Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
Add more filters

Publication year range
1.
Stem Cells ; 39(3): 280-295, 2021 03.
Article in English | MEDLINE | ID: mdl-33373496

ABSTRACT

Quiescence and self-renewal of human corneal epithelial progenitor/stem cells (LEPC) are regulated by the limbal niche, presumably through close interaction with limbal (stromal) niche cells (LNC). Paired box homeotic gene 6 (Pax6), a conserved transcription factor essential for eye development, is essential for proper differentiation of limbal and corneal epithelial stem cells. Pax6 haploinsufficiency causes limbal stem cell deficiency, which leads to subsequent corneal blindness. We previously reported that serial passage of nuclear Pax6+ LNC resulted in the gradual loss of nuclear Pax6+ and neural crest progenitor status, the latter of which was reverted upon recovery of Pax6. These findings suggest Pax6 plays a pivotal role in supporting the self-renewal of LEPC in limbal niche. Herein, we show that HC-HA/PTX3, a unique matrix purified from amniotic membrane (AM) and consists of heavy chain 1of inter-α-trypsin inhibitor covalently linked to hyaluronic acid and complexed with pentraxin 3, is capable of reverting senescent LNC to nuclear Pax6+ neural crest progenitors that support self-renewal of LEPC. Such reversion is causally linked to early cell aggregation mediated by activation of C-X-C chemokine receptor type 4 (CXCR4)-mediated signaling followed by activation of bone morphogenetic protein (BMP) signaling. Furthermore, CXCR4-mediated signaling, but not BMP signaling, controls recovery of the nuclear Pax6+ neural crest progenitors. These findings not only explain why AM helps in vivo and ex vivo expansion of human LEPC, but they also illuminate the potential role of HC-HA/PTX3 as a surrogate matrix niche that complements stem cell-based therapies in regenerative medicine.


Subject(s)
C-Reactive Protein/metabolism , Limbus Corneae/cytology , PAX6 Transcription Factor/metabolism , Serum Amyloid P-Component/metabolism , Stem Cell Niche/physiology , Aged , Cell Differentiation/physiology , Cells, Cultured , Corneal Diseases/genetics , Epithelial Cells/metabolism , Epithelium, Corneal/cytology , Humans , Middle Aged , Neural Crest/cytology , Stem Cells/metabolism
2.
Exp Eye Res ; 199: 108181, 2020 10.
Article in English | MEDLINE | ID: mdl-32795525

ABSTRACT

Homeostasis of the corneal epithelium is ultimately maintained by stem cells that reside in a specialized microenvironment within the corneal limbus termed palisades of Vogt. This limbal niche nourishes, protects, and regulates quiescence, self-renewal, and fate decision of limbal epithelial stem/progenitor cells (LEPCs) toward corneal epithelial differentiation. This review focuses on our current understanding of the mechanism by which limbal (stromal) niche cells (LNCs) regulate the aforementioned functions of LEPCs. Based on our discovery and characterization of a unique extracellular matrix termed HC-HA/PTX3 (Heavy chain (HC1)-hyaluronan (HA)/pentraxin 3 (PTX3) complex, "-" denotes covalent linkage; "/" denotes non-covalent binding) in the birth tissue, i.e., amniotic membrane and umbilical cord, we put forth a new paradigm that HC-HA/PTX3 serves as a surrogate matrix niche by maintaining the in vivo nuclear Pax6+ neural crest progenitor phenotype to support quiescence and self-renewal but prevent corneal fate decision of LEPCs. This new paradigm helps explain how limbal stem cell deficiency (LSCD) develops in aniridia due to Pax6-haplotype deficiency and further explains why transplantation of HC-HA/PTX3-containing amniotic membrane prevents LSCD in acute chemical burns and Stevens Johnson syndrome, augments the success of autologous LEPCs transplantation in patients suffering from partial or total LSCD, and assists ex vivo expansion (engineering) of a graft containing LEPCs. We thus envisage that this new paradigm based on regenerative matrix HC-HA/PTX3 as a surrogate niche can set a new standard for regenerative medicine in and beyond ophthalmology.


Subject(s)
C-Reactive Protein/metabolism , Corneal Diseases/genetics , Limbus Corneae/metabolism , Serum Amyloid P-Component/metabolism , Stem Cell Niche , Cell Differentiation , Cells, Cultured , Corneal Diseases/metabolism , Corneal Diseases/pathology , Humans , Limbus Corneae/pathology
3.
Wound Repair Regen ; 28(4): 526-531, 2020 07.
Article in English | MEDLINE | ID: mdl-32386343

ABSTRACT

An open label, multicenter 16-week trial of cryopreserved human umbilical cord (TTAX01) was previously undertaken in 32 subjects presenting with a Wagner grade 3 or 4 diabetic foot ulcer, with 16 (50%) of these having confirmed closure following a median of one product application (previous study). All but two subjects (30/32; 94%) consented to participate in this follow-up study to 1-year postexposure. No restrictions were placed on treatments for open wounds. At 8-week intervals, subjects were evaluated for adverse events (AEs) and wound status (open or closed). Average time from initial exposure to end of follow-up was 378 days (range 343-433), with 29 of 30 (97%) subjects completing a full year. AEs were all typical for the population under study, and none were attributed to prior exposure to TTAX01. One previously healed wound re-opened, one previously unconfirmed closed wound remained healed, and nine new wound closures occurred, giving 25 of 29 (86.2%) healed in the ITT population. Three of the new closures followed the use of various tissue-based products. Three subjects whose wounds were healed required subsequent minor amputations due to osteomyelitis, one of which progressed to a major amputation (1/29; 3.4%). One additional subject underwent two minor amputations prior to healing. Overall, the study found TTAX01 to be safe in long-term follow-up and associated with both a low rate of major amputation and a higher than expected rates of healing.


Subject(s)
Biological Products/therapeutic use , Cryopreservation , Diabetic Foot/therapy , Umbilical Cord/transplantation , Wound Healing , Adult , Aged , Amputation, Surgical/statistics & numerical data , Female , Follow-Up Studies , Humans , Male , Middle Aged , Severity of Illness Index , Treatment Outcome
4.
Wound Repair Regen ; 27(6): 680-686, 2019 11.
Article in English | MEDLINE | ID: mdl-31376297

ABSTRACT

Clinical trials of potential new therapies for diabetic foot ulcers rarely enroll patients whose wounds extend to muscle, fascia, or bone with clinical and radiographic evidence of underlying osteomyelitis. An open-label, multicenter trial of cryopreserved human umbilical cord (TTAX01) was undertaken in 32 subjects presenting with such complex wounds with a mean duration of 6.1 ± 9.0 (range: 0.2-47.1) months and wound area at screening of 3.8 ± 2.9 (range: 1.0-9.6) cm2 . Aggressive surgical debridement at baseline resulted in 17 minor amputations and an increase in mean wound area to 7.4 ± 5.8 (range: 1.1-28.6) cm2 . All subjects were placed on systemic antibiotics for at least 6 weeks in conjunction with baseline application of TTAX01. Repeat applications were made at no less than 4-week intervals over the 16-week trial. Initial closure occurred in 18 of 32 (56%) wounds, with 16 (50%) of these having confirmed closure in 16 weeks with a median of one-product application. Cases with biopsy confirmed osteomyelitis (n = 20) showed initial closure in 12 (60%) wounds and confirmed closure in 10 (50%) wounds. Four of the five ulcers presenting as recurrences experienced confirmed closure. Mean overall time to healing was 12.8 ± 4.3 weeks. Mean wound area reduction from baseline was 91% for all wounds. Of the 16 wounds without confirmed closure during the 16-week treatment period, five (31.3%) achieved 99-100% wound area reduction by their final visit. The product was well tolerated. Two minor amputations occurred during the study period due to recurrent or persistent osteomyelitis; however, there were no major amputations.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Diabetic Foot/therapy , Osteomyelitis/therapy , Umbilical Cord/transplantation , Wound Healing/physiology , Adult , Aged , Cryopreservation/methods , Debridement/methods , Diabetes Mellitus, Type 2/complications , Diabetic Foot/complications , Diabetic Foot/diagnosis , Female , Humans , Male , Middle Aged , Osteomyelitis/complications , Osteomyelitis/diagnostic imaging , Pilot Projects , Prognosis , Risk Assessment , Severity of Illness Index
5.
Int Ophthalmol ; 38(5): 2219-2222, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29101724

ABSTRACT

PURPOSE: To present a case of successful treatment of immune-related severe dry eye disease (DED) by self-retained cryopreserved amniotic membrane (CAM) in conjunction with conventional and systemic immunotherapy. OBSERVATIONS: A 48-year-old female with a 10-year history of rheumatoid arthritis under systemic immunomodulation developed non-resolved severe ocular dryness, pain, photophobia and blurred vision due to corneal epithelial keratopathy OD much worse than OS despite topical artificial tears, steroids, cyclosporine, autologous serum drops, punctal plugs and scleral lens for the last 3 years. During the first year, she received punctal cauterization and a total of 6 CAM, each for an average of 7.2 ± 2.3 days, for recurrent diffuse superficial punctate keratitis (SPK) with filaments to achieve an average symptom-free period of 2.4 ± 0.9 months and visual acuity improvement from 20/400 to 20/200. During the next 2 years, she received surgical closure of puncta for recurrent punctal reopening, additional systemic immunomodulation and a total of 4 CAM, each for an average of 8.5 ± 2 days, for recurrent scattered SPK, to achieve an average symptom-free period of 6.4 ± 1 months and visual acuity improvement from 20/200 to 20/70. CONCLUSIONS: Self-retained CAM can be an adjunctive treatment for immune-related DED, which is refractory to conventional and systemic immunotherapies.


Subject(s)
Amnion/transplantation , Autoimmune Diseases/therapy , Cryopreservation , Cyclosporine/therapeutic use , Dry Eye Syndromes/therapy , Visual Acuity , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Dry Eye Syndromes/diagnosis , Dry Eye Syndromes/immunology , Female , Humans , Immunosuppressive Agents/therapeutic use , Lubricant Eye Drops , Middle Aged
6.
Prenat Diagn ; 37(5): 473-481, 2017 May.
Article in English | MEDLINE | ID: mdl-28295455

ABSTRACT

BACKGROUND: Prenatal spina bifida (SB) repair with a regenerative patch may improve neurological outcomes by decreasing inflammatory scarring. OBJECTIVE: This study aims to compare cryopreserved human umbilical cord (HUC) and biocellulose film (BCF) patches sutured over SB lesions for regeneration of native cells and inflammatory response. STUDY DESIGN: Sprague-Dawley rats were gavaged with retinoic acid (RA) on embryonic day 10 to induce SB. Hysterotomy was performed on embryonic day 20 and on HUC or BCF patches sutured over the defect. Pups were harvested 30 to 34 h later, and hematoxylin and eosin staining and Trichrome staining assessed basic cellular migration. Immunohistochemistry demonstrated the exact nature of the cellular migration. Patches and surrounding exudates were evaluated with microscopy and cells quantified. RESULTS: Histology showed cellular migration with all HUC patches compared with none with BCF patches. Epithelial cells were noted migrating over the dorsal HUC surface, astrocytes were noted along the HUC surface adjacent to the lesion, and endothelial cells were noted within the HUC. HUC patches showed minimal inflammatory cells. Exudates surrounding the HUC patches had fewer inflammatory cells than exudates around BCF patches. CONCLUSION: HUC promotes cellular migration of native cells with minimal inflammatory response compared with BCF. HUC may be the superior patch material for prenatal SB repair. © 2017 John Wiley & Sons, Ltd.


Subject(s)
Cellulose/therapeutic use , Fetoscopy/methods , Skin, Artificial , Spinal Dysraphism/surgery , Umbilical Cord/transplantation , Animals , Cells, Cultured , Cryopreservation , Disease Models, Animal , Female , Humans , Membranes, Artificial , Mesenchymal Stem Cell Transplantation , Pregnancy , Rats , Rats, Sprague-Dawley , Spinal Dysraphism/pathology
7.
Stem Cells ; 33(11): 3341-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26148958

ABSTRACT

To explore how limbal niche cells (LNCs) may control quiescence, self-renewal, and corneal epithelial lineage commitment/differentiation of limbal epithelial progenitor/stem cells (LEPCs), we have established an in vitro sphere assay by reunion between the two cell types in three-dimensional Matrigel. The resultant sphere exhibits inhibition of corneal epithelial lineage commitment/differentiation and marked clonal growth of LEPCs, of which the latter is correlated with activation of canonical Wnt signaling. Herein, we have created a similar reunion assay in immobilized heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3), which is purified from amniotic membrane (AM) and consists of a complex formed by hyaluronic covalently linked to heavy chain 1 of inter-α-inhibitor and noncovalently linked to pentraxin 3. The resultant spheres exhibited similar suppression of corneal epithelial lineage commitment/differentiation but upregulation of quiescence markers including nuclear translocation of Bmi-1, and negligible clonal growth of LEPCs. This outcome was correlated with the suppression of canonical Wnt but activation of noncanonical (Planar cell polarity) Wnt signaling as well as BMP signaling in both LEPCs and LNCs. The activation of BMP signaling in LNCs was pivotal because nuclear translocation of pSmad1/5/8 was prohibited in hLEPCs when reunioned with mLNCs of conditionally deleted Bmpr1a;Acvr1(DCKO) mice. Furthermore, ablation of BMP signaling in LEPCs led to upregulation of cell cycle genes, downregulation of Bmi-1, nuclear exclusion of phosphorylated Bmi-1, and marked promotion of the clonal growth of LEPCs. Hence, HC-HA/PTX3 uniquely upregulates BMP signaling in LNCs which leads to BMP signaling in LEPCs to achieve quiescence, helping explain how AM transplantation is clinically useful to be used as a matrix for ex vivo expansion of LEPCs and to treat corneal blindness caused by limbal stem cells deficiency.


Subject(s)
Amnion/metabolism , Bone Morphogenetic Proteins/biosynthesis , C-Reactive Protein/biosynthesis , Epithelial Cells/metabolism , Hyaluronic Acid/biosynthesis , Serum Amyloid P-Component/biosynthesis , Stem Cell Niche/physiology , 3T3 Cells , Animals , C-Reactive Protein/isolation & purification , Cell Differentiation/physiology , Cells, Cultured , Humans , Hyaluronic Acid/isolation & purification , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/isolation & purification , Mice , Mice, Transgenic , Serum Amyloid P-Component/isolation & purification , Signal Transduction/physiology , Stem Cells/metabolism
8.
Nature ; 463(7284): E10-1; discussion E11, 2010 Feb 25.
Article in English | MEDLINE | ID: mdl-20182462

ABSTRACT

The longstanding concept that corneal epithelial stem cells reside mainly in the limbus is supported by the absence of major corneal epithelial differentiation markers, that is, K3 and K12 keratins, in limbal basal cells (these markers are expressed, however, in corneal basal cells, thus distinguishing the mode of keratin expression in corneal epithelium from that of all other stratified epithelia), the centripetal migration of corneal epithelial cells, the exclusive location of slow-cycling cells in the limbal basal layer, the superior in vitro proliferative potential of limbal epithelial cells, and the transplanted limbal cells' ability to reconstitute corneal epithelium in vivo (reviewed in refs 1-4). Moreover, previous data indicate that corneal and conjunctival epithelia represent two separate cell lineages (reviewed in refs 1-4). Majo et al. suggested, however, that corneal and conjunctival epithelia are equipotent, and that identical oligopotent stem cells are present throughout the corneal, limbal and conjunctival epithelia. We point out here that these suggestions are inconsistent with many known growth, differentiation and cell migration properties of the anterior ocular epithelia.


Subject(s)
Cell Movement , Epithelium, Corneal/cytology , Limbus Corneae/cytology , Stem Cells/cytology , Animals , Cattle , Cell Differentiation , Cell Lineage , Cell Proliferation , Conjunctiva/cytology , Goblet Cells/cytology , Humans , Mice , Models, Biological , Rabbits , Reproducibility of Results , Sheep , Swine
9.
J Biol Chem ; 289(19): 13531-42, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24652286

ABSTRACT

Heavy chain (HC)-hyaluronan (HA), a complex formed by the covalent linkage between HC1 from the inter-α-trypsin inhibitor (IαI) and HA, purified from the human amniotic membrane (AM), is responsible for the anti-inflammatory, antiscarring, and antiangiogenic actions of the AM. This HC-HA complex is produced by constitutive expression of TNF-stimulated gene 6 and endogenous production of IαI by AM cells. Pentraxin 3 (PTX3), a prototypic long pentraxin that plays a non-redundant role in innate immunity against selected pathogens, also helps stabilize HC-HA to ensure female fertility. Here we noted strong positive PTX3 staining in the AM epithelium and compact stroma. PTX3 was constitutively expressed and secreted by cultured AM epithelial and stromal cells and, further, greatly up-regulated by TNF and IL-1ß. Using an agarose overlay to trap the HA-containing matrix, the HC-HA-PTX3 complex was formed, as analyzed by Western blot analysis, by AM cells but not human skin fibroblasts, despite being cultured in the presence of serum and TNF. However, exogenous PTX3 helps human skin fibroblasts form the HC-HA-PTX3 complex with an agarose overlay. Furthermore, PTX3 can be coimmunoprecipitated with the HC-HA complex from agarose-overlaid AM cell extracts by an anti-human IαI antibody. Such a HC-HA-PTX3 complex can be reconstituted in vitro and exhibit similar effects as those reported for AM HC-HA-PTX3 on polarization of M2 macrophages. The tight binding between PTX3 and AM HC-HA withstands four runs of CsCl ultracentrifugation in the presence of 4 m GnHCl. These results indicate that PTX3 is constitutively expressed and secreted by AM cells as an integral component of the AM HC-HA-PTX3 complex and contributes to the biological function of AM HC-HA-PTX3.


Subject(s)
Amnion/metabolism , C-Reactive Protein/biosynthesis , Gene Expression Regulation/physiology , Hyaluronic Acid/metabolism , Serum Amyloid P-Component/biosynthesis , Adult , Amnion/cytology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Curr Opin Ophthalmol ; 26(4): 295-300, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26058028

ABSTRACT

PURPOSE OF REVIEW: To summarize recent advances on ocular Demodex infestation. RECENT FINDINGS: Demodex infestation is a potential cause of ocular surface inflammation. The pathogenesis of Demodex in eliciting ocular surface inflammation has been further clarified. Cliradex is currently the treatment of choice, it comprises the most active ingredient of tea tree oil, that is terpinen-4-ol, which helps eradicate Demodex mites and reduce ocular surface inflammation. SUMMARY: Ocular demodicosis is a common but overlooked eye disease that manifests a number of morbidities. Demodex folliculorum causes chronic anterior blepharitis whereas Demodex brevis causes posterior blepharitis, meibomian gland dysfunction, recurrent chalazia, and refractory keratoconjunctivitis. The lash sampling and microscopic counting method and in-vivo confocal microscopy are key diagnostic methods. Cliradex shows promising potential to reduce Demodex counts with additional antibacterial, antifungal, and anti-inflammatory actions.


Subject(s)
Blepharitis/diagnosis , Eye Infections, Parasitic/diagnosis , Meibomian Glands/parasitology , Mite Infestations/diagnosis , Animals , Anti-Infective Agents, Local/therapeutic use , Blepharitis/parasitology , Blepharitis/therapy , Eye Infections, Parasitic/parasitology , Eye Infections, Parasitic/therapy , Humans , Hygiene , Mite Infestations/parasitology , Mite Infestations/therapy , Mites , Tea Tree Oil/therapeutic use
11.
J Biol Chem ; 288(36): 25792-25803, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23878196

ABSTRACT

Despite the known anti-inflammatory effect of amniotic membrane, its action mechanism remains largely unknown. HC-HA complex (HC-HA) purified from human amniotic membrane consists of high molecular weight hyaluronic acid (HA) covalently linked to the heavy chain (HC) 1 of inter-α-trypsin inhibitor. In this study, we show that soluble HC-HA also contained pentraxin 3 and induced the apoptosis of both formyl-Met-Leu-Phe or LPS-activated neutrophils and LPS-activated macrophages while not affecting the resting cells. This enhanced apoptosis was caused by the inhibition of cell adhesion, spreading, and proliferation caused by HC-HA binding of LPS-activated macrophages and preventing adhesion to the plastic surface. Preferentially, soluble HC-HA promoted phagocytosis of apoptotic neutrophils in resting macrophages, whereas immobilized HC-HA promoted phagocytosis in LPS-activated macrophages. Upon concomitant LPS stimulation, immobilized HC-HA but not HA polarized macrophages toward the M2 phenotype by down-regulating IRF5 protein and preventing its nuclear localization and by down-regulating IL-12, TNF-α, and NO synthase 2. Additionally, IL-10, TGF-ß1, peroxisome proliferator-activated receptor γ, LIGHT (TNF superfamily 14), and sphingosine kinase-1 were up-regulated, and such M2 polarization was dependent on TLR ligation. Collectively, these data suggest that HC-HA is a unique matrix component different from HA and uses multiple mechanisms to suppress M1 while promoting M2 phenotype. This anti-inflammatory action of HC-HA is highly desirable to promote wound healing in diseases heightened by unsuccessful transition from M1 to M2 phenotypes.


Subject(s)
Hyaluronic Acid/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/metabolism , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Cell Line , Cytokines/biosynthesis , Down-Regulation/drug effects , Humans , Hyaluronic Acid/pharmacology , Interferon Regulatory Factors/biosynthesis , Macrophages/cytology , Mice , Neutrophils/cytology , Neutrophils/metabolism , Nitric Oxide Synthase Type II/biosynthesis , PPAR gamma/biosynthesis , Phagocytosis/drug effects , Toll-Like Receptors/agonists , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
12.
J Cell Sci ; 125(Pt 15): 3636-48, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22505615

ABSTRACT

Contact inhibition ubiquitously exists in non-transformed cells that are in contact with neighboring cells. This phenomenon explains the poor regenerative capacity of in vivo human corneal endothelial cells during aging, injury and surgery. This study demonstrated that the conventional approach of expanding human corneal endothelial cells by disrupting contact inhibition with EDTA followed by bFGF activated canonical Wnt signaling and lost the normal phenotype to endothelial-mesenchymal transition, especially if TGFß1 was added. By contrast, siRNA against p120 catenin (CTNND1) also uniquely promoted proliferation of the endothelial cells by activating trafficking of p120 catenin to the nucleus, thus relieving repression by nuclear Kaiso. This nuclear p120-catenin-Kaiso signaling is associated with activation of RhoA-ROCK signaling, destabilization of microtubules and inhibition of Hippo signaling, but not with activation of Wnt-ß-catenin signaling. Consequently, proliferating human corneal endothelial cells maintained a hexagonal shape, with junctional expression of N-cadherin, ZO-1 and Na(+)/K(+)-ATPase. Further expansion of human corneal endothelial monolayers with a normal phenotype and a higher density was possible by prolonging treatment with p120 catenin siRNA followed by its withdrawal. This new strategy of perturbing contact inhibition by selective activation of p120-catenin-Kaiso signaling without disrupting adherent junction could be used to engineer surgical grafts containing normal human corneal endothelial cells to meet a global corneal shortage and for endothelial keratoplasties.


Subject(s)
Adherens Junctions/physiology , Catenins/physiology , Contact Inhibition/physiology , Cornea/cytology , Endothelium, Corneal/cytology , Mitosis/physiology , Adherens Junctions/drug effects , Adolescent , Adult , Aged , Aged, 80 and over , Catenins/genetics , Catenins/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cells, Cultured , Contact Inhibition/drug effects , Cornea/drug effects , Edetic Acid/pharmacology , Endothelium, Corneal/drug effects , Endothelium, Corneal/ultrastructure , Epithelial-Mesenchymal Transition/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Middle Aged , Mitosis/drug effects , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Transcription Factors/metabolism , Transfection , Transforming Growth Factor beta1/pharmacology , Wnt Proteins/metabolism , Young Adult , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Delta Catenin
13.
Tissue Cell ; 91: 102535, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39217785

ABSTRACT

The heavy chain (HC)-hyaluronan (HA)/pentraxin 3 (HC-HA/PTX3) complex is formed by tumor necrosis factor-stimulated gene-6 (TSG-6) catalyzing the covalent (ester bond) transfer of HC1 from inter-α-trypsin inhibitor (IαI) to HA followed by tight binding of PTX3. The presence of such a complex has been found in human amniotic membrane (AM) and is considered to be a major matrix component responsible for its anti­inflammatory and anti­scarring properties to promote regenerative healing. Because the therapeutic potentials of AM and umbilical cord (UC) are similar, we herein evaluated whether human UC also contains HC-HA/PTX3. Immunostaining of UC cross-sections showed abundant PTX3, HC1, HA, TSG-6, and bikunin. Western blot analysis suggested the presence of HC1 complex bound via a NaOH-sensitive bond and tightly bound to PTX3 multimer in UC and AM extracts but not in chorion and placenta extracts. HC-HA/PTX3 was purified from UC extract by successive runs of density gradient ultracentrifugation and verified the presence of HC1 but not HC2 or HC3 based on western blot analysis. These results suggest the presence of HC-HA/PTX3 complex in UC is similar to AM.

14.
bioRxiv ; 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-38826432

ABSTRACT

Pain after surgery causes significant suffering. Opioid analgesics cause severe side effects and accidental death. Therefore, there is an urgent need to develop non-opioid therapies for managing post-surgical pain. Local application of Clarix Flo (FLO), a human amniotic membrane (AM) product, attenuated established post-surgical pain hypersensitivity without exhibiting known side effects of opioid use in mice. This effect was achieved through direct inhibition of nociceptive dorsal root ganglion (DRG) neurons via CD44-dependent pathways. We further purified the major matrix component, the heavy chain-hyaluronic acid/pentraxin 3 (HC-HA/PTX3) from human AM that has greater purity and water solubility than FLO. HC-HA/PTX3 replicated FLO-induced neuronal and pain inhibition. Mechanistically, HC-HA/PTX3 induced cytoskeleton rearrangements to inhibit sodium current and high-voltage activated calcium current on nociceptive neurons, suggesting it is a key bioactive component mediating pain relief. Collectively, our findings highlight the potential of naturally derived biologics from human birth tissues as an effective non-opioid treatment for post-surgical pain. Moreover, we unravel the underlying mechanisms of pain inhibition induced by FLO and HC-HA/PTX3.

15.
J Biol Chem ; 287(15): 12433-44, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22351758

ABSTRACT

Recently, we reported HC-HA, a covalent complex formed between heavy chains (HCs) of inter-α-inhibitor (IαI) and hyaluronan (HA) by the catalytic action of tumor necrosis factor (TNF)-stimulated gene-6 (TSG-6), is responsible for human amniotic membrane (AM) anti-inflammatory, anti-scarring, and anti-angiogenic actions. At the present time, the only well characterized source of IαI is serum being produced by the liver. This study showed that AM epithelial and stromal cells and stromal matrix all stained positively for HA, HC 1, 2, and 3, bikunin, and TSG-6. TSG-6 mRNA and protein were constitutively expressed by cultured AM epithelial and stromal cells without being up-regulated by TNF. In serum-free conditions, these cells expressed IαI, leading to the formation of HC-HA complex that contained both HC1 and HC2. In contrast, only HC1 was found in the HC-HA complex purified from AM. Local production of IαI, the HC-TSG-6 intermediate complex, and HC-HA were abolished when cells were treated with siRNA to HC1, HC2, bikunin (all of which impair the biosynthesis of IαI), or TSG-6 but not to HC3. Collectively, these results indicate that AM is another tissue in addition to the liver to constitutively produce IαI and that the HC-HA complex made by this tissue is different from that found at inflammatory sites (e.g. in asthma and arthritis) and in the matrix of the cumulus oocyte complex.


Subject(s)
Alpha-Globulins/metabolism , Amnion/cytology , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Hyaluronic Acid/metabolism , Stromal Cells/metabolism , Alpha-Globulins/genetics , Amnion/metabolism , Cell Adhesion Molecules/genetics , Cells, Cultured , Culture Media, Serum-Free , Gene Expression , Humans , Macromolecular Substances/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
16.
J Cell Physiol ; 228(1): 225-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22674610

ABSTRACT

Epithelial-mesenchymal transition (EMT), via activation of Wnt signaling, is prevailing in embryogenesis, but postnatally it only occurs in pathological processes, such as in tissue fibrosis and tumor metastasis. Our prior studies led us to speculate that EMT might be involved in the loss of limbal epithelial stem cells in explant cultures. To examine this hypothesis, we successfully grew murine corneal/limbal epithelial progenitors by prolonging the culture time and by seeding at a low density in a serum-free medium. Single cell-derived clonal growth was accompanied by a gradient of Wnt signaling activity, from the center to the periphery, marked by a centrifugal loss of E-cadherin and ß-catenin from intercellular junctions, coupled with nuclear translocation of ß-catenin and LEF-1. Large-colony-forming efficiency at central location of colony was higher than peripheral location. Importantly, there was also progressive centrifugal differentiation, with positive K14 keratin expression and the loss of p63 and PCNA nuclear staining, and irreversible EMT, evidenced by cytoplasmic expression of α-SMA and nuclear localization of S100A4; and by nuclear translocation of Smad4. Furthermore, cytoplasmic expression of α-SMA was promoted by high-density cultures and their conditioned media, which contained cell density-dependent levels of TGF-ß1, TGF-ß2, GM-CSF, and IL-1α. Exogenous TGF-ß1 induced α-SMA positive cells in a low-density culture, while TGF-ß1 neutralizing antibody partially inhibited α-SMA expression in a high-density culture. Collectively, these results indicate that irreversible EMT emerges in the periphery of clonal expansion where differentiation and senescence of murine corneal/limbal epithelial progenitors occurs as a result of Smad-mediated TGF-ß-signaling.


Subject(s)
Cornea/cytology , Epithelial Cells/cytology , Mesenchymal Stem Cells/cytology , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism , Actins/genetics , Actins/metabolism , Animals , Antibodies, Neutralizing , Cell Differentiation/physiology , Cells, Cultured , Gene Expression Regulation/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Mesenchymal Stem Cells/physiology , Mice , Smad Proteins/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta2/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
17.
Ophthalmology ; 120(8): 1681-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583167

ABSTRACT

PURPOSE: To determine whether conjunctivochalasis (CCh) interferes with tear flow from the fornix to the tear meniscus and depletes the fornix tear reservoir. DESIGN: Comparative case series. PARTICIPANTS: The study group of 24 CCh patients (8 asymptomatic and 16 symptomatic), 9 of whom underwent operative correction, was compared with a control group of 13 normal subjects. METHODS: After instilling a 5-µl fluorescein drop into the inferior fornix, the inferior tear meniscus was depleted using a capillary tube. The tear meniscus height, with and without blinking, was recorded and calculated by video meniscometer from sequential captured images. MAIN OUTCOME MEASURES: The recovery rate of the original meniscus height was compared among groups at each time point after maximal depletion. RESULTS: The recovery rate of the tear meniscus was significantly slower in symptomatic than asymptomatic CCh patients when compared with normal subjects. Blinking 5 times facilitated such recovery in normal subjects and in asymptomatic CCh patients to the same extent as the normal, but not in symptomatic CCh patients. Deepening of the inferior fornix by removing degenerated Tenon's and reconstruction by cryopreserved amniotic membrane improved the recovery rate in symptomatic CCh patients to the same extent as normal subjects. CONCLUSIONS: The tear reservoir in the fornix rapidly replenishes the meniscus under normal circumstances. Conjunctivochalasis obliterates tears not only in the meniscus, but also in the reservoir, explaining how symptoms develop in CCh patients. Blinking is an effective compensatory mechanism to distinguish the severity of CCh. Surgical correction should not only restore the tear meniscus, but also deepen the fornix in CCh patients. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found after the references.


Subject(s)
Conjunctival Diseases/physiopathology , Tears/physiology , Adult , Aged , Aged, 80 and over , Blinking/physiology , Conjunctival Diseases/diagnosis , Conjunctival Diseases/surgery , Female , Fluorescein , Fluorescent Dyes , Fluorophotometry , Humans , Male , Middle Aged , Tears/chemistry
18.
Ophthalmology ; 120(7): 1341-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23664471

ABSTRACT

PURPOSE: To evaluate ocular demodicosis as a potential risk factor in pterygium recurrence. DESIGN: Cross-sectional study to correlate clinical findings with laboratory data. PARTICIPANTS: We retrospectively reviewed 94 patients (43 with primary and 51 with recurrent pterygia), among whom 68 patients received surgical correction, and prospectively enrolled another 23 pterygium patients and 14 nonpterygium controls for measuring the tear level of interleukin (IL)-17. METHODS: All patients had microscopically confirmed ocular demodicosis. Statistical correlations were analyzed among age, sex, aqueous tear deficiency, dry eye, ocular demodicosis, follow-up period, surgical outcome, and tear levels of IL-17 measured by enzyme-linked immunosorbent assay. MAIN OUTCOME MEASURES: Correlation between ocular demodicosis or IL-17 levels and pterygium recurrence. RESULTS: Among 94 patients, ocular demodicosis was more prevalent in patients with recurrent pterygium than those with primary pterygium (P = 0.015). During follow-up of 16.5 ± 11.5 months, 68 postsurgical patients developed 7 corneal recurrences, which constituted 7.4% of primary and 12.2% of recurrent pterygium (P = 0.820). They also developed 8 conjunctival recurrences. Kaplan-Meier survival analysis showed combined (P = 0.000), corneal (P = 0.044), and conjunctival (P = 0.002) recurrence was significantly higher among patients with demodicosis than those without. Conjunctival recurrence occurred within 6 months in eyes without demodicosis but extended beyond 6 months in eyes with demodicosis. In 34 postsurgical patients with demodicosis, the mite count of 14 patients with recurrence was significantly higher than that of 20 without (P = 0.005). The IL-17 level was significantly higher in patients with either pterygium or demodicosis than controls (P = 0.049 and 0.040, respectively), and the IL-17 level was further elevated in patients with both pterygium and demodicosis (all P<0.05). CONCLUSIONS: Ocular demodicosis is a risk factor for pterygium recurrence, especially for conjunctival recurrence, presumably by perpetuating chronic inflammation mediated by T-helper (Th)17 lymphocytes.


Subject(s)
Eye Infections, Parasitic/complications , Mite Infestations/complications , Pterygium/etiology , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Eye Infections, Parasitic/diagnosis , Eye Infections, Parasitic/metabolism , Eye Proteins/metabolism , Female , Follow-Up Studies , Humans , Interleukin-17/metabolism , Male , Middle Aged , Mite Infestations/diagnosis , Mite Infestations/metabolism , Ophthalmologic Surgical Procedures , Prospective Studies , Pterygium/diagnosis , Pterygium/metabolism , Recurrence , Retrospective Studies , Risk Factors , Tears/metabolism
19.
Taiwan J Ophthalmol ; 13(1): 49-54, 2023.
Article in English | MEDLINE | ID: mdl-37252158

ABSTRACT

PURPOSE: To assess the extent of inferior fornix shortening in conjunctivochalasis (CCh) and to evaluate whether fornix deepening reconstruction can restore the fornix tear reservoir in patients with CCh. MATERIALS AND METHODS: This was a retrospective review of five patients (3 unilateral and 2 bilateral eyes, total 7 eyes) with CCh who underwent fornix deepening reconstruction with conjunctival recession and amniotic membrane transplantation. Postsurgical outcome measures included changes in fornix depth with correlation to basal tear volumes, symptoms, corneal staining, and conjunctival inflammation. RESULTS: For the three patients with unilateral surgery, both the fornix depth (8.3 ± 1.5 mm) and wetting length (9.3 ± 8.5 mm) of the operative eyes were less than the fellow eyes (10.3 ± 1.5 mm and 10.3 ± 8.5 mm, respectively). At 5.3 ± 2.7 months (range 1.7-8.7) postoperatively, the fornix depth increased significantly by 2.0 ± 1.1 mm (P = 0.02). Deepening of the fornix depth was accompanied by overwhelming symptomatic relief (91.5%) that could be subdivided into complete relief (87.5%) and partial relief (4%) of symptoms, with blurred vision being the most notably relieved symptom (P = 0.03). Furthermore, superficial punctate keratitis and conjunctival inflammation were significantly improved at follow-up (P = 0.008 and 0.05, respectively). CONCLUSION: Deepening of the fornix to restore the tear reservoir is an important surgical objective that may change the tear hydrodynamic state to provide a stable tear film and improve outcomes in CCh.

20.
Ocul Surf ; 29: 301-310, 2023 07.
Article in English | MEDLINE | ID: mdl-37268293

ABSTRACT

PURPOSE: Human corneal endothelial cells (HCECs) play a significant role in maintaining visual function. However, these cells are notorious for their limited proliferative capacity in vivo. Current treatment of corneal endothelial dysfunction resorts to corneal transplantation. Herein we describe an ex vivo engineering method to manufacture HCEC grafts suitable for transplantation through reprogramming into neural crest progenitors. METHODS: HCECs were isolated by collagenase A from stripped Descemet membrane of cadaveric corneoscleral rims, and induced reprogramming via knockdown with p120 and Kaiso siRNAs on collagen IV-coated atelocollagen. Engineered HCEC grafts were released after assessing their identity, potency, viability, purity and sterility. Phase contrast was used for monitoring cell shape, graft size, and cell density. Immunostaining was used to determine the normal HCEC phenotype with expression of N-cadherin, ZO-1, ATPase, acetyl-α-tubulin, γ-tubulin, p75NTR, α-catenin, ß-catenin, and F-actin. Stability of manufactured HCEC graft was evaluated after transit and storage for up to 3 weeks. The pump function of HCEC grafts was measured by lactate efflux. RESULTS: One HCEC graft suitable for corneal transplantation was generated from 1/8th of the donor corneoscleral rim with normal hexagonal cell shape, density, and phenotype. The manufactured grafts were stable for up to 3 weeks at 37 °C or up to 1 week at 22 °C in MESCM medium and after transcontinental shipping at room temperature by retaining normal morphology (hexagonal, >2000 cells/mm2, >8 mm diameter), phenotype, and pump function. CONCLUSIONS: This regenerative strategy through knockdown with p120 and Kaiso siRNAs can be used to manufacture HCEC grafts with normal phenotype, morphology and pump function following prolonged storage and shipping.


Subject(s)
Corneal Transplantation , Endothelium, Corneal , Humans , Endothelium, Corneal/metabolism , Endothelium, Corneal/transplantation , Endothelial Cells , Cells, Cultured , Cornea
SELECTION OF CITATIONS
SEARCH DETAIL