1
|
Jeon KI, Kumar A, Brookes PS, Nehrke K, Huxlin KR. Manipulating mitochondrial pyruvate carrier function causes metabolic remodeling in corneal myofibroblasts that ameliorates fibrosis. Redox Biol 2024; 75:103235. [PMID: 38889622 PMCID: PMC11231598 DOI: 10.1016/j.redox.2024.103235] [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: 05/16/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Myofibroblasts are key cellular effectors of corneal wound healing from trauma, surgery, or infection. However, their persistent deposition of disorganized extracellular matrix can also cause corneal fibrosis and visual impairment. Recent work showed that the PPARγ agonist Troglitazone can mitigate established corneal fibrosis, and parallel in vitro data suggested this occurred through inhibition of the mitochondrial pyruvate carrier (MPC) rather than PPARγ. In addition to oxidative phosphorylation (Ox-Phos), pyruvate and other mitochondrial metabolites provide carbon for the synthesis of biological macromolecules. However, it is currently unclear how these roles selectively impact fibrosis. Here, we performed bioenergetic, metabolomic, and epigenetic analyses of corneal fibroblasts treated with TGF-β1 to stimulate myofibroblast trans-differentiation, with further addition of Troglitazone or the MPC inhibitor UK5099, to identify MPC-dependencies that may facilitate remodeling and loss of the myofibroblast phenotype. Our results show that a shift in energy metabolism is associated with, but not sufficient to drive cellular remodeling. Metabolites whose abundances were sensitive to MPC inhibition suggest that sustained carbon influx into the Krebs' cycle is prioritized over proline synthesis to fuel collagen deposition. Furthermore, increased abundance of acetyl-CoA and increased histone H3 acetylation suggest that epigenetic mechanisms downstream of metabolic remodeling may reinforce cellular phenotypes. Overall, our results highlight a novel molecular target and metabolic vulnerability that affects myofibroblast persistence in the context of corneal wounding.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Ankita Kumar
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA
| | - Paul S Brookes
- Dept. Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Keith Nehrke
- Dept. Medicine-Nephrology Division, University of Rochester, Rochester, NY, USA
| | - Krystel R Huxlin
- Dept. Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
2
|
Zhu Y, Jin X, Fu N, Li J. Medrysone promotes corneal injury repair by promoting M2-like polarization of macrophages. BMC Ophthalmol 2023; 23:503. [PMID: 38082280 PMCID: PMC10712160 DOI: 10.1186/s12886-023-03234-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Accumulated evidence suggests that M2-like polarized macrophages plays an important role in reducing inflammation, promoting and accelerating wound healing process and tissue repair. Thus, M2-like TAMs (Tumour-associated macrophages) was an appealing target for therapy intervention. METHOD Flow cytometry and RT-PCR assay were used to detect the polarization of macrophages induced by Medrysone, and the rat corneal mechanical injury model was established to evaluate the efficacy of Medrysone in cornel repair. RESULTS Here we found that Medrysone enhanced IL-4 induced M2 polarization of macrophages, as illustrated by increased expression of CD206, up-regulation of M2 marker mRNAs. Medrysone promoted VEGF and CCL2 secretion in IL-4 induced M2-like polarization. IL-4 triggered STAT6 activation was further enhanced by Medrysone and silencing of STAT6 partially abrogated the stimulatory effect of Medrysone. Medrysone improved migration-promoting feature of M2-like macrophages, as indicated by increased migration of endothelial cells. Further, Medrysone promoted corneal injury repair by inducing M2 polarization of macrophages in vivo. CONCLUSION Our study suggest that Medrysone promotes corneal injury repair by inducing the M2 polarization of macrophages, providing a theoretical basis for the application of Medrysone in the treatment of corneal injury.
Collapse
Affiliation(s)
- Yaqin Zhu
- Hangzhou Aier Eye Hospital, Hangzhou, 310000, Zhejiang, China
| | - Xiaohong Jin
- Hangzhou Aier Eye Hospital, Hangzhou, 310000, Zhejiang, China
| | - Ning Fu
- Sir Run Run Shaw Hospital Hangzhou, Hangzhou, 310000, China
| | - Jiuke Li
- Hangzhou Aier Eye Hospital, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
3
|
Peng X, Du YL, Liu ST, Chen H, Wang JS, Wang C, Xie HT, Zhang MC. Isolation and Culture of Human Meibomian Gland Ductal Cells. Invest Ophthalmol Vis Sci 2023; 64:29. [PMID: 38133507 PMCID: PMC10746932 DOI: 10.1167/iovs.64.15.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Purpose Hyperkeratinization of meibomian gland (MG) ducts is currently recognized as the primary pathologic mechanism of meibomian gland dysfunction (MGD). This research figured out a method to isolate the MG ducts and established a novel system to culture the human meibomian gland ductal cells (HMGDCs) for investigating the process of MGD. Methods The MG ducts were obtained from the eyelids of recently deceased donors and subjected to enzymatic digestion. The acini were then removed to isolate independent ducts. These MG ducts were subsequently cultivated on Matrigel-coated wells and covered with a glass plate to obtain HMGDCs. The HMGDCs were further cultivated until passage 2, and when they reached 60% confluence, they were treated with IL-1β and rosiglitazone for a duration of 48 hours. Immunofluorescence staining and Western blot techniques were employed to identify ductal cells and analyze the effects of IL-1β on HMGDCs in an in vitro setting. Results Ophthalmic micro-forceps and insulin needles can be employed for the purpose of isolating ducts. Within this particular culture system, the rapid expansion of HMGDCs occurred in close proximity to the duct tissue. MG ducts specifically expressed keratin 6 (Krt6) and hardly synthesized lipids. Furthermore, the expression of Krt6 was significantly higher (P < 0.0001) in HMGDCs compared to human meibomian gland cells. Upon treatment with IL-1β, HMGDCs exhibited an overexpression of keratin 1, which was effectively blocked by the administration of rosiglitazone. Conclusions The present study successfully isolated human MG ducts and cultured HMGDCs, providing a valuable in vitro model for investigating the mechanism of MGD. Additionally, the potential therapeutic efficacy of rosiglitazone in treating hyperkeratinization of ducts in patients with MGD was identified.
Collapse
Affiliation(s)
- Xi Peng
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Li Du
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-Ting Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Chen
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Song Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Jeon KI, Kumar A, Callan CL, DeMagistris M, MacRae S, Nehrke K, Huxlin KR. Blocking Mitochondrial Pyruvate Transport Alters Corneal Myofibroblast Phenotype: A New Target for Treating Fibrosis. Invest Ophthalmol Vis Sci 2023; 64:36. [PMID: 37870848 PMCID: PMC10599161 DOI: 10.1167/iovs.64.13.36] [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: 06/12/2023] [Accepted: 09/21/2023] [Indexed: 10/24/2023] Open
Abstract
Purpose The purpose of this study was to critically test the hypothesis that mitochondrial pyruvate carrier (MPC) function is essential for maintenance of the corneal myofibroblast phenotype in vitro and in vivo. Methods Protein and mRNA for canonical profibrotic markers were assessed in cultured cat corneal myofibroblasts generated via transforming growth factor (TGF)-β1 stimulation and treated with either the thiazolidinedione (TZD) troglitazone or the MPC inhibitor alpha-cyano-beta-(1-phenylindol-3-yl) acrylate (UK-5099). RNA sequencing was used to gain insight into signaling modules related to instructive, permissive, or corollary changes in gene expression following treatment. A feline photorefractive keratectomy (PRK) model of corneal wounding was used to test the efficacy of topical troglitazone at reducing α-smooth muscle actin (SMA)-positive staining when applied 2 to 4 weeks postoperatively, during peak fibrosis. Results Troglitazone caused cultured myofibroblasts to adopt a fibroblast-like phenotype through a noncanonical, peroxisome proliferator-activated receptor (PPAR)-γ-independent mechanism. Direct MPC inhibition using UK-5099 recapitulated this effect, but classic inhibitors of oxidative phosphorylation (OXPHOS) did not. Gene Set Enrichment Analysis (GSEA) of RNA sequencing data converged on energy substrate utilization and the Mitochondrial Permeability Transition pore as key players in myofibroblast maintenance. Finally, troglitazone applied onto an established zone of active fibrosis post-PRK significantly reduced stromal α-SMA expression. Conclusions Our results provide empirical evidence that metabolic remodeling in myofibroblasts creates selective vulnerabilities beyond simply mitochondrial energy production, and that these are critical for maintenance of the myofibroblast phenotype. For the first time, we provide proof-of-concept data showing that this remodeling can be exploited to treat existing corneal fibrosis via inhibition of the MPC.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Ankita Kumar
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Christine L Callan
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Margaret DeMagistris
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Scott MacRae
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Keith Nehrke
- Department of Medicine-Nephrology Division, University of Rochester, Rochester, New York, United States
| | - Krystel R Huxlin
- Department of Ophthalmology, Flaum Eye Institute and Center for Visual Science, University of Rochester, Rochester, New York, United States
| |
Collapse
|
5
|
Ung L, Chodosh J. Urgent unmet needs in the care of bacterial keratitis: An evidence-based synthesis. Ocul Surf 2023; 28:378-400. [PMID: 34461290 PMCID: PMC10721114 DOI: 10.1016/j.jtos.2021.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/21/2022]
Abstract
Bacterial corneal infections, or bacterial keratitis (BK), are ophthalmic emergencies that frequently lead to irreversible visual impairment. Though increasingly recognized as a major cause of global blindness, modern paradigms of evidence-based care in BK have remained at a diagnostic and therapeutic impasse for over half a century. Current standards of management - based on the collection of corneal cultures and the application of broad-spectrum topical antibiotics - are beset by important yet widely underrecognized limitations, including approximately 30% of all patients who will develop moderate to severe vision loss in the affected eye. Though recent advances have involved a more clearly defined role for adjunctive topical corticosteroids, and novel therapies such as corneal crosslinking, overall progress to improve patient and population-based outcomes remains incommensurate to the chronic morbidity caused by this disease. Recognizing that the care of BK is guided by the clinical axiom, "time equals vision", this chapter offers an evidence-based synthesis for the clinical management of these infections, underscoring critical unmet needs in disease prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Lawson Ung
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Infectious Disease Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Talpan D, Salla S, Seidelmann N, Walter P, Fuest M. Antifibrotic Effects of Caffeine, Curcumin and Pirfenidone in Primary Human Keratocytes. Int J Mol Sci 2023; 24:ijms24021461. [PMID: 36674976 PMCID: PMC9862324 DOI: 10.3390/ijms24021461] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
We evaluated the small molecules (AFM) caffeine, curcumin and pirfenidone to find non-toxic concentrations reducing the transformation of activated human corneal stromal keratocytes (aCSK) to scar-inducing myofibroblasts (MYO-SF). CSK were isolated from 16 human corneas unsuitable for transplantation and expanded for three passages in control medium (0.5% FBS). Then, aCSK were exposed to concentrations of caffeine of 0−500 μM, curcumin of 0−200 μM, pirfenidone of 0−2.2 nM and the profibrotic cytokine TGF-β1 (10 ng/mL) for 48 h. Alterations in viability and gene expression were evaluated by cell viability staining (FDA/PI), real-time polymerase chain reaction (RT-PCR) and immunocytochemistry. We found that all AFMs reduced cell counts at high concentrations. The highest concentrations with no toxic effect were 100 µM of caffeine, 20 µM of curcumin and 1.1 nM of pirfenidone. The addition of TGF-β1 to the control medium effectively transformed aCSK into myofibroblasts (MYO-SF), indicated by a 10-fold increase in α-smooth muscle actin (SMA) expression, a 39% decrease in lumican (LUM) expression and a 98% decrease in ALDH3A1 expression (p < 0.001). The concentrations of 100 µM of caffeine, 20/50 µM of curcumin and 1.1 nM of pirfenidone each significantly reduced SMA expression under TGF-β1 stimulation (p ≤ 0.024). LUM and ALDH3A1 expression remained low under TGF-β1 stimulation, independently of AFM supplementation. Immunocytochemistry showed that 100 µM of caffeine, 20 µM of curcumin and 1.1 nM of pirfenidone reduce the conversion rate of aCSK to SMA+ MYO-SF. In conclusion, in aCSK, 100 µM of caffeine, 20 µM of curcumin and 1.1 nM of pirfenidone significantly reduced SMA expression and MYO-SF conversion under TGF-β1 stimulation, with no influence on cell counts. However, the AFMs were unable to protect aCSK from characteristic marker loss.
Collapse
Affiliation(s)
- Delia Talpan
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
| | - Sabine Salla
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Nina Seidelmann
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
| | - Peter Walter
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Matthias Fuest
- Department of Ophthalmology, RWTH Aachen University, 52074 Aachen, Germany
- Cornea Bank Aachen, RWTH Aachen University, 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
7
|
Jeon KI, Kumar A, Wozniak KT, Nehrke K, Huxlin KR. Defining the Role of Mitochondrial Fission in Corneal Myofibroblast Differentiation. Invest Ophthalmol Vis Sci 2022; 63:2. [PMID: 35377925 PMCID: PMC8994166 DOI: 10.1167/iovs.63.4.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Fibrosis caused by corneal wounding can lead to scar formation, impairing vision. Although preventing fibroblast-to-myofibroblast differentiation has therapeutic potential, effective mechanisms for doing so remain elusive. Recent work shows that mitochondria contribute to differentiation in several tissues. Here, we tested the hypothesis that mitochondrial dynamics, and specifically fission, are key for transforming growth factor (TGF)-β1-induced corneal myofibroblast differentiation. Methods Mitochondrial fission was inhibited pharmacologically in cultured primary cat corneal fibroblasts. We measured its impact on molecular markers of myofibroblast differentiation and assessed changes in mitochondrial morphology through fluorescence imaging. The phosphorylation status of established regulatory proteins, both of myofibroblast differentiation and mitochondrial fission, was assessed by Western analysis. Results Pharmacological inhibition of mitochondrial fission suppressed TGF-β1-induced increases in alpha-smooth muscle actin, collagen 1, and fibronectin expression, and prevented phosphorylation of c-Jun N-terminal kinase (JNK), but not small mothers against decapentaplegic 3, p38 mitogen-activated protein kinase (p38), extracellular signal-regulated kinase 1 (ERK1), or protein kinase B (AKT). TGF-β1 increased phosphorylation of dynamin-related protein 1 (DRP1), a mitochondrial fission regulator, and caused fragmentation of the mitochondrial network. Although inhibition of JNK, ERK1, or AKT prevented phosphorylation of DRP1, none sufficed to independently suppress TGF-β1-induced fragmentation. Conclusions Mitochondrial dynamics play a key role in early corneal fibrogenesis, acting together with profibrotic signaling. This is consistent with mitochondria's role as signaling hubs that coordinate metabolic decision-making. This suggests a feed-forward cascade through which mitochondria, at least in part through fission, reinforce noncanonical TGF-β1 signaling to attain corneal myofibroblast differentiation.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Department of Ophthalmology, University of Rochester, Rochester, New York, United States.,Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Ankita Kumar
- Department of Ophthalmology, University of Rochester, Rochester, New York, United States
| | - Kaitlin T Wozniak
- Department of Ophthalmology, University of Rochester, Rochester, New York, United States.,Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Keith Nehrke
- Department of Medicine - Nephrology Division, University of Rochester, Rochester, New York, United States
| | - Krystel R Huxlin
- Department of Ophthalmology, University of Rochester, Rochester, New York, United States.,Center for Visual Science, University of Rochester, Rochester, New York, United States
| |
Collapse
|
8
|
Singh RB, Das S, Chodosh J, Sharma N, Zegans ME, Kowalski RP, Jhanji V. Paradox of complex diversity: Challenges in the diagnosis and management of bacterial keratitis. Prog Retin Eye Res 2021; 88:101028. [PMID: 34813978 DOI: 10.1016/j.preteyeres.2021.101028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Bacterial keratitis continues to be one of the leading causes of corneal blindness in the developed as well as the developing world, despite swift progress since the dawn of the "anti-biotic era". Although, we are expeditiously developing our understanding about the different causative organisms and associated pathology leading to keratitis, extensive gaps in knowledge continue to dampen the efforts for early and accurate diagnosis, and management in these patients, resulting in poor clinical outcomes. The ability of the causative bacteria to subdue the therapeutic challenge stems from their large genome encoding complex regulatory networks, variety of unique virulence factors, and rapid secretion of tissue damaging proteases and toxins. In this review article, we have provided an overview of the established classical diagnostic techniques and therapeutics for keratitis caused by various bacteria. We have extensively reported our recent in-roads through novel tools for accurate diagnosis of mono- and poly-bacterial corneal infections. Furthermore, we outlined the recent progress by our group and others in understanding the sub-cellular genomic changes that lead to antibiotic resistance in these organisms. Finally, we discussed in detail, the novel therapies and drug delivery systems in development for the efficacious management of bacterial keratitis.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, Leiden University Medical Center, 2333, ZA Leiden, the Netherlands
| | - Sujata Das
- Cornea and Anterior Segment Services, LV Prasad Eye Institute, Bhubaneshwar, India
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Namrata Sharma
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Michael E Zegans
- Department of Ophthalmology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Regis P Kowalski
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; The Charles T Campbell Ophthalmic Microbiology Laboratory, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Vishal Jhanji
- Department of Ophthalmology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; The Charles T Campbell Ophthalmic Microbiology Laboratory, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Nuwormegbe S, Park NY, Kim SW. Lobeglitazone attenuates fibrosis in corneal fibroblasts by interrupting TGF-beta-mediated Smad signaling. Graefes Arch Clin Exp Ophthalmol 2021; 260:149-162. [PMID: 34468828 DOI: 10.1007/s00417-021-05370-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Transforming growth factor beta 1 (TGF-β1) is an important cytokine released after ocular surface injury to promote wound healing. However, its persistence at the injury site triggers a fibrotic response that leads to corneal scarring and opacity. Thiazolidinediones (TZDs) are synthetic peroxisome proliferator-activated receptor gamma (PPAR-γ) ligands used to regulate glucose and lipid metabolism in the management of type 2 diabetes. Studies have also showed TZDs have antifibrotic effect. In this study, we investigated the antifibrotic effect of the TZD lobeglitazone on TGF-β1-induced fibrosis in corneal fibroblasts. METHODS Human primary corneal fibroblasts were cultivated and treated with TGF-β1 (5 ng/mL) to induce fibrosis, with or without pre-treatments with different concentrations of lobeglitazone. Myofibroblast differentiation and extracellular matrix (ECM) protein expression was evaluated by western blotting, immunofluorescence, real-time PCR, and collagen gel contraction assay. The effect of lobeglitazone on TGF-β1-induced reactive oxygen species (ROS) generation was evaluated by DCFDA-cellular ROS detection assay kit. Signaling proteins were evaluated by western blotting to determine the mechanism underlying the antifibrotic effect. RESULTS Our results showed lobeglitazone attenuated TGF-β1-induced ECM synthesis and myofibroblast differentiation of corneal fibroblasts. This antifibrotic effect appeared to be independent of PPAR signaling and rather due to the inhibition of the TGF-β1-induced Smad signaling. Lobeglitazone also blocked TGF-β1-induced ROS generation and nicotinamide adenine dinucleotide phosphate oxidase (Nox) 4 transcription. CONCLUSION These findings indicate that lobeglitazone may be a promising therapeutic agent for corneal scarring. KEY MESSAGES.
Collapse
Affiliation(s)
- Selikem Nuwormegbe
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Ilsan-ro, Gangwon-do, 26426, Republic of Korea
| | - Na-Young Park
- Department of Ophthalmology, Wonju College of Medicine, Yonsei University, Wonju, Ilsan-ro, Gangwon-do, 26426, Republic of Korea
| | - Sun Woong Kim
- Department of Ophthalmology, Wonju College of Medicine, Yonsei University, Wonju, Ilsan-ro, Gangwon-do, 26426, Republic of Korea.
| |
Collapse
|
10
|
Escandon P, Vasini B, Whelchel AE, Nicholas SE, Matlock HG, Ma JX, Karamichos D. The role of peroxisome proliferator-activated receptors in healthy and diseased eyes. Exp Eye Res 2021; 208:108617. [PMID: 34010603 PMCID: PMC8594540 DOI: 10.1016/j.exer.2021.108617] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/23/2022]
Abstract
Peroxisome Proliferator-Activated Receptors (PPARs) are a family of nuclear receptors that play essential roles in modulating cell differentiation, inflammation, and metabolism. Three subtypes of PPARs are known: PPAR-alpha (PPARα), PPAR-gamma (PPARγ), and PPAR-beta/delta (PPARβ/δ). PPARα activation reduces lipid levels and regulates energy homeostasis, activation of PPARγ results in regulation of adipogenesis, and PPARβ/δ activation increases fatty acid metabolism and lipolysis. PPARs are linked to various diseases, including but not limited to diabetes, non-alcoholic fatty liver disease, glaucoma and atherosclerosis. In the past decade, numerous studies have assessed the functional properties of PPARs in the eye and key PPAR mechanisms have been discovered, particularly regarding the retina and cornea. PPARγ and PPARα are well established in their functions in ocular homeostasis regarding neuroprotection, neovascularization, and inflammation, whereas PPARβ/δ isoform function remains understudied. Naturally, studies on PPAR agonists and antagonists, associated with ocular pathology, have also gained traction with the development of PPAR synthetic ligands. Studies on PPARs has significantly influenced novel therapeutics for diabetic eye disease, ocular neuropathy, dry eye, and age-related macular degeneration (AMD). In this review, therapeutic potentials and implications will be highlighted, as well as reported adverse effects. Further investigations are necessary before any of the PPARs ligands can be utilized, in the clinics, to treat eye diseases. Future research on the prominent role of PPARs will help unravel the complex mechanisms involved in order to prevent and treat ocular diseases.
Collapse
Affiliation(s)
- Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Brenda Vasini
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Amy E Whelchel
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Sarah E Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - H Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, 940 Stanton L Young, Oklahoma City, OK, USA; Harold Hamm Oklahoma Diabetes Center, 1000 N Lincoln Blvd, Oklahoma City, OK, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA; Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
11
|
Jhanji V, Billig I, Yam GHF. Cell-Free Biological Approach for Corneal Stromal Wound Healing. Front Pharmacol 2021; 12:671405. [PMID: 34122095 PMCID: PMC8193853 DOI: 10.3389/fphar.2021.671405] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Corneal opacification is the fourth most common cause of blindness globally behind cataracts, glaucoma, and age-related macular degeneration. The standard treatment of serious corneal scarring is corneal transplantation. Though it is effective for restoring vision, the treatment outcome is not optimal, due to limitations such as long-term graft survival, lifelong use of immunosuppressants, and a loss of corneal strength. Regulation of corneal stromal wound healing, along with inhibition or downregulation of corneal scarring is a promising approach to prevent corneal opacification. Pharmacological approaches have been suggested, however these are fraught with side effects. Tissue healing is an intricate process that involves cell death, proliferation, differentiation, and remodeling of the extracellular matrix. Current research on stromal wound healing is focused on corneal characteristics such as the immune response, angiogenesis, and cell signaling. Indeed, promising new technologies with the potential to modulate wound healing are under development. In this review, we provide an overview of cell-free strategies and some approaches under development that have the potential to control stromal fibrosis and scarring, especially in the context of early intervention.
Collapse
Affiliation(s)
- Vishal Jhanji
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Isabelle Billig
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gary Hin-Fai Yam
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
12
|
Temporal evolution of the biological response to laser-induced refractive index change (LIRIC) in rabbit corneas. Exp Eye Res 2021; 207:108579. [PMID: 33864783 DOI: 10.1016/j.exer.2021.108579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/22/2021] [Accepted: 04/10/2021] [Indexed: 02/06/2023]
Abstract
Laser-induced refractive index change (LIRIC) is a new, non-incisional, non-ablative, femtosecond photo-modification technique being developed for vision correction in humans. Prior, exvivo studies showed intra-tissue refractive index change to induce minimal cell death, restricted to the laser focal zone in the corneal stroma, and with no observable damage to the epithelium or endothelium. Here, we used live rabbits to ascertain longer-term consequences of LIRIC in vivo. Specifically, we assessed cell death, fibrosis, corneal nerve distribution, endothelial cell density, and corneal structure for up to 3 months after LIRIC. A +2.5 D gradient-index LIRIC Fresnel lens was inscribed inside 20 applanated corneas of Dutch Belted rabbits, over a circular region of the mid-stroma measuring 4.5 mm in diameter. Twelve additional rabbit eyes were used as applanation-only controls to differentiate the effects of laser treatment and suction applanation on biological and structural parameters. In vivo optical measurements were performed pre-operatively, then immediately, 2, 4, and 12 weeks after the procedure, to measure endothelial cell density and changes in corneal structure. Groups of four rabbits were sacrificed at 4 hours, 2, 4, and 12 weeks after LIRIC for histological determinations; the TUNEL assay was used to evaluate cell death, H&E staining was used to assess inflammatory infiltration, and immunostaining for α-smooth muscle actin (α-SMA) and βIII tubulin (Tuj-1) was performed to assess myofibroblast differentiation and corneal nerve distribution, respectively. Consistent with prior ex vivo data, only minimal cell death was observed in the laser focal zone, with TUNEL-positive cells restricted to the stromal region of refractive index change 4 h after LIRIC. No TUNEL-positive cells were evident anywhere in the cornea 2, 4, or 12 weeks after LIRIC. Applanation-only corneas were completely TUNEL-negative. Neither LIRIC-treated nor applanation-only eyes exhibited α-SMA-positive staining or altered corneal nerve distributions at any of the time points examined. In vivo confocal imaging revealed normal endothelial cell densities in all eyes (whether LIRIC-treated or applanation-only) at all time points. Optical coherence tomography showed suction applanation to cause a temporary decrease in central corneal thickness, which returned to normal within 4 h. Corneas into which LIRIC Fresnel lenses were written while applanated did not undergo major structural or shape changes beyond the temporary thinning already described for suction applanation. The present findings suggest that LIRIC patterns, which generated a clinically-relevant refractive correction in the mid-stromal region of live rabbit corneas, induced little-to-no disruption to corneal structure and biology for 3 months after the procedure. This affirms the relative safety of LIRIC and predicts that compared to traditional laser vision correction surgeries, common post-operative complications such as dry eye, haze, or patient discomfort may be entirely avoided.
Collapse
|
13
|
Chen J, Zhu J, Zhu T, Cui J, Deng Z, Chen K, Chang C, Geng Y, Chen F, Ouyang K, Xiong J, Wang M, Wang D, Zhu W. Pathological changes of frozen shoulder in rat model and the therapeutic effect of PPAR-γ agonist. J Orthop Res 2021; 39:891-901. [PMID: 33222263 DOI: 10.1002/jor.24920] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/07/2020] [Accepted: 11/19/2020] [Indexed: 02/04/2023]
Abstract
Frozen shoulder is a common shoulder disorder characterized by a gradual increase of pain and a limited range of motion. However, its pathophysiologic mechanisms remain unclear and there is no consensus as to the most effective treatment. The purpose of the study was to investigate the effect of transforming growth factor-β (TGF-β) on fibrosis and inflammatory response of the shoulder joint of rat models and to explore the therapeutic effect of the peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist. In the study, the effect of PPAR-γ agonist CDDO-IM treatment on cell proliferation, migration, and extracellular matrix proteins synthesis (vimentin, α-smooth muscle actin, collagen I, and collagen III) were tested by cell proliferation test, scratches test, real-time quantitative polymerase chain reaction, and Western blot analysis. The frozen shoulder was also established on the rat model by injecting adenovirus-TGF-β1 into rats' shoulder capsule. Pathological changes of the frozen shoulder tissue of the experimental group and PPAR-γ agonist treatment group were evaluated. The stiffness of joints of the three groups was tested. Inflammatory mediators' expression including cyclooxygenase-1, interleukin-1β, and tumor necrosis factor-α of the shoulder was tested by enzyme-linked immunosorbent assay, and the expression of extracellular matrix proteins was evaluated by hematoxylin and eosin staining and immunohistochemistry. The results showed that pathological changes of the frozen shoulder in the rat model include an abnormal proliferation of fibroblasts, infiltration of inflammatory cells, and disorder of fibrous structure, while rosiglitazone reduced the severity of the frozen shoulder in the treatment group. Clinically, PPAR-γ agonists may be a promising target for the treatment of the frozen shoulder.
Collapse
Affiliation(s)
- Jinfu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Junjun Zhu
- Department of Mechanical Engineering and Material Science, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tianfei Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jiaming Cui
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Kang Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Chongfei Chang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Yiyun Geng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Fei Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Kan Ouyang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Jianyi Xiong
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Manyi Wang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Daping Wang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
14
|
Silencing TLR4/MyD88/NF-κB Signaling Pathway Alleviated Inflammation of Corneal Epithelial Cells Infected by ISE. Inflammation 2020; 44:633-644. [PMID: 33174138 DOI: 10.1007/s10753-020-01363-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/07/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
The regulatory role of toll-like receptor 4 (TLR4) in the inactivate staphylococcus epidermidis (ISE)-induced cornea inflammation is not well investigated. Here, TLR4 silence could decrease inflammatory cytokines in corneal epithelial cells treated with ISE. The mouse corneal epithelial cells were exposed to ISE for 24 h, either alone or with the NF-κB inhibitor, TLR4 lentivirus to bilaterally (knock-down or and overexpression). The expression of TLR4 in mouse corneal epithelial cells was investigated using western blot and qRT-PCR assay. The inflammatory cytokine levels were evaluated by qRT-PCR and ELISA, respectively. The relative impact factors of TLR4-mediated NF-κB signaling detected using western blot assay. Results show the expression levels of TLR4 and some inflammatory cytokines were significantly increased in corneal epithelial cells treated with ISE. TLR4 Silence markedly decreased ISE-induced production of IL12, TNF-α, CCL5, and CCL9 in corneal epithelial cells. Furthermore, the nuclear translocation of NF-κB p65 and myeloid differentiation protein 88 (MyD88) in the cells treated with ISE were further reduced by silencing TLR4. Inhibition of TLR4-mediated NF-κB signaling by using BAY11-7082 also alleviated ISE-induced inflammation. In the rescue experiment, transfected the stable TLR4 silenced corneal epithelial cells with TLR4 overexpression lentivirus, we found that TLR4 overexpression can restore the down-regulation of TLR4 and inflammatory cytokines (IL12, TNF-α, CCL9) caused by TLR4 knocked down. Therefore, ISE-induced cornea inflammation was due to the activation of the TLR4/MyD88/NF-κB signaling pathway, and dramatically stimulated IL12, TNF-α, CCL9 secretion. TLR4 silence presented mitigates damage in corneal epithelial cells treated with ISE.
Collapse
|
15
|
Kwok SS, Wong FSY, Shih KC, Chan YK, Bu Y, Chan TCY, Ng ALK, Lo ACY, Tong L, Yam GHF, Jhanji V. Lycium barbarum Polysaccharide Suppresses Expression of Fibrotic Proteins in Primary Human Corneal Fibroblasts. J Clin Med 2020; 9:jcm9113572. [PMID: 33171906 PMCID: PMC7694544 DOI: 10.3390/jcm9113572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/24/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Objective: To study the anti-fibrotic effects of Lycium barbarum polysaccharides (LBP) on corneal stromal fibroblasts and assess LBP’s effect on cell viability. (2) Methods: Primary human corneal keratocytes of passage 3 to 6 were used for all experiments. Cells are pretreated with LBP solution for 24 h and then transforming growth factor beta 1 (TGFβ1) for 48 h and collected for experiments. Fibrotic protein analysis was performed using immunofluorescence and Western blot. The effect of LBP on cell viability was assessed using the MTS assay. (3) Results: LBP significantly reduced the expression of fibrotic proteins, including α-SMA and extracellular matrix proteins (collagen type I and III). LBP significantly decreased the viability of myofibroblasts but not the fibroblasts. Conclusions: In this study, LBP was effective in the prevention of fibrosis gene expression. Further studies to assess the underlying mechanism and pharmacological properties will facilitate the formation of a topical LBP solution for in vivo studies.
Collapse
Affiliation(s)
- Sum Sum Kwok
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Francisca Siu-Yin Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Kendrick Co Shih
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
- Correspondence:
| | - Yau-Kei Chan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Yashan Bu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Tommy Chung-Yan Chan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Alex Lap-Ki Ng
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Amy Cheuk-Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong and Hong Kong SAR, Hong Kong, China; (S.S.K.); (F.S.-Y.W.); (Y.-K.C.); (Y.B.); (T.C.-Y.C.); (A.L.-K.N.); (A.C.-Y.L.)
| | - Louis Tong
- Cornea and External Eye Disease Service, Singapore National Eye Centre, Singapore 168751, Singapore;
- Ocular Surface Research Group, Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Gary Hin-Fai Yam
- Department of Ophthalmology, University of Pittsburgh Medical Centre, Pittsburgh, PA 15213, USA; (G.H.-F.Y.); (V.J.)
| | - Vishal Jhanji
- Department of Ophthalmology, University of Pittsburgh Medical Centre, Pittsburgh, PA 15213, USA; (G.H.-F.Y.); (V.J.)
| |
Collapse
|
16
|
Kamil S, Mohan RR. Corneal stromal wound healing: Major regulators and therapeutic targets. Ocul Surf 2020; 19:290-306. [PMID: 33127599 DOI: 10.1016/j.jtos.2020.10.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/16/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022]
Abstract
Corneal stromal wound healing is a complex event that occurs to restore the transparency of an injured cornea. It involves immediate apoptosis of keratocytes followed by their activation, proliferation, migration, and trans-differentiation to myofibroblasts. Myofibroblasts contract to close the wound and secrete extracellular matrix and proteinases to remodel it. Released proteinases may degenerate the basement membrane allowing an influx of cytokines from overlying epithelium. Immune cells infiltrate the wound to clear cellular debris and prevent infections. Gradually basement membrane regenerates, myofibroblasts and immune cells disappear, abnormal matrix is resorbed, and transparency of the cornea is restored. Often this cascade deregulates and corneal opacity results. Factors that prevent corneal opacity after an injury have always intrigued the researchers. They hold clinical relevance as they can guide the outcomes of corneal surgeries. Studies in the past have shed light on the role of various factors in stromal healing. TGFβ (transforming growth factor-beta) signaling is the central player guiding stromal responses. Other major regulators include myofibroblasts, basement membrane, collagen fibrils, small leucine-rich proteoglycans, biophysical cues, proteins derived from extracellular matrix, and membrane channels. The knowledge about their roles helped to develop novel therapies to prevent corneal opacity. This article reviews the role of major regulators that determine the outcome of stromal healing. It also discusses emerging therapies that modulate the role of these regulators to prevent stromal opacity.
Collapse
Affiliation(s)
- Sabeeh Kamil
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; One-Health Vision Research Program, Department of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Rajiv R Mohan
- Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA; One-Health Vision Research Program, Department of Veterinary Medicine & Surgery and Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA; Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
17
|
Systematic Review on Therapeutic Strategies to Minimize Corneal Stromal Scarring After Injury. Eye Contact Lens 2020; 45:347-355. [PMID: 30724841 DOI: 10.1097/icl.0000000000000584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To evaluate recent studies on available and experimental therapies in preventing or minimizing corneal stromal scarring after injury. METHODS We performed an Entrez PubMed literature search using keywords "cornea," "scarring," "haze," "opacity," "ulcer," "treatments," "therapies," "treatment complications," and "pathophysiology" resulting in 390 articles of which 12 were analyzed after filtering, based on English language and publication within 8 years, and curation for relevance by the authors. RESULTS The 12 articles selected included four randomized control trials (RCTs) (two were double-blinded placebo-controlled RCTs, one was a prospective partially masked RCT, and one was an open-label RCT), two retrospective observational studies, and six laboratory-based studies including two studies having in vivo and in vitro experiments, one was in vivo study, one was ex vivo study, and the last two were in vitro studies. The current mainstay for preventing or minimizing corneal scarring involves the use of topical corticosteroids and local application of mitomycin C. However, supportive evidence for their use in clinical practice from well-designed RCTs is lacking. Laboratory studies on topical rosiglitazone therapy, vitamin C prophylaxis, gene therapy, and stem cell therapy have shown promising results but have yet to be translated to clinical research. CONCLUSION There is a need for more robust randomized controlled trials to support treatments using topical corticosteroids and mitomycin C. Furthermore, their clinical efficacy and safety profile should be compared with new treatments that have shown promising results in the laboratory setting. Ultimately, the goal should be to personalize cornea scarring treatment according to the most effective treatment for the specific underlying pathology.
Collapse
|
18
|
Shen T, Zheng Q, Luo H, Li X, Chen Z, Song Z, Zhou G, Hong C. Exosomal miR-19a from adipose-derived stem cells suppresses differentiation of corneal keratocytes into myofibroblasts. Aging (Albany NY) 2020; 12:4093-4110. [PMID: 32112551 PMCID: PMC7093196 DOI: 10.18632/aging.102802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 01/19/2020] [Indexed: 04/11/2023]
Abstract
In this study, we investigated the effects of exosomal microRNAs (miRNAs) from adipose-derived stem cells (ADSCs) on the differentiation of rabbit corneal keratocytes. Keratocytes grown in 10% FBS differentiated into myofibroblasts by increasing HIPK2 kinase levels and activity. HIPK2 enhanced p53 and Smad3 pathways in FBS-induced keratocytes. Keratocytes grown in 10% FBS also showed increased levels of pro-fibrotic proteins, including collagen III, MMP9, fibronectin, and α-SMA. These effects were reversed by knocking down HIPK2. Moreover, ADSCs and exosomes derived from ADSCs (ADSCs-Exo) suppressed FBS-induced differentiation of keratocytes into myofibroblasts by inhibiting HIPK2. Quantitative RT-PCR analysis showed that ADSCs-Exos were significantly enriched in miRNA-19a as compared to ADSCs. Targetscan and dual luciferase reporter assays confirmed that the HIPK2 3'UTR is a direct binding target of miR-19a. Keratocytes treated with 10% FBS and ADSCs-Exo-miR-19a-agomir or ADSCs-Exo-NC-antagomir showed significantly lower levels of HIPK2, phospho-Smad3, phospho-p53, collagen III, MMP9, fibronectin and α-SMA than those treated with 10% FBS plus ADSCs-Exo-NC-agomir or ADSCs-Exo-miR-19a-antagomir. Thus, exosomal miR-19a derived from the ADSCs suppresses FBS-induced differentiation of rabbit corneal keratocytes into myofibroblasts by inhibiting HIPK2 expression. This suggests their potential use in the treatment of corneal fibrosis.
Collapse
Affiliation(s)
- Ting Shen
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital and People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, P. R. China
| | - Qingqing Zheng
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital and People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, P. R. China
| | - Hongbo Luo
- Department of Ophthalmology, Zhejiang Provincial People’s Hospital and People’s Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, P. R. China
| | - Xin Li
- Wenzhou School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Zhuo Chen
- Bengbu Medical College, Bengbu 233030, Anhui, P. R. China
| | - Zeyu Song
- Wenzhou School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Guanfang Zhou
- Bengbu Medical College, Bengbu 233030, Anhui, P. R. China
| | - Chaoyang Hong
- Wenzhou School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
- Department of Ophthalmology, Zhejiang Hospital, Hangzhou 310007, Zhejiang, P. R. China
| |
Collapse
|
19
|
Jeon KI, Nehrke K, Huxlin KR. Semaphorin 3A potentiates the profibrotic effects of transforming growth factor-β1 in the cornea. Biochem Biophys Res Commun 2020; 521:333-339. [PMID: 31668808 DOI: 10.1016/j.bbrc.2019.10.107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/12/2019] [Indexed: 11/16/2022]
Abstract
Corneal scarring is a major cause of blindness worldwide with few effective therapeutic options. Finding a treatment would be of tremendous public health benefit, but requires a thorough understanding of the complex interactions that underlie this phenomenon. Here, we tested the hypothesis that the large increase in expression of Semaphorin 3A (SEMA3A) in corneal wounds contributes to the development of stromal fibrosis. We first verified this increased expression in vivo, in a cat model of photorefractive keratectomy-induced corneal wounding. We then examined the impact of adding exogenous SEMA3A to cultured corneal fibroblasts, and assessed how this affected the ability of transforming growth factor-beta1 (TGF-β1) to induce their differentiation into myofibroblasts. Finally, we examined how siRNA knockdown of endogenous SEMA3A affected these same phenomena. We found exogenous SEMA3A to significantly potentiate TGF-β1's profibrotic effects, with only a minimal contribution from cell-intrinsic SEMA3A. Our results suggest a previously unrecognized interaction between SEMA3A and TGF-β1 in the wounded cornea, and a possible contribution of SEMA3A to the regulation of tissue fibrosis and remodeling in this transparent organ.
Collapse
Affiliation(s)
| | - Keith Nehrke
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
20
|
Hyperlipidemia induces meibomian gland dysfunction. Ocul Surf 2019; 17:777-786. [PMID: 31201956 DOI: 10.1016/j.jtos.2019.06.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/28/2019] [Accepted: 06/10/2019] [Indexed: 11/20/2022]
Abstract
PURPOSE To investigate the pathological changes of the meibomian gland (MG) and ocular surface in Apolipoprotein E knockout (ApoE-/-) mice and to investigate the association of meibomian gland dysfunction (MGD) with hyperlipidemia. METHODS Total plasma cholesterol was measured in different ages of ApoE-/- and wild type (WT) mice, whilst the ocular surfaces were observed by slit-lamp biomicroscopy. MG sections were subjected to H&E staining, Oil Red O staining, TUNEL assay and immunostaining. Quantitate RT-PCR and Western blot analyses were performed to detect the relative gene expression in MGs. The 5-month-old ApoE-/- mice were administered with rosiglitazone or GW9662 + rosiglitazone via oral gavage for 2 months to determine their effect on MG pathological change. RESULTS We found eyelid abnormality, MG dropout, abnormal MG acinar morphology, dilated MG duct and plugging of the MG orifice in ApoE-/- mice. MG acini in ApoE-/- mice showed exaggerated lipid accumulation. Abnormal keratinization increased in MG duct, accompanied with decreased proliferation and increased apoptosis in ApoE-/- mice. Inflammatory cells infiltrated into the surrounding microenvironment of MG acini, and the NF-κB signaling pathway was activated in MG acinar cells. Oxidative stress was evident in MG acinar cells of ApoE-/- mice. Further investigation showed downregulation of PPAR-γ in MG acinar cells of ApoE-/- mice. PPAR-γ agonist rosiglitazone treatment reduced the morbidity of eyelid, as well as corneal pathological changes and MG inflammation in ApoE-/- mice. CONCLUSION MGD and hyperlipidemia are closely associated in ApoE-/- mice, which represent a new model to study the pathophysiology of MGD related to dyslipidemia.
Collapse
|
21
|
Hindman HB, DeMagistris M, Callan C, McDaniel T, Bubel T, Huxlin KR. Impact of topical anti-fibrotics on corneal nerve regeneration in vivo. Exp Eye Res 2019; 181:49-60. [PMID: 30660507 PMCID: PMC6443430 DOI: 10.1016/j.exer.2019.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 12/12/2022]
Abstract
Recent work in vitro has shown that fibroblasts and myofibroblasts have opposing effects on neurite outgrowth by peripheral sensory neurons. Here, we tested a prediction from this work that dampening the fibrotic response in the early phases of corneal wound healing in vivo could enhance reinnervation after a large, deep corneal injury such as that induced by photorefractive keratectomy (PRK). Since topical steroids and Mitomycin C (MMC) are often used clinically for mitigating corneal inflammation and scarring after PRK, they were ideal to test this prediction. Twenty adult cats underwent bilateral, myopic PRK over a 6 mm optical zone followed by either: (1) intraoperative MMC (n = 12 eyes), (2) intraoperative prednisolone acetate (PA) followed by twice daily topical application for 14 days (n = 12 eyes), or (3) no post-operative treatment (n = 16 eyes). Anti-fibrotic effects of MMC and PA were verified optically and histologically. First, optical coherence tomography (OCT) performed pre-operatively and 2, 4 and 12 weeks post-PRK was used to assess changes in corneal backscatter reflectivity. Post-mortem immunohistochemistry was then performed at 2, 4 and 12 weeks post-PRK, using antibodies against α-smooth muscle actin (α-SMA). Finally, immunohistochemistry with antibodies against βIII-tubulin (Tuj-1) was performed in the same corneas to quantify changes in nerve distribution relative to unoperated, control cat corneas. Two weeks after PRK, untreated corneas exhibited the greatest amount of staining for α-SMA, followed by PA-treated and MMC-treated eyes. This was matched by higher OCT-based stromal reflectivity values in untreated, than PA- and MMC-treated eyes. PA treatment appeared to slow epithelial healing and although normal epithelial thickness was restored by 12 weeks-post-PRK, intra-epithelial nerve length only reached ∼1/6 normal values in PA-treated eyes. Even peripheral cornea (outside the ablation zone) exhibited depressed intra-epithelial nerve densities after PA treatment. Stromal nerves were abundant under the α-SMA zone, but appeared to largely avoid it, creating an area of sub-epithelial stroma devoid of nerve trunks. In turn, this may have led to the lack of sub-basal and intra-epithelial nerves in the ablation zone of PA-treated eyes 4 weeks after PRK, and their continuing paucity 12 weeks after PRK. Intra-operative MMC, which sharply decreased α-SMA staining, was followed by rapid restoration of nerve densities in all corneal layers post-PRK compared to untreated corneas. Curiously, stromal nerves appeared unaffected by the development of large, stromal, acellular zones in MMC-treated corneas. Overall, it appears that post-PRK treatments that were most effective at reducing α-SMA-positive cells in the early post-operative period benefited nerve regeneration the most, resulting in more rapid restoration of nerve densities in all corneal layers of the ablation zone and of the corneal periphery.
Collapse
Affiliation(s)
- Holly B Hindman
- The Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA; Center for Visual Science, University of Rochester, Rochester, NY, 14627, USA
| | | | - Christine Callan
- The Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
| | - Thurma McDaniel
- The Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA
| | - Tracy Bubel
- Center for Visual Science, University of Rochester, Rochester, NY, 14627, USA
| | - Krystel R Huxlin
- The Flaum Eye Institute, University of Rochester, Rochester, NY, 14642, USA; Center for Visual Science, University of Rochester, Rochester, NY, 14627, USA.
| |
Collapse
|
22
|
Corneal myofibroblasts inhibit regenerating nerves during wound healing. Sci Rep 2018; 8:12945. [PMID: 30154512 PMCID: PMC6113331 DOI: 10.1038/s41598-018-30964-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
Abnormal nerve regeneration often follows corneal injury, predisposing patients to pain, dry eye and vision loss. Yet, we lack a mechanistic understanding of this process. A key event in corneal wounds is the differentiation of keratocytes into fibroblasts and scar-forming myofibroblasts. Here, we show for the first time that regenerating nerves avoid corneal regions populated by myofibroblasts in vivo. Recreating this interaction in vitro, we find neurite outgrowth delayed when myofibroblasts but not fibroblasts, are co-cultured with sensory neurons. After neurites elongated sufficiently, contact inhibition was observed with myofibroblasts, but not fibroblasts. Reduced neurite outgrowth in vitro appeared mediated by transforming growth factor beta 1 (TGF-β1) secreted by myofibroblasts, which increased phosphorylation of collapsin response mediating protein 2 (CRMP2) in neurons. The significance of this mechanism was further tested by applying Mitomycin C after photorefractive keratectomy to decrease myofibroblast differentiation. This generated earlier repopulation of the ablation zone by intra-epithelial and sub-basal nerves. Our findings suggest that attaining proper, rapid corneal nerve regeneration after injury may require blocking myofibroblast differentiation and/or TGF-β during wound healing. They also highlight hitherto undefined myofibroblast-neuron signaling processes capable of restricting neurite outgrowth in the cornea and other tissues where scars and nerves co-exist.
Collapse
|
23
|
Liu K, Zou C, Qin B. The association between nuclear receptors and ocular diseases. Oncotarget 2018; 8:27603-27615. [PMID: 28187442 PMCID: PMC5432361 DOI: 10.18632/oncotarget.15178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/24/2017] [Indexed: 01/18/2023] Open
Abstract
Nuclear hormone receptors (NRs) are one of the most abundant transcription factors in the human cells. They regulate expression of genes via interactions with corresponding ligands, co-activators, and co-repressors. These molecular pathways play important roles in the development, cell differentiation, and physiologic and metabolic processes. Increasingly, targeting nuclear receptors is becoming a promising strategy for new drug development. The aim of this review is to discuss the association between nuclear receptors and eye development, and expand their role in various ocular diseases such as keratitis, cataract, glaucoma, uveitis, retinopathy, and ophthalmic tumors. Recent studies in this area are highlighted as well as future research directions and potential clinical applications. Finally, various strategies will be elucidated to inspire more targeted therapies for ocular diseases through the use of nuclear receptors.
Collapse
Affiliation(s)
- Ke Liu
- Jinan University, Guangzhou, China.,Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry of Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Ocular Trauma Treatment and Stem Cell Differentiation Public Service Platform of Shenzhen, Shenzhen, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical College, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Bo Qin
- Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Jinan University, Joint College of Optometry of Shenzhen University, Shenzhen Key Laboratory of Ophthalmology, Ocular Trauma Treatment and Stem Cell Differentiation Public Service Platform of Shenzhen, Shenzhen, China
| |
Collapse
|
24
|
Zada M, Pattamatta U, White A. Modulation of Fibroblasts in Conjunctival Wound Healing. Ophthalmology 2017; 125:179-192. [PMID: 29079272 DOI: 10.1016/j.ophtha.2017.08.028] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/18/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022] Open
Abstract
Modulating conjunctival wound healing has the potential to improve outcomes after glaucoma filtration surgery and for several ocular disorders, including ocular cicatrial pemphigoid, vernal keratoconjunctivitis, and pterygium. Although anti-inflammatories and antimetabolites have been used with success, these nonspecific agents are not without their complications. The search for novel and more targeted means to control conjunctival fibrosis without such limitations has brought much attention to the regulation of fibroblast proliferation, differentiation, extracellular matrix production, and apoptosis. This review provides an update on where we stand with current antifibrotic agents and outlines the strategies that novel agents use, as they evolve from the bench to the bedside.
Collapse
Affiliation(s)
- Mark Zada
- Glaucoma Cell Biology Group, The Westmead Institute for Medical Research, NSW, Australia; Discipline of Ophthalmology, Sydney Medical School, University of Sydney, NSW, Australia.
| | - Ushasree Pattamatta
- Glaucoma Cell Biology Group, The Westmead Institute for Medical Research, NSW, Australia; Discipline of Ophthalmology, Sydney Medical School, University of Sydney, NSW, Australia
| | - Andrew White
- Glaucoma Cell Biology Group, The Westmead Institute for Medical Research, NSW, Australia; Discipline of Ophthalmology, Sydney Medical School, University of Sydney, NSW, Australia; Save Sight Institute, University of Sydney, NSW, Australia
| |
Collapse
|
25
|
Jeon KI, Phipps RP, Sime PJ, Huxlin KR. Antifibrotic Actions of Peroxisome Proliferator-Activated Receptor γ Ligands in Corneal Fibroblasts Are Mediated by β-Catenin-Regulated Pathways. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1660-1669. [PMID: 28606794 DOI: 10.1016/j.ajpath.2017.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 01/08/2023]
Abstract
Wound healing after corneal injury typically involves fibrosis, with transforming growth factor β1 (TGF-β1) as one of its strongest mediators. A class of small molecules-peroxisome proliferator-activated receptor γ (PPARγ) ligands-exert potent antifibrotic effects in the cornea by blocking phosphorylation of p38 mitogen-activated protein kinase (MAPK). However, why this blocks fibrosis remains unknown. Herein, we show that PPARγ ligands (rosiglitazone, troglitazone, and 15-deoxy-Δ12,14-prostaglandin J2) decrease levels of β-catenin. We also show that β-catenin siRNA and the Wingless/integrated (Wnt) inhibitor pyrvinium block the ability of corneal fibroblasts to up-regulate synthesis of α-smooth muscle actin (α-SMA), collagen 1 (COL1), and fibronectin (FN) in response to TGF-β1. Activation of TGF-β receptors and p38 MAPK increased glycogen synthase kinase 3β (GSK3β) phosphorylation, whereas a chemical inhibitor of p38 MAPK (SB203580) reduced the phosphorylation of GSK3β, decreasing active β-catenin levels in both cytoplasmic and nuclear fractions. Finally, lithium chloride, a GSK3 inhibitor, also attenuated the TGF-β1-induced increase in α-SMA, COL1, and FN expression. All in all, our results suggest that TGF-β1 stimulation increases active β-catenin concentration in cultured corneal fibroblasts through p38 MAPK regulation of canonical Wnt/β-catenin signaling, increasing α-SMA, COL1, and FN synthesis. Thus, PPARγ ligands, by blocking TGF-β1-induced p38 MAPK phosphorylation, prevent increases in both total and active β-catenin through p38 MAPK-GSK3β signaling.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Richard P Phipps
- Flaum Eye Institute, University of Rochester, Rochester, New York; Department of Medicine, University of Rochester, Rochester, New York; Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Patricia J Sime
- Department of Medicine, University of Rochester, Rochester, New York; Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Krystel R Huxlin
- Flaum Eye Institute, University of Rochester, Rochester, New York; Center for Visual Science, University of Rochester, Rochester, New York.
| |
Collapse
|
26
|
An E, Park H, Lee ARC. Inhibition of fibrotic contraction by C-phycocyanin through modulation of connective tissue growth factor and α-smooth muscle actin expression. Tissue Eng Regen Med 2016; 13:388-395. [PMID: 30603420 DOI: 10.1007/s13770-015-0104-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 12/22/2022] Open
Abstract
The effects of C-phycocyanin (C-pc), a phycobiliprotein, on the expression of pro-fibrotic mediators in hyper-tropic scarring such as connective tissue growth factor (CTGF) and α-smooth muscle actins (α-SMA) were investigated in relation to trans-differentiation of fibroblast to myo-fibroblast, an icon of scar formation. C-pc was isolated from Spirulina Platensis extract using sonication method and C-pc concentration was determined by Bennet and Bogorad equation. α-SMA and CTGF levels in wounded primary human dermal fibroblasts were determined by western blot analysis and immuno-fluorescence confocal microscope was employed. Fibroblast contractility was examined by three-dimensional collagen lattice contraction assay. There was an elevation of α-SMA (121%) and CTGF (143%) levels in wound cells as compared with non-wound cells. The does-response profiles of down regulation demonstrated that the maximum inhibitions of α-SMA by 63% (p<0.05) and CTGF by 50% (p<0.1) were achieved by C-pc (6 nM) treated cells. In confocal assay, non-wound fibroblasts exhibited basal level of α-SMA staining, while wounded cells without C-pc treatment showed strong up-regulation of α-SMA by 147% (p<0.05). C-pc (6 nM) inhibited α-SMA expression by 70% (p<0.05) and reduced collagen contraction by 29% (p<0.05). C-pc seemed to lessen the over expression of CTGF, α-SMA, subsequently alleviating the fibrotic contracture. This study suggests the potential application of C-pc to regulation of the expression of pro-fibrotic mediators in scarring process and its potential usage as an efficient means for anti-fibrosis therapy.
Collapse
Affiliation(s)
- Eunjin An
- 1College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Hyunju Park
- 1College of Pharmacy, Duksung Women's University, Seoul, Korea
| | - Ae-Ri Cho Lee
- 1College of Pharmacy, Duksung Women's University, Seoul, Korea.,2College of Pharmacy, Duksung Women's University, 33 Samyang-ro 144-gil, Dobong-gu, Seoul, 01369 Korea
| |
Collapse
|
27
|
Abstract
Corneal wound healing is a complex process involving cell death, migration, proliferation, differentiation, and extracellular matrix remodeling. Many similarities are observed in the healing processes of corneal epithelial, stromal and endothelial cells, as well as cell-specific differences. Corneal epithelial healing largely depends on limbal stem cells and remodeling of the basement membrane. During stromal healing, keratocytes get transformed to motile and contractile myofibroblasts largely due to activation of transforming growth factor-β (TGF-β) system. Endothelial cells heal mostly by migration and spreading, with cell proliferation playing a secondary role. In the last decade, many aspects of wound healing process in different parts of the cornea have been elucidated, and some new therapeutic approaches have emerged. The concept of limbal stem cells received rigorous experimental corroboration, with new markers uncovered and new treatment options including gene and microRNA therapy tested in experimental systems. Transplantation of limbal stem cell-enriched cultures for efficient re-epithelialization in stem cell deficiency and corneal injuries has become reality in clinical setting. Mediators and course of events during stromal healing have been detailed, and new treatment regimens including gene (decorin) and stem cell therapy for excessive healing have been designed. This is a very important advance given the popularity of various refractive surgeries entailing stromal wound healing. Successful surgical ways of replacing the diseased endothelium have been clinically tested, and new approaches to accelerate endothelial healing and suppress endothelial-mesenchymal transformation have been proposed including Rho kinase (ROCK) inhibitor eye drops and gene therapy to activate TGF-β inhibitor SMAD7. Promising new technologies with potential for corneal wound healing manipulation including microRNA, induced pluripotent stem cells to generate corneal epithelium, and nanocarriers for corneal drug delivery are discussed. Attention is also paid to problems in wound healing understanding and treatment, such as lack of specific epithelial stem cell markers, reliable identification of stem cells, efficient prevention of haze and stromal scar formation, lack of data on wound regulating microRNAs in keratocytes and endothelial cells, as well as virtual lack of targeted systems for drug and gene delivery to select corneal cells.
Collapse
Affiliation(s)
- Alexander V Ljubimov
- Eye Program, Board of Governors Regenerative Medicine Institute, Departments of Biomedical Sciences and Neurosurgery, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
| | - Mehrnoosh Saghizadeh
- Eye Program, Board of Governors Regenerative Medicine Institute, Departments of Biomedical Sciences and Neurosurgery, Cedars-Sinai Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
28
|
Jeon KI, Phipps RP, Sime PJ, Huxlin KR. Inhibitory effects of PPARγ ligands on TGF-β1-induced CTGF expression in cat corneal fibroblasts. Exp Eye Res 2015; 138:52-8. [PMID: 26142957 DOI: 10.1016/j.exer.2015.06.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/24/2015] [Accepted: 06/30/2015] [Indexed: 12/16/2022]
Abstract
Ligands of Peroxisome Proliferator Activated Receptor gamma (PPARγ) possess strong anti-fibrotic properties in the cornea and several other body tissues. In the cornea, we recently showed this class of molecules to prevent stromal myofibroblast differentiation partially by blocking the actions of p38 mitogen-activated protein kinase (MAPK). However, given the important role assigned to connective tissue growth factor (CTGF) in mediating corneal fibrosis, here we asked whether PPARγ ligands also act by affecting transforming growth factor-β (TGF-β) 1-induced expression of CTGF in cultured corneal fibroblasts. Corneal keratocytes were isolated from young, adult cats and early passage cells were exposed to TGF-β1 with or without the PPARγ ligands Rosiglitazone, Troglitazone and 15d-PGJ2. Western blots were used to assay levels of CTGF and alpha smooth muscle actin (αSMA), a marker of myofibroblast differentiation. CTGF siRNA demonstrated a critical role for CTGF in TGF-β1-mediated myofibroblast differentiation, while exogenously applied CTGF potentiated the pro-fibrogenic effects of TGF-β1. TGF-β1-mediated increases in CTGF and αSMA expression were strongly inhibited by all three PPARγ ligands tested, and by a c-jun N-terminal kinase (JNK) inhibitor. However, while extracellular signal-regulated kinase (ERK) 1/2, protein kinase B (AKT) and p38 MAPK inhibitors also blocked TGF-β1-induced αSMA induction, they did not dampen TGF-β1-induced increases in levels of CTGF. Thus, we conclude that PPARγ ligands block TGF-β1-induced increases in CTGF levels in cat corneal fibroblasts. They appear to do this in addition to their anti-fibrotic effect on p38 MAPK, providing a second intracellular pathway by which PPARγ ligands block αSMA induction.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Flaum Eye Institute, University of Rochester, Rochester, NY, USA
| | - Richard P Phipps
- Flaum Eye Institute, University of Rochester, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester, Rochester, NY, USA; Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Patricia J Sime
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA; Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Krystel R Huxlin
- Flaum Eye Institute, University of Rochester, Rochester, NY, USA; Center for Visual Science, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
29
|
Role of Peroxisome Proliferator-Activated Receptor γ in Ocular Diseases. J Ophthalmol 2015; 2015:275435. [PMID: 26146566 PMCID: PMC4471377 DOI: 10.1155/2015/275435] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/19/2015] [Indexed: 01/14/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPAR γ), a member of the nuclear receptor superfamily, is a ligand-activated transcription factor that plays an important role in the control of a variety of physiological processes. The last decade has witnessed an increasing interest for the role played by the agonists of PPAR γ in antiangiogenesis, antifibrosis, anti-inflammation effects and in controlling oxidative stress response in various organs. As the pathologic mechanisms of major blinding diseases, such as age-related macular degeneration (AMD), diabetic retinopathy (DR), keratitis, and optic neuropathy, often involve neoangiogenesis and inflammation- and oxidative stress-mediated cell death, evidences are accumulating on the potential benefits of PPAR γ to improve or prevent these vision threatening eye diseases. In this paper we describe what is known about the role of PPAR γ in the ocular pathophysiological processes and PPAR γ agonists as novel adjuvants in the treatment of eye diseases.
Collapse
|
30
|
Science and Art of Cell-Based Ocular Surface Regeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 319:45-106. [DOI: 10.1016/bs.ircmb.2015.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
The Antifibrosis Effects of Peroxisome Proliferator-Activated Receptor δ on Rat Corneal Wound Healing after Excimer Laser Keratectomy. PPAR Res 2014; 2014:464935. [PMID: 25477952 PMCID: PMC4248330 DOI: 10.1155/2014/464935] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/17/2014] [Indexed: 12/16/2022] Open
Abstract
Corneal stromal fibrosis characterized by myofibroblasts and abnormal extracellular matrix (ECM) is usually the result of inappropriate wound healing. The present study tested the hypothesis that the ligand activation of peroxisome proliferator-activated receptor (PPAR) δ had antifibrosis effects in a rat model of corneal damage. Adult Sprague-Dawley rats underwent bilateral phototherapeutic keratectomy (PTK). The eyes were randomized into four groups: PBS, GW501516 (a selective agonist of PPARδ), GSK3787 (a selective antagonist of PPARδ), or GW501516 combined with GSK3787. The agents were subconjunctivally administered twice a week until sacrifice. The cellular aspects of corneal wound healing were evaluated with in vivo confocal imaging and postmortem histology. A myofibroblast marker (α-smooth muscle actin) and ECM production (fibronectin, collagen type III and collagen type I) were examined by immunohistochemistry and RT-PCR. At the early stages of wound healing, GW501516 inhibited reepithelialization and promoted angiogenesis. During the remodeling phase of wound healing, GW501516 attenuated the activation and proliferation of keratocytes, which could be reversed by GSK3787. GW501516 decreased transdifferentiation from keratocytes into myofibroblasts, ECM synthesis, and corneal haze. These results demonstrate that GW501516 controls corneal fibrosis and suggest that PPARδ may potentially serve as a therapeutic target for treating corneal scars.
Collapse
|
32
|
Savage DE, Brooks DR, DeMagistris M, Xu L, MacRae S, Ellis JD, Knox WH, Huxlin KR. First demonstration of ocular refractive change using blue-IRIS in live cats. Invest Ophthalmol Vis Sci 2014; 55:4603-12. [PMID: 24985471 DOI: 10.1167/iovs.14-14373] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine the efficacy of intratissue refractive index shaping (IRIS) using 400-nm femtosecond laser pulses (blue light) for writing refractive structures directly into live cat corneas in vivo, and to assess the longevity of these structures in the eyes of living cats. METHODS Four eyes from two adult cats underwent Blue-IRIS. Light at 400 nm with 100-femtosecond (fs) pulses were tightly focused into the corneal stroma of each eye at an 80-MHz repetition rate. These pulses locally increased the refractive index of the corneal stroma via an endogenous, two-photon absorption process and were used to inscribe three-layered, gradient index patterns into the cat corneas. The optical effects of the patterns were then tracked using optical coherence tomography (OCT) and Shack-Hartmann wavefront sensing. RESULTS Blue-IRIS patterns locally changed ocular cylinder by -1.4 ± 0.3 diopters (D), defocus by -2.0 ± 0.5 D, and higher-order root mean square (HORMS) by 0.31 ± 0.04 μm at 1 month post-IRIS, without significant changes in corneal thickness or curvature. Refractive changes were maintained for the duration they were tracked, 12 months post-IRIS in one eye, and just more than 3 months in the remaining three eyes. CONCLUSIONS Blue-IRIS can be used to inscribe refractive structures into live cat cornea in vivo that are stable for at least 12 months, and are not associated with significant alterations in corneal thicknesses or radii of curvature. This result is a critical step toward establishing Blue-IRIS as a promising technique for noninvasive vision correction.
Collapse
Affiliation(s)
- Daniel E Savage
- The Institute of Optics, University of Rochester, Rochester, New York, United States Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Daniel R Brooks
- The Institute of Optics, University of Rochester, Rochester, New York, United States
| | - Margaret DeMagistris
- Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| | - Lisen Xu
- The Institute of Optics, University of Rochester, Rochester, New York, United States
| | - Scott MacRae
- Center for Visual Science, University of Rochester, Rochester, New York, United States Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| | - Jonathan D Ellis
- The Institute of Optics, University of Rochester, Rochester, New York, United States Department of Mechanical Engineering, University of Rochester, Rochester, New York, United States
| | - Wayne H Knox
- The Institute of Optics, University of Rochester, Rochester, New York, United States Center for Visual Science, University of Rochester, Rochester, New York, United States
| | - Krystel R Huxlin
- Center for Visual Science, University of Rochester, Rochester, New York, United States Flaum Eye Institute, University of Rochester, Rochester, New York, United States
| |
Collapse
|
33
|
Karamichos D, Hutcheon AEK, Zieske JD. Reversal of fibrosis by TGF-β3 in a 3D in vitro model. Exp Eye Res 2014; 124:31-6. [PMID: 24800655 DOI: 10.1016/j.exer.2014.04.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 03/16/2014] [Accepted: 04/25/2014] [Indexed: 11/26/2022]
Abstract
Corneal scarring following moderate to severe injury is inevitable. Despite significant advancements in the field, current treatments following these types of injuries are limited, and often, the visual recovery is poor. One of the problems and limitations is that corneal wound healing is a complex process, involving corneal cells, extracellular matrix components and growth factors. Therefore, further understanding is required, along with new treatments and techniques to reduce or prevent corneal scarring following injury. Two isoforms of transforming growth factor-beta (TGF-β), TGF-β1 and -β3 (T1 and T3, respectively), are associated with corneal wound healing. T1 has been shown to drive the corneal keratocytes to differentiate into myofibroblasts; whereas, T3 has been found to inhibit fibrotic markers. In the current study, we examined whether the fibrotic characteristics expressed by human corneal fibroblasts (HCF) in our 3-dimensional (3D) construct following T1 stimulation could be reversed by introducing T3 to the in vitro system. To do this, HCF were isolated and cultured in 10% serum, and when they reached confluence, the cells were stimulated with a stable Vitamin C (VitC) derivative for 4 weeks, which allowed them to secrete a self-assembled matrix. Three conditions were tested: (1) CONTROL: 10% serum (S) only, (2) T1: 10%S + T1, or (3) Rescue: 10%S + T1 for two weeks and then switched to 10%S + T3 for another two weeks. At the end of 4 weeks, the constructs were processed for analysis by indirect-immunofluorescence (IF) and transmission electron microscopy (TEM). Different collagens that are normally present in healthy corneas in vivo, such as Type I and V, as well as Type III, which is a fibrotic indicator, were examined. In addition, we examined smooth muscle actin (SMA), a marker of myofibroblasts, and thrombospondin-1 (TSP-1), a multifunctional matrix protein known to activate the latent complex of TGF-β and appear upon wounding in vivo. Our data showed high expression of collagens type I and V under all conditions throughout the 3D constructs; however, type III and SMA expression were higher in the constructs that were stimulated with T1 and reduced to almost nothing in the Rescue samples. A similar pattern was seen with TSP-1, where TSP-1 expression following "rescue" was decreased considerably. Overall, this data is in agreement with our previous observations that T3 has a significant non-fibrotic effect on HCFs, and presents a novel model for the "rescue" of both cellular and matrix fibrotic components with a single growth factor.
Collapse
Affiliation(s)
- D Karamichos
- Schepens Eye Research Institute/Massachusetts Eye and Ear and the Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, USA.
| | - A E K Hutcheon
- Schepens Eye Research Institute/Massachusetts Eye and Ear and the Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, USA.
| | - J D Zieske
- Schepens Eye Research Institute/Massachusetts Eye and Ear and the Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, USA.
| |
Collapse
|
34
|
Jeon KI, Kulkarni A, Woeller CF, Phipps RP, Sime PJ, Hindman HB, Huxlin KR. Inhibitory effects of PPARγ ligands on TGF-β1-induced corneal myofibroblast transformation. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1429-45. [PMID: 24650561 DOI: 10.1016/j.ajpath.2014.01.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/08/2014] [Accepted: 01/16/2014] [Indexed: 01/11/2023]
Abstract
Corneal scarring, whether caused by trauma, laser refractive surgery, or infection, remains a significant problem for humans. Certain ligands for peroxisome proliferator-activated receptor gamma (PPARγ) have shown promise as antiscarring agents in a variety of body tissues. In the cornea, their relative effectiveness and mechanisms of action are still poorly understood. Here, we contrasted the antifibrotic effects of three different PPARγ ligands (15-deoxy-Δ12,14-prostaglandin J2, troglitazone, and rosiglitazone) in cat corneal fibroblasts. Western blot analyses revealed that all three compounds reduced transforming growth factor (TGF)-β1-driven myofibroblast differentiation and up-regulation of α-smooth muscle actin, type I collagen, and fibronectin expression. Because these effects were independent of PPARγ, we ascertained whether they occurred by altering phosphorylation of Smads 2/3, p38 mitogen-activated protein kinase, stress-activated protein kinase, protein kinase B, extracellular signal-regulated kinase, and/or myosin light chain 2. Only p38 mitogen-activated protein kinase phosphorylation was significantly inhibited by all three PPARγ ligands. Finally, we tested the antifibrotic potential of troglitazone in a cat model of photorefractive keratectomy-induced corneal injury. Topical application of troglitazone significantly reduced α-smooth muscle actin expression and haze in the stromal ablation zone. Thus, the PPARγ ligands tested here showed great promise as antifibrotics, both in vitro and in vivo. Our results also provided new evidence for the signaling pathways that may underlie these antifibrotic actions in corneal fibroblasts.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Ajit Kulkarni
- Department of Medicine, University of Rochester, Rochester, New York
| | - Collynn F Woeller
- Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Richard P Phipps
- Flaum Eye Institute, University of Rochester, Rochester, New York; Department of Medicine, University of Rochester, Rochester, New York; Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Patricia J Sime
- Department of Medicine, University of Rochester, Rochester, New York; Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Holly B Hindman
- Flaum Eye Institute, University of Rochester, Rochester, New York; Center for Visual Science, University of Rochester, Rochester, New York
| | - Krystel R Huxlin
- Flaum Eye Institute, University of Rochester, Rochester, New York; Center for Visual Science, University of Rochester, Rochester, New York.
| |
Collapse
|
35
|
Stepp MA, Zieske JD, Trinkaus-Randall V, Kyne BM, Pal-Ghosh S, Tadvalkar G, Pajoohesh-Ganji A. Wounding the cornea to learn how it heals. Exp Eye Res 2014; 121:178-93. [PMID: 24607489 DOI: 10.1016/j.exer.2014.02.007] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 02/07/2014] [Accepted: 02/10/2014] [Indexed: 12/12/2022]
Abstract
Corneal wound healing studies have a long history and rich literature that describes the data obtained over the past 70 years using many different species of animals and methods of injury. These studies have lead to reduced suffering and provided clues to treatments that are now helping patients live more productive lives. In spite of the progress made, further research is required since blindness and reduced quality of life due to corneal scarring still happens. The purpose of this review is to summarize what is known about different types of wound and animal models used to study corneal wound healing. The subject of corneal wound healing is broad and includes chemical and mechanical wound models. This review focuses on mechanical injury models involving debridement and keratectomy wounds to reflect the authors' expertise.
Collapse
Affiliation(s)
- Mary Ann Stepp
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, DC 20037, USA; Department of Ophthalmology, The George Washington University Medical Center, Washington, DC 20037, USA.
| | - James D Zieske
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA 02114-2500, USA
| | - Vickery Trinkaus-Randall
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA 02118, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| | - Briana M Kyne
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, DC 20037, USA
| | - Sonali Pal-Ghosh
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, DC 20037, USA
| | - Gauri Tadvalkar
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, DC 20037, USA
| | - Ahdeah Pajoohesh-Ganji
- Department of Anatomy and Regenerative Biology, The George Washington University Medical Center, Washington, DC 20037, USA
| |
Collapse
|