent LASIK and SMILE to elucidate global changes to the proteomic profile during the period the patient cornea undergoes healing. In this study, 10 patients underwent LASIK and SMILE surgery with a contralateral paired eye design. Tear samples were collected using Schirmer's strips preoperatively, at 1 month, 3 months and 6 months postoperatively. Quantitative ITRAQ labeled proteomics was performed and the tear protein ratios were normalized to pre-operative protein levels for each subject. Whole proteomics identified 1345 proteins in tears from LASIK and 1584 proteins in SMILE across time points. About 67 proteins were common in LASIK and SMILE tears across all the time points. Wound healing responses were differentially regulated between two refractive surgeries (SMILE and LASIK). The proteins Ceruloplasmin, Clusterin, Serotransferrin were upregulated at 1 month and 3 months and downregulated at 6 months post operatively in LASIK surgery where as in SMILE these were downregulated. Galectin 3 binding protein showed upregulation at 1 month and the levels decreased at 3 months and 6 months postop in LASIK tears whereas the levels increased at 3 months and 6 months post-op in SMILE tears. The levels of proteins that protect from oxidative stress were higher in SMILE as compared to LASIK postoperatively. The extracellular matrix proteins showed an increase in expression at 6 months in SMILE tears and was stabilized at 6 months in LASIK tears post operatively. Different refractive surgeries induce distinct wound healing responses as identified in tears. This study has implications in targeting key proteins for improving the clinical outcome postrefractive surgery.
Collapse
Affiliation(s)
| | - Pooja Khamar
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore, India
| | - Rohit Shetty
- Cornea and Refractive Services, Narayana Nethralaya, Bangalore, India
| | - Ramaraj Kannan
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | - Nikhil Ashok
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India
| | | | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| | - Vrushali Deshpande
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, India.
| |
Collapse
|
2
|
Wang A, Zhang H, Li X, Zhao Y. Annexin A1 in the nervous and ocular systems. Neural Regen Res 2024; 19:591-597. [PMID: 37721289 PMCID: PMC10581565 DOI: 10.4103/1673-5374.380882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/06/2023] [Accepted: 06/02/2023] [Indexed: 09/19/2023] Open
Abstract
The therapeutic potential of Annexin A1, an important member of the Annexin superfamily, has become evident in results of experiments with multiple human systems and animal models. The anti-inflammatory and pro-resolving effects of Annexin A1 are characteristic of pathologies involving the nervous system. In this review, we initially describe the expression sites of Annexin A1, then outline the mechanisms by which Annexin A1 maintains the neurological homeostasis through either formyl peptide receptor 2 or other molecular approaches; and, finally, we discuss the neuroregenerative potential qualities of Annexin A1. The eye and the nervous system are anatomically and functionally connected, but the association between visual system pathogenesis, especially in the retina, and Annexin A1 alterations has not been well summarized. Therefore, we explain the beneficial effects of Annexin A1 for ocular diseases, especially for retinal diseases and glaucoma on the basis of published findings, and we explore present and future delivery strategies for Annexin A1 to the retina.
Collapse
Affiliation(s)
- Aijia Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xing Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
3
|
Liu H, Zhang X, Tan Q, Ge L, Lu J, Ren C, Bian B, Li Y, Liu Y. A moderate dosage of prostaglandin E2-mediated annexin A1 upregulation promotes alkali-burned corneal repair. iScience 2023; 26:108565. [PMID: 38144456 PMCID: PMC10746505 DOI: 10.1016/j.isci.2023.108565] [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: 09/07/2023] [Revised: 10/14/2023] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
Corneal alkali burn remains a clinical challenge in ocular emergency, necessitating the development of effective therapeutic drugs. Here, we observed the arachidonic acid metabolic disorders of corneas induced by alkali burns and aimed to explore the role of Prostaglandin E2 (PGE2), a critical metabolite of arachidonic acid, in the repair of alkali-burned corneas. We found a moderate dosage of PGE2 promoted the alkali-burned corneal epithelial repair, whereas a high dosage of PGE2 exhibited a contrary effect. This divergent effect is attributed to different dosages of PGE2 regulating ANXA1 expression differently. Mechanically, a high dosage of PGE2 induced higher GATA3 expression, followed by enhanced GATA3 binding to the ANXA1 promoter to inhibit ANXA1 expression. In contrast, a moderate dosage of PGE2 increased CREB1 phosphorylation and reduced GATA3 binding to the ANXA1 promoter, promoting ANXA1 expression. We believe PGE2 and its regulatory target ANXA1 could be potential drugs for alkali-burned corneas.
Collapse
Affiliation(s)
- Hongling Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Xue Zhang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Qiang Tan
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Lingling Ge
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Jia Lu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Chunge Ren
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Baishijiao Bian
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Third Military Medical University (Army Medical University), Shigatse 857000, China
- State Key Laboratory of Trauma, Burns, and Combined Injury, Department of Trauma Medical Center, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Yijian Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
4
|
Lyngstadaas AV, Olsen MV, Bair J, Yang M, Hodges RR, Utheim TP, Serhan CN, Dartt DA. Anti-Inflammatory and Pro-Resolving Actions of the N-Terminal Peptides Ac2-26, Ac2-12, and Ac9-25 of Annexin A1 on Conjunctival Goblet Cell Function. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1817-1832. [PMID: 37423551 PMCID: PMC10616711 DOI: 10.1016/j.ajpath.2023.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 07/11/2023]
Abstract
Annexin A1 (AnxA1) is the primary mediator of the anti-inflammatory actions of glucocorticoids. AnxA1 functions as a pro-resolving mediator in cultured rat conjunctival goblet cells to ensure tissue homeostasis through stimulation of intracellular [Ca2+] ([Ca2+]i) and mucin secretion. AnxA1 has several N-terminal peptides with anti-inflammatory properties of their own, including Ac2-26, Ac2-12, and Ac9-25. The increase in [Ca2+]i caused by AnxA1 and its N-terminal peptides in goblet cells was measured to determine the formyl peptide receptors used by the compounds and the action of the peptides on histamine stimulation. Changes in [Ca2+]i were determined by using a fluorescent Ca2+ indicator. AnxA1 and its peptides each activated formyl peptide receptors in goblet cells. AnxA1 and Ac2-26 at 10-12 mol/L and Ac2-12 at 10-9 mol/L inhibited the histamine-stimulated increase in [Ca2+]i, as did resolvin D1 and lipoxin A4 at 10-12 mol/L, whereas Ac9-25 did not. AnxA1 and Ac2-26 counter-regulated the H1 receptor through the p42/p44 mitogen-activated protein kinase/extracellular regulated kinase 1/2, β-adrenergic receptor kinase, and protein kinase C pathways, whereas Ac2-12 counter-regulated only through β-adrenergic receptor kinase. In conclusion, current data show that the N-terminal peptides Ac2-26 and Ac2-12, but not Ac9-25, share multiple functions with the full-length AnxA1 in goblet cells, including inhibition of histamine-stimulated increase in [Ca2+]i and counter-regulation of the H1 receptor. These actions suggest a potential pharmaceutical application of the AnxA1 N-terminal peptides Ac2-26 and Ac2-12 in homeostasis and ocular inflammatory diseases.
Collapse
Affiliation(s)
- Anne V Lyngstadaas
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Markus V Olsen
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jeffrey Bair
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Menglu Yang
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Robin R Hodges
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Tor P Utheim
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway; Department of Plastic and Reconstructive Surgery, University of Oslo, Oslo, Norway
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesia, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Darlene A Dartt
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts; Department of Plastic and Reconstructive Surgery, University of Oslo, Oslo, Norway.
| |
Collapse
|
5
|
Zheng Y, Li Y, Li S, Hu R, Zhang L. Annexin A1 (Ac2-26)-dependent Fpr2 receptor alleviates sepsis-induced acute kidney injury by inhibiting inflammation and apoptosis in vivo and in vitro. Inflamm Res 2023; 72:347-362. [PMID: 36544058 PMCID: PMC9925514 DOI: 10.1007/s00011-022-01640-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Excessive inflammatory responses and apoptosis are critical pathologies that contribute to sepsis-induced acute kidney injury (SI-AKI). Annexin A1 (ANXA1), a member of the calcium-dependent phospholipid-binding protein family, protects against SI-AKI through its anti-inflammatory and antiapoptotic effects, but the underlying mechanisms are still largely unknown. METHODS In vivo, SI-AKI mouse models were established via caecal ligation and puncture (CLP) and were then treated with the Ac2-26 peptide of ANXA1 (ANXA1 (Ac2-26)), WRW4 (Fpr2 antagonist) or both. In vitro, HK-2 cells were induced by lipopolysaccharide (LPS) and then treated with ANXA1 (Ac2-26), Fpr2-siRNA or both. RESULTS In the present study, we found that the expression levels of ANXA1 were decreased, and the expression levels of TNF-α, IL-1β, IL-6, cleaved caspase-3, cleaved caspase-8 and Bax were significantly increased, accompanied by marked kidney tissue apoptosis in vivo. Moreover, we observed that ANXA1 (Ac2-26) significantly reduced the levels of TNF-α, IL-1β and IL-6 and cleaved caspase-3, cleaved caspase-8, FADD and Bax and inhibited apoptosis in kidney tissue and HK-2 cells, accompanied by pathological damage to kidney tissue. Seven-day survival, kidney function and cell viability were significantly improved in vivo and in vitro, respectively. Furthermore, the administration of ANXA1 (Ac2-26) inhibited the CLP- or LPS-induced phosphorylation of PI3K and AKT and downregulated the level of NF-κB in vivo and in vitro. Moreover, our data demonstrate that blocking the Fpr2 receptor by the administration of WRW4 or Fpr2-siRNA reversed the abovementioned regulatory role of ANXA1, accompanied by enhanced phosphorylation of PI3K and AKT and upregulation of the level of NF-κB in vivo and in vitro. CONCLUSIONS Taken together, this study provides evidence that the protective effect of ANXA1 (Ac2-26) on SI-AKI largely depends on the negative regulation of inflammation and apoptosis via the Fpr2 receptor.
Collapse
Affiliation(s)
- Yanlei Zheng
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079 China
| | - Yan Li
- Department of Critical Care Medicine, Xiangyang Central Hospital, Xiangyang, 440121 China
| | - Shi Li
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079 China
| | - Ronghua Hu
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079 China
| | - Li Zhang
- Department of Critical Care Medicine, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China.
| |
Collapse
|
6
|
André da Silva R, Moraes de Paiva Roda V, Philipe de Souza Ferreira L, Oliani SM, Paula Girol A, Gil CD. Annexins as potential targets in ocular diseases. Drug Discov Today 2022; 27:103367. [PMID: 36165812 DOI: 10.1016/j.drudis.2022.103367] [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] [Received: 01/31/2022] [Revised: 08/05/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022]
Abstract
Annexins (AnxAs) are Ca2+/phospholipid-binding proteins extensively studied and generally involved in several diseases. Although evidence exists regarding the distribuition of AnxAs in the visual system, their exact roles and the exact cell types of the eye where these proteins are expressed are not well-understood. AnxAs have pro-resolving roles in infectious, autoimmune, degenerative, fibrotic and angiogenic conditions, making them an important target in ocular tissue homeostasis. This review summarizes the current knowledge on the distribution and function of AnxA1-8 isoforms under normal and pathological conditions in the visual system, as well as perspectives for ophthalmologic treatments, including the potential use of the AnxA1 recombinant and/or its mimetic peptide Ac2-26.
Collapse
Affiliation(s)
- Rafael André da Silva
- Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP 15054-000, Brazil
| | - Vinicius Moraes de Paiva Roda
- Life Systems Biology Graduate Program, Institute of Biomedical Sciences, Universidade de São Paulo (USP), São Paulo, SP 05508-000, Brazil
| | - Luiz Philipe de Souza Ferreira
- Structural and Functional Biology Graduate Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04023-900, Brazil
| | - Sonia M Oliani
- Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP 15054-000, Brazil; Structural and Functional Biology Graduate Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04023-900, Brazil; Advanced Research Center in Medicine (CEPAM) Unilago, São José do Rio Preto, SP 15030-070, Brazil
| | - Ana Paula Girol
- Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP 15054-000, Brazil; Structural and Functional Biology Graduate Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04023-900, Brazil; Centro Universitário Padre Albino (UNIFIPA), Catanduva, SP 15809-144, Brazil
| | - Cristiane D Gil
- Biosciences Graduate Program, Institute of Biosciences, Letters and Exact Sciences, Universidade Estadual Paulista (UNESP), São José do Rio Preto, SP 15054-000, Brazil; Structural and Functional Biology Graduate Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP 04023-900, Brazil.
| |
Collapse
|
7
|
Li W, Jiang H, Bai C, Yu S, Pan Y, Wang C, Li H, Li M, Sheng Y, Chu F, Wang J, Chen Y, Li J, Jiang J. Ac2-26 attenuates hepatic ischemia-reperfusion injury in mice via regulating IL-22/IL-22R1/STAT3 signaling. PeerJ 2022; 10:e14086. [PMID: 36193422 PMCID: PMC9526407 DOI: 10.7717/peerj.14086] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/29/2022] [Indexed: 01/27/2023] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is one of the major sources of mortality and morbidity associated with hepatic surgery. Ac2-26, a short peptide of Annexin A1 protein, has been proved to have a protective effect against IRI. However, whether it exerts a protective effect on HIRI has not been reported. The HIRI mice model and the oxidative damage model of H2O2-induced AML12 cells were established to investigate whether Ac2-26 could alleviate HIRI by regulating the activation of IL-22/IL-22R1/STAT3 signaling. The protective effect of Ac2-26 was measured by various biochemical parameters related to liver function, apoptosis, inflammatory reaction, mitochondrial function and the expressions of IL-22, IL-22R1, p-STAT3Tyr705. We discovered that Ac2-26 reduced the Suzuki score and cell death rate, and increased the cell viability after HIRI. Moreover, we unraveled that Ac2-26 significantly decreased the number of apoptotic hepatocytes, and the expressions of cleaved-caspase-3 and Bax/Bcl-2 ratio. Furthermore, HIRI increased the contents of malondialdehyde (MDA), NADP+/NADPH ratio and reactive oxygen species (ROS), whereas Ac2-26 decreased them significantly. Additionally, Ac2-26 remarkably alleviated mitochondria dysfunction, which was represented by an increase in the adenosine triphosphate (ATP) content and mitochondrial membrane potential, a decrease in mitochondrial DNA (mtDNA) damage. Finally, we revealed that Ac2-26 pretreatment could significantly inhibit the activation of IL-22/IL22R1/STAT3 signaling. In conclusion, this work demonstrated that Ac2-26 ameliorated HIRI by reducing oxidative stress and inhibiting the mitochondrial apoptosis pathway, which might be closely related to the inhibition of the IL-22/IL22R1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Wanzhen Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Hongxin Jiang
- Morphology Lab, Weifang Medical University, Weifang, Shandong, China
| | - Chen Bai
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Shuna Yu
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Yitong Pan
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Chenchen Wang
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Huiting Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Ming Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Yaxin Sheng
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Fangfang Chu
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Jie Wang
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Yuting Chen
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Jianguo Li
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| | - Jiying Jiang
- Department of Anatomy, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
8
|
Yu C, Chen P, Xu J, Wei S, Cao Q, Guo C, Wu X, Di G. Corneal Epithelium-Derived Netrin-1 Alleviates Dry Eye Disease via Regulating Dendritic Cell Activation. Invest Ophthalmol Vis Sci 2022; 63:1. [PMID: 35648640 PMCID: PMC9172049 DOI: 10.1167/iovs.63.6.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose To investigate the expression of corneal epithelium-derived netrin-1 (NTN-1) and its immunoregulatory function in dry eye disease (DED) using a DED mouse model. Methods We generated DED mouse models with desiccating stress under scopolamine treatment. RNA sequencing was performed to identify differentially expressed genes (DEGs) in the corneal epithelium of DED mice. NTN-1 expression was analyzed via real-time PCR, immunofluorescence staining, and immunoblotting. The DED mice were then treated with recombinant NTN-1 or neutralizing antibodies to investigate the severity of the disease, dendritic cell (DC) activation, and inflammatory cytokine expression. Results A total of 347 DEGs (292 upregulated and 55 downregulated) were identified in the corneal epithelium of DED mice: corneal epithelium-derived NTN-1 expression was significantly decreased in DED mice compared to that in control mice. Topical recombinant NTN-1 application alleviated the severity of the disease, accompanied by restoration of tear secretion and goblet cell density. In addition, NTN-1 decreased the number of DCs, inhibited the activation of the DCs and Th17 cells, and reduced the expression of inflammatory factors in DED mice. In contrast, blocking endogenous NTN-1 activity with an anti-NTN-1 antibody aggravated the disease, enhanced DC activation, and upregulated the inflammatory factors in the conjunctivae of DED mice. Conclusions We identified decreased NTN-1 expression in the corneal epithelium of DED mice. Our findings elucidate the role of NTN-1 in alleviating DED and impeding DC activation, thereby indicating its therapeutic potential in suppressing ocular inflammation in DED.
Collapse
Affiliation(s)
- Chaoqun Yu
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Peng Chen
- Department of Anthropotomy and Histo-Embryology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jing Xu
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Susu Wei
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qilong Cao
- Qingdao Haier Biotech Co. Ltd., Qingdao, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xianggen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Guohu Di
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Wgealla MMAMA, Liang H, Chen R, Xie Y, Li F, Qin M, Zhang X. Amniotic fluid derived stem cells promote skin regeneration and alleviate scar formation through exosomal miRNA-146a-5p via targeting CXCR4. J Cosmet Dermatol 2022; 21:5026-5036. [PMID: 35364624 DOI: 10.1111/jocd.14956] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/27/2022] [Accepted: 03/18/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Regenerative medicine is promising in wound healing. Exosomes derived from human amniotic fluid derived stem cells(hAFS) have become an important area of research for many diseases as a key paracrine factor,but its effects in wound healing remains unknown. In this study, we investigated the possible role and possible mechanisms of hAFS in skin wound healing. METHODS hAFS were isolated from human amniotic fluid via routine amniocentesis. The mice were randomly divided into 2 groups: control group and hAFS group treated with 1.25×106 hAFS cells. immunohistochemistry staining was performed for histological analysis and qRT-PCR for assessment of gene levels. Luciferase Reporter Assay was performed for verification of target gene. RESULTS Our results demonstrated that hAFS accelerated wound closure. hAFS alleviated scar formation via promoting ECM remodeling, upregulating molecular of immune response, enhancing anti-fibrotic activity and decreasing the secretion of inflammation-associated cytokines through exosomal miRNA-146a-5p via targeting CXCR4. CONCLUSIONS Taken together, hAFS was a promising cell source for wound healing. The findings in this study provide vital references and pave the way for future research.
Collapse
Affiliation(s)
- Mutwakil Mub Arak Mohammed Ali Wgealla
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China.,The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China
| | - Hansi Liang
- Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ruihua Chen
- The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China.,Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yufei Xie
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - Fang Li
- Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - Mingde Qin
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu Province, China.,The Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province (The State Key Laboratory Incubation Base), Soochow University, Suzhou, Jiangsu Province, China
| | - Xueguang Zhang
- Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
10
|
Shi Q, Zhao G, Wei S, Guo C, Wu X, Zhao RC, Di G. Pterostilbene alleviates liver ischemia/reperfusion injury via PINK1-mediated mitophagy. J Pharmacol Sci 2022; 148:19-30. [PMID: 34924126 DOI: 10.1016/j.jphs.2021.09.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury contributes to morbidity and mortality during liver resection or transplantation, with limited effective treatments available. Here, we investigated the potential benefits and underlying mechanisms of pterostilbene (Pt), a natural component of blueberries and grapes, in preventing hepatic I/R injury. Male C57BL/6 mice subjected to partial warm hepatic I/R and human hepatocyte cell line L02 cells exposed to anoxia/reoxygenation (A/R) were used as in vivo and in vitro models, respectively. Our findings showed that pretreatment with Pt ameliorated hepatic I/R injury by improving liver histology, decreasing hepatocyte apoptosis, and reducing plasma ALT and AST levels. Likewise, cell apoptosis, mitochondrial membrane dysfunction, and mitochondrial ROS overproduction in L02 cells triggered by the A/R challenge in vitro were reduced due to Pt administration. Mechanistically, Pt treatment efficiently enhanced mitophagy and upregulated PINK1, Parkin, and LC3B expression. Notably, the protective effect of Pt was largely abrogated after cells were transfected with PINK1 siRNA. Moreover, Pt pretreatment promoted hepatocyte proliferation and liver regeneration in the late phase of hepatic I/R. In conclusion, our findings provide evidence that Pt exerts hepatoprotective effects in hepatic I/R injury by upregulating PINK1-mediated mitophagy.
Collapse
Affiliation(s)
- Qiangqiang Shi
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Guangfen Zhao
- Department of Medicine, The Liaocheng Third People's Hospital, Liaocheng, China
| | - Susu Wei
- College of Basic Medicine, Qingdao University, Qingdao, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xianggen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | | | - Guohu Di
- College of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
11
|
Lopes AG, de Almeida GC, Miola MP, Teixeira RM, Pires FCBL, Miani RA, de Mattos LC, Brandão CC, Castiglioni L. Absence of significant genetic alterations in the VSX1, SOD1, TIMP3, and LOX genes in Brazilian patients with Keratoconus. Ophthalmic Genet 2021; 43:73-79. [PMID: 34802378 DOI: 10.1080/13816810.2021.1992785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE To identify inherited or acquired mutations in the VSX1, SOD1, TIMP3 and LOX genes from the combined analysis of corneal and blood samples from patients with Keratoconus. METHODS The casuistry was consisted of samples of peripheral blood and corneal epithelium from 35 unrelated patients with Keratoconus who were submitted to corneal crosslink treatment. Also, blood and corneal epithelium samples from 89 non-keratoconic patients were used to compose the control group. Ophthalmologic evaluations included a clinical examination, topography and tomography. DNA samples were extracted from peripheral blood and from corneal epithelium in both groups and all coding regions of the VSX1, SOD1, TIMP3 and LOX genes were amplified by polymerase chain reaction, denatured and subjected to polyacrylamide gel electrophoresis. Mutational screening was performed by single-strand conformation polymorphism and direct DNA sequencing. RESULTS No pathogenic variant was found in all coding regions of VSX1, SOD1, TIMP3 and LOX genes, we detected only few SNPs (single-nucleotide polymorphisms). Among the polymorphisms stand out three of them, corresponding to the synonymous exchange of amino acids: exon 3 of VSX1 Ala182Ala and exon 3 of TIMP3 His83His and Ser87Ser; in patients with Keratoconus and also in control subjects. All the polymorphisms were found in samples of corneal epithelium and corresponding blood. CONCLUSION There is absence of KC pathogenic related to mutations in the VSX1, SOD1, TIMP3 and LOX genes in the studied patients.
Collapse
Affiliation(s)
- Alessandro Garcia Lopes
- Biology Department, Instituto De Biociências, Letras E, Universidade Estadual Paulista "Júlio De Mesquita Filho", São José do Rio Preto, São Paulo, Brazil.,Immunogenetics Laboratory, Molecular Biology Department, Faculdade De Medicina De São José Do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | - Gildásio Castello de Almeida
- Ophthalmology Outpatient Clinic, Hospital De Base Da Fundação Faculdade Regional De Medicina (HB-, São José do Rio Preto, São Paulo, Brazil
| | - Marcos Paulo Miola
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade De Medicina De São José Do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | - Ronan Marques Teixeira
- Biology Department, Instituto De Biociências, Letras E, Universidade Estadual Paulista "Júlio De Mesquita Filho", São José do Rio Preto, São Paulo, Brazil
| | | | | | - Luiz Carlos de Mattos
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade De Medicina De São José Do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | - Cinara Cássia Brandão
- Immunogenetics Laboratory, Molecular Biology Department, Faculdade De Medicina De São José Do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | - Lilian Castiglioni
- Biology Department, Instituto De Biociências, Letras E, Universidade Estadual Paulista "Júlio De Mesquita Filho", São José do Rio Preto, São Paulo, Brazil.,Epidemiology and Collective Health, Faculdade De Medicina De São José Do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
12
|
Qin X, He L, Fan D, Liang W, Wang Q, Fang J. Targeting the resolution pathway of inflammation using Ac2-26 peptide-loaded PEGylated lipid nanoparticles for the remission of rheumatoid arthritis. Asian J Pharm Sci 2021; 16:483-493. [PMID: 34703497 PMCID: PMC8520054 DOI: 10.1016/j.ajps.2021.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/03/2021] [Accepted: 03/05/2021] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by joint inflammation and immune dysfunction. Although various therapeutic approaches have been utilized for the treatment of RA in clinical applications, the low responsiveness of RA patients and undesired systemic toxicity are still unresolved problems. Targeting the resolution pathway of inflammation with pro-resolving mediators would evoke the protective actions of patient for combating the inflammation. Ac2-26, a 25-amino acid peptide derived from Annexin A (a pro-resolving mediator), has shown good efficacy in the treatment of inflammatory disorders. However, the low bioavailability of Ac2-26 peptides hinders their efficacy in vivo. In this paper, we formed PEGylated lipid nanoparticles (LDNPs) by the co-assembly of l-ascorbyl palmitate (L-AP) and N-(carbonyl methoxypolyethylene glycol-2000)-1,2-distearoyl-sn‑glycero-3-phosphoethanolamine (DSPE-PEG2k) to encapsulate and deliver Ac2-26 peptides to the arthritic rats. They showed good stability and biocompatibility. After being intravenously administrated, Ac2-26 peptide-loaded PEGylated lipid nanoparticles (ADNPs) showed the prolonged in vivo circulation time and enhanced accumulation in inflamed sites. In vivo therapeutic evaluations revealed that ADNPs could attenuate synovial inflammation and improve joint pathology. Therefore, the pro-resolving therapeutic strategy using ADNPs is effective in RA treatment.
Collapse
Affiliation(s)
- Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Liming He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Donghao Fan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenlang Liang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiyu Fang
- Advanced Materials Processing and Analysis Center and Department of Materials Science and Engineering, University of Central Florida, Florida 32816, United State
| |
Collapse
|
13
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
14
|
Vital SA, Senchenkova EY, Ansari J, Gavins FNE. Targeting AnxA1/Formyl Peptide Receptor 2 Pathway Affords Protection against Pathological Thrombo-Inflammation. Cells 2020; 9:cells9112473. [PMID: 33202930 PMCID: PMC7697101 DOI: 10.3390/cells9112473] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is a leading cause of death and disability globally and is associated with a number of co-morbidities including sepsis and sickle cell disease (SCD). Despite thrombo-inflammation underlying these co-morbidities, its pathogenesis remains complicated and drug discovery programs aimed at reducing and resolving the detrimental effects remain a major therapeutic challenge. The objective of this study was to assess whether the anti-inflammatory pro-resolving protein Annexin A1 (AnxA1) was able to reduce inflammation-induced thrombosis and suppress platelet activation and thrombus formation in the cerebral microvasculature. Using two distinct models of pathological thrombo-inflammation (lipopolysaccharide (LPS) and sickle transgenic mice (STM)), thrombosis was induced in the murine brain using photoactivation (light/dye) coupled with intravital microscopy. The heightened inflammation-induced microvascular thrombosis present in these two distinct thrombo-inflammatory models was inhibited significantly by the administration of AnxA1 mimetic peptide AnxA1Ac2-26 (an effect more pronounced in the SCD model vs. the endotoxin model) and mediated by the key resolution receptor, Fpr2/ALX. Furthermore, AnxA1Ac2-26 treatment was able to hamper platelet aggregation by reducing platelet stimulation and aggregation (by moderating αIIbβ3 and P-selectin). These findings suggest that targeting the AnxA1/Fpr2/ALX pathway represents an attractive novel treatment strategy for resolving thrombo-inflammation, counteracting e.g., stroke in high-risk patient cohorts.
Collapse
Affiliation(s)
- Shantel A. Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
| | - Elena Y. Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
| | - Junaid Ansari
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
| | - Felicity N. E. Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA; (S.A.V.); (E.Y.S.); (J.A.)
- Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
- Correspondence: ; Tel.: +44-(0)-1895-267151
| |
Collapse
|
15
|
hADSCs derived extracellular vesicles inhibit NLRP3inflammasome activation and dry eye. Sci Rep 2020; 10:14521. [PMID: 32884023 PMCID: PMC7471690 DOI: 10.1038/s41598-020-71337-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 08/14/2020] [Indexed: 11/18/2022] Open
Abstract
The present study was set out to address the therapeutic efficacy of human adipose tissue stem cells derived extracellular vesicles (hADSC-Evs) in a mouse model of dry eye disease and to investigate the underlying mechanisms involved. hADSC-Evs eye drops were topically administered to mice that subjected to desiccating stress (DS). Clinical parameters of ocular surface damage were assessed with fluorescein staining, tear production and PAS staining. For in vitro studies, cell viability assay and TUNEL staining were performed in human corneal epithelial cells (HCECs) treated with hADSC-Evs under hyperosmotic media. In addition, immunofluorescent staining, Real-time PCR (qRT-PCR) and Western blots were used to evaluated NLRP3, ASC, caspase-1, and IL-1β expression levels. Compared with vehicle control mice, topical hADSC-Evs treated mice showed decreased corneal epithelial defects, increased tear production, decreased goblet cell loss, as well as reduced inflammatory cytokines production. In vitro, hADSC-Evs could protect HCECs against hyperosmotic stress-induced cell apoptosis. Mechanistically, hADSC-Evs treatment suppressed the DS induced rises in NLRP3 inflammasome formation, caspase-1 activation and IL-1β maturation. In conclusion, hADSC-Evs eye drops effectively suppress NLRP3 inflammatory response and alleviate ocular surface damage in dry eye disease.
Collapse
|