Pathogenesis of Alkali Injury-Induced Limbal Stem Cell Deficiency: A Literature Survey of Animal Models

Limbal stem cell deficiency (LSCD) is a debilitating ocular surface disease that eventuates from a depleted or dysfunctional limbal epithelial stem cell (LESC) pool, resulting in corneal epithelial failure and blindness. The leading cause of LSCD is a chemical burn, with alkali substances being the most common inciting agents. Characteristic features of alkali-induced LSCD include corneal conjunctivalization, inflammation, neovascularization and fibrosis. Over the past decades, animal models of corneal alkali burn and alkali-induced LSCD have been instrumental in improving our understanding of the pathophysiological mechanisms responsible for disease development. Through these paradigms, important insights have been gained with regards to signaling pathways that drive inflammation, neovascularization and fibrosis, including NF-κB, ERK, p38 MAPK, JNK, STAT3, PI3K/AKT, mTOR and WNT/β-catenin cascades. Nonetheless, the molecular and cellular events that underpin re-epithelialization and those that govern long-term epithelial behavior are poorly understood. This review provides an overview of the current mechanistic insights into the pathophysiology of alkali-induced LSCD. Moreover, we highlight limitations regarding existing animal models and knowledge gaps which, if addressed, would facilitate development of more efficacious therapeutic strategies for patients with alkali-induced LSCD.

[1]  Valeria Villabona-Martinez,et al.  Standardization of corneal alkali burn methodology in rabbits. , 2023, Experimental eye research.

[2]  S. Wilson The Yin and Yang of Mesenchymal Cells in the Corneal Stromal Fibrosis Response to Injury: The Cornea as a Model of Fibrosis in Other Organs , 2022, Biomolecules.

[3]  Mijeong Park,et al.  Cell identity changes in ocular surface Epithelia , 2022, Progress in Retinal and Eye Research.

[4]  E. Pandzic,et al.  Plasticity of ocular surface epithelia: Using a murine model of limbal stem cell deficiency to delineate metaplasia and transdifferentiation , 2022, Stem cell reports.

[5]  P. Rama,et al.  The two-faced effects of nerves and neuropeptides in corneal diseases , 2021, Progress in Retinal and Eye Research.

[6]  S. Saika,et al.  Roles of Epithelial and Mesenchymal TRP Channels in Mediating Inflammatory Fibrosis , 2022, Frontiers in Immunology.

[7]  R. Costi,et al.  Understanding Drivers of Ocular Fibrosis: Current and Future Therapeutic Perspectives , 2021, International journal of molecular sciences.

[8]  N. Di Girolamo,et al.  Evaluating the clinical translational relevance of animal models for limbal stem cell deficiency: A systematic review. , 2021, The ocular surface.

[9]  N. Mikhailova,et al.  Derivation and Characterization of EGFP-Labeled Rabbit Limbal Mesenchymal Stem Cells and Their Potential for Research in Regenerative Ophthalmology , 2021, Biomedicines.

[10]  U. Schlötzer-Schrehardt,et al.  Melanocytes as emerging key players in niche regulation of limbal epithelial stem cells. , 2021, The ocular surface.

[11]  Yanning Yang,et al.  The potential protective effects of miR-497 on corneal neovascularization are mediated via macrophage through the IL-6/STAT3/VEGF signaling pathway. , 2021, International immunopharmacology.

[12]  Y. Savir,et al.  Discrete limbal epithelial stem cell populations mediate corneal homeostasis and wound healing. , 2021, Cell stem cell.

[13]  S. Wilson TGF beta -1, -2 and -3 in the modulation of fibrosis in the cornea and other organs. , 2021, Experimental eye research.

[14]  L. Yin,et al.  Protective roles of the TIR/BB-loop mimetic AS-1 in alkali-induced corneal neovascularization by inhibiting ERK phosphorylation. , 2021, Experimental eye research.

[15]  Steven E. Wilson,et al.  Interleukin-1 and Transforming Growth Factor Beta: Commonly Opposing, but Sometimes Supporting, Master Regulators of the Corneal Wound Healing Response to Injury , 2021, Investigative ophthalmology & visual science.

[16]  Q. Lan,et al.  Epigenetic Landscape Analysis of the Long Non-Coding RNA and Messenger RNA in a Mouse Model of Corneal Alkali Burns , 2021, Investigative ophthalmology & visual science.

[17]  Dong Hyun Kim,et al.  Comparison of therapeutic effects between topical 8-oxo-2′-deoxyguanosine and corticosteroid in ocular alkali burn model , 2021, Scientific Reports.

[18]  N. Di Girolamo,et al.  Neuronal-epithelial cell alignment: A determinant of health and disease status of the cornea. , 2021, The ocular surface.

[19]  A. Wylęgała,et al.  Impact of corneal parameters, refractive error and age on density and morphology of the subbasal nerve plexus fibers in healthy adults , 2021, Scientific Reports.

[20]  V. Singh,et al.  Long term observation of ocular surface alkali burn in rabbit models: Quantitative analysis of corneal haze, vascularity and self-recovery. , 2021, Experimental eye research.

[21]  P. Lu,et al.  Inhibition of PDGF-BB reduces alkali-induced corneal neovascularization in mice , 2021, Molecular medicine reports.

[22]  A. Shimizu,et al.  Effects of Selective Peroxisome Proliferator Activated Receptor Agonists on Corneal Epithelial Wound Healing , 2021, Pharmaceuticals.

[23]  Chaoqun Yu,et al.  Therapeutic effect of secretome from TNF-α stimulated mesenchymal stem cells in an experimental model of corneal limbal stem cell deficiency. , 2021, International journal of ophthalmology.

[24]  Pengfei Yang,et al.  Rapamycin ameliorates corneal injury after alkali burn through methylation modification in mouse TSC1 and mTOR genes. , 2020, Experimental eye research.

[25]  Xin Liu,et al.  Subconjunctival Injection of Transdifferentiated Oral Mucosal Epithelial Cells for Limbal Stem Cell Deficiency in Rats , 2020, The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society.

[26]  Qinke Yao,et al.  Subconjunctival Injection of Regulatory T Cells Potentiates Corneal Healing Via Orchestrating Inflammation and Tissue Repair After Acute Alkali Burn , 2020, Investigative ophthalmology & visual science.

[27]  J. Chen,et al.  Gelatin methacryloyl hydrogel eye pad loaded with amniotic extract prevents symblepharon in rabbit eyes. , 2020, European review for medical and pharmacological sciences.

[28]  G. Dusting,et al.  Wound Healing After Alkali Burn Injury of the Cornea Involves Nox4-Type NADPH Oxidase , 2020, Investigative ophthalmology & visual science.

[29]  S. Wilson Corneal myofibroblasts and fibrosis. , 2020, Experimental eye research.

[30]  Hongbo Hu,et al.  Targeting NF-κB pathway for the therapy of diseases: mechanism and clinical study , 2020, Signal Transduction and Targeted Therapy.

[31]  Xiaorong Wu,et al.  S100A4 Silencing Facilitates Corneal Wound Healing After Alkali Burns by Promoting Autophagy via Blocking the PI3K/Akt/mTOR Signaling Pathway , 2020, Investigative ophthalmology & visual science.

[32]  R. Dana,et al.  Global Consensus on the Management of Limbal Stem Cell Deficiency. , 2020, Cornea.

[33]  H. Dua,et al.  Chemical eye injury: pathophysiology, assessment and management , 2020, Eye.

[34]  S. Wilson Corneal wound healing. , 2020, Experimental eye research.

[35]  S. W. Kim,et al.  AMPK Activation by 5-Amino-4-Imidazole Carboxamide Riboside-1-β-D-Ribofuranoside Attenuates Alkali Injury-Induced Corneal Fibrosis , 2020, Investigative ophthalmology & visual science.

[36]  M. Debacker Chemical injuries , 2020, Handbook of Disaster Medicine.

[37]  Jin Yao,et al.  SKLB1002, a potent inhibitor of VEGF receptor 2 signaling, inhibits endothelial angiogenic function in vitro and ocular angiogenesis in vivo , 2020, Molecular medicine reports.

[38]  J. Jung,et al.  A Novel CD147 Inhibitor, SP-8356, Attenuates Pathological Fibrosis in Alkali-Burned Rat Cornea , 2020, International journal of molecular sciences.

[39]  Hai-Feng Xu,et al.  LRG1 promotes corneal angiogenesis and lymphangiogenesis in a corneal alkali burn mouse model. , 2020, International journal of ophthalmology.

[40]  Jiucheng He,et al.  Novel RvD6 stereoisomer induces corneal nerve regeneration and wound healing post-injury by modulating trigeminal transcriptomic signature , 2020, Scientific Reports.

[41]  M. Stepp,et al.  Corneal Epithelial “Neuromas”: A Case of Mistaken Identity? , 2020, Cornea.

[42]  Li-Ying Tang,et al.  Keratocytes promote corneal neovascularization through VEGFr3 induced by PPARα-inhibition. , 2020, Experimental eye research.

[43]  A. Sada,et al.  Defining compartmentalized stem cell populations with distinct cell division dynamics in the ocular surface epithelium , 2020, Development.

[44]  Qi Hui,et al.  Comparative Analysis of KGF-2 and bFGF in Prevention of Excessive Wound Healing and Scar Formation in a Corneal Alkali Burn Model. , 2019, Cornea.

[45]  Qiong Yi,et al.  The Wound Healing Effect of Doxycycline after Corneal Alkali Burn in Rats , 2019, Journal of ophthalmology.

[46]  P. Cidad,et al.  Dynamic changes of the extracellular matrix during corneal wound healing. , 2019, Experimental eye research.

[47]  H. Mao,et al.  Nanofiber-Reinforced Decellularized Amniotic Membrane Improves Limbal Stem Cell Transplantation in a Rabbit Model of Corneal Epithelial Defect. , 2019, Acta biomaterialia.

[48]  S. Yamagami,et al.  Pro-inflammatory role of NLRP3 inflammasome in experimental sterile corneal inflammation , 2019, Scientific Reports.

[49]  V. Sangwan,et al.  Inflammation, vascularization and goblet cell differences in LSCD: Validating animal models of corneal alkali burns. , 2019, Experimental eye research.

[50]  C. Paterson,et al.  Assessment of corneal substrate biomechanics and its effect on epithelial stem cell maintenance and differentiation , 2019, Nature Communications.

[51]  R. Donahue,et al.  Latent Sensitization in a Mouse Model of Ocular Neuropathic Pain , 2019, Translational vision science & technology.

[52]  Federico Castro-Muñozledo,et al.  (-)-Epigallocatechin-3-gallate, reduces corneal damage secondary from experimental grade II alkali burns in mice. , 2019, Burns : journal of the International Society for Burn Injuries.

[53]  R. Dana,et al.  Global Consensus on Definition, Classification, Diagnosis, and Staging of Limbal Stem Cell Deficiency , 2018, Cornea.

[54]  A. Shimizu,et al.  PPARα Agonist Suppresses Inflammation after Corneal Alkali Burn by Suppressing Proinflammatory Cytokines, MCP-1, and Nuclear Translocation of NF-κB , 2018, Molecules.

[55]  Y. Deng,et al.  The long-term effect of tacrolimus on alkali burn-induced corneal neovascularization and inflammation surpasses that of anti-vascular endothelial growth factor , 2018, Drug design, development and therapy.

[56]  J. Larrick,et al.  Anti-angiogenic effect of a humanized antibody blocking the Wnt/β-catenin signaling pathway. , 2018, Microvascular research.

[57]  L. Ye,et al.  Xanthatin inhibits corneal neovascularization by inhibiting the VEGFR2-mediated STAT3/PI3K/Akt signaling pathway , 2018, International journal of molecular medicine.

[58]  Shuangling Chen,et al.  Human limbal niche cells are a powerful regenerative source for the prevention of limbal stem cell deficiency in a rabbit model , 2018, Scientific Reports.

[59]  Qian Wang,et al.  Resolvin D1 promotes corneal epithelial wound healing and restoration of mechanical sensation in diabetic mice , 2018, Molecular vision.

[60]  P. Rama,et al.  Substance P Modulation of Human and Murine Corneal Neovascularization. , 2018, Investigative ophthalmology & visual science.

[61]  D. Soteriou,et al.  Corneal-Committed Cells Restore the Stem Cell Pool and Tissue Boundary following Injury. , 2018, Cell reports.

[62]  A. Shimizu,et al.  Peroxisome proliferator-activated receptor alpha agonist suppresses neovascularization by reducing both vascular endothelial growth factor and angiopoietin-2 in corneal alkali burn , 2017, Scientific Reports.

[63]  W. Verri,et al.  Naringenin Eye Drops Inhibit Corneal Neovascularization by Anti-Inflammatory and Antioxidant Mechanisms. , 2017, Investigative ophthalmology & visual science.

[64]  P. Rama,et al.  Growth inhibition of formed corneal neovascularization following Fosaprepitant treatment , 2017, Acta ophthalmologica.

[65]  S. Swamynathan,et al.  Inhibition of HUVEC tube formation via suppression of NF&kgr;B suggests an anti‐angiogenic role for SLURP1 in the transparent cornea , 2017, Experimental eye research.

[66]  A. Ljubimov,et al.  Concise Review: Stem Cells for Corneal Wound Healing , 2017, Stem cells.

[67]  Shao-Cong Sun,et al.  NF-κB signaling in inflammation , 2017, Signal Transduction and Targeted Therapy.

[68]  Bumseok Kim,et al.  Therapeutic effects of zerumbone in an alkali‐burned corneal wound healing model , 2017, International immunopharmacology.

[69]  J. Won,et al.  Comprehensive Modeling of Corneal Alkali Injury in the Rat Eye , 2017, Current eye research.

[70]  E. A. Volpe,et al.  Inhibition of NLRP3 Inflammasome Pathway by Butyrate Improves Corneal Wound Healing in Corneal Alkali Burn , 2017, International journal of molecular sciences.

[71]  S. Chanda,et al.  Inhibition of NUCKS Facilitates Corneal Recovery Following Alkali Burn , 2017, Scientific Reports.

[72]  F. Jakobiec,et al.  Sustained Subconjunctival Delivery of Infliximab Protects the Cornea and Retina Following Alkali Burn to the Eye , 2017, Investigative ophthalmology & visual science.

[73]  Ting Wu,et al.  MK2 inhibitor reduces alkali burn-induced inflammation in rat cornea , 2016, Scientific Reports.

[74]  Xiao-Jian Han,et al.  Involvement of NADPH oxidases in alkali burn-induced corneal injury , 2016, International journal of molecular medicine.

[75]  N. Girolamo Moving epithelia: Tracking the fate of mammalian limbal epithelial stem cells , 2015, Progress in Retinal and Eye Research.

[76]  A. Zajícová,et al.  A Comparative Study of the Therapeutic Potential of Mesenchymal Stem Cells and Limbal Epithelial Stem Cells for Ocular Surface Reconstruction , 2015, Stem cells translational medicine.

[77]  Y. Shao,et al.  Therapeutic Effects of Topical Netrin-4 Inhibits Corneal Neovascularization in Alkali-Burn Rats , 2015, PloS one.

[78]  Won‐Kyo Jung,et al.  Inhibitory effects of the platelet-activating factor receptor antagonists, CV-3988 and Ginkgolide B, on alkali burn-induced corneal neovascularization , 2015, Cutaneous and ocular toxicology.

[79]  S. Seo,et al.  Effects of topical chondrocyte-derived extracellular matrix treatment on corneal wound healing, following an alkali burn injury. , 2015, Molecular medicine reports.

[80]  P. Rama,et al.  NK1 receptor antagonists as a new treatment for corneal neovascularization. , 2014, Investigative ophthalmology & visual science.

[81]  He Wang,et al.  Keratocytes create stromal spaces to promote corneal neovascularization via MMP13 expression. , 2014, Investigative ophthalmology & visual science.

[82]  W. Wee,et al.  Bone Marrow-derived Mesenchymal Stem Cells Affect Immunologic Profiling of Interleukin-17-secreting Cells in a Chemical Burn Mouse Model , 2014, Korean journal of ophthalmology : KJO.

[83]  Y. Fukuda,et al.  An ophthalmic solution of a peroxisome proliferator-activated receptor gamma agonist prevents corneal inflammation in a rat alkali burn model , 2013, Molecular vision.

[84]  K. Yi,et al.  Chemical injury-induced corneal opacity and neovascularization reduced by rapamycin via TGF-β1/ERK pathways regulation. , 2013, Investigative ophthalmology & visual science.

[85]  R. Dana,et al.  Nerves and neovessels inhibit each other in the cornea. , 2013, Investigative ophthalmology & visual science.

[86]  Shi-you Zhou,et al.  Minocycline Inhibits Alkali Burn-Induced Corneal Neovascularization in Mice , 2012, PloS one.

[87]  Y. Shao,et al.  Netrin-1 simultaneously suppresses corneal inflammation and neovascularization. , 2012, Investigative ophthalmology & visual science.

[88]  W. Su,et al.  Doxycycline enhances the inhibitory effects of bevacizumab on corneal neovascularization and prevents its side effects. , 2011, Investigative ophthalmology & visual science.

[89]  A. Murakami,et al.  Role of the IL-6 classic- and trans-signaling pathways in corneal sterile inflammation and wound healing. , 2011, Investigative ophthalmology & visual science.

[90]  N. Mukaida,et al.  Critical role of SDF-1α-induced progenitor cell recruitment and macrophage VEGF production in the experimental corneal neovascularization , 2011, Molecular vision.

[91]  K. Tsubota,et al.  Hydrogen and N-acetyl-L-cysteine rescue oxidative stress-induced angiogenesis in a mouse corneal alkali-burn model. , 2011, Investigative ophthalmology & visual science.

[92]  S. Saika,et al.  Endothelial Mesenchymal Transition: A Therapeutic Target in Retrocorneal Membrane , 2010, Cornea.

[93]  N. Mukaida,et al.  Opposite Roles of CCR2 and CX3CR1 Macrophages in Alkali-Induced Corneal Neovascularization , 2009, Cornea.

[94]  S. Saika,et al.  Sonic hedgehog: its expression in a healing cornea and its role in neovascularization , 2009 .

[95]  T. Fuchsluger,et al.  Limbal Stem Cell Deficiency and Corneal Neovascularization , 2009, Seminars in ophthalmology.

[96]  N. Mukaida,et al.  Essential contribution of CCL3 to alkali-induced corneal neovascularization by regulating vascular endothelial growth factor production by macrophages , 2008, Molecular vision.

[97]  N. Mukaida,et al.  Protective Roles of the Fractalkine/CX3CL1-CX3CR1 Interactions in Alkali-Induced Corneal Neovascularization through Enhanced Antiangiogenic Factor Expression1 , 2008, The Journal of Immunology.

[98]  A. Joussen,et al.  The role of integrin α5β1 in the regulation of corneal neovascularization , 2007 .

[99]  G. Melles,et al.  Descemet membrane endothelial keratoplasty (DMEK). , 2006, Cornea.

[100]  K. Flanders,et al.  Expression of Smad7 in mouse eyes accelerates healing of corneal tissue after exposure to alkali. , 2005, The American journal of pathology.

[101]  K. Flanders,et al.  Therapeutic effect of topical administration of SN50, an inhibitor of nuclear factor-kappaB, in treatment of corneal alkali burns in mice. , 2005, The American journal of pathology.

[102]  K. Flanders,et al.  Therapeutic effects of adenoviral gene transfer of bone morphogenic protein-7 on a corneal alkali injury model in mice , 2005, Laboratory Investigation.

[103]  K. Ando,et al.  Redox Regulation of NF-κB Activation: Distinct Redox Regulation Between the Cytoplasm and the Nucleus , 2005 .

[104]  Nathan Efron,et al.  Corneal nerve tortuosity in diabetic patients with neuropathy. , 2004, Investigative ophthalmology & visual science.

[105]  N. Mitsiades,et al.  VEGF-dependent conjunctivalization of the corneal surface. , 2003, Investigative ophthalmology & visual science.

[106]  D. Archer,et al.  The inflammatory milieu associated with conjunctivalized cornea and its alteration with IL-1 RA gene therapy. , 2002, Investigative ophthalmology & visual science.

[107]  B. Kirchhof,et al.  Inhibition of inflammatory corneal angiogenesis by TNP-470. , 2001, Investigative ophthalmology & visual science.

[108]  C. Sotozono,et al.  Cytokine expression in the alkali-burned cornea. , 1997, Current eye research.

[109]  W. Kao,et al.  Expression of collagen I, smooth muscle alpha-actin, and vimentin during the healing of alkali-burned and lacerated corneas. , 1993, Investigative ophthalmology & visual science.