1
|
Nakhla MN, Patel R, Crowley E, Li Y, Peiris TB, Brocks D. Utilizing PROSE as a Drug Delivery Device for Preservative-Free Cyclosporine 0.05% for the Treatment of Dry Eye Disease: A Pilot Study. Clin Ophthalmol 2024; 18:3203-3213. [PMID: 39539800 PMCID: PMC11559178 DOI: 10.2147/opth.s487369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose To evaluate the tolerability of utilizing Prosthetic Replacement of the Ocular Surface Ecosystem (PROSE) as a drug delivery device for preservative-free cyclosporine 0.05% for the treatment of dry eye disease. Patients and Methods Fourteen current daily PROSE wearers were enrolled, with four screen failures and one subject that did not complete the study protocol due to burning and stinging. Nine subjects, 18 eyes completed the study protocol. All participants were instructed to instill one drop of preservative-free cyclosporine 0.05% in the PROSE reservoir and then fill the rest of the reservoir with preservative-free normal saline. After applying the PROSE and wearing for 6 hours, the PROSE was removed, and the protocol was repeated for at least another 4 additional hours of wear. Baseline, 1 week and 1 month symptom and sign data were collected. Results At one month, OSDI improved by an average of 3.83 ± 6.87 from baseline (p = 0.07) and there was no statistically significant change in best corrected visual acuity. Without comparing with placebo, there was statistically significant (p < 0.05) improvement in mean per subject and mean per eye corneal fluorescein staining, conjunctival lissamine staining, and conjunctival hyperemia by slit lamp examination at one-month follow-up. Conclusion Utilizing PROSE as a drug delivery system for non-preserved cyclosporine 0.05% was well tolerated in regard to both ocular symptoms and ocular surface signs. Results from this pilot study are suggestive of efficacy. The results of this study support progressing this protocol to a larger scale randomized controlled double blinded prospective clinical trial.
Collapse
Affiliation(s)
| | - Ria Patel
- Tufts University School of Medicine, Boston, MA, USA
| | | | - Yichen Li
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Thelge Buddika Peiris
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Daniel Brocks
- Department of Ophthalmology, BostonSight, Needham, MA, USA
| |
Collapse
|
2
|
Li Y, Chen A, Hong A, Xiong S, Chen X, Xie Q. Shark Cartilage-Derived Anti-Angiogenic Peptide Inhibits Corneal Neovascularization. Bioengineering (Basel) 2024; 11:693. [PMID: 39061775 PMCID: PMC11273382 DOI: 10.3390/bioengineering11070693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Corneal neovascularization is a significant cause of vision loss, often resulting in corneal clouding and chronic inflammation. Shark cartilage is widely recognized as a significant natural source of anti-angiogenic compounds. Our previous studies have shown that a polypeptide from white-spotted catshark (Chiloscyllium plagiosum Bonnet) has the potential to inhibit the angiogenesis of breast tumors. This study applied this peptide (SAIF) to a corneal alkali injury model to assess its effect on corneal neovascularization. Results revealed that SAIF inhibits endothelial cell proliferation, migration, and tube formation. SAIF inhibited VEGF-induced angiogenesis in the matrigel plug. Using the corneal alkali injury model, SAIF significantly inhibited corneal vascular neovascularization in mice. We found that SAIF not only significantly inhibited the upregulation of pro-angiogenic factors such as VEGF, bFGF, and PDGF expression induced by alkali injury, but also promoted the expression of anti-angiogenesis factor PEDF. Moreover, we also analyzed the MMPs and TIMPs involved in extracellular matrix (ECM) remodeling, angiogenesis, and lymphangiogenesis. We found that SAIF treatment inhibited the expression of pro-angiogenic factors like MMP1, MMP2, MMP3, MMP9, MMP13, and MMP14, and promoted the expression of anti-angiogenesis factors such as MMP7, TIMP1, TIMP2, and TIMP3. In conclusion, SAIF acts as an anti-angiogenic factor to inhibit the proliferation, migration, and tube formation of endothelial cells, inhibit pro-angiogenic factors, promote anti-angiogenic factors, and regulate the expression of MMPs, ultimately inhibiting corneal neovascularization.
Collapse
Affiliation(s)
- Yunxian Li
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (A.H.); (S.X.)
| | - Aoke Chen
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China;
- Guangdong Jida Engineering Research Center of Genetic Medicine Co., Ltd., Guangzhou 510535, China
| | - An Hong
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (A.H.); (S.X.)
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China;
| | - Sheng Xiong
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (A.H.); (S.X.)
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China;
| | - Xiaojia Chen
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (A.H.); (S.X.)
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China;
| | - Qiuling Xie
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China; (Y.L.); (A.H.); (S.X.)
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China;
| |
Collapse
|
3
|
Wu H, Ye J, Zhang M, Zhang L, Lin S, Li Q, Liu Y, Han Y, Huang C, Wu Y, Cheng Y, Cai S, Ke L, Liu G, Li W, Chu C. A SU6668 pure nanoparticle-based eyedrops: toward its high drug Accumulation and Long-time treatment for corneal neovascularization. J Nanobiotechnology 2024; 22:290. [PMID: 38802884 PMCID: PMC11129376 DOI: 10.1186/s12951-024-02510-8] [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: 10/31/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Corneal neovascularization (CNV) is one of the common blinding factors worldwide, leading to reduced vision or even blindness. However, current treatments such as surgical intervention and anti-VEGF agent therapy still have some shortcomings or evoke some adverse effects. Recently, SU6668, an inhibitor targeting angiogenic tyrosine kinases, has demonstrated growth inhibition of neovascularization. But the hydrophobicity and low ocular bioavailability limit its application in cornea. Hereby, we proposed the preparation of SU6668 pure nanoparticles (NanoSU6668; size ~135 nm) using a super-stable pure-nanomedicine formulation technology (SPFT), which possessed uniform particle size and excellent aqueous dispersion at 1 mg/mL. Furthermore, mesenchymal stem cell membrane vesicle (MSCm) was coated on the surface of NanoSU6668, and then conjugated with TAT cell penetrating peptide, preparing multifunctional TAT-MSCm@NanoSU6668 (T-MNS). The T-MNS at a concentration of 200 µg/mL was treated for CNV via eye drops, and accumulated in blood vessels with a high targeting performance, resulting in elimination of blood vessels and recovery of cornea transparency after 4 days of treatment. Meanwhile, drug safety test confirmed that T-MNS did not cause any damage to cornea, retina and other eye tissues. In conclusion, the T-MNS eye drop had the potential to treat CNV effectively and safely in a low dosing frequency, which broke new ground for CNV theranostics.
Collapse
Affiliation(s)
- Han Wu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Jinfa Ye
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Minjie Zhang
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Department of Rheumatology and Clinical Immunology, School of Medicine, the First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, XM, 361000, China
- Municipal Clinical Research Center for Immune Diseases, Xiamen, XM, 361000, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, XM, 361000, China
| | - Lingyu Zhang
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Sijie Lin
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Qingjian Li
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yanbo Liu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yun Han
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Caihong Huang
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yiming Wu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Yuhang Cheng
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Shundong Cai
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Lang Ke
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Physical Chemistry of Solid Surfaces, The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361002, China.
- Shen Zhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| | - Wei Li
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China.
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China.
| | - Chengchao Chu
- School of Medicine, Xiamen University Affiliated Xiamen Eye Center, Eye Institute of Xiamen University, Xiamen University, Xiamen, 361102, China.
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Xiamen, 361102, China.
- Shen Zhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
4
|
He J, Pham TL, Kakazu AH, Ponnath A, Do KV, Bazan HEP. Lipoxin A4 (LXA4) Reduces Alkali-Induced Corneal Inflammation and Neovascularization and Upregulates a Repair Transcriptome. Biomolecules 2023; 13:831. [PMID: 37238701 PMCID: PMC10216426 DOI: 10.3390/biom13050831] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
PURPOSE To investigate the anti-inflammatory and anti-angiogenic effects of the bioactive lipid mediator LXA4 on a rat model of severe corneal alkali injury. METHODS To induce a corneal alkali injury in the right eyes of anesthetized Sprague Dawley rats. They were injured with a Φ 4 mm filter paper disc soaked in 1 N NaOH placed on the center of the cornea. After injury, the rats were treated topically with LXA4 (65 ng/20 μL) or vehicle three times a day for 14 days. Corneal opacity, neovascularization (NV), and hyphema were recorded and evaluated in a blind manner. Pro-inflammatory cytokine expression and genes involved in cornel repair were assayed by RNA sequencing and capillary Western blot. Cornea cell infiltration and monocytes isolated from the blood were analyzed by immunofluorescence and by flow cytometry. RESULTS Topical treatment with LXA4 for two weeks significantly reduced corneal opacity, NV, and hyphema compared to the vehicle treatment. RNA-seq and Western blot results showed that LXA4 decreased the gene and protein expression of pro-inflammatory cytokines interleukin (IL)-1β and IL-6 and pro-angiogenic mediators matrix metalloproteinase (MMP)-9 and vascular endothelial growth factor (VEGFA). It also induces genes involved in keratinization and ErbB signaling and downregulates immune pathways to stimulate wound healing. Flow cytometry and immunohistochemistry showed significantly less infiltration of neutrophils in the corneas treated with LXA4 compared to vehicle treatment. It also revealed that LXA4 treatment increases the proportion of type 2 macrophages (M2) compared to M1 in blood-isolated monocytes. CONCLUSIONS LXA4 decreases corneal inflammation and NV induced by a strong alkali burn. Its mechanism of action includes inhibition of inflammatory leukocyte infiltration, reduction in cytokine release, suppression of angiogenic factors, and promotion of corneal repair gene expression and macrophage polarization in blood from alkali burn corneas. LXA4 has potential as a therapeutic candidate for severe corneal chemical injuries.
Collapse
Affiliation(s)
- Jiucheng He
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (J.H.)
- Department of Ophthalmology, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA
| | - Thang L. Pham
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (J.H.)
- HENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, Hanoi 11313, Vietnam
| | - Azucena H. Kakazu
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (J.H.)
| | - Abhilash Ponnath
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (J.H.)
| | - Khanh V. Do
- Faculty of Medicine, PHENIKAA University, Hanoi 12116, Vietnam
| | - Haydee E. P. Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA; (J.H.)
- Department of Ophthalmology, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA 70112, USA
| |
Collapse
|
5
|
Abdi B, Mofidfar M, Hassanpour F, Kirbas Cilingir E, Kalajahi SK, Milani PH, Ghanbarzadeh M, Fadel D, Barnett M, Ta CN, Leblanc RM, Chauhan A, Abbasi F. Therapeutic contact lenses for the treatment of corneal and ocular surface diseases: advances in extended and targeted drug delivery. Int J Pharm 2023; 638:122740. [PMID: 36804524 DOI: 10.1016/j.ijpharm.2023.122740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 02/21/2023]
Abstract
The eye is one of the most important organs in the human body providing critical information on the environment. Many corneal diseases can lead to vision loss affecting the lives of people around the world. Ophthalmic drug delivery has always been a major challenge in the medical sciences. Since traditional methods are less efficient (∼ 5%) at delivering drugs to ocular tissues, contact lenses have generated growing interest in ocular drug delivery due to their potential to enhance drug bioavailability in ocular tissues. The main techniques used to achieve sustained release are discussed in this review, including soaking in drug solutions, incorporating drug into multilayered contact lenses, use of vitamin E barriers, molecular imprinting, nanoparticles, micelles and liposomes. The most clinically relevant results on different eye pathologies are presented. In addition, this review summarizes the benefits of contact lenses over eye drops, strategies for incorporating drugs into lenses to achieve sustained release, results of in vitro and in vivo studies, and the recent advances in the commercialization of therapeutic contact lenses for allergic conjunctivitis.
Collapse
Affiliation(s)
- Behnam Abdi
- Institute of Polymeric Materials (IPM), Sahand University of Technology, New Town of Sahand, Tabriz, Iran; Faculty of Polymer Engineering, Sahand University of Technology, New Town of Sahand, Tabriz, Iran
| | - Mohammad Mofidfar
- Department of Chemistry, Stanford University, Stanford, CA, USA; School of Medicine, Stanford University, Stanford, CA, USA
| | - Fatemeh Hassanpour
- Institute of Polymeric Materials (IPM), Sahand University of Technology, New Town of Sahand, Tabriz, Iran; Faculty of Polymer Engineering, Sahand University of Technology, New Town of Sahand, Tabriz, Iran
| | | | - Sepideh K Kalajahi
- Institute of Polymeric Materials (IPM), Sahand University of Technology, New Town of Sahand, Tabriz, Iran; Faculty of Polymer Engineering, Sahand University of Technology, New Town of Sahand, Tabriz, Iran
| | - Paria H Milani
- Institute of Polymeric Materials (IPM), Sahand University of Technology, New Town of Sahand, Tabriz, Iran; Faculty of Polymer Engineering, Sahand University of Technology, New Town of Sahand, Tabriz, Iran
| | - Mahsa Ghanbarzadeh
- Institute of Polymeric Materials (IPM), Sahand University of Technology, New Town of Sahand, Tabriz, Iran; Faculty of Polymer Engineering, Sahand University of Technology, New Town of Sahand, Tabriz, Iran
| | - Daddi Fadel
- Center for Ocular Research & Education (CORE), School of Optometry & Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Melissa Barnett
- University of California, Davis Eye Center, Sacramento, CA, USA
| | - Christopher N Ta
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, Coral Gables, FL, USA.
| | - Anuj Chauhan
- Chemical and Biological Engineering Department, Colorado School of Mines, CO, USA.
| | - Farhang Abbasi
- Institute of Polymeric Materials (IPM), Sahand University of Technology, New Town of Sahand, Tabriz, Iran; Faculty of Polymer Engineering, Sahand University of Technology, New Town of Sahand, Tabriz, Iran.
| |
Collapse
|
6
|
Impaired Autophagy Causes Severe Corneal Neovascularization. Cells 2022; 11:cells11233895. [PMID: 36497153 PMCID: PMC9737787 DOI: 10.3390/cells11233895] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
PURPOSE To investigate the role of macrophage autophagy in the process of corneal neovascularization (CNV). METHODS In vivo, mice CNV was induced by alkali injury and compared with rapamycin-treated alkaline burn mice. Western blot was used to determine the autophagic status of the macrophages. We quantified the levels of macrophage polarization markers (CD86, INOS, CD163, CD206) by RT-qPCR and measured inflammatory factors through ELISA (IL-6 and TNF-α) in the early phase after injury. In vitro, the human umbilical vein endothelial cells (HUVECs) were co-cultured with macrophage-conditioned medium (MCM) induced by the THP-1 cell line to simulate the neovascular microenvironment. The vascularization capacity of HUVECs was examined using the CCK-8 assay kit, tube formation assay, and scratch wound-healing assay. RESULTS In vivo, the mRNA expression of Beclin-1 and ATG5 was increased, together with the upregulation of M1 macrophage markers (CD86 and INOS) in corneas after early alkali injury. The area of CNV is effectively relieved in the rapamycin-treated mice. In vitro, upregulation of autophagy level by pretreatment with 3-methyladenine (3-MA) could increase the mRNA expression of the M1 markers. Macrophage-conditioned medium with impaired autophagy contains more IL-6 and TNF-α compared to the M1 macrophage-conditioned medium, promoting HUVEC proliferation, migration, and tube formation capacity. Enhancing the autophagy level with rapamycin (RAPA) could reverse this phenomenon. CONCLUSIONS Impaired autophagy promoted macrophage polarization toward M1 type and increased the expression of IL-6 and TNF-α, which led to severe CNV. Using the autophagy activator (RAPA) could effectively alleviate CNV by promoting autophagy.
Collapse
|
7
|
Cui Y, Huo Y, Li Z, Qiu Y, Yang Q, Chen Z, Fan S, Huang X, Hao J, Kang L, Liang G. VEGF-targeted scFv inhibits corneal neovascularization via STAT3 pathway in alkali burn model. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.130764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
8
|
Li S, Shi S, Xia F, Luo B, Ha Y, Luisi J, Gupta PK, Merkley KH, Motamedi M, Liu H, Zhang W. CXCR3 deletion aggravates corneal neovascularization in a corneal alkali-burn model. Exp Eye Res 2022; 225:109265. [PMID: 36206861 PMCID: PMC10191246 DOI: 10.1016/j.exer.2022.109265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/25/2022] [Accepted: 09/21/2022] [Indexed: 01/16/2023]
Abstract
Corneal neovascularization can cause devastating consequences including vision impairment and even blindness. Corneal inflammation is a crucial factor for the induction of corneal neovascularization. Current anti-inflammatory approaches are of limited value with poor therapeutic effects. Therefore, there is an urgent need to develop new therapies that specifically modulate inflammatory pathways and inhibit neovascularization in the cornea. The interaction of chemokines and their receptors plays a key role in regulating leukocyte migration during inflammatory response. CXCR3 is essential for mediating the recruitment of activated T cells and microglia/macrophages, but the role of CXCR3 in the initiation and promotion of corneal neovascularization remains unclear. Here, we showed that the expression of CXCL10 and CXCR3 was significantly increased in the cornea after alkali burn. Compared with WT mice, CXCR3-/- mice exhibited significantly increased corneal hemangiogenesis and lymphangiogenesis after alkali burn. In addition, exaggerated leukocyte infiltration and leukostasis, and elevated expression of inflammatory cytokines and angiogenic factor were also found in the corneas of CXCR3-/- mice subjected to alkali burn. With bone marrow (BM) transplantation, we further demonstrated that the deletion of CXCR3 in BM-derived leukocytes plays a key role in the acceleration of alkali burn-induced corneal neovascularization. Taken together, our results suggest that upregulation of CXCR3 does not exhibit its conventional action as a proinflammatory cytokine but instead serves as a self-protective mechanism for the modulation of inflammation and maintenance of corneal avascularity after corneal alkali burn.
Collapse
Affiliation(s)
- Shengguo Li
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Shuizhen Shi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Fan Xia
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Ban Luo
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Yonju Ha
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan Luisi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Praveena K Gupta
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Kevin H Merkley
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Massoud Motamedi
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Hua Liu
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA.
| | - Wenbo Zhang
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neuroscience, Cell Biology & Anatomy, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
9
|
Yang L, Guo J, He J, Shao J. Skin grafting treatment of adolescent lower limb avulsion injury. Front Surg 2022; 9:953038. [PMID: 36189402 PMCID: PMC9521200 DOI: 10.3389/fsurg.2022.953038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Background Under the influence of various factors, the number of lower extremity avulsion injuries in adolescents is increasing year by year. The main modality of treatment is skin grafting. There are many types of skin grafting. Although many studies on skin grafting after avulsion injuries have been published in the past few decades, there are differences in the treatment options for adolescents with post avulsion injuries. Main body Thorough debridement and appropriate skin grafts are essential for the surgical management of avulsion injuries for optimal prognosis. In the acquisition of grafts, progress has been made in equipment for how to obtain different depths of skin. The severity of the avulsion injury varies among patients on admission, and therefore the manner and type of skin grafting will vary. Especially in adolescents, graft survival and functional recovery are of great concern to both patients and physicians. Therefore, many efforts have been made to improve survival rate and activity. Conclusion This review summarizes the principles of treatment of avulsion injuries, the historical development of skin grafts, and the selection of skin grafts, hoping to be helpful for future research.
Collapse
|
10
|
Wang P, Hao P, Chen X, Li L, Zhou Y, Zhang X, Zhu L, Ying M, Han R, Wang L, Li X. Targeting HMGB1-NFκb Axis and miR-21 by Glycyrrhizin: Role in Amelioration of Corneal Injury in a Mouse Model of Alkali Burn. Front Pharmacol 2022; 13:841267. [PMID: 35586052 PMCID: PMC9108160 DOI: 10.3389/fphar.2022.841267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Corneal neovascularization (CNV) is a sight-threatening condition usually associated with various inflammatory settings including chemical injury. High mobility group box 1 (HMGB1) is identified as an inflammatory alarmin in diverse tissue damage. Here, we evaluate the expression of HMGB1 and the consequences of its inhibition through its selective inhibitor glycyrrhizin (GLY) in alkali burn-induced corneal inflammation and neovascularization. GLY effectively attenuated alkali burn-induced HMGB1 expression at both mRNA and protein levels. Furthermore, slit-lamp analysis, ink perfusion, H&E staining, and CD31 histochemical staining showed that GLY relieved corneal neovascularization, while GLY attenuated VEGF expression via inhibiting HMGB1/NF-κB/HIF-1α signal pathway. In addition, GLY treatment decreased the cytokine expression of CCL2 and CXCL5, accompanied by the reduction of their receptors of CCR2 and CXCR2. GLY diminished the inflammatory cell infiltration of the cornea, as well as reduced the expression of IL-1β, IL-6, and TNF-α. Moreover, treatment with GLY reduced the degree of cornea opacity through inactivating extracellular HMGB1 function, which otherwise induces TGF-β1 release and myofibroblast differentiation. Furthermore, we found that GLY treatment attenuated the upregulation of miR-21 levels in alkali burned cornea; while inhibition of miR-21in keratocytes in vitro, significantly inhibited TGF-β1-induced myofibroblast differentiation. Collectively, our results suggested that targeting HMGB1-NFκb axis and miR-21 by GLY could introduce a therapeutic approach to counter CNV.
Collapse
Affiliation(s)
- Peihong Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Peng Hao
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
- Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Xi Chen
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
- Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Linghan Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Yongying Zhou
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Xiaohan Zhang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Lin Zhu
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Ming Ying
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
- Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Ruifang Han
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
- Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Liming Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
- Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Xuan Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
- Nankai University Affiliated Eye Hospital, Tianjin, China
- *Correspondence: Xuan Li,
| |
Collapse
|
11
|
Corneal stromal repair and regeneration. Prog Retin Eye Res 2022; 91:101090. [DOI: 10.1016/j.preteyeres.2022.101090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/02/2023]
|
12
|
Therapeutic Applications of Adeno-Associated Virus (AAV) Gene Transfer of HLA-G in the Eye. Int J Mol Sci 2022; 23:ijms23073465. [PMID: 35408825 PMCID: PMC8998501 DOI: 10.3390/ijms23073465] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 01/09/2023] Open
Abstract
The purpose of this paper is to review human leukocyte antigen G (HLA-G) in the eye, its role in immune tolerance, and the potential therapeutic use of AAV gene transfer and expression of HLA-G in various ocular tissues. Several studies are reviewed that demonstrate efficacy in animal models of disease, including intracorneal delivery of AAV-HLA-G to treat corneal inflammation and prevent corneal graft rejection, subconjunctival injection of AAV-HLA-G for ocular graft vs. host disease and potentially dry eye disease, and intravitreal injection of AAV-HLA-G to inhibit uveitis. Furthermore, due to the anti-vascular function of HLA-G, AAV-HLA-G may be an effective therapy for posterior ocular diseases, such as neovascular age-related macular degeneration, diabetic retinopathy, and choroidal neovascularization. Therefore, AAV-mediated gene transfer of HLA-G may be an effective treatment for common immune-mediated, inflammatory, and neovascular diseases of the eye.
Collapse
|
13
|
Daphnetin inhibits corneal inflammation and neovascularization on a mouse model of corneal alkali burn. Int Immunopharmacol 2021; 103:108434. [PMID: 34920334 DOI: 10.1016/j.intimp.2021.108434] [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: 09/26/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/21/2022]
Abstract
Alkali burn is a significant contributor to corneal injury. Alkali burn-induced corneal inflammation often causes vision loss due to corneal neovascularization. Daphnetin (DAP) has been studied for its anti-inflammatory and antiangiogenic properties with encouraging results. Driven by those encouraging results, we sought to explore the effects of DAP in treating alkali burn-induced corneal inflammation and neovascularization and its mechanism of action. We found that the angiogenesis processes of human umbilical vein endothelial cells (HUVECs) induced by vascular endothelial growth factor A (VEGF-A) were primarily attenuated by treatment with DAP, including proliferation, migration, and tube formation. Treatment of DAP significantly suppressed the VEGF-A-induced protein expression of VEGF receptor2 (VEGFR2), as well as the activation of downstream signal transducer and activator of transcription 3 (STAT3), AKT, and extracellular signal-regulated kinase (ERK) signaling. In the mouse corneal alkali burn model, the inflammatory cell infiltrations and neovascularization in the cornea caused by alkali burn were inhibited by 10 µM DAP eye drops. Alkali burn-induced corneal protein expression of VEGF-A, VEGFR2, phosphorylated (p-)STAT3, p-AKT, and p-ERK in corneal tissue were reduced mainly by DAP. Moreover, the upregulation of inflammatory caused by alkali burn in the pathological process was significantly neutralized by DAP. Mechanistically, the inflammatory response could be alleviated by DAP in the way of inhibiting the expression levels of TLR4, p-NF-κB, NLRP3, ASC, Cleaved-caspase-1 (p20), mature-IL-1β (p17), and N-GSDM. In conclusion, our findings confirmed that the corneal inflammation and neovascularization caused by alkali burn could be inhibited by DAP in vitro and in vivo, elucidating the underlying mechanisms of its protective effects. DAP may have tremendous therapeutic potential for the treatment of corneal alkali burn.
Collapse
|
14
|
Wakamatsu TH, Dos Santos MS, Barreiro TP, Sant'Anna AEBPP, Murta F, da Costa AX, Marculino LGC, de Alcântara RJA, de Farias CC, Gomes JÁP. Clinical Aspects of Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis With Severe Ocular Complications in Brazil. Front Med (Lausanne) 2021; 8:649369. [PMID: 34222274 PMCID: PMC8252916 DOI: 10.3389/fmed.2021.649369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/19/2021] [Indexed: 12/01/2022] Open
Abstract
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are acute and potentially fatal inflammatory vesiculobullous reactions that affect the skin and mucous membranes, and which are most often triggered by particular medications and infections. In Brazil, the drugs most frequently associated with TEN and SJS include cold medicine such as dipyrone and NSAIDs, followed by carbamazepine, phenobarbital, penicillin, and allopurinol. Genetic variations have been found to increase the risk of SJS/TEN in response to triggering factors such as medications. The most closely associated genes found in Brazilian cold-medicine-related SJS/TEN patients with severe ocular complications are HLA-A*66:01 in those of mixed African and European ancestry and HLA-B*44:03 and HLA-C*12:03 in those of solely European ancestry. Our classification system for grading ocular surface complication severity in SJS/TEN patients revealed the most severe complications to be limbal stem cell deficiency and dry eye. Changes to the conjunctival flora have also been observed in SJS/TEN patients. Our group identified bacterial colonization in 95% of the eyes (55.5% of which were gram-positive cocci, 25.5% of which were gram-negative bacilli, and 19% of which were gram-positive bacilli). Several new treatment options in the acute and chronic ocular management of the SJS/TEN patients have been described. This article highlights some Brazilian institutions' contributions to ocular surface care in both the acute phase (including the use of amniotic membrane transplantation) and the chronic phase (such as eyelid margin and fornix reconstruction, minor salivary gland transplantation, amniotic membrane and limbal transplantation, scleral contact lenses, anti-angiogenic eyedrops for corneal neovascularization, ex-vivo cultivated limbal epithelium transplantation, conjunctival-limbal autografting, oral mucosa transplantation, and keratoprosthesis).
Collapse
Affiliation(s)
| | | | | | | | - Fabíola Murta
- Department of Ophthalmology, Federal University of São Paulo, São Paulo, Brazil.,Moorfields Eye Hospital, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
15
|
Lužnik Z, Anchouche S, Dana R, Yin J. Regulatory T Cells in Angiogenesis. THE JOURNAL OF IMMUNOLOGY 2021; 205:2557-2565. [PMID: 33168598 DOI: 10.4049/jimmunol.2000574] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/06/2020] [Indexed: 12/20/2022]
Abstract
Regulatory T cells (Tregs) are crucial mediators of immune homeostasis. They regulate immune response by suppressing inflammation and promoting self-tolerance. In addition to their immunoregulatory role, a growing body of evidence highlights the dynamic role of Tregs in angiogenesis, the process of forming new blood vessels. Although angiogenesis is critically important for normal tissue regeneration, it is also a hallmark of pathological processes, including malignancy and chronic inflammation. Interestingly, the role of Tregs in angiogenesis has been shown to be highly tissue- and context-specific and as a result can yield either pro- or antiangiogenic effects. For these reasons, there is considerable interest in determining the molecular underpinnings of Treg-mediated modulation of angiogenesis in different disease states. The present review summarizes the role of Tregs in angiogenesis and mechanisms by which Tregs regulate angiogenesis and discusses how these mechanisms differ in homeostatic and pathological settings.
Collapse
Affiliation(s)
- Zala Lužnik
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114.,Eye Hospital, University Medical Centre, 1000 Ljubljana, Slovenia; and
| | - Sonia Anchouche
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114.,Faculty of Medicine, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Reza Dana
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114;
| | - Jia Yin
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114;
| |
Collapse
|
16
|
Pishavar E, Luo H, Bolander J, Atala A, Ramakrishna S. Nanocarriers, Progenitor Cells, Combinational Approaches, and New Insights on the Retinal Therapy. Int J Mol Sci 2021; 22:1776. [PMID: 33579019 PMCID: PMC7916765 DOI: 10.3390/ijms22041776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Progenitor cells derived from the retinal pigment epithelium (RPECs) have shown promise as therapeutic approaches to degenerative retinal disorders including diabetic retinopathy, age-related macular degeneration and Stargardt disease. However, the degeneration of Bruch's membrane (BM), the natural substrate for the RPE, has been identified as one of the major limitations for utilizing RPECs. This degeneration leads to decreased support, survival and integration of the transplanted RPECs. It has been proposed that the generation of organized structures of nanofibers, in an attempt to mimic the natural retinal extracellular matrix (ECM) and its unique characteristics, could be utilized to overcome these limitations. Furthermore, nanoparticles could be incorporated to provide a platform for improved drug delivery and sustained release of molecules over several months to years. In addition, the incorporation of tissue-specific genes and stem cells into the nanostructures increased the stability and enhanced transfection efficiency of gene/drug to the posterior segment of the eye. This review discusses available drug delivery systems and combination therapies together with challenges associated with each approach. As the last step, we discuss the application of nanofibrous scaffolds for the implantation of RPE progenitor cells with the aim to enhance cell adhesion and support a functionally polarized RPE monolayer.
Collapse
Affiliation(s)
- Elham Pishavar
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 91735, Iran;
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Hongrong Luo
- Engineering Research Center in Biomaterials, Sichuan University, Chengdu 610064, China;
| | - Johanna Bolander
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Antony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology, National University of Singapore, Singapore 117581, Singapore
| |
Collapse
|
17
|
Huang Y, Pan L, Wu T. Improvement of cerebral ischemia-reperfusion injury by L-3-n-butylphthalide through promoting angiogenesis. Exp Brain Res 2020; 239:341-350. [PMID: 33180154 DOI: 10.1007/s00221-020-05978-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022]
Abstract
Cerebral ischemia/reperfusion (I/R) injury may lead to a poor prognosis for ischemic stroke patients after reperfusion therapy, and currently, lacks effective therapeutic intervention. This study aimed to investigate the effects of L-3-n-butylphthalide (L-NBP) on cerebral I/R injury in rats. Rat models of cerebral I/R injury were established using the middle cerebral artery occlusion/refusion (MACO/R) surgery and were administrated intragastrically with L-NBP or vehicle. We found that L-NBP attenuated the histological damages and reduced the brain hematoma in MACO/R rats. L-NBP also significantly improved the neurological function, alleviated the brain edema, and reduced the permeability of blood-brain barrier of MACO/R rats. Moreover, we detected that L-NBP considerably facilitated microvessel formation in the lesion area of brain in MACO/R rats. Finally, we found that L-NBP significantly increased the protein and mRNA expression levels of Nrf2, HIF-1α, and VEGF in the brain of MACO/R rats. In conclusion, our results demonstrated that L-NBP exerted significant beneficial effects on cerebral I/R injury in rats through promoting angiogenesis, which may be associated with the activation of Nrf2/HIF-1α/VEGF signaling pathway. Our results suggested that L-NBP could be a potential therapeutic drug for cerebral I/R injury.
Collapse
Affiliation(s)
- Ying Huang
- Department of Pharmacy, The People's Hospital of Yichun City, Yichun, 336000, People's Republic of China
| | - Lishou Pan
- Department of Neurology, The People's Hospital of Yichun City, Yichun, 336000, People's Republic of China
| | - Ting Wu
- Department of Pharmacy, The People's Hospital of Yichun City, Yichun, 336000, People's Republic of China.
| |
Collapse
|
18
|
Zhao M, Zhang H, Zhen D, Huang M, Li W, Li Z, Liu Y, Xie Y, Zeng B, Wang Z, Huang B. Corneal Recovery Following Rabbit Peripheral Blood Mononuclear Cell-Amniotic Membrane Transplantation with Antivascular Endothelial Growth Factor in Limbal Stem Cell Deficiency Rabbits. Tissue Eng Part C Methods 2020; 26:541-552. [PMID: 33019886 DOI: 10.1089/ten.tec.2020.0209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Limbal stem cell deficiency (LSCD) is a refractory ocular surface disorder characterized by progressive corneal epithelial degeneration, conjunctivalization, and neovascularization, potentially leading to blindness. There are currently no effective therapeutic options for patients experiencing routine symptomatic treatment failure. Transplantation of amniotic membrane (AM) with adherent stem cells (but not bare AM transplantation alone) has shown promise in preclinical studies for ocular surface restoration. A major limitation, however, is finding a reliable stem cell source. Stem cells can be isolated from the peripheral blood mononuclear cell (PBMC) population, and these PBMC-derived stem cells have numerous advantages over allogeneic and other autologous stem cell types for therapeutic application, including relative ease of acquisition, nonimmunogenicity, and the absence of ethical issues associated with embryonic stem cells. Experiment: We examined the efficacy of autologous PBMC-AM sheet cultures combined with postoperative antiangiogenesis treatment for corneal restoration in LSCD model rabbits. Rabbit PBMCs (rPBMCs) were isolated, labeled with EdU for in vivo tracing, and then cultured on AMs in conditioned medium before transplantation. Rabbits were transplanted with bare AMs (group 1), rPBMC-AM sheets (group 2), or rPBMC-AM sheets plus postoperative treatment with the vascular endothelial growth factor antagonist bevacizumab (group 3). Corneal opacity and neovascularization were monitored by slit-lamp imaging for 8 weeks and corneas were examined histologically at 1 and 2 months. Results: Corneal opacity decreased in all three groups over 8 weeks, but was significantly lower in group 2 and even lower in group 3. Corneal neovascularization was significantly higher in group 1 throughout the observation period, and significantly lower in group 3 than group 1 and 2 by 8 weeks post-transplant. At 4 weeks, the corneal surface completed epithelialization (although thinner than normal) in group 3 but still patchy in groups 1 and 2. By 8 weeks, the epithelium in group 3 was complete and smooth, resembling a normal epithelium. Integrin β1 as a progenitor marker was also generally higher in groups 2 and 3. Conclusions: Autologous rPBMC-AM sheets with post-transplant topical bevacizumab can effectively facilitate corneal epithelium recovery in a LSCD model, suggesting clinical utility for LSCD-related ocular surface diseases. Impact statement Limbal stem cell deficiency (LSCD) increases corneal opacity and vascularization, resulting in severe visual impairment or even blindness. Traditional surgical limbal transplant is currently the main treatment option for LSCD, but carries the risks of rejection and immunosuppressant side effects. Autologous stem cell-based therapy is a promising alternative approach, but a reliable stem cell source is a major limitation. We report that transplantation of autologous rabbit peripheral blood mononuclear cell-amniotic membrane sheets plus antivascular endothelial growth factor restored avascular transparent cornea in a rabbit LSCD model. These results demonstrate a potentially effective approach for ocular surface reconstruction in bilateral LSCD.
Collapse
Affiliation(s)
- Minglei Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Hening Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Dongqin Zhen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | | | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Zhiquan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Ying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Yaojue Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Baozhu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Zhichong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| | - Bing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, GuangZhou, China
| |
Collapse
|
19
|
Giannaccare G, Pellegrini M, Bovone C, Spena R, Senni C, Scorcia V, Busin M. Anti-VEGF Treatment in Corneal Diseases. Curr Drug Targets 2020; 21:1159-1180. [PMID: 32189591 DOI: 10.2174/1389450121666200319111710] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/31/2019] [Accepted: 01/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Corneal neovascularization (CN) is a clue feature of different ocular pathological conditions and can lead to corneal edema and opacification with subsequent vision loss. Vascular endothelial growth factor (VEGF), which plays a key role in new vessels formation, proliferation and migration, was found to be up-regulated in these conditions. Nowadays, it is possible to downregulate the angiogenic process by using anti-VEGF agents administered by different routes. OBJECTIVE To evaluate the efficacy, safety and possible future directions of anti-VEGF agents used for the treatment of CNV owing to different aetiologies. METHODS A computerized search of articles dealing with the topic of anti-VEGF therapy in CN was conducted in PubMed, Scopus and Medline electronic databases. The following key phrases were used: anti-VEGF agents, corneal neovascularization, bevacizumab, ranibizumab, vascular endothelial growth factor, angiogenesis. RESULTS The use of anti-VEGF therapy in the treatment of CN reduced pathological vessel density without causing significant side effects. Various administration routes such as topical, subconjunctival and intrastromal ones are available, and the choice depends on patient and disease characteristics. Much more effectiveness is achieved in case of early administration before mature and wellestablished vessels take place. A combined approach between various drugs including anti-VEGF agents should be adopted in those cases at higher risk of neovascularization recurrence such as chronic long-standing diseases where ischemic and inflammatory stimuli are not definitively reversed. CONCLUSION The efficacy and safety of anti-VEGF agents support their adoption into the daily clinical practice for the management of CN.
Collapse
Affiliation(s)
- Giuseppe Giannaccare
- Department of Ophthalmology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | | | - Cristina Bovone
- Department of Ophthalmology, Ospedale Privato "Villa Igea", Forli, Italy
| | - Rossella Spena
- Department of Ophthalmology, Ospedale Privato "Villa Igea", Forli, Italy
| | - Carlotta Senni
- Ophthalmology Unit, University of Bologna, Bologna, Italy
| | - Vincenzo Scorcia
- Department of Ophthalmology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Massimo Busin
- Department of Ophthalmology, Ospedale Privato "Villa Igea", Forli, Italy
| |
Collapse
|
20
|
Shang Q, Chu Y, Li Y, Han Y, Yu D, Liu R, Zheng Z, Song L, Fang J, Li X, Cao L, Gong Z, Zhang L, Chen Y, Wang Y, Shao C, Shi Y. Adipose-derived mesenchymal stromal cells promote corneal wound healing by accelerating the clearance of neutrophils in cornea. Cell Death Dis 2020; 11:707. [PMID: 32848141 PMCID: PMC7450061 DOI: 10.1038/s41419-020-02914-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
The dome-shaped cornea is a transparent, non-vascularized, and epithelialized highly organized tissue. Physical and chemical injuries may trigger corneal wound healing (CWH) response and result in neovascularization that impairs the visual function. CWH involves not only migration, proliferation, and differentiation of the cells in different layers of cornea, but also the mobilization of immune cells. We demonstrated here that human adipose-derived mesenchymal stromal cells (ADSCs) could effectively inhibit neovascularization during ethanol-induced injury in mouse cornea. Importantly, we found that while neutrophils are essential for CWH, excessive and prolonged neutrophil retention during the granulation stage contributes to neovascularization. ADSCs were found to promote the clearance of neutrophils in the cornea during the granulation stage, likely via increasing the reverse transendothelial cell migration of CXCR4high neutrophils from cornea to the lung. Our results demonstrate that ADSCs are effective in treating CWH-induced neovascularization and modulation of neutrophil clearance could be novel strategies for better vision recovery after injury.
Collapse
Affiliation(s)
- Qianwen Shang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Yunpeng Chu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Yanan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Yuyi Han
- Department of Ophthalmology, The Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214062, China
| | - Daojiang Yu
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Zhiyuan Zheng
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Lin Song
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Xiaolei Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Lijuan Cao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Zheng Gong
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Liying Zhang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Yongjing Chen
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China
| | - Ying Wang
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, 200025, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China.
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou, Jiangsu, 215123, China.
- Key Laboratory of Stem Cell Biology, Shanghai Jiao Tong University School of Medicine and Shanghai Institutes for Biological Science, Chinese Academy of Sciences, Shanghai, 200025, China.
| |
Collapse
|
21
|
Sunitinib malate-loaded biodegradable microspheres for the prevention of corneal neovascularization in rats. J Control Release 2020; 327:456-466. [PMID: 32822742 DOI: 10.1016/j.jconrel.2020.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 01/10/2023]
Abstract
Corneal neovascularization (NV) predisposes patients to compromised corneal transparency and visional acuity. Sunitinib malate (Sunb-malate) targeting against multiple receptor tyrosine kinases, exerts potent antiangiogenesis. However, the rapid clearance of Sunb-malate eye drops administered through topical instillation limits its therapeutic efficacy and poses a challenge for potential patient compliance. Sunb-malate, the water-soluble form of sunitinib, was shown to have higher intraocular penetration through transscleral diffusion following subconjunctival (SCT) injection in comparison to its sunitinib free base formulation. However, it is difficult to load highly water-soluble drugs and achieve sustained drug release. We developed Sunb-malate loaded poly(D,L-lactic-co-glycolic acid) (PLGA) microspheres (Sunb-malate MS) with a particle size of approximately 15 μm and a drug loading of 7 wt%. Sunb-malate MS sustained the drug release for 30 days under the in vitro infinite sink condition. Subconjunctival (SCT) injection of Sunb-malate MS provided a prolonged ocular drug retention and did not cause ocular toxicity at a dose of 150 μg of active agent. Sunb-malate MS following SCT injection more effectively suppressed the suture-induced corneal NV than either Sunb-malate free drug or the placebo MS. Local sustained release of Sunb-malate through the SCT injection of Sunb-malate MS mitigated the proliferation of vascular endothelial cells and the recruitment of mural cells into the cornea. Moreover, the gene upregulation of proangiogenic factors induced by the pathological process was greatly neutralized by SCT injection of Sunb-malate MS. Our findings provide a sustained release platform for local delivery of tyrosine kinase inhibitors to treat corneal NV.
Collapse
|
22
|
Jabbehdari S, Memar OM, Caughlin B, Djalilian AR. Update on the pathogenesis and management of ocular rosacea: an interdisciplinary review. Eur J Ophthalmol 2020; 31:22-33. [PMID: 32586107 DOI: 10.1177/1120672120937252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Rosacea is one of the most common conditions affecting the ocular surface. The purpose of this review is to provide an update on the pathogenesis and treatment of rosacea based on the dermatology and ophthalmology literatures. METHODS Literature searches were conducted for rosacea and ocular rosacea. Preference was given to systematic reviews, meta-analysis, case-controlled studies, and documented case reports while excluding poorly documented case studies and commentaries. The data were examined and independently analyzed by more than two of the authors. RESULTS Rosacea is a complex inflammatory condition involving the pilosebaceous unit. Its underlying mechanism involves an interplay of the microbiome, innate immunity, adaptive immunity, environmental triggers, and neurovascular sensitivity. The latest classification of rosacea includes three dermatologic subgroups and a fourth subgroup, ocular rosacea. Ocular rosacea clinically displays many features that are analogous to the cutaneous disease, such as lid margin telangiectasia and phlyctenulosis. The role of environmental triggers in the exacerbation of ocular rosacea appears to be understudied. While lid hygiene and systemic treatment with tetracycline drugs remain the mainstay of treatment for ocular rosacea, newer dermatologic targets and therapies may have potential application for the eye disease. CONCLUSIONS Ocular rosacea appears to embody many of the manifestation of the dermatologic disease. Hence, the basic pathophysiologic mechanisms of the ocular and cutaneous disease are likely to be shared. Better understanding of the ocular surface microbiome and the immunologic mechanisms, may lead to novel approaches in the management of ocular rosacea.
Collapse
Affiliation(s)
- Sayena Jabbehdari
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Benjamin Caughlin
- Department of Surgery/Division of Otolaryngology, Jr. Hospital of Cook County, Chicago, IL, USA.,Hospital of Cook County, Chicago, IL, USA.,Division of Facial Plastic and Reconstructive Surgery, Jesse Brown VA Medical Center, Chicago, IL, USA.,Division of Facial Plastic and Reconstructive Surgery, University of Illinois Health Hospital System, Chicago, IL, USA
| | - Ali R Djalilian
- Department of Ophthalmology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
23
|
Abstract
The use of contact lenses as ocular drug delivery systems has been considered intuitive for decades. However, at this time, there are no approved products using such systems. In this article, we review the challenges with current therapies, pharmacokinetics, and pharmacodynamics of different drug classes and the patient population. In addition, we note the relative lack of clinical studies, and list potential products in active development at this time. In particular, we address the alignment of time course of the therapeutic need, the pharmacokinetics of the molecule, and the delivery characteristics of the systems (e.g., pulsatile vs. zero-order). We also discuss the needs of various populations including the elderly (who may have motor and cognitive issues as well as presbyopia) and the young. While a contact lens delivery system may also provide refractive correction, to date, most of the studies have used noncorrective (plano) lenses. We also considered nanotechnology-based carrier systems. We generalize the development of contact lens delivery systems to all ocular delivery systems in which there are relatively few product approvals and long development times.
Collapse
Affiliation(s)
- Gary D Novack
- Department of Ophthalmology & Visual Sciences, University of California, Davis, California, USA
- PharmaLogic Development, Inc., San Rafael, California, USA
| | - Melissa Barnett
- Department of Ophthalmology & Visual Sciences, University of California, Davis, California, USA
| |
Collapse
|
24
|
Akbari M, Soltani Moghadam R, Elmi R, Nosrati A, Taghiabadi E, Aghdami N. Topical Tacrolimus as an adjunct to Conventional Therapy for Stromal Herpetic Keratitis: a Randomized Clinical Trial. J Ophthalmic Vis Res 2019; 14:400-411. [PMID: 31875094 PMCID: PMC6825688 DOI: 10.18502/jovr.v14i4.5437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/10/2019] [Indexed: 02/05/2023] Open
Abstract
Purpose This study investigates the effects of 0.05% topical tacrolimus as an adjunct therapy for patients with non-necrotizing herpetic stromal keratitis (HSK). Methods Patients with non-necrotizing HSK, referred to the Cornea Clinic at Hospital in Rasht, Iran, between September 2016 and February 2018, were randomly assigned to two groups. The case group (N = 25) and the control group (N = 25) received conventional treatment with systemic acyclovir and topical prednisolone. The case group (N = 25) additionally received 0.05% tacrolimus eye drops four times a day for one month. Complete ocular examinations, including best-corrected visual acuity (BCVA) assessment, intraocular pressure (IOP) measurement, slit lamp biomicroscopy, and photo slit lamp imaging, were performed before treatment, and 3, 7, 14, 21, and 28 days after the intervention. Results The mean age of the patients was 46.2 ± 12.9 years, and 70% of the patients were male. There was no difference between the groups in terms of age, sex, and baseline ocular measurements (P> 0.05). The case group had a lower mean logarithm of the minimum angle of resolution (LogMAR) for BCVA, lower grading scores, and steeper decreasing trends for corneal haziness, edema, neovascularization, and epitheliopathy compared to the control group after the second week (P< 0.05), while IOP remained unchanged between groups (P> 0.05). Conclusion The addition of 0.05% topical tacrolimus enhances visual acuity and reduces corneal inflammation, neovascularization, and scarring; thus, it can used as an appropriate adjunct treatment for patients with HSK.
Collapse
Affiliation(s)
- Mitra Akbari
- Eye Research Center, Amiralmomenin Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Soltani Moghadam
- Eye Research Center, Amiralmomenin Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ramin Elmi
- Legal Medicine Organization, Rasht, Iran
| | - Amir Nosrati
- Eye Research Center, Amiralmomenin Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Taghiabadi
- Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Biomedicine at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
25
|
Lin CJ, Lan YM, Ou MQ, Ji LQ, Lin SD. Expression of miR-217 and HIF-1α/VEGF pathway in patients with diabetic foot ulcer and its effect on angiogenesis of diabetic foot ulcer rats. J Endocrinol Invest 2019; 42:1307-1317. [PMID: 31079353 DOI: 10.1007/s40618-019-01053-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 04/30/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the expression of miR-217 and HIF-1α/VEGF pathway in patients with diabetic foot ulcer (DFU) and its effect on angiogenesis in DFU rats. METHODS The serum levels of miR-217, HIF-1α and VEGF were detected in DFU and simple diabetes mellitus (DM) patients, and healthy controls. DFU rat models were established and treated with miR-217 inhibitors and/or HIF-1α siRNA. The ulcer healing of DFU rats was observed. Besides, ELISA method was performed to detect the serum level of HIF-1α, VEGF and inflammatory factors, immunohistochemical (IHC) method to test the micro-vessel density (MVD), as well as qRT-PCR and Western blot to determine expressions of miR-217, HIF-1α, VEGF, VEGFR2, eNOS, MMP-2, and MMP-9 in tissues. RESULTS The serum levels of miR-217 were up-regulated while HIF-1α and VEGF were down-regulated in DFU patients and rats when compared with DM and healthy controls (all P < 0.05). Dual-luciferase reporter gene assay confirmed that HIF-1α was the direct target gene of miR-217. DFU rats treated with miR-217 inhibitors had decreased foot ulcer area and accelerated ulcer healing, with significantly reduced inflammatory factors (IL-1β, TNF-α and IL-6), as well as elevated HIF-1α and VEGF (all P < 0.05); meanwhile, they remarkably increased the MVD in foot dorsum wound tissues and the protein expressions of HIF-1α, VEGF, VEGFR2, eNOS, MMP-2, and MMP-9 (all P < 0.05). CONCLUSION Inhibiting miR-217 could up-regulate HIF-1α/VEGF pathway to promote angiogenesis and ameliorate inflammation of DFU rats, thereby effectively advancing the healing of ulcerated area.
Collapse
Affiliation(s)
- C-J Lin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China.
| | - Y-M Lan
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China
| | - M-Q Ou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China
| | - L-Q Ji
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China
| | - S-D Lin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, People's Republic of China
| |
Collapse
|
26
|
Brash JT, Denti L, Ruhrberg C, Bucher F. VEGF188 promotes corneal reinnervation after injury. JCI Insight 2019; 4:130979. [PMID: 31672940 PMCID: PMC6948769 DOI: 10.1172/jci.insight.130979] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) induces angiogenesis and vascular hyperpermeability in ocular tissues and is therefore a key therapeutic target for eye conditions in which these processes are dysregulated. In contrast, the therapeutic potential of VEGF's neurotrophic roles in the eye has remained unexploited. In particular, it is not known whether modulating levels of any of the 3 major alternatively spliced VEGF isoforms might provide a therapeutic approach to promote neural health in the eye without inducing vascular pathology. Here, we have used a variety of mouse models to demonstrate differences in overall VEGF levels and VEGF isoform ratios across tissues in the healthy eye. We further show that VEGF isoform expression was differentially regulated in retinal versus corneal disease models. Among the 3 major isoforms - termed VEGF120, VEGF164, and VEGF188 - VEGF188 was upregulated to the greatest extent in injured cornea, where it was both necessary and sufficient for corneal nerve regeneration. Moreover, topical VEGF188 application further promoted corneal nerve regeneration without inducing pathological neovascularization. VEGF isoform modulation should therefore be explored further for its potential in promoting neural health in the eye.
Collapse
Affiliation(s)
- James T Brash
- UCL Institute of Ophthalmology, London, United Kingdom
| | - Laura Denti
- UCL Institute of Ophthalmology, London, United Kingdom
| | | | - Franziska Bucher
- UCL Institute of Ophthalmology, London, United Kingdom.,Department of Ophthalmology, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
27
|
|
28
|
Fadel D, Kramer E. Potential contraindications to scleral lens wear. Cont Lens Anterior Eye 2019; 42:92-103. [DOI: 10.1016/j.clae.2018.10.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/28/2018] [Accepted: 10/30/2018] [Indexed: 12/11/2022]
|
29
|
Sun MM, Chan AM, Law SM, Duarte S, Diaz-Aguilar D, Wadehra M, Gordon LK. Epithelial Membrane Protein-2 (EMP2) Antibody Blockade Reduces Corneal Neovascularization in an In Vivo Model. Invest Ophthalmol Vis Sci 2019; 60:245-254. [PMID: 30646013 PMCID: PMC6336205 DOI: 10.1167/iovs.18-24345] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 12/03/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose Pathologic corneal neovascularization is a major cause of blindness worldwide, and treatment options are currently limited. VEGF is one of the critical mediators of corneal neovascularization but current anti-VEGF therapies have produced limited results in the cornea. Thus, additional therapeutic agents are needed to enhance the antiangiogenic arsenal. Our group previously demonstrated epithelial membrane protein-2 (EMP2) involvement in pathologic angiogenesis in multiple cancer models including breast cancer and glioblastoma. In this paper, we investigate the efficacy of anti-EMP2 immunotherapy in the prevention of corneal neovascularization. Methods An in vivo murine cornea alkali burn model was used to study pathologic neovascularization. A unilateral corneal burn was induced using NaOH, and subconjunctival injection of either anti-EMP2 antibody, control antibody, or sterile saline was performed after corneal burn. Neovascularization was clinically scored at 7 days postalkali burn, and eyes were enucleated for histologic analysis and immunostaining including VEGF, CD31, and CD34 expression. Results Anti-EMP2 antibody, compared to control antibody or vehicle, significantly reduced neovascularization as measured by clinical score and central cornea thickness, as well as by histologic reduction of neovascularization, decreased CD34 staining, and decreased CD31 staining. Incubation of corneal limbal cells in vitro with anti-EMP2 blocking antibody significantly decreased EMP2 expression, VEGF expression and secretion, and cell migration. Conclusions This work demonstrates the effectiveness of EMP2 as a novel target in pathologic corneal neovascularization in an animal model and supports additional investigation into EMP2 antibody blockade as a potential new therapeutic option.
Collapse
Affiliation(s)
- Michel M. Sun
- Department of Ophthalmology, Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States
| | - Ann M. Chan
- Department of Ophthalmology, Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States
| | - Samuel M. Law
- Department of Ophthalmology, Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States
| | - Sergio Duarte
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Daniel Diaz-Aguilar
- Department of Ophthalmology, Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States
| | - Madhuri Wadehra
- Departments of Pathology and Laboratory Medicine, and Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Lynn K. Gordon
- Department of Ophthalmology, Jules Stein Eye Institute, University of California Los Angeles, Los Angeles, California, United States
| |
Collapse
|
30
|
Yin J, Jacobs DS. Long-term outcome of using Prosthetic Replacement of Ocular Surface Ecosystem (PROSE) as a drug delivery system for bevacizumab in the treatment of corneal neovascularization. Ocul Surf 2019; 17:134-141. [PMID: 30468876 PMCID: PMC6340761 DOI: 10.1016/j.jtos.2018.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/12/2018] [Accepted: 11/19/2018] [Indexed: 01/04/2023]
Abstract
PURPOSE To report the long-term outcome of Prosthetic Replacement of the Ocular Surface Ecosystem (PROSE) for delivery of bevacizumab in the treatment of corneal neovascularization (KNV). METHODS Retrospective, non-comparative, interventional case series of 13 sequential patients treated for KNV at the BostonSight between 2006 and 2017. In all cases, PROSE treatment was initiated for management of ocular surface disease and patients wore PROSE consistently on a daily wear basis prior to bevacizumab treatment. Patients applied a drop of 1% preservative free bevacizumab to the reservoir of PROSE device twice daily. Patients continued with daily wear of the device during treatment and afterwards. RESULTS 13 patients (8 female and mean age of 45 years) are included with a mean follow-up of 5.1 years (range 6 months-11 years). Underlying ocular diagnoses included Stevens-Johnson syndrome (7), ocular chronic graft-versus-host disease (2), corneal transplant (2), contact lens-related corneal ulcer and limbal stem cell deficiency (1), and familial dysautonomia (1). Median duration of bevacizumab use was 6 months (range 3 months-10 years). Twelve cases (92%) had regression of KNV and 10 cases (77%) had improved best-corrected visual acuity (BCVA) with treatment. Median BCVA improved from -1.1 (LogMAR) at baseline, to -0.66 at end of bevacizumab treatment, and remained -0.63 at last follow-up (P = 0.047). KNV progressed in one eye after discontinuation of bevacizumab. There were no ophthalmic or systemic complications. CONCLUSIONS Topical bevacizumab used in PROSE is effective in treating KNV and improving vision. Long-term follow-up reveals durable response and no complications.
Collapse
Affiliation(s)
- Jia Yin
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, USA.
| | - Deborah S Jacobs
- Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 243 Charles Street, Boston, MA, 02114, USA
| |
Collapse
|
31
|
Roshandel D, Eslani M, Baradaran-Rafii A, Cheung AY, Kurji K, Jabbehdari S, Maiz A, Jalali S, Djalilian AR, Holland EJ. Current and emerging therapies for corneal neovascularization. Ocul Surf 2018; 16:398-414. [PMID: 29908870 DOI: 10.1016/j.jtos.2018.06.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/10/2018] [Accepted: 06/12/2018] [Indexed: 02/08/2023]
Abstract
The cornea is unique because of its complete avascularity. Corneal neovascularization (CNV) can result from a variety of etiologies including contact lens wear; corneal infections; and ocular surface diseases due to inflammation, chemical injury, and limbal stem cell deficiency. Management is focused primarily on the etiology and pathophysiology causing the CNV and involves medical and surgical options. Because inflammation is a key factor in the pathophysiology of CNV, corticosteroids and other anti-inflammatory medications remain the mainstay of treatment. Anti-VEGF therapies are gaining popularity to prevent CNV in a number of etiologies. Surgical options including vessel occlusion and ocular surface reconstruction are other options depending on etiology and response to medical therapy. Future therapies should provide more effective treatment options for the management of CNV.
Collapse
Affiliation(s)
- Danial Roshandel
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Medi Eslani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA; Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Alireza Baradaran-Rafii
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Albert Y Cheung
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Khaliq Kurji
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA
| | - Sayena Jabbehdari
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Alejandra Maiz
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Setareh Jalali
- Ocular Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Edward J Holland
- Cincinnati Eye Institute, Edgewood, KY/ University of Cincinnati, Department of Ophthalmology, Cincinnati, OH, USA.
| |
Collapse
|
32
|
Selective IKK2 inhibitor IMD0354 disrupts NF-κB signaling to suppress corneal inflammation and angiogenesis. Angiogenesis 2018; 21:267-285. [PMID: 29332242 PMCID: PMC5878206 DOI: 10.1007/s10456-018-9594-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022]
Abstract
Corneal neovascularization is a sight-threatening condition caused by angiogenesis in the normally avascular cornea. Neovascularization of the cornea is often associated with an inflammatory response, thus targeting VEGF-A alone yields only a limited efficacy. The NF-κB signaling pathway plays important roles in inflammation and angiogenesis. Here, we study consequences of the inhibition of NF-κB activation through selective blockade of the IKK complex IκB kinase β (IKK2) using the compound IMD0354, focusing on the effects of inflammation and pathological angiogenesis in the cornea. In vitro, IMD0354 treatment diminished HUVEC migration and tube formation without an increase in cell death and arrested rat aortic ring sprouting. In HUVEC, the IMD0354 treatment caused a dose-dependent reduction in VEGF-A expression, suppressed TNFα-stimulated expression of chemokines CCL2 and CXCL5, and diminished actin filament fibers and cell filopodia formation. In developing zebrafish embryos, IMD0354 treatment reduced expression of Vegf-a and disrupted retinal angiogenesis. In inflammation-induced angiogenesis in the rat cornea, systemic selective IKK2 inhibition decreased inflammatory cell invasion, suppressed CCL2, CXCL5, Cxcr2, and TNF-α expression and exhibited anti-angiogenic effects such as reduced limbal vessel dilation, reduced VEGF-A expression and reduced angiogenic sprouting, without noticeable toxic effect. In summary, targeting NF-κB by selective IKK2 inhibition dampened the inflammatory and angiogenic responses in vivo by modulating the endothelial cell expression profile and motility, thus indicating an important role of NF-κB signaling in the development of pathologic corneal neovascularization.
Collapse
|
33
|
Zhang YK, Li JM, Qin L. Suppression of corneal neovascularization by curcumin via inhibition of Wnt/β-catenin pathway activation. Int J Ophthalmol 2017; 10:1791-1797. [PMID: 29259894 DOI: 10.18240/ijo.2017.12.01] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022] Open
Abstract
AIM To investigate whether curcumin suppressed corneal neovascularization (CNV) formation via inhibiting activation of Wnt/β-catenin pathway. METHODS Suture-induced CNV was established on Sprague-Dawley (SD) rats. Curcumin were daily administrated by subconjunctival injection. Phosphorylation of low-density lipoprotein receptor-related protein 6 (LRP6) and nuclear accumulation of β-catenin, two indicators of activated Wnt/β-catenin pathway, were determined by Western-blot analysis in subconfluent/proliferating human microvascular endothelial cells (HMEC) and neovascularized corneas. Wnt3a conditioned medium (WCM) were harvested from Wnt3a expressing cells. WCM-induced cell proliferation and endothelial tubular formation capacity was measured by MTT assay and Matrigel assay, respectively. RESULTS Phosphorylation of LRP6 and nuclear accumulation of β-catenin was significantly increased in subconfluent/proliferating endothelial cells. Activation of Wnt/β-catenin pathway by WCM markedly promotes HMEC proliferation and tubular formation. Curcumin inhibited LRP6 phosphorylation and nuclear accumulation of β-catenin. In addition, curcumin attenuated WCM-induced HMEC proliferation and disrupted tubular structure of endothelial cells on Matrigel. Meanwhile curcumin suppressed suture-induced CNV and inhibited LRP6 phosphorylation as well as β-catenin accumulation in SD rats. CONCLUSION Taken together, activation of Wnt/β-catenin pathway could be involved in endothelial proliferation during suture-induced CNV formation and curcumin attenuated CNV formation via inhibition of Wnt/β-catenin pathway activation.
Collapse
Affiliation(s)
- Yong-Kang Zhang
- Department of Ophthalmology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.,Department of Ophthalmology, Shaanxi Kangfu Hospital, Xi'an 710065, Shaanxi Province, China
| | - Jing-Ming Li
- Department of Ophthalmology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Li Qin
- Department of Ophthalmology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| |
Collapse
|
34
|
Feizi S, Azari AA, Safapour S. Therapeutic approaches for corneal neovascularization. EYE AND VISION 2017; 4:28. [PMID: 29234686 PMCID: PMC5723406 DOI: 10.1186/s40662-017-0094-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Angiogenesis refers to new blood vessels that originate from pre-existing vascular structures. Corneal neovascularization which can lead to compromised visual acuity occurs in a wide variety of corneal pathologies. A large subset of measures has been advocated to prevent and/or treat corneal neovascularization with varying degrees of success. These approaches include topical corticosteroid administration, laser treatment, cautery, and fine needle diathermy. Since the imbalance between proangiogenic agents and antiangiogenic agents primarily mediate the process of corneal neovascularization, recent therapies are intended to disrupt the different steps in the synthesis and actions of proangiogenic factors. These approaches, however, are only partially effective and may lead to several side effects. The aim of this article is to review the most relevant treatments for corneal neovascularization available so far.
Collapse
Affiliation(s)
- Sepehr Feizi
- Ophthalmic Research Center, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, 16666 Iran
| | - Amir A Azari
- Ophthalmic Research Center, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, 16666 Iran
| | - Sharareh Safapour
- Ophthalmic Research Center, Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, 16666 Iran
| |
Collapse
|
35
|
Cakmak H, Gokmen E, Bozkurt G, Kocaturk T, Ergin K. Effects of sunitinib and bevacizumab on VEGF and miRNA levels on corneal neovascularization. Cutan Ocul Toxicol 2017; 37:191-195. [DOI: 10.1080/15569527.2017.1375943] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Harun Cakmak
- Department of Ophthalmology, Adnan Menderes University Medical Faculty, Aydin, Turkey
| | - Esra Gokmen
- Department of Histology and Embryology, Adnan Menderes University Medical Faculty, Aydin, Turkey
| | - Gokay Bozkurt
- Department of Medical Genetics, Adnan Menderes University Medical Faculty, Aydin, Turkey
| | - Tolga Kocaturk
- Department of Ophthalmology, Adnan Menderes University Medical Faculty, Aydin, Turkey
| | - Kemal Ergin
- Department of Histology and Embryology, Adnan Menderes University Medical Faculty, Aydin, Turkey
| |
Collapse
|
36
|
Adiponectin promotes VEGF-A-dependent angiogenesis in human chondrosarcoma through PI3K, Akt, mTOR, and HIF-α pathway. Oncotarget 2017; 6:36746-61. [PMID: 26468982 PMCID: PMC4742208 DOI: 10.18632/oncotarget.5479] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/02/2015] [Indexed: 01/16/2023] Open
Abstract
Chondrosarcoma is a type of highly malignant tumor with a potent capacity to invade locally and cause distant metastasis. Adiponectin is a protein hormone secreted predominantly by differentiated adipocytes. On the other hand, angiogenesis is a critical step in tumor growth and metastasis. However, the relationship of adiponectin with vascular endothelial growth factor-A (VEGF-A) expression and angiogenesis in human chondrosarcoma is mostly unknown. In this study we first demonstrated that the expression of adiponectin was correlated with tumor stage of human chondrosarcoma tissues. In addition, we also found that adiponectin increased VEGF-A expression in human chondrosarcoma cells and subsequently induced migration and tube formation in human endothelial progenitor cells (EPCs). Adiponectin promoted VEGF-A expression through adiponectin receptor (AdipoR), phosphoinositide 3 kinase (PI3K), Akt, mammalian target of rapamycin (mTOR), and hypoxia-inducible factor-1α (HIF)-1α signaling cascades. Knockdown of adiponectin decreased VEGF-A expression and also abolished chondrosarcoma conditional medium-mediated tube formation in EPCs in vitro as well as angiogenesis effects in the chick chorioallantoic membrane and Matrigel plug nude mice model in vivo. Therefore, adiponectin is crucial for tumor angiogenesis and growth, which may represent a novel target for anti-angiogenic therapy in human chondrosarcoma.
Collapse
|
37
|
Ntumba K, Akla N, Oh SP, Eichmann A, Larrivée B. BMP9/ALK1 inhibits neovascularization in mouse models of age-related macular degeneration. Oncotarget 2016; 7:55957-55969. [PMID: 27517154 PMCID: PMC5302889 DOI: 10.18632/oncotarget.11182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/13/2016] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in aging populations of industrialized countries. The drawbacks of inhibitors of vascular endothelial growth factor (VEGFs) currently used for the treatment of AMD, which include resistance and potential serious side-effects, require the identification of new therapeutic targets to modulate angiogenesis. BMP9 signaling through the endothelial Alk1 serine-threonine kinase receptor modulates the response of endothelial cells to VEGF and promotes vessel quiescence and maturation during development. Here, we show that BMP9/Alk1 signaling inhibits neovessel formation in mouse models of pathological ocular angiogenesis relevant to AMD. Activating Alk1 signaling in laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR) inhibited neovascularization and reduced the volume of vascular lesions. Alk1 signaling was also found to interfere with VEGF signaling in endothelial cells whereas BMP9 potentiated the inhibitory effects of VEGFR2 signaling blockade, both in OIR and laser-induced CNV. Together, our data show that targeting BMP9/Alk1 efficiently prevents the growth of neovessels in AMD models and introduce a new approach to improve conventional anti-VEGF therapies.
Collapse
Affiliation(s)
- Kalonji Ntumba
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Naoufal Akla
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - S. Paul Oh
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, New Haven, CT, USA
- Inserm U970, Paris Cardiovascular Research Center, Paris, France
| | - Bruno Larrivée
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Molecular Biology, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, Quebec, Canada
| |
Collapse
|
38
|
Guzel H, Bakbak B, Koylu MT, Gonul S, Ozturk B, Gedik S. The effect and safety of intravitreal injection of ranibizumab and bevacizumab on the corneal endothelium in the treatment of diabetic macular edema. Cutan Ocul Toxicol 2016; 36:5-8. [DOI: 10.3109/15569527.2016.1140177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Çakmak H, Ergin K, Bozkurt G, Kocatürk T, Evliçoğlu GE. The effects of topical everolimus and sunitinib on corneal neovascularization. Cutan Ocul Toxicol 2015; 35:97-103. [PMID: 25864572 DOI: 10.3109/15569527.2015.1034360] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE To evaluate the effects of topical everolimus and sunitinib on corneal neovascularization (CNV). METHODS CNV was induced by application of silver nitrate to the cornea for all groups. Rats were divided into four groups of 10 rats each, and two corneas were obtained from each rat. Group I received 1 mg/ml everolimus, Group II received 0.5 mg/ml sunitinib, Group IV received no treatment (control group) and Group IV received 1% Dimethylsulfoxide (DMSO). All treatments were administrated twice daily for 2 weeks. The right corneas were used for extracellular signal-regulated kinase 1/2 (ERK 1/2) protein analysis by western blot analysis and the left corneas were used for ERK 1/2 and vascular endothelial growth factor-receptor (VEGFR-2) gene expression analysis by quantitative real-time PCR. RESULTS VEGFR-2 mRNA expression levels (ΔCt, median, min-max) were reduced in the everolimus 1.0 (0.25-1.81) and sunitinib 1.06 (0.24-2.68) treated groups compared with the control 4.74 (1.02-14.74) and DMSO groups 7.41 (0.72-13.10). The expression of ERK 1/2 protein and mRNA levels were reduced in everolimus group compared with the control group (p < 0.05). These differences were not seen between the sunitinib and control groups. CONCLUSION Topical administration of both everolimus and sunitinib reduced VEGFR-2 levels and inhibited CNV. In additon, everolimus reduced ERK 1/2 levels and seems to be more effective than sunitinib on CNV.
Collapse
Affiliation(s)
| | - Kemal Ergin
- b Department of Histology and Embryology , and
| | - Gökay Bozkurt
- c Department of Genetics, Adnan Menderes University Medical Faculty , Aytepe , Aydin , Turkey
| | | | | |
Collapse
|
40
|
CCL5 promotes vascular endothelial growth factor expression and induces angiogenesis by down-regulating miR-199a in human chondrosarcoma cells. Cancer Lett 2014; 357:476-87. [PMID: 25444917 DOI: 10.1016/j.canlet.2014.11.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/28/2014] [Accepted: 11/09/2014] [Indexed: 01/22/2023]
Abstract
Chondrosarcoma is a primary malignant bone cancer, with a potent capacity to invade locally and cause distant metastasis. Angiogenesis is a critical step in tumor growth and metastasis. Chemokine CCL5 (previously called RANTES) has been shown to facilitate tumor progression and metastasis. However, the relationship of CCL5 with vascular endothelial growth factor (VEGF) expression and angiogenesis in human chondrosarcoma is mostly unknown. In this study, CCL5 increased VEGF expression and also promoted chondrosarcoma medium-mediated angiogenesis in vitro as well as angiogenesis effects in the chick chorioallantoic membrane and Matrigel plug nude mice model in vivo. MicroRNA analysis was performed in CCL5-treated chondrosarcoma cells versus control cells to investigate the mechanism of CCL5-mediated promotion of chondrosarcoma angiogenesis. Among the miRNAs regulated by CCL5, miR-199a was the most downregulated miRNA after CCL5 treatment. In addition, co-transfection with miR-199a mimic reversed the CCL5-mediated VEGF expression and angiogenesis in vitro and in vivo. Moreover, overexpression of CCL5 increased tumor-associated angiogenesis and tumor growth by downregulating miR-199a in the xenograft tumor angiogenesis model. Taken together, these results demonstrated that CCL5 promotes VEGF-dependent angiogenesis in human chondrosarcoma cells by downregulating miR-199a.
Collapse
|
41
|
Lin CY, Hung SY, Chen HT, Tsou HK, Fong YC, Wang SW, Tang CH. Brain-derived neurotrophic factor increases vascular endothelial growth factor expression and enhances angiogenesis in human chondrosarcoma cells. Biochem Pharmacol 2014; 91:522-33. [PMID: 25150213 DOI: 10.1016/j.bcp.2014.08.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 01/07/2023]
Abstract
Chondrosarcomas are a type of primary malignant bone cancer, with a potent capacity for local invasion and distant metastasis. Brain-derived neurotrophic factor (BDNF) is commonly upregulated during neurogenesis. The aim of the present study was to examine the mechanism involved in BDNF-mediated vascular endothelial growth factor (VEGF) expression and angiogenesis in human chondrosarcoma cells. Here, we knocked down BDNF expression in chondrosarcoma cells and assessed their capacity to control VEGF expression and angiogenesis in vitro and in vivo. We found knockdown of BDNF decreased VEGF expression and abolished chondrosarcoma conditional medium-mediated angiogenesis in vitro as well as angiogenesis effects in vivo in the chick chorioallantoic membrane and Matrigel plug nude mouse models. In addition, in the xenograft tumor angiogenesis model, the knockdown of BDNF significantly reduced tumor growth and tumor-associated angiogenesis. BDNF increased VEGF expression and angiogenesis through the TrkB receptor, PLCγ, PKCα, and the HIF-1α signaling pathway. Finally, we analyzed samples from chondrosarcoma patients by immunohistochemical staining. The expression of BDNF and VEGF protein in 56 chondrosarcoma patients was significantly higher than in normal cartilage. In addition, the high level of BDNF expression correlated strongly with VEGF expression and tumor stage. Taken together, our results indicate that BDNF increases VEGF expression and enhances angiogenesis through a signal transduction pathway that involves the TrkB receptor, PLCγ, PKCα, and the HIF-1α. Therefore, BDNF may represent a novel target for anti-angiogenic therapy for human chondrosarcoma.
Collapse
Affiliation(s)
- Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan
| | - Shih-Ya Hung
- Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hsien-Te Chen
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Materials Science and Engineering, Feng Chia University, Taichung, Taiwan
| | - Hsi-Kai Tsou
- Department of Materials Science and Engineering, Feng Chia University, Taichung, Taiwan; Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Early Childhood Care and Education, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Yi-Chin Fong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
42
|
Efficacy of intralesional bevacizumab administration in primary pterygium: a quantitative analysis. Eye Contact Lens 2014; 40:46-50. [PMID: 24335454 DOI: 10.1097/icl.0000000000000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To analyze the regression of the pterygium tissue quantitatively after intralesional bevacizumab administration in patients with primary pterygium. METHODS Thirty-three eyes of 33 patients with primary pterygium who underwent intralesional bevacizumab (1.25 mg per 0.05 mL) administration were included in this prospective study. The ocular irritation, best-corrected visual acuity (BCVA), slit-lamp and funduscopic examinations, corneal astigmatism, the horizontal length, and the thickness of the pterygium were assessed before and after 2 months of treatment. To evaluate the corneal astigmatism and the thickness of the pterygium, the Pentacam comprehensive eye scanner (Pentacam CES; Oculus GmbH) was used. RESULTS After 2 months of bevacizumab administration, the average BCVA was improved (P=0.003). The average ocular irritation score, horizontal length, and the thickness of the pterygium were statistically significantly decreased (P<0.001, P<0.001, P<0.001, respectively). CONCLUSION After 2 months of intralesional bevacizumab administration, the average amount of decrease in the horizontal length and thickness of the pterygium was 0.6 mm and 37 μm, respectively. Intralesional bevacizumab administration is useful in the management of primary pterygium without having any local or systemic adverse effects; however, repeated administrations are needed to provide clinically more significant results.
Collapse
|
43
|
Bradykinin promotes vascular endothelial growth factor expression and increases angiogenesis in human prostate cancer cells. Biochem Pharmacol 2013; 87:243-53. [PMID: 24225154 DOI: 10.1016/j.bcp.2013.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/15/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy in men and shows a tendency for metastasis to distant organs. Angiogenesis is required for metastasis. Bradykinin (BK) is an inflammatory mediator involved in tumor growth and metastasis, but its role in vascular endothelial growth factor (VEGF) expression and angiogenesis in human prostate cancer remains unknown. The aim of this study was to examine whether BK promotes prostate cancer angiogenesis via VEGF expression. We found that exogenous BK increased VEGF expression in prostate cancer cells and further promoted tube formation in endothelial progenitor cells and human umbilical vein endothelial cells. Pretreatment of prostate cancer with B2 receptor antagonist or small interfering RNA (siRNA) reduced BK-mediated VEGF production. The Akt and mammalian target of rapamycin (mTOR) pathways were activated after BK treatment, and BK-induced VEGF expression was abolished by the specific inhibitor and siRNA of the Akt and mTOR cascades. BK also promoted nuclear factor-κB (NF-κB) and activator protein 1 (AP-1) activity. Importantly, BK knockdown reduced VEGF expression and abolished prostate cancer cell conditional medium-mediated angiogenesis. Taken together, these results indicate that BK operates through the B2 receptor, Akt, and mTOR, which in turn activate NF-κB and AP-1, activating VEGF expression and contributing to angiogenesis in human prostate cancer cells.
Collapse
|
44
|
Liang P, Jiang B, Lv C, Huang X, Sun L, Zhang P, Huang X. The expression and proangiogenic effect of nucleolin during the recovery of heat-denatured HUVECs. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1830:4500-12. [PMID: 23726991 DOI: 10.1016/j.bbagen.2013.05.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/27/2013] [Accepted: 05/20/2013] [Indexed: 11/25/2022]
Abstract
BACKGROUND The present study aims to examine the expression patterns and roles of nucleolin during the recovery of heat-denatured human umbilical vein endothelial cells (HUVECs). METHODS Deep partial thickness burn model in Sprague-Dawley rats and the heat denatured cell model (52°C, 35s) were used. The expression of nucleolin was measured using Western blot analysis and real-time PCR. Angiogenesis was assessed using in vitro parameters including endothelial cell proliferation, transwell migration assay, and scratched wound healing. Gene transfection and RNA interference approaches were employed to investigate the roles of nucleolin. RESULTS Nucleolin mRNA and protein expression showed a time-dependent increase during the recovery of heat-denatured dermis and HUVECs. Heat-denaturation time-dependently promoted cell growth, adhesion, migration, scratched wound healing and formation of tube-like structures in HUVECs. These effects of heat denaturation on endothelial wound healing and formation of tube-like structures were prevented by knockdown of nucleolin, whereas over-expression of nucleolin increased cell growth, migration, and formation of tube-like structures in cultured HUVEC endothelial cells. In addition, we found that the expression of vascular endothelial growth factor (VEGF) increased during the recovery of heat-denatured dermis and HUVECs, and nucleolin up-regulated VEGF in HUVECs. CONCLUSIONS The present study reveals that the expression of nucleolin is up-regulated, and plays a pro-angiogenic role during the recovery of heat-denatured dermis and its mechanism is probably dependent on production of VEGF. GENERAL SIGNIFICANCE We find a novel and important pro-angiogenic role of nucleolin during the recovery of heat-denatured dermis.
Collapse
Affiliation(s)
- Pengfei Liang
- Department of Burns and Plastic Surgery, Central South University, Changsha, Hunan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
45
|
Tsai CH, Chiang YC, Chen HT, Huang PH, Hsu HC, Tang CH. High glucose induces vascular endothelial growth factor production in human synovial fibroblasts through reactive oxygen species generation. Biochim Biophys Acta Gen Subj 2013; 1830:2649-58. [PMID: 23274526 DOI: 10.1016/j.bbagen.2012.12.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 12/03/2012] [Accepted: 12/16/2012] [Indexed: 11/28/2022]
Abstract
BACKGROUND Diabetes is an independent risk factor of osteoarthritis (OA). Angiogenesis is essential for the progression of OA. Here, we investigated the intracellular signaling pathways involved in high glucose (HG)-induced vascular endothelial growth factor (VEGF) expression in human synovial fibroblast cells. METHODS HG-mediated VEGF expression was assessed with qPCR and ELISA. The mechanisms of action of HG in different signaling pathways were studied using Western blotting. Knockdown of proteins was achieved by transfection with siRNA. Chromatin immunoprecipitation assays were used to study in vivo binding of c-Jun to the VEGF promoter. RESULTS Stimulation of OA synovial fibroblasts (OASF) with HG induced concentration- and time-dependent increases in VEGF expression. Treatment of OASF with HG increased reactive oxygen species (ROS) generation. Pretreatment with NADPH oxidase inhibitor (APO or DPI), ROS scavenger (NAC), PI3K inhibitor (Ly294002 or wortmannin), Akt inhibitor, or AP-1 inhibitor (curcumin or tanshinone IIA) blocked the HG-induced VEGF production. HG also increased PI3K and Akt activation. Treatment of OASF with HG increased the accumulation of phosphorylated c-Jun in the nucleus, AP-1-luciferase activity, and c-Jun binding to the AP-1 element on the VEGF promoter. CONCLUSIONS Our results suggest that the HG increases VEGF expression in human synovial fibroblasts via the ROS, PI3K, Akt, c-Jun and AP-1 signaling pathway. GENERAL SIGNIFICANCE We link high glucose on VEGF expression in osteoarthritis.
Collapse
Affiliation(s)
- Chun-Hao Tsai
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Endothelin-1 promotes vascular endothelial growth factor-dependent angiogenesis in human chondrosarcoma cells. Oncogene 2013; 33:1725-35. [DOI: 10.1038/onc.2013.109] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/25/2013] [Accepted: 02/07/2013] [Indexed: 01/01/2023]
|
47
|
Mayer WJ, Grüterich M, Kook D, Sigg W, Kernt M, Messmer EM, Haritoglou C, Kampik A, Wolf A. Modification of Amniotic Membrane as a Depot Carrier for Bevacizumab – AnIn-VitroModel for a Slow Release Mechanism. Curr Eye Res 2013; 38:445-50. [DOI: 10.3109/02713683.2012.757326] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Zhou AY, Bai YJ, Zhao M, Yu WZ, Li XX. KH902, a Recombinant Human VEGF Receptor Fusion Protein, Reduced the Level of Placental Growth Factor in Alkali Burn Induced-Corneal Neovascularization. Ophthalmic Res 2013; 50:180-6. [DOI: 10.1159/000353437] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 05/08/2013] [Indexed: 11/19/2022]
|
49
|
Tzeng HE, Tsai CH, Chang ZL, Su CM, Wang SW, Hwang WL, Tang CH. Interleukin-6 induces vascular endothelial growth factor expression and promotes angiogenesis through apoptosis signal-regulating kinase 1 in human osteosarcoma. Biochem Pharmacol 2012; 85:531-40. [PMID: 23219526 DOI: 10.1016/j.bcp.2012.11.021] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 11/23/2012] [Accepted: 11/27/2012] [Indexed: 12/17/2022]
Abstract
Osteosarcoma is characterized by a high malignant and metastatic potential. Angiogenesis is essential for the caner metastasis. Interleukin-6 (IL-6) is a multifunctional cytokine that is associated with the disease status and outcomes of cancers. However, the relationship between IL-6 and vascular endothelial growth factor (VEGF) expression in human osteosarcoma is mostly unknown. Here we found that the IL-6 and VEGF expression was correlated with tumor stage and significantly higher than that in normal bone. Incubation of osteosarcoma cells with IL-6 increased VEGF mRNA and protein expression. Pretreatment of cells with IL-6R antibody reduced IL-6-mediated VEGF production. The apoptosis signal-regulating kinase 1 (ASK1)/p38/AP-1 pathway was activated after IL-6 treatment, and IL-6-induced VEGF expression was abolished by the specific inhibitor and siRNA of ASK1, p38, and AP-1 cascades. Importantly, knockdown IL-6 reduced VEGF expression and abolished osteosarcoma conditional medium-mediated angiogenesis. Taken together, these results indicate that IL-6 occurs through ASK1 and p38, which in turn activates AP-1, resulting in the activations of VEGF expression and contributing the angiogenesis of human osteosarcoma cells.
Collapse
Affiliation(s)
- Huey-En Tzeng
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
50
|
Lin YM, Huang YL, Fong YC, Tsai CH, Chou MC, Tang CH. Hepatocyte growth factor increases vascular endothelial growth factor-A production in human synovial fibroblasts through c-Met receptor pathway. PLoS One 2012; 7:e50924. [PMID: 23209838 PMCID: PMC3508989 DOI: 10.1371/journal.pone.0050924] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/26/2012] [Indexed: 11/19/2022] Open
Abstract
Background Angiogenesis is essential for the progression of osteoarthritis (OA). Hepatocyte growth factor (HGF) is an angiogenic mediator, and it shows elevated levels in regions of OA. However, the relationship between HGF and vascular endothelial growth factor (VEGF-A) in OA synovial fibroblasts (OASFs) is mostly unknown. Methodology/Principal Findings Here we found that stimulation of OASFs with HGF induced concentration- and time-dependent increases in VEGF-A expression. Pretreatment with PI3K inhibitor (Ly294002), Akt inhibitor, or mTORC1 inhibitor (rapamycin) blocked the HGF-induced VEGF-A production. Treatment of cells with HGF also increased PI3K, Akt, and mTORC1 phosphorylation. Furthermore, HGF increased the stability and activity of HIF-1 protein. Moreover, the use of pharmacological inhibitors or genetic inhibition revealed that c-Met, PI3K, Akt, and mTORC1 signaling pathways were potentially required for HGF-induced HIF-1α activation. Conclusions/Significance Taken together, our results provide evidence that HGF enhances VEGF-A expression in OASFs by an HIF-1α-dependent mechanism involving the activation of c-Met/PI3K/Akt and mTORC1 pathways.
Collapse
Affiliation(s)
- Yu-Min Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Medicine and Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|