Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 65(6): 39, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38935032

ABSTRACT

Purpose: This study aimed to explore protective effects and potential mechanism of ectoine, a natural osmoprotectant, on ocular surface mucin production in dry eye disease. Methods: A dry eye model was established in C57BL/6 mice exposed to desiccating stress (DS) with untreated (UT) mice as controls. DS mice were topically treated with 2.0% ectoine or PBS vehicle. Corneal epithelial defects were assessed by Oregon Green Dextran (OGD) fluorescent staining. Conjunctival goblet cells, ocular mucins, and T help (Th) cytokines were evaluated by immunofluorescent staining or ELISA, and RT-qPCR. Results: Compared with UT mice, corneal epithelial defects were detected as strong punctate OGD fluorescent staining in DS mice with vehicle, whereas ectoine treatment largely reduced OGD staining to near-normal levels. Conjunctival goblet cell density and cell size decreased markedly in DS mice, but was significantly recovered by ectoine treatment. The protein production and mRNA expression of two gel-forming secreted MUC5AC and MUC2, and 4 transmembrane mucins, MUC1, MUC4, MUC16, and MUC15, largely decreased in DS mice, but was restored by ectoine. Furthermore, Th2 cytokine IL-13 was inhibited, whereas Th1 cytokine IFN-γ was stimulated at protein and mRNA levels in conjunctiva and draining cervical lymph nodes (CLNs) of DS mice, leading to decreased IL-13/IFN-γ ratio. Interestingly, 2.0% ectoine reversed their alternations and restored IL-13/IFN-γ balance. Conclusions: Our findings demonstrate that topical ectoine significantly reduces corneal damage, and enhances goblet cell density and mucin production through restoring imbalanced IL-13/IFN-γ signaling in murine dry eye model. This suggests therapeutic potential of natural osmoprotectant ectoine for dry eye disease.


Subject(s)
Disease Models, Animal , Dry Eye Syndromes , Goblet Cells , Interferon-gamma , Interleukin-13 , Mice, Inbred C57BL , Mucins , Animals , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/drug therapy , Mice , Goblet Cells/metabolism , Goblet Cells/drug effects , Goblet Cells/pathology , Interferon-gamma/metabolism , Mucins/metabolism , Mucins/biosynthesis , Mucins/genetics , Interleukin-13/metabolism , Conjunctiva/metabolism , Conjunctiva/drug effects , Conjunctiva/pathology , Enzyme-Linked Immunosorbent Assay , Female , Epithelium, Corneal/metabolism , Epithelium, Corneal/drug effects , Real-Time Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Amino Acids, Diamino
2.
Ocul Surf ; 32: 182-191, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38490477

ABSTRACT

PURPOSE: To explore novel role and molecular mechanism of a natural osmoprotectant ectoine in protecting corneal epithelial cell survival and barrier from hyperosmotic stress. METHODS: Primary human corneal epithelial cells (HCECs) were established from donor limbus. The confluent cultures in isosmolar medium were switched to hyperosmotic media (400-500 mOsM), with or without ectoine or rhIL-37 for different time periods. Cell viability and proliferation were evaluated by MTT or WST assay. The integrity of barrier proteins and the expression of cytokines and cathepsin S were evaluated by RT-qPCR, ELISA, and immunostaining with confocal microscopy. RESULTS: HCECs survived well in 450mOsM but partially damaged in 500mOsM medium. Ectoine well protected HCEC survival and proliferation at 500mOsM. The integrity of epithelial barrier was significantly disrupted in HCECs exposed to 450mOsM, as shown by 2D and 3D confocal immunofluorescent images of tight junction proteins ZO-1 and occludin. Ectoine at 5-20 mM well protected these barrier proteins under hyperosmotic stress. The expression of TNF-α, IL-1ß, IL-6 and IL-8 were dramatically stimulated by hyperosmolarity but significantly suppressed by Ectoine at 5-40 mM. Cathepsin S, which was stimulated by hyperosmolarity, directly disrupted epithelial barrier. Interestingly, anti-inflammatory cytokine IL-37 was suppressed by hyperosmolarity, but restored by ectoine at mRNA and protein levels. Furthermore, rhIL-37 suppressed cathepsin S and rescued cell survival and barrier in HCECs exposed to hyperosmolarity. CONCLUSION: Our findings demonstrate that ectoine protects HCEC survival and barrier from hyperosmotic stress by promoting IL-37. This provides new insight into pathogenesis and therapeutic potential for dry eye disease.


Subject(s)
Amino Acids, Diamino , Cell Survival , Epithelium, Corneal , Osmotic Pressure , Humans , Cell Survival/drug effects , Epithelium, Corneal/metabolism , Epithelium, Corneal/drug effects , Epithelium, Corneal/pathology , Cells, Cultured , Amino Acids, Diamino/pharmacology , Interleukin-1/metabolism , Interleukin-1/pharmacology , Enzyme-Linked Immunosorbent Assay , Microscopy, Confocal , Cell Proliferation/drug effects , Cytokines/metabolism
3.
Pharmaceutics ; 16(2)2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38399289

ABSTRACT

Ectoine, a novel natural osmoprotectant, protects bacteria living in extreme environments. This study aimed to explore the therapeutic effect of ectoine for dry eye disease. An experimental dry eye model was created in C57BL/6 mice exposed to desiccating stress (DS) with untreated mice as controls (UT). DS mice were dosed topically with 0.5-2.0% of ectoine or a vehicle control. Corneal epithelial defects were detected via corneal smoothness and Oregon Green dextran (OGD) fluorescent staining. Pro-inflammatory cytokines and chemokines were evaluated using RT-qPCR and immunofluorescent staining. Compared with UT mice, corneal epithelial defects were observed as corneal smoothness irregularities and strong punctate OGD fluorescent staining in DS mice with vehicle. Ectoine treatment protected DS mice from corneal damage in a concentration-dependent manner, and ectoine at 1.0 and 2.0% significantly restored the corneal smoothness and reduced OGD staining to near normal levels. Expression of pro-inflammatory cytokines (TNF-α, IL-1ß, and IL-6) and chemokines CCL3 and CXCL11 was significantly elevated in the corneas and conjunctivas of DS mice, whereas 1.0 and 2.0% ectoine suppressed these inflammatory mediators to near normal levels. Our findings demonstrate that ectoine can significantly reduce the hallmark pathologies associated with dry eye and may be a promising candidate for treating human disease.

4.
Ocul Surf ; 26: 234-243, 2022 10.
Article in English | MEDLINE | ID: mdl-36208723

ABSTRACT

PURPOSE: To explore novel role and molecular mechanism of a natural anti-inflammatory cytokine interleukin (IL) 37 in preventing corneal epithelial barrier disruption from hyperosmolar stress as can occur in dry eye disease. METHODS: Primary human corneal epithelial cells (HCECs) were cultured from fresh donor limbal explants. An in vitro dry eye model with hyperosmolar stress was established by switching HCECs from isosmolar (312mOsM) to hyperosmolar medium (350-500 mOsM), and some cells were treated with rhIL-37 or rhTNF-α, for different periods (2-48 h). The expression of cytokines and cathepsin S, and barrier protein integrity were evaluated by RT-qPCR, ELISA, and immunofluorescent staining with confocal microscopy. RESULTS: The integrity of epithelial barrier was significantly disrupted in HCECs exposed to hyperosmolar medium, as shown by immunofluorescent images of tight junction (TJ, ZO-1, occludin and claudin-1) and adheren junction (E-cadherin) proteins. TNF-α accentuated hyperosmolar-induced disruption of TJ barrier functional integrity whereas exposure to IL-37 blunted or even reversed these changes. Cathepsin S, encoded by CTSS gene, was found to directly disrupt epithelial barrier integrity. Interestingly, CTSS expression was significantly induced by TNF-α and hyperosmolarity, while exogenous rhIL-37 inhibited TNF-α and CTSS expression at mRNA and protein levels following hyperosmolar stress. Furthermore, rhIL-37 restored barrier protein integrity, observed in 2D and 3D confocal immunofluorescent images, in HCECs under hyperosmolar stress. CONCLUSION: Our findings demonstrate a novel signaling pathway by which anti-inflammatory cytokine IL-37 prevents corneal epithelial barrier disruption under hyperosmotic stress via suppressing TNF-α and CTSS expression. This study provides new insight into mechanisms protecting corneal barrier in dry eye disease.


Subject(s)
Dry Eye Syndromes , Epithelium, Corneal , Interleukin-1 , Humans , Cathepsins/metabolism , Cells, Cultured , Cytokines/metabolism , Dry Eye Syndromes/metabolism , Epithelium, Corneal/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Interleukin-1/metabolism
5.
Exp Eye Res ; 214: 108895, 2022 01.
Article in English | MEDLINE | ID: mdl-34910926

ABSTRACT

Cathepsin S (Ctss) is a protease that is proinflammatory on epithelial cells. The purpose of this study was to investigate the role of Ctss in age-related dry eye disease. Ctss-/- mice [in a C57BL/6 (B6) background] of different ages were compared to B6 mice. Ctss activity in tears and lacrimal gland (LG) lysates was measured. The corneal barrier function was investigated in naïve mice or after topical administration of Ctss eye drops 5X/day for two days. Eyes were collected, and conjunctival goblet cell density was measured in PAS-stained sections. Immunoreactivity of the tight junction proteins, ZO-1 and occludin, was investigated in primary human cultured corneal epithelial cells (HCEC) without or with Ctss, with or without a Ctss inhibitor. A significant increase in Ctss activity was observed in the tears and LG lysates in aged B6 compared to young mice. This was accompanied by higher Ctss transcripts and protein expression in LG and spleen. Compared to B6, 12 and 24-month-old Ctss-/- mice did not display age-related corneal barrier disruption and goblet cell loss. Treatment of HCEC with Ctss for 48 h disrupted occludin and ZO-1 immunoreactivity compared to control cells. This was prevented by the Ctss inhibitor LY3000328 or Ctss-heat inactivation. Topical reconstitution of Ctss in Ctss-/- mice for two days disrupted corneal barrier function. Aging on the ocular surface is accompanied by increased expression and activity of the protease Ctss. Our results suggest that cathepsin S modulation might be a novel target for age-related dry eye disease.


Subject(s)
Aging/physiology , Cathepsins/metabolism , Dry Eye Syndromes/metabolism , Lacrimal Apparatus/metabolism , Tears/metabolism , Animals , Cells, Cultured , Conjunctiva/metabolism , Drug Delivery Systems , Dry Eye Syndromes/drug therapy , Epithelium, Corneal/metabolism , Goblet Cells/metabolism , Mice , Mice, Inbred C57BL , Occludin/metabolism , Spleen/metabolism , Tight Junction Proteins/metabolism , Zonula Occludens-1 Protein/metabolism
6.
Ocul Surf ; 22: 163-171, 2021 10.
Article in English | MEDLINE | ID: mdl-34428579

ABSTRACT

PURPOSE: To explore the distinct expression and diverse roles of IL-36 cytokines in dry eye disease using an in vitro hyperosmolarity model of human corneal epithelial cells (HCECs). METHODS: Primary HCECs were cultured from fresh donor limbal explants. Hyperosmolarity model was established by switching HCECs from isosmotic (312 mOsM) to hyperosmotic medium (350-500 mOsM) alone or with addition of rhIL-36RA or rhIL-38 for 2-48 h. Some cultures were treated with IL-36α (1-10 ng/ml) with or without rhIL-36RA or rhIL-38. Gene expression was detected by RT-qPCR; and protein production and barrier disruption were evaluated by ELISA and/or immunofluorescent staining. RESULTS: IL-36 cytokines were differential expressed in primary HCECs. Among 3 pro-inflammatory agonists, IL-36α, but not IL-36ß and IL-36γ, was distinctly induced at osmolarity-dependent manner while two antagonist IL-36RA and IL-38 were significantly suppressed in HCECs exposed to hyperosmotic stress. IL-36α increased to 4.4-fold in mRNA and 6.9-fold at protein levels (116.0 ± 36.33 pg/ml vs 16.79 ± 6.51 pg/ml in controls) by 450 mOsM, but dramatically inhibited by addition of rhIL-36RA or rhIL-38. Exogenous rhIL-36α stimulated expression of TNF-α and IL-1ß at mRNA and protein levels and disrupted tight junction proteins ZO-1 and occludin. However, rhIL-36RA or rhIL-38 suppressed TNF-α and IL-1ß production and protected HCECs from barrier disruption in response to IL-36α or hyperosmolarity. CONCLUSIONS: Our findings demonstrate that the stimulated pro-inflammatory IL-36α with the suppressed antagonists IL-36RA and IL-38 is a novel mechanism by which hyperosmolarity induces inflammation in dry eye. IL-36RA and IL-38 may have a therapeutic potential in dry eye.


Subject(s)
Dry Eye Syndromes , Epithelium, Corneal , Cytokines , Epithelial Cells , Humans , Inflammation , Interleukins , Signal Transduction
7.
Invest Ophthalmol Vis Sci ; 62(9): 36, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34297801

ABSTRACT

Purpose: Differentiated from adult stem cells (ASCs), transit-amplifying cells (TACs) play an important role in tissue homeostasis, development, and regeneration. This study aimed to characterize the gene expression profile of a candidate TAC population in limbal basal epithelial cells using single-cell RNA sequencing (scRNA-seq). Methods: Single cells isolated from the basal corneal limbus were subjected to scRNA-seq using the 10x Genomics platform. Cell types were clustered by graph-based visualization methods and unbiased computational analysis. BrdU proliferation assays, immunofluorescent staining, and real-time reverse transcription quantitative polymerase chain reaction were performed using multiple culture models of primary human limbal epithelial cells to characterize the TAC pool. Results: Single-cell transcriptomics of 16,360 limbal basal cells revealed 12 cell clusters. A unique cluster (3.21% of total cells) was identified as a TAC entity, based on its less differentiated progenitor status and enriched exclusive proliferation marker genes, with 98.1% cells in S and G2/M phases. The cell cycle-dependent genes were revealed to be largely enriched by the TAC population. The top genes were characterized morphologically and functionally at protein and mRNA levels. The specific expression patterns of RRM2, TK1, CENPF, NUSAP1, UBE2C, and CDC20 were well correlated in a time- and cycle-dependent manner with proliferation stages in the cell growth and regeneration models. Conclusions: For the first time, to the best of our knowledge, we have identified a unique TAC entity and uncovered a group of cell cycle-dependent genes that serve as TAC signature markers. The findings provide insight into ASCs and TACs and lay the foundation for understanding corneal homeostasis and diseases.


Subject(s)
Epithelium, Corneal/cytology , Limbus Corneae/cytology , Transcriptome/genetics , Cell Count , Cell Cycle , Cell Differentiation , Cell Proliferation , Cells, Cultured , Epithelium, Corneal/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Female , Humans , Limbus Corneae/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
8.
Ocul Surf ; 20: 20-32, 2021 04.
Article in English | MEDLINE | ID: mdl-33388438

ABSTRACT

PURPOSE: This study aimed to uncover novel cell types in heterogenous basal limbus of human cornea for identifying LSC at single cell resolution. METHODS: Single cells of human limbal basal epithelium were isolated from young donor corneas. Single-cell RNA-Sequencing was performed using 10x Genomics platform, followed by clustering cell types through the graph-based visualization method UMAP and unbiased computational informatic analysis. Tissue RNA in situ hybridization with RNAscope, immunofluorescent staining and multiple functional assays were performed using human corneas and limbal epithelial culture models. RESULTS: Single-cell transcriptomics of 16,360 limbal basal cells revealed 12 cell clusters belonging to three lineages. A smallest cluster (0.4% of total cells) was identified as LSCs based on their quiescent and undifferentiated states with enriched marker genes for putative epithelial stem cells. TSPAN7 and SOX17 are discovered and validated as new LSC markers based on their exclusive expression pattern and spatial localization in limbal basal epithelium by RNAscope and immunostaining, and functional role in cell growth and tissue regeneration models with RNA interference in cultures. Interestingly, five cell types/states mapping a developmental trajectory of LSC from quiescence to proliferation and differentiation are uncovered by Monocle3 and CytoTRACE pseudotime analysis. The transcription factor networks linking novel signaling pathways are revealed to maintain LSC stemness. CONCLUSIONS: This human corneal scRNA-Seq identifies the LSC population and uncovers novel cell types mapping the differentiation trajectory in heterogenous limbal basal epithelium. The findings provide insight into LSC concept and lay the foundation for understanding the corneal homeostasis and diseases.


Subject(s)
Epithelium, Corneal , Limbus Corneae , Cell Differentiation , Cornea , Humans , Stem Cells , Transcriptome
9.
Int J Mol Sci ; 21(23)2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33255884

ABSTRACT

Inflammation is the main pathophysiology of dry eye, characterized by tear film instability and hyperosmolarity. The aim of this study was to investigate the association of inflammation and cellular autophagy using an in vitro dry eye model with primary cultured human corneal epithelial cells (HCECs). Primary HCECs cultured with fresh limbal explants from donors were switched to a hyperosmotic medium (450 mOsM) by adding sodium chloride into the culture medium. We observed the stimulated inflammatory mediators, TNF-α, IL-1ß, IL-6 and IL-8, as well as the increased expression of autophagy related genes, Ulk1, Beclin1, Atg5 and LC3B, as evaluated by RT-qPCR and ELISA. The immunofluorescent staining of LC3B and Western blotting revealed the activated autophagosome formation and autophagic flux, as evidenced by the increased LC3B autophagic cells with activated Beclin1, Atg5, Atg7 and LC3B proteins, and the decreased levels of P62 protein in HCECs. Interestingly, the autophagy activation was later at 24 h than inflammation induced at 4 h in HCECs exposed to 450 mOsM. Furthermore, application of rapamycin enhanced autophagy activation also reduced the inflammatory mediators and restored cell viability in HCECs exposed to the hyperosmotic medium. Our findings for the first time demonstrate that the autophagy activation is a late phase response to hyperosmotic stress, and is enhanced by rapamycin, which protects HCECs by suppressing inflammation and promoting cells survival, suggesting a new therapeutic potential to treat dry eye diseases.


Subject(s)
Autophagy , Dry Eye Syndromes/pathology , Inflammation/pathology , Models, Biological , Adolescent , Adult , Aged , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Survival/drug effects , Cells, Cultured , Cytoprotection/drug effects , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelium, Corneal/pathology , Humans , Inflammation Mediators/metabolism , Middle Aged , Osmotic Pressure , Sirolimus/pharmacology , Time Factors , Young Adult
10.
Invest Ophthalmol Vis Sci ; 61(10): 26, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32785678

ABSTRACT

Purpose: Autophagy plays an important role in balancing the inflammatory response to restore homeostasis. The aim of this study was to explore the mechanism by which trehalose suppresses inflammatory cytokines via autophagy activation in primary human corneal epithelial cells (HCECs) exposed to hyperosmotic stress. Methods: An in vitro dry eye model was used in which HCECs were cultured in hyperosmolar medium with the addition of sodium chloride (NaCl). Trehalose was applied in different concentrations. The levels of TNF-α, IL-1ß, IL-6, and IL-8 were detected using RT-qPCR and ELISA. Cell viability assays, immunofluorescent staining of LC3B, and western blots of Beclin1, Atg5, Atg7, LC3B, and P62 were conducted. The key factors in upstream signaling pathways of autophagy activation were measured: P-Akt, Akt, and transcription factor EB (TFEB). Results: Trehalose reduced the proinflammatory mediators TNF-α, IL-1ß, IL-6, and IL-8 in primary HCECs at 450 mOsM. This effect was osmolarity dependent, and a level of 1.0% trehalose showed the most suppression. Trehalose promoted autophagosome formation and autophagic flux, as evidenced by increased production of Beclin1, Atg5, and Atg7, as well as higher LC3B I protein turnover to LC3B II, with decreased protein levels of P62/SQSTM1. The addition of 3-methyladenine blocked autophagy activation and increased the release of proinflammatory cytokines. Trehalose further activated TFEB, with translocation from cytoplasm to the nucleus, but diminished Akt activity. Conclusions: Our findings demonstrate that trehalose, functioning as an autophagy enhancer, suppresses the inflammatory response by promoting autophagic flux via TFEB activation in primary HCECs exposed to hyperosmotic stress, a process that is beneficial to dry eye.


Subject(s)
Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Epithelium, Corneal/drug effects , Signal Transduction/drug effects , Trehalose/pharmacology , Adolescent , Adult , Aged , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Epithelium, Corneal/cytology , Epithelium, Corneal/metabolism , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Middle Aged , Osmotic Pressure , Real-Time Polymerase Chain Reaction , Trehalose/administration & dosage , Tumor Necrosis Factor-alpha/metabolism , Young Adult
11.
Mucosal Immunol ; 13(6): 919-930, 2020 11.
Article in English | MEDLINE | ID: mdl-32358573

ABSTRACT

This study was to explore a novel IL-33/ST2/IL-9/IL-9R signaling pathway that disrupts ocular surface barrier and amplifies allergic inflammation. Two murine models of experimental allergic conjunctivitis (EAC) and IL-9 topical challenge in wild type Balb/c and ST2-/- mice, and two culture models of primarily human corneal epithelial cells (HCECs) and mouse CD4+ T cells were performed. Clinical manifestations, Oregon-Green Dextran (OGD) staining, the apical junction complexes (AJCs), IL-33/ST2 and IL-9/IL-9R signaling molecules were evaluated in ocular surface and its draining cervical lymph nodes (CLNs) by RT-qPCR, immunostaining and ELISA. The typical allergic signs, enhanced OGD staining intensity, disrupted morphology of AJCs, including ZO-1, claudin 1, occludin, and E-cadherin, and the stimulated signaling of IL-33/ST2 and IL-9/IL-9R were observed in ocular mucosa and draining CLNs in EAC-Balb/c mice, but significantly reduced or eliminated in EAC-ST2-/- mice. Topical challenge of IL-9 resulted in the obvious OGD staining and disrupted ocular surface AJCs in Balb/c mice and in HCECs in vitro. IL-9 production was found to be stimulated by IL-33 in CD4+ cells from Balb/c mice in vitro. Our findings uncovered a novel phenomenon and mechanism by which ocular surface barrier integrity is disrupted in allergic conjunctivitis by IL-33/ST2/IL-9/IL-9R signaling pathway, which may amplify the allergic inflammation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Conjunctivitis, Allergic/immunology , Epithelium, Corneal/metabolism , Eye/metabolism , Inflammation/immunology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Interleukin-9/metabolism , Receptors, Interleukin-9/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Epithelium, Corneal/pathology , Eye/pathology , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , Tight Junction Proteins/metabolism
12.
J Biochem ; 167(1): 41-48, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31598678

ABSTRACT

The study aimed to investigate the regulatory effect of miR-146a in proliferation, invasion and migration of breast cancer and its possible mechanism via NM23-H1. The expression levels of miR-146a in breast cancer with different pathological classification were significantly increased, while the expression levels of NM23-H1 were significantly decreased, which were closely correlated. Double luciferase reporter gene was used to verify the target regulatory relationship between miR-146 and NM23-H1 on a human breast cancer cell line. miR-146a was closely related to the proliferation and metastasis of breast cancer. miR-146a also promoted the growth of breast cancer in vivo via targeting NM23-H1. In conclusion, miR-146 can promote the proliferation and invasion of breast cancer by targeting NM23-H1.


Subject(s)
Breast Neoplasms/metabolism , MicroRNAs/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Tumor Cells, Cultured
13.
Mucosal Immunol ; 12(5): 1141-1149, 2019 09.
Article in English | MEDLINE | ID: mdl-31350466

ABSTRACT

This study was to explore the role and mechanism of macrophages in pollen-triggered allergic inflammation. A murine model of short ragweed (SRW) pollen-induced experimental allergic conjunctivitis (EAC), and bone marrow (BM)-macrophages cultures were used. Typical allergic manifestations and TSLP-stimulated Th2 hyperresponse were observed in ocular surface of EAC model in wild-type (WT) mice induced by SRW. The M2 phenotype markers, Arg1, Ym1 and FIZZ1, were highly expressed by conjunctiva and draining cervical lymph nodes (CLNs) of WT-EAC mice when compared with controls, as evaluated by RT-qPCR and Immunofluorescent double staining with macrophage marker F4/80. The stimulated expression of TSLPR and OX40L by macrophage was detected in conjunctiva and CLNs by RT-qPCR, double staining, and flow cytometry. M2 macrophages were found to produce TARC and MDC. In contrast, EAC model with TSLPR-/- mice did not show allergic signs and any increase of Th2 cytokines (IL-4, IL-5 and IL-13) and M2 markers. In vitro cultures confirmed that SRW extract stimulates expression of TSLPR, OX40L, TARC, MDC, and three M2 markers by BM-macrophages from WT mice, but not from TSLPR-/- mice. These findings demonstrate that SRW pollen primes macrophage polarization toward to M2 phenotype via TSLP/TSLPR/OX40L signaling to amplify allergic inflammation.


Subject(s)
Antigens, Plant/immunology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Plant Extracts/immunology , Signal Transduction , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Immunoglobulins/metabolism , Mice , Mice, Knockout , OX40 Ligand/metabolism , Phenotype , Receptors, Cytokine/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Thymic Stromal Lymphopoietin
14.
Allergy ; 74(5): 910-921, 2019 05.
Article in English | MEDLINE | ID: mdl-30515838

ABSTRACT

BACKGROUND: While most studies focus on pro-allergic cytokines, the protective role of immunosuppressive cytokines in allergic inflammation is not well elucidated. This study was to explore a novel anti-inflammatory role and cellular/molecular mechanism of IL-27 in allergic inflammation. METHODS: A murine model of experimental allergic conjunctivitis (EAC) was induced in BALB/c, C57BL/6 or IL-27Rα-deficient (WSX-1-/- ) mice by short ragweed pollen, with untreated or PBS-treated mice as controls. The serum, eyeballs, conjunctiva, cervical lymph nodes (CLNs) were used for study. Gene expression was determined by RT-qPCR, and protein production and activation were evaluated by immunostaining, ELISA and Western blotting. RESULTS: Typical allergic manifestations and stimulated thymic stromal lymphopoietin (TSLP) signaling and Th2 responses were observed in ocular surface of EAC models in BALB/c and C57BL/6 mice. The decrease of IL-27 at mRNA (IL-27/EBI3) and protein levels were detected in serum, conjunctiva and CLN, as evaluated by RT-qPCR, immunofluorescent staining, ELISA and Western blotting. EAC induced in WSX-1-/- mice showed aggravated allergic signs with higher TSLP-driven Th2-dominant inflammation, accompanied by stimulated Th17 responses, including IL-17A, IL-17F, and transcription factor RORγt. In contrast, Th1 cytokine IFNγ and Treg marker IL-10, with their respective transcription factors T-bet and foxp3, were largely suppressed. Interestingly, imbalanced activation between reduced phosphor (P)-STAT1 and stimulated P-STAT6 were revealed in EAC, especially WSX-1-/- -EAC mice. CONCLUSION: These findings demonstrated a natural protective mechanism by IL-27, of which signaling deficiency develops a Th17-type hyperresponse that further aggravates Th2-dominant allergic inflammation.


Subject(s)
Conjunctivitis, Allergic/etiology , Conjunctivitis, Allergic/metabolism , Disease Susceptibility , Interleukin-27/metabolism , Signal Transduction , Th17 Cells/metabolism , Th2 Cells/metabolism , Animals , Biomarkers , Biopsy , Conjunctivitis, Allergic/pathology , Cytokines/metabolism , Disease Models, Animal , Immunohistochemistry , Inflammation Mediators/metabolism , Mice , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/immunology , Th2 Cells/immunology
15.
Int Immunol ; 30(10): 457-470, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30010888

ABSTRACT

Conjunctival goblet cell loss in ocular surface diseases is accompanied by increased number of interleukin-12 (IL-12)-producing antigen-presenting cells (APCs) and increased interferon-γ (IFN-γ) expression. This study tested the hypothesis that mouse conjunctival goblet cells produce biologically active retinoic acid (RA) that suppresses CD86 expression and IL-12 production by myeloid cells. We found that conditioned media from cultured conjunctival goblet cells (CjCM) suppressed stimulated CD86 expression, NF-κB p65 activation and IL-12 and IFN-γ production in unstimulated and lipopolysaccharide-stimulated cultured bone marrow-derived cells (BMDCs) containing a mixed population of APCs. Goblet cell-conditioned, ovalbumin-loaded APCs suppressed IFN-γ production and increased IL-13 production in co-cultured OTII cells. The goblet cell suppressive activity is due in part to their ability to synthesize RA from retinol. Conjunctival goblet cells had greater expression of aldehyde dehydrogenases Aldh1a1 and a3 and ALDEFLUOR activity than cornea epithelium lacking goblet cells. The conditioning activity was lost in goblet cells treated with an ALDH inhibitor, and a retinoid receptor alpha antagonist blocked the suppressive effects of CjCM on IL-12 production. Similar to RA, CjCM increased expression of suppressor of cytokine signaling 3 (SOCS3) in BMDCs. SOCS3 silencing reversed the IL-12-suppressive effects of CjCM. Our findings indicate that conjunctival goblet cells are capable of synthesizing RA from retinol secreted by the lacrimal gland into tears that can condition APCs. Evidence suggests goblet cell RA may function in maintaining conjunctival immune tolerance and loss of conjunctival goblet cells may contribute to increased Th1 priming in dry eye.


Subject(s)
B7-2 Antigen/biosynthesis , Bone Marrow Cells/metabolism , Goblet Cells/metabolism , Interleukin-12/biosynthesis , Tretinoin/metabolism , Animals , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , Benzoates/pharmacology , Bone Marrow Cells/immunology , Cells, Cultured , Chromans/pharmacology , Female , Goblet Cells/chemistry , Goblet Cells/immunology , Interleukin-12/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Tretinoin/chemistry
16.
Cell Physiol Biochem ; 47(1): 316-329, 2018.
Article in English | MEDLINE | ID: mdl-29768259

ABSTRACT

BACKGROUND/AIMS: The rejuvenation properties of nanofat grafting have been described in recent years. However, it is not clear whether the clinical efficacy of the procedure is attributable to stem cells or linked to other components of adipose tissue. In this study we isolated nanofat-derived stem cells (NFSCs) to observe their biological characteristics and evaluate the efficacy of precise intradermal injection of nanofat combined with platelet-rich fibrin (PRF) in patients undergoing facial rejuvenation treatment. METHODS: Third-passage NFSCs were isolated and cultured using a mechanical emulsification method and their surface CD markers were analyzed by flow cytometry. The adipogenic and osteogenic nature and chondrogenic differentiation capacity of NFSCs were determined using Oil Red O staining, alizarin red staining, and Alcian blue staining, respectively. Paracrine function of NFSCs was evaluated by enzyme-linked immunosorbent assay (ELISA) at 1, 3, 7, 14, and 28 days after establishing the culture. Then, the effects of PRF on NFSC proliferation were assessed in vitro. Finally, we compared the outcome in 103 patients with facial skin aging who underwent both nanofat and intradermal PRF injection (treatment group) and 128 patients who underwent hyaluronic acid (HA) injection treatment (control group). Outcomes in the two groups were compared by assessing pictures taken at the same angle before and after treatment, postoperative recovery, incidence of local absorption and cysts, and skin quality before treatment, and at 1, 12, 24 months after treatment using the VISIA Skin Image Analyzer and a SOFT5.5 skin test instrument. RESULTS: NFSCs expressed CD29, CD44, CD49d, CD73, CD90, and CD105, but did not express CD34, CD45, and CD106. NFSCs also differentiated into adipocytes, osteoblasts, and chondrocytes under appropriate induction conditions. NFSCs released large amounts of growth factors such as VEGF, bFGF, EGF, and others, and growth factor levels increased in a time-dependent manner. At the same time, PRF enhanced proliferation of NFSCs in vitro in a dose-dependent manner, and the growth curves under different concentrations of PRF all showed plateaus 6d after seeding. Facial skin texture was improved to a greater extent after combined injection of nanofat and PRF than after control injection of HA. The nanofat-PRF group had a higher satisfaction rate. Neither treatment caused any complications such as infection, anaphylaxis, or paresthesia during long-term follow-up. CONCLUSION: NFSCs demonstrate excellent multipotential differentiation and paracrine function, and PRF promotes proliferation of NFSCs during the early stage after seeding. Both nanofat-PRF and HA injection improve facial skin status without serious complications, but the former was associated with greater patient satisfaction, implying that nanofat-PRF injection is a safe, highly effective, and long-lasting method for skin rejuvenation.


Subject(s)
Adipose Tissue/cytology , Platelet-Rich Fibrin/metabolism , Rejuvenation , Skin Aging , Skin Physiological Phenomena , Stromal Cells/cytology , Stromal Cells/transplantation , Adult , Cell Proliferation , Cells, Cultured , Face , Female , Humans , Injections, Intradermal , Intercellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Stromal Cells/metabolism , Young Adult
17.
Sci Rep ; 7(1): 5169, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701781

ABSTRACT

Human Corneal epithelial stem cells (CESCs) have been identified to reside in limbus for more than 2 decades. However, the precise location of CESCs in other mammalian remains elusive. This study was to identify differential localization of putative CESCs in mice. Through a series of murine corneal cross-sections from different directions, we identified that anatomically and morphologically the murine limbus is composed of the thinnest epithelium and the thinnest stroma without any palisades of Vogt-like niche structure. The cells expressing five of stem/progenitor cell markers are localized in basal layer of entire murine corneal epithelium. BrdU label-retaining cells, a key characteristic of epithelial stem cells, are detected in both limbal and central cornea of mouse eye. Functionally, corneal epithelium can be regenerated in cultures from central and limbal explants of murine cornea. Such a distribution of mouse CESCs is different from human cornea, where limbal stem cell concept has been well established and accepted. We are aware that some new evidence supports limbal stem cell concept in mouse recently. However, it is important to know that central cornea may provide an alternative source of stem cells when one utilizes mice as animal model for corneal research.


Subject(s)
Cell Differentiation , Epithelium, Corneal/cytology , Phenotype , Stem Cells/cytology , Stem Cells/metabolism , Animals , Biomarkers , Humans , Immunohistochemistry , Limbus Corneae/cytology , Mice
18.
Oncotarget ; 8(29): 46875-46890, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28423354

ABSTRACT

Fat flap transplantation is frequently performed in patients suffering from soft tissue defects resulting from disease or trauma. This study explored the feasibility of constructing vascularized fat flaps using rabbit adipose-derived stem cells (rASCs) and collagen scaffolds in a rabbit model. We evaluated rASCs proliferation, paracrine function, adipogenesis, vascularization, and CD54 expression, with or without HIF-1α transfection in vitro and in vivo. We observed that adipogenic differentiation potential was greater in rASCs with high CD54 expression (CD54+rASCs) than in those with low expression (CD54-rASCs), both in vitro and in vivo. HIF-1α overexpression not only augmented this effect, but also enhanced cell proliferation and paracrine function in vitro. We also demonstrated that HIF-1α-transfected CD54+rASCs showed enhanced paracrine function and adipogenic capacity, and that paracrine function increases expression of angiogenesis-related markers. Thus, CD54+rASCs overexpressing HIF-1α enhanced large volume vascularized fat flap regeneration in rabbits, suggesting CD54 may be an ideal candidate marker for ASCs adipogenic differentiation.


Subject(s)
Adipose Tissue/cytology , Free Tissue Flaps , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Adipogenesis/genetics , Animals , Biomarkers , Cell Differentiation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunophenotyping , Models, Animal , Neovascularization, Physiologic , Paracrine Communication , Rabbits , Wound Healing/genetics
19.
Int J Mol Sci ; 18(3)2017 Mar 05.
Article in English | MEDLINE | ID: mdl-28273882

ABSTRACT

Epithelial cells are involved in the regulation of innate and adaptive immunity in response to different stresses. The purpose of this study was to investigate if alkali-injured corneal epithelia activate innate immunity through the nucleotide-binding oligomerization domain-containing protein (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome pathway. A unilateral alkali burn (AB) was created in the central cornea of C57BL/6 mice. Mice received either no topical treatment or topical treatment with sodium butyrate (NaB), ß-hydroxybutyric acid (HBA), dexamethasone (Dex), or vehicle (balanced salt solution, BSS) quater in die (QID) for two or five days (d). We evaluated the expression of inflammasome components including NLRP3, apoptosis-associated speck-like protein (ASC), and caspase-1, as well as the downstream cytokine interleukin (IL)-1ß. We found elevation of NLRP3 and IL-1ß messenger RNA (mRNA) transcripts, as well as levels of inflammasome component proteins in the alkali-injured corneas compared to naïve corneas. Treatment with NLRP3 inhibitors using NaB and HBA preserved corneal clarity and decreased NLRP3, caspase-1, and IL-1ß mRNA transcripts, as well as NLRP3 protein expression on post-injury compared to BSS-treated corneas. These findings identified a novel innate immune signaling pathway activated by AB. Blocking the NLRP3 pathway in AB mouse model decreases inflammation, resulting in greater corneal clarity. These results provide a mechanistic basis for optimizing therapeutic intervention in alkali injured eyes.


Subject(s)
Burns, Chemical/drug therapy , Butyrates/therapeutic use , Corneal Injuries/drug therapy , Eye Burns/drug therapy , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Wound Healing/drug effects , Alkalies/toxicity , Animals , Apoptosis Regulatory Proteins/metabolism , Burns, Chemical/metabolism , Butyrates/pharmacology , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cornea/drug effects , Cornea/metabolism , Corneal Injuries/chemically induced , Corneal Injuries/metabolism , Eye Burns/chemically induced , Eye Burns/metabolism , Female , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL
20.
J Autoimmun ; 80: 65-76, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28238526

ABSTRACT

The concept of innate immunity has been expanded to recognize environmental pathogens other than microbial components. However, whether and how the innate immunity is initiated by epithelium in response to environmental physical challenges such as low humidity and high osmolarity in an autoimmune disease, dry eye, is still largely unknown. Using two experimental dry eye models, primary human corneal epithelial cultures exposed to hyperosmolarity and mouse ocular surface facing desiccating stress, we uncovered novel innate immunity pathway by ocular surface epithelium, where oxidized mitochondrial DNA induces imbalanced activation of NLRP3/NLRP6 inflammasomes via stimulation of caspase-8 and BRCC36 in response to environmental stress. Activated NLRP3 with suppressed NLRP6 stimulates caspase-1 activation that leads to IL-1ß and IL-18 maturation and secretion. NLRP3-independent caspase-8 noncanonically activates caspase-1 via reciprocal regulation of NLRP3/NLRP6-mediated inflammasomes. Reactive oxygen species-induced mitochondrial DNA oxidative damage and BRCC36 deubiquitinating activity provide a missing link and mechanism by which innate immunity responds to environmental stress via caspase-8-involved NLRP3/NLRP6 inflammasomes.


Subject(s)
Caspase 8/metabolism , DNA, Mitochondrial/metabolism , Dry Eye Syndromes/immunology , Epithelium, Corneal/immunology , Inflammasomes/metabolism , Membrane Proteins/metabolism , Adolescent , Adult , Aged , Animals , Autoimmunity , Cells, Cultured , DNA Damage , DNA, Mitochondrial/genetics , Deubiquitinating Enzymes , Environmental Exposure/adverse effects , Epithelium, Corneal/pathology , Female , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Models, Animal , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...