Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Lab Invest ; 103(5): 100061, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36801638

RESUMEN

Corneal injury-associated inflammation could induce inward-growing neovascularization from the periphery of the tissue. Such neovascularization could cause stromal opacification and curvature disturbance, and both potentially impair visual function. In this study, we determined the effects of the loss of transient receptor potential vanilloid 4 (TRPV4) expression on the development of neovascularization in the corneal stroma in mice by producing a cauterization injury in the central area of the cornea. New vessels were immunohistochemically labeled with anti-TRPV4 antibodies. TRPV4 gene knockout suppressed the growth of such CD31-labeled neovascularization in association with the suppression of infiltration of macrophages and tissue messenger RNA expression of the vascular endothelial cell growth factor A level. Treatment of cultured vascular endothelial cells with supplementation of HC-067047 (0.1 µM, 1 µM, or 10 µM), a TRPV4 antagonist, attenuated the formation of a tube-like structure with sulforaphane (15 µM, for positive control) that modeled the new vessel formation. Therefore, the TRPV4 signal is involved in injury-induced macrophagic inflammation and neovascularization activity by vascular endothelial cells in a mouse corneal stroma. TRPV4 could be a therapeutic target to prevent unfavorable postinjury neovascularization in the cornea.


Asunto(s)
Canales de Potencial de Receptor Transitorio , Ratones , Animales , Canales de Potencial de Receptor Transitorio/metabolismo , Células Endoteliales/metabolismo , Neovascularización Patológica/metabolismo , Córnea/metabolismo , Macrófagos/metabolismo , Inflamación/metabolismo , Cationes/metabolismo , Cationes/farmacología
2.
Lab Invest ; 103(11): 100256, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37797886

RESUMEN

We examined the effects of gene ablation and chemical inhibition of transient receptor potential ankyrin 1 (TRPA1) on the growth of experimental argon laser-induced choroidal neovascularization (CNV) in mice. CNV was induced in the eyes of 6- to 8-week-old TRPA1-null (knockout [KO]) and wild-type (WT) mice by argon laser irradiation. Gene expression analysis was performed in laser-injured tissues at days 1 and 3. CNV growth was evaluated at day 14. Reciprocal bone marrow transplantation was performed between each genotype to identify the components responsible for either recipient tissue or bone marrow-derived inflammatory cells. Our results show that laser irradiation successfully induced CNV growth at the site of laser injury. The size of induced CNV was significantly smaller in KO mice than in WT mice at day 14, as determined by angiography with fluorescein isothiocyanate-dextran. Invasion of neutrophils, but not macrophages, was suppressed in association with suppression of the expression of transforming growth factor ß1 and interleukin 6 in laser-irradiated KO tissue. Bone marrow transplantation indicated that the genotype of the recipient mouse, but not of inflammatory cells, is attributable to the KO phenotype. Systemic administration of a TRPA1 antagonist also reduced the CNV in a WT mouse. In conclusion, TRPA1 signaling in local cells is involved in growth of laser-induced CNV. The phenotype was not attributable to vascular endothelial cells and inflammatory cells. Blocking TRPA1 signal may therefore be a potential treatment strategy for CNV-related ocular diseases.


Asunto(s)
Neovascularización Coroidal , Factor de Crecimiento Transformador beta1 , Animales , Ratones , Argón , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Rayos Láser , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Factor de Crecimiento Transformador beta1/genética
3.
J Cell Mol Med ; 26(20): 5315-5325, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36127870

RESUMEN

We investigated the effects of lacking TNFα on the development and regression of Argon-laser-induced choroidal neovascularization (CNV) in mice. We lasered ocular fundus for induction of CNV in both wild-type (WT) and TNFα-null (KO) mice. Fluorescence angiography was performed to examine the size of CNV lesions. Gene expression pattern of wound healing-related components was examined. The effects of exogenous TNFα on apoptosis of human retinal microvascular endothelial cells (HRMECs) and on the tube-like structure of the cells were investigated in vitro. The results showed that Argon-laser irradiation-induced CNV was significantly larger in KO mice than WT mice on Day 21, but not at other timepoints. Lacking TNFα increased neutrophil population in the lesion. The distribution of cleaved caspase3-labelled apoptotic cells was more frequently observed in the laser-irradiated tissue in a WT mouse as compared with a KO mouse. Exogenous TNFα induced apoptosis of HRMECs and accelerated regression of tube-like structure of HRMECs in cell culture. Taken together, TNFα gene knockout delays the regression of laser-induced CNV in mice. The mechanism underlying the phenotype might include the augmentation of neutrophil population in the treated tissue and attenuation of vascular endothelial cell apoptosis.


Asunto(s)
Neovascularización Coroidal , Animales , Argón , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Humanos , Rayos Láser , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Necrosis Tumoral alfa
4.
Lab Invest ; 101(2): 245-257, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33199821

RESUMEN

Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid generated through sphingosine kinase1 (SPK1)-mediated phosphorylation of sphingosine. We show here that injury-induced S1P upregulation increases corneal neovascularization through stimulating S1PR3, a cognate receptor. since this response was suppressed in S1PR3-knockout mice. Furthermore, Cayman10444, a selective S1PR3 inhibitor, reduced this response in WT mice. Such reductions in neovascularization were associated with reduced vascular endothelial growth factor A (VEGF-A) mRNA expression levels in WT TKE2 corneal epithelial cells and macrophages treated with CAY10444 as well as macrophages isolated from S1PR3 KO mice. S1P increased tube-like vessel formation in human vascular endothelial cells (HUVEC) and human retinal microvascular endothelial cells (HRMECs) cells expressing S1PR3. In S1PR3 KO mice, TGFß1-induced increases in αSMA gene expression levels were suppressed relative to those in the WT counterparts. In S1PR3 deficient macrophages, VEGF-A expression levels were lower than in WT macrophages. Transforming growth factor ß1(TGFß1) upregulated SPK1 expression levels in ocular fibroblasts and TKE2 corneal epithelial cells. CAY10444 blocked S1P-induced increases in VEGF-A mRNA expression levels in TKE2 corneal epithelial cells. Endogenous S1P signaling upregulated VEGF-A and VE-cadherin mRNA expression levels in HUVEC. Unlike in TKE2 cells, SIS3 failed to block TGFß1-induced VEGF-A upregulation in ocular fibroblasts. Taken together, these results indicate that injury-induced TGFß1 upregulation increases S1P generation through increases in SPK1 activity. The rise in S1P formation stimulates the S1PR3-linked signaling pathway, which in turn increases VEGF-A expression levels and angiogenesis in mouse corneas.


Asunto(s)
Córnea , Lesiones de la Cornea/metabolismo , Neovascularización Fisiológica/genética , Receptores de Esfingosina-1-Fosfato , Animales , Células Cultivadas , Córnea/citología , Córnea/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Receptores de Esfingosina-1-Fosfato/genética , Receptores de Esfingosina-1-Fosfato/metabolismo , Tiazolidinas/farmacología
5.
Lab Invest ; 101(6): 690-700, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33782532

RESUMEN

The purpose of the study was to uncover the role of tenascin X in modulation of healing in mouse corneas subjected to epithelium debridement. Healing in corneas with an epithelial defect was evaluated at the levels of gene and protein expression. Wound healing-related mediators and inflammatory cell infiltration were detected by histology, immunohistochemistry and real-time RT-PCR. Tenascin X protein was upregulated in the wounded wild-type (WT) corneal epithelium. The lack of tenascin X impaired closure of an epithelial defect and accelerated infiltration of neutrophils into the wound periphery as compared to the response in WT tissue. Expression of wound healing-related proinflammatory and reparative components, i.e., interleukin-6, transforming growth factor ß, matrix metalloproteinases, were unaffected by the loss of tenascin X expression. Marked accumulation of malondialdehyde (a lipid peroxidation-derived product) was observed in KO healing epithelia as compared with its WT counterpart. Neutropenia induced by systemic administration of a specific antibody rescued the impairment of epithelial healing in KO corneas, with reduction of malondialdehyde levels in the epithelial cells. Finally, we showed that a chemical scavenging reactive oxygen species reversed the impairment of attenuation of epithelial repair with a reduction of tissue levels of malondialdehyde. In conclusion, loss of tenascin X prolonged corneal epithelial wound healing and increased neutrophilic inflammatory response to debridement in mice. Tenascin X contributes to the control of neutrophil infiltration needed to support the regenerative response to injury and prevent the oxidative stress mediators from rising to cytotoxic levels.


Asunto(s)
Córnea/inmunología , Infiltración Neutrófila , Especies Reactivas de Oxígeno/metabolismo , Tenascina/fisiología , Cicatrización de Heridas/inmunología , Animales , Córnea/metabolismo , Ratones Noqueados
6.
Lab Invest ; 99(2): 210-230, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30413814

RESUMEN

In order to understand the pathobiology of neurotrophic keratopathy, we established a mouse model by coagulating the first branch of the trigeminal nerve (V1 nerve). In our model, the sensory nerve in the central cornea disappeared and remaining fibers were sparse in the peripheral limbal region. Impaired corneal epithelial healing in the mouse model was associated with suppression of both cell proliferation and expression of stem cell markers in peripheral/limbal epithelium as well as a reduction of transient receptor potential vanilloid 4 (TRPV4) expression in tissue. TRPV4 gene knockout also suppressed epithelial repair in mouse cornea, although it did not seem to directly modulate migration of epithelium. In a co-culture experiment, TRPV4-introduced KO trigeminal ganglion upregulated nerve growth factor (NGF) in cultured corneal epithelial cells, but ganglion with a control vector did not. TRPV4 gene introduction into a damaged V1 nerve rescues the impairment of epithelial healing in association with partial recovery of the stem/progenitor cell markers and upregulation of cell proliferation and of NGF expression in the peripheral/limbal epithelium. Gene transfer of TRPV4 did not accelerate the regeneration of nerve fibers. Sensory nerve TRPV4 is critical to maintain stemness of peripheral/limbal basal cells, and is one of the major mechanisms of homeostasis maintenance of corneal epithelium.


Asunto(s)
Epitelio Corneal , Células Madre , Canales Catiónicos TRPV/metabolismo , Nervio Trigémino/metabolismo , Cicatrización de Heridas/fisiología , Animales , Células Cultivadas , Epitelio Corneal/citología , Epitelio Corneal/lesiones , Epitelio Corneal/metabolismo , Técnicas de Inactivación de Genes , Ratones , Células Madre/citología , Células Madre/metabolismo , Canales Catiónicos TRPV/genética , Nervio Trigémino/química
7.
Exp Eye Res ; 181: 90-97, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30633924

RESUMEN

Corneal neovascularization and inflammatory fibrosis induced by severe injury or infection leads to tissue opacification and even blindness. Transient receptor potential (TRP) channel subtypes contribute to mediating these maladaptive responses through their interactions with other receptors. TRPV1 is one of the contributing channel isoforms inducing neovascularization in an alkali burn mouse wound healing model. VEGF-A upregulation contributes to neovascularization through interaction with its cognate receptors (VEGFR). Since the TRP isoform in this tissue, TRPA1, is also involved, we determined here if one of the pathways mediating neovascularization and immune cell infiltration involve an interaction between VEGFR and TRPA1 in a cauterization corneal mouse wound healing model. Localization of TRPA1 and endothelial cell (EC) CD31 immunostaining pattern intensity determined if TRPA1 expression was EC delimited during cauterization induced angiogenesis. Quantitative RT-PCR evaluated the effects of the absence of TRPA1 function on VEGF-A and TGF-ß1 mRNA expression during this process. Macrophage infiltration increased based on rises in F4/80 antigen immunoreactivity. TRPA1 immunostaining was absent on CD31-immunostained EC cells undergoing neovascularization, but it was present on other cell type(s) adhering to EC in vivo. Absence of TRPA1 expression suppressed both stromal neovascularization and inhibited macrophage infiltration. Similarly, the increases occurring in both VEGF-A and TGF-ß1 mRNA expression levels in WT tissue were blunted in the TRPA1-/- counterpart. On the other hand, in the macrophages their levels were invariant and their infiltration was inhibited. To determine if promotion by TRPA1 of angiogenesis was dependent on its expression on other unidentified cell types, the effects were compared of pharmacological manipulation of TRPA1 activity on EC proliferation tube formation and migration. In the presence and absence of a fibroblast containing feeder layer. Neither VEGF-induced increases in human vascular endothelial cell (HUVEC) proliferation nor migration were changed by a TRPA1 antagonist HC-030031 in the absence of a feeder layer. However, on a fibroblast feeder layer this antagonist suppressed HUVEC tube formation. In conclusion, during corneal wound healing transactivation by VEGFR of TRPA1 contributes to mediating neovascularization and macrophage infiltration. Such crosstalk is possible because of close proximity between VEGFR delimited expression on EC and TRPA1 expression restricted to cell types adhering to EC.


Asunto(s)
Neovascularización de la Córnea/fisiopatología , Sustancia Propia/patología , Canal Catiónico TRPA1/deficiencia , Animales , Neovascularización de la Córnea/metabolismo , Sustancia Propia/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Canal Catiónico TRPA1/antagonistas & inhibidores , Canal Catiónico TRPA1/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/fisiología
8.
J Cell Mol Med ; 22(2): 948-956, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29160014

RESUMEN

To determine the contribution by tenascin X (Tnx) gene expression to corneal stromal angiogenesis, the effects were determined of its loss on this response in TNX knockout (KO) mice. In parallel, the effects of such a loss were evaluated on vascular endothelial growth factor (VEGF) and transforming growth factor ß1 (TGFß1) gene and protein expression in fibroblasts and macrophages in cell culture. Histological, immunohistochemical and quantitative RT-PCR changes determined if Tnx gene ablation on angiogenic gene expression, inflammatory cell infiltration and neovascularization induced by central corneal stromal cauterization. The role was determined of Tnx function in controlling VEGF-A or TGFß1 gene expression by comparing their expression levels in ocular fibroblasts and macrophages obtained from wild-type (WT) and body-wide Tnx KO mice. Tnx was up-regulated in cauterized cornea. In Tnx KO, macrophage invasion was attenuated, VEGF-A and its cognate receptor mRNA expression along with neovascularization were lessened in Tnx KOs relative to the changes occurring in their WT counterpart. Loss of Tnx instead up-regulated in vivo mRNA expression of anti-angiogenic VEGF-B but not VEGF-A. On the other hand, TGFß1 mRNA expression declined in Tnx KO cultured ocular fibroblasts. Loss of Tnx gene expression caused VEGF-A expression to decline in macrophages. Tnx gene expression contributes to promoting TGFß1 mRNA expression in ocular fibroblasts and VEGF-A in macrophages, macrophage invasion, up-regulation of VEGF-A expression and neovascularization in an injured corneal stroma. On the other hand, it suppresses anti-angiogenic VEGF-B mRNA expression in vivo.


Asunto(s)
Neovascularización de la Córnea/genética , Sustancia Propia/irrigación sanguínea , Sustancia Propia/lesiones , Tenascina/deficiencia , Tenascina/genética , Animales , Cauterización , Neovascularización de la Córnea/patología , Citocinas/metabolismo , Regulación de la Expresión Génica , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Miofibroblastos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Exp Eye Res ; 163: 78-84, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28950941

RESUMEN

Dysfunction of the meibomian glands alters secreted meibum quantitatively and qualitatively that can lead to damage to the ocular surface epithelium. In response to an unstable tear film cause by meibomian gland dysfunction, ocular surface epithelium is damaged and expresses inflammatory cytokines leading to secondary ocular inflammation. In turn, inflammatory disorders of the palpebral conjunctiva and lid margin may affect the structure and function of meibomian gland. The disorders include allergic conjunctivitis, long-term usage of contact lenses, dermatological diseases that affect conjunctival homeostasis, Stevens-Johnson's syndrome or chemical burning of the ocular surface and lid margin.


Asunto(s)
Enfermedades de la Conjuntiva/complicaciones , Epitelio Corneal/metabolismo , Enfermedades de los Párpados/complicaciones , Glándulas Tarsales/metabolismo , Lentes de Contacto/efectos adversos , Edema Corneal/metabolismo , Citocinas/metabolismo , Humanos , Lágrimas/química , Lágrimas/metabolismo
11.
Lab Invest ; 96(6): 641-51, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26950486

RESUMEN

We evaluated the effects of the loss of Smad3 on the development of experimental argon laser-induced choroidal neovascularization (CNV) in mice. An in vitro angiogenesis model was also used to examine the role of transforming growth factor-ß1 (TGFß1)/Smad3 signaling in vessel-like tube formation by human umbilical vein endothelial cells (HUVECs). CNV was induced in eyes of 8-12-week-old B6.129-background Smad3-deficient (KO) mice (n=47) and wild-type (WT) mice (n=47) by argon laser irradiation. Results showed that the size of the CNV induced was significantly smaller in KO mice as compared with WT mice at day 14 as revealed by high-resolution angiography with fluorescein isothiocyanate-dextran. Immunohistochemistry and real-time reverse transcription-polymerase chain reaction of RNA extracted from laser-irradiated choroidal tissues were conducted on specimens at specific timepoints. Invasion of macrophages (F4/80+), but not neutrophils (myeloperoxidase+), and appearance of myofibroblasts (α-smooth muscle actin+) were suppressed in laser-irradiated KO tissues. mRNA expression of inflammation-related factors, that is, vascular endothelial growth factor (VEGF), macrophage-chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6) and TGFß1 in choroidal tissues was suppressed by the loss of Smad3. We then examined the effects of adding a Smad3 inhibitor, SIS3, or an ALK5 inhibitor, SB431542, on tube formation promoted by TGFß1 or VEGF in HUVECs cocultured with fibroblast feeder. Further addition of SIS3 or SB431542 augmented vessel-like tube formation by HUVECs in the presence of TGFß1 or VEGF. In conclusion, lack of Smad3 attenuated the growth of laser-induced CNV with suppression of inflammation by macrophages in mice. Because blocking TGFß1/Smad3 signal stimulated the activity of angiogenesis of HUVECs in vitro, the reduction of CNV in vivo in KO mice is attributed to a decrease in growth factor levels in the tissue by the loss of Smad3.


Asunto(s)
Neovascularización Coroidal/prevención & control , Macrófagos/patología , Proteína smad3/deficiencia , Animales , Neovascularización Coroidal/etiología , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mediadores de Inflamación/metabolismo , Isoquinolinas/farmacología , Láseres de Gas/efectos adversos , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Piridinas/farmacología , Pirroles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína smad3/antagonistas & inhibidores , Proteína smad3/genética , Factor de Crecimiento Transformador beta1
12.
Lab Invest ; 93(2): 207-17, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23207449

RESUMEN

We investigated the effects of loss of tenascin C on the healing of the stroma using incision-injured mice corneas. Tenascin C was upregulated in the stroma following incision injury to the cornea. Wild-type (WT) and tenascin C-null (knockout (KO)) mice on a C57BL/6 background were used. Cell culture experiments were also conducted to determine the effects of the lack of tenascin C on fibrogenic gene expression in ocular fibroblasts. Histology, immunohistochemistry and real-time reverse transcription PCR were employed to evaluate the healing process in the stroma. The difference in the incidence of wound closure was statistically analyzed in hematoxylin and eosin-stained samples between WT and KO mice in addition to qualitative observation. Healing of incision injury in corneal stroma was delayed, with less appearance of myofibroblasts, less invasion of macrophages and reduction in expression of collagen Iα1, fibronectin and transforming growth factor ß1 (TGFß1) in KO mice compared with WT mice. In vitro experiments showed that the loss of tenascin C counteracted TGFß1 acceleration of mRNA expression of TGFß1, and of collagen Iα1 and of myofibroblast conversion in ocular fibroblasts. These results indicate that tenascin C modulates wound healing-related fibrogenic gene expression in ocular fibroblasts and is required for primary healing of the corneal stroma.


Asunto(s)
Lesiones de la Cornea , Sustancia Propia/fisiopatología , Regulación de la Expresión Génica/genética , Tenascina/deficiencia , Cicatrización de Heridas/fisiología , Análisis de Varianza , Animales , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Fibronectinas/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/metabolismo
13.
Exp Anim ; 72(2): 224-232, 2023 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-36436910

RESUMEN

Transient Receptor Potential (TRP) ion channels mediate the influx of cations into cells responding to chemical or physical stimuli. TRP vanilloid 1 (TRPV1) regulates cutaneous functions. Its function in cutaneous wound healing, however, has not been clarified. The current study elucidated the role of TRPV1 in cutaneous wound healing of dorsal circular excisional injury using Trpv1-null (KO) and wild type (WT) male/female C57BL/6 mice. Macroscopic observation showed that the remaining cutaneous lesion was significantly larger in KO than that of WT at postoperative days (POD) 7 and 10. Histological analysis showed significantly delayed re-epithelialization in KO at POD7. The number of macrophages in KO and WT similarly returned to the reduced state from POD4 to POD7. Whereas, the number of neutrophils in KO did not significantly return to the reduced state, in contrast to WT. Of note, The H3Cit-labeled NETs (Neutrophil Extracellular Traps) formation of KO was prominently increased both in POD4 and 7. The current results suggest that the loss of TRPV1 induces prolonged neutrophilic inflammation and NETs formation, retarding murine cutaneous wound healing in vivo. This study provides a possible link with TRPV1 and neutrophilic regulation in cutaneous wound healing.


Asunto(s)
Canales Catiónicos TRPV , Cicatrización de Heridas , Ratones , Femenino , Masculino , Animales , Ratones Noqueados , Ratones Endogámicos C57BL , Cicatrización de Heridas/genética , Canales Catiónicos TRPV/genética
14.
Arch Plast Surg ; 50(5): 446-451, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37808334

RESUMEN

Background Elderly patients often have complications of blepharoptosis surgery that can result in the appearance or exacerbation of superficial punctate keratopathy (SPK). However, postoperative changes to SPK status have not been previously reported. We used subjective assessment of symptoms and measurement of SPK scale classification to investigate the safety and efficacy of blepharoptosis surgery in elderly patients. Methods Included in this prospective study were 22 patients (44 eyes) with bilateral blepharoptosis that underwent surgery. Patients comprised 8 males and 14 females with a mean (±standard deviation) age of 75.7 ± 8.2 years (range: 61-89). Blepharoptosis surgery consisted of transcutaneous levator advancement and blepharoplasty including resection of soft tissue (skin, subcutaneous tissue, and the orbicularis oculi muscle). Margin reflex distance-1 (MRD-1) measurement, a questionnaire survey of symptoms and SPK scale classification, was administered preoperatively and 3 months postoperatively for evaluation. Results The median MRD-1 was 1 mm preoperatively and 2.5 mm postoperatively, representing a significant postoperative improvement. SPK area and density scores were found to increase when the MRD-1 increase was more than 2.5 mm with surgery. All 10 items on the questionnaire tended have increased scores after surgery, and significant differences were observed in 7 items (poor visibility, ocular fatigue, heavy eyelid, foreign body sensation, difficulty in focusing, headaches, and stiff shoulders). Conclusion Blepharoptosis surgery was found to be a safe and effective way to maintain the increase in MRD-1 within 2.0 mm. Despite the benefits, surgeons must nonetheless be aware that blepharoptosis surgery is a delicate procedure in elderly people.

15.
Ocul Surf ; 30: 286-294, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37972853

RESUMEN

PURPOSE: We investigated healing pattern of an incisional wound in corneal stroma of lumican-null (KO) mice. METHODS: C57BL/6 mice (wild-type, WT) and lumican-null (knockout, KO) mice were used. A linear full-thickness incision was produced in one cornea of each mouse. After intervals of healing, the corneas were processed for the following analyses. Histology was employed to measure the distance between each edge of the disrupted Descemet's membrane at the center of the cornea. Immunohistochemistry and real-time RT-PCR were employed to evaluate the expression of wound healing-related components in the tissue. Cultured ocular fibroblasts were obtained from cornea and sclera of WT and KO postnatal day 1 pups. The cells were subjected to examination for cell proliferation and expression of wound healing-related gene products. In vitro gel contraction assay was used to asses cell contractile activity of WT and KO cells. RESULTS: At day 5 of incision, the distance between the disrupted Descemet's membrane was larger in a KO mouse as compared with a WT mouse. Myofibroblast appearance in the wound was suppressed by the loss of lumican. The loss of lumican downregulated TGFß1's effects on mRNA expression of α-smooth muscle actin and collagen Ia1 in cultured ocular fibroblasts. Cell proliferation rate increased in injured stroma, which was further supported by in vitro datum of cell proliferation augmentation by the loss of lumican. Loss of lumican suppressed cell-mediated gel contraction. CONCLUSION: Loss of lumican perturbs the healing of penetrating incision in mouse corneal stroma in association with suppression of myofibroblast generation.


Asunto(s)
Sustancia Propia , Cicatrización de Heridas , Animales , Ratones , Sustancia Propia/patología , Lumican/metabolismo , Ratones Endogámicos C57BL , Cicatrización de Heridas/fisiología , Córnea/patología
16.
Pathogens ; 11(5)2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35631047

RESUMEN

Background: Corneal infection of Colletotrichum gloeosporioides is uncommon and usually limited to the anterior stroma. However, we observed a case of corneal stromal perforation caused by this fungus under a compromised condition. Case: A 73-year-old woman consulted us with a severe corneal ulceration. She was a tangerine orange farmer who suffered from rheumatoid arthritis for more than ten years. Before consultation with us, she received pterygium excision in her right eye. She then developed a corneal ulceration and received topical glucocorticoid therapy upon diagnosis of rheumatoid arthritis-related stromal ulcer in the eye. At the first consultation with us, a corneal ulceration was observed in the inferotemporal area of her right cornea. Biological examination detected a filamentous fungus, Colletotrichum gloeosporioides. Topical and systemic antifungal treatments were not significantly effective. Fourteen days after consultation, the lesion grew worse, leading to stromal perforation, which was treated by therapeutic penetrating keratoplasty using a preserved corneal button. Conclusions: Topical glucocorticoid could accelerate the growth of Colletotrichum gloeosporioides before diagnosis, even though the primary cause of corneal ulceration development might be rheumatoid arthritis.

17.
Ocul Surf ; 26: 111-127, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35988880

RESUMEN

Spinster 2 (Spns2) is a transporter that pumps sphingosine-1-phosphate (S1P), a bioactive lipid mediator synthesized in the cytoplasm, out of cells into the inter cellular space. S1P is a signal that modulates cellular behavior during embryonic development, inflammation and tissue repair, etc. A Spns2-null (KO) mouse is born with failure of eyelid closure (eyelid-open-at birth; EOB) and develop corneal fibrosis in adulthood. It remains elusive whether corneal lesion is caused by exposure to keratitis (lagophthalmos) of EOB phenotype or the loss of Spns2 directly perturbs the corneal tissue morphogenesis and intra-eyelid structures. Therefore, we investigated differences between the cornea and ocular adnexa morphogenesis in KO and wild-type (WT) embryos and adults as well. The loss of Spns2 perturbs cornea morphogenesis during embryonic development as early as E16.5 besides EOB phenotype. Histology showed that the corneal stroma was thinner with less extracellular matrix accumulation, e.g., collagen and keratocan in the KO mouse. Epithelial stratification, expression of keratin 12 and formation of desmosomes and hemidesmosomes were also perturbed in these KO corneas. Lacking Spns2 impaired morphogenesis of the Meibomian glands and of orbicularis oculi muscles. KO glands were labeled for ELOVL4 and PPARγ and were Oil-Red O-positive, suggesting KO acinar cells possessed functionality as the glands. This is the first report on the roles of Spns2 in corneal and Meibomian gland morphogenesis. Corneal tissue destruction in an adult KO mouse might be due to not only lagophthalmos but also to an impaired morphogenesis of cornea, Meibomian glands, and orbicularis oculi muscle.


Asunto(s)
Enfermedades de la Córnea , Enfermedades de los Párpados , Embarazo , Femenino , Ratones , Animales , Ratones Noqueados , Lisofosfolípidos/metabolismo , Córnea/metabolismo , Glándulas Tarsales/metabolismo , Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo
18.
Ocul Surf ; 15(4): 713-722, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28442381

RESUMEN

PURPOSE: To examine effects of alkali injury of the ocular surface on meibomian gland pathology in mice. METHODS: Three µL of 1 N NaOH were applied under general anesthesia to the right eye of 10-week-old BALB/c (n = 54) mice to produce a total ocular surface alkali burn. The meibomian gland morphology was examined at days 1, 2, 5, 10, and 20 by stereomicroscopy and non-contact infrared meibography. Mice were then sacrificed and eyelids processed for histology with hematoxylin-eosin and immunohistochemistry for ELOVL4, PPARγ, myeloperoxidase (a neutrophil marker) and F4/80 macrophage antigen, as well as TUNEL staining. Another set of specimens was processed for cryosectioning and Oil red O staining. RESULTS: Alkali injury to the ocular surface produced cellular apoptosis, infiltration of neutrophils and macrophages, degeneration of the meibomian gland, and ductal dilation. Inflammation in and destruction of acunal stricture seemed more prominent in the lower eyelid, while duct dilation was more frequently observed in the upper eyelid during healing. Surviving acinar cells were labeled for ELOVL4 and PPARγ. Oil red O staining showed that the substance in the dilated duct contained predominantly neutral lipid. CONCLUSIONS: Alkali injury to the ocular surface results in damage and destruction of the eyelid meibomian glands. The pattern of the tissue damage differs between glands of the upper and lower eyelids.


Asunto(s)
Glándulas Tarsales , Álcalis , Animales , Lesiones Oculares , Enfermedades de los Párpados , Lípidos , Ratones , PPAR gamma
20.
Gene Expr Patterns ; 3(2): 193-7, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12711548

RESUMEN

The winged-helix/forkhead transcription factor gene family has been shown to play important roles in the development of the central nervous system (CNS) as well as heart, lung, and liver. Recently, we have identified Foxp1, a novel subfamily of winged-helix/forkhead genes, which was abundant in the lung and brain of adult mice. Here we analyzed the expression pattern of Foxp1 in the developing CNS using in situ hybridization. The expression of Foxp1 mRNA was first detected in the ventral horn of the spinal cord at 9.5 days postcoitum. During the late-stage of development, its gene expression was not detectable in neuroepithelia, but was clearly observed in the postmitotic neurons of various CNS regions, including caudate-putamen, neocortex, several brainstem nuclei, and cerebellum. In neonates, its gene expression was persisted in these motor-related regions.


Asunto(s)
Sistema Nervioso Central/metabolismo , Proteínas Represoras/genética , Animales , Sistema Nervioso Central/embriología , Factores de Transcripción Forkhead , Perfilación de la Expresión Génica , Ratones , Proteínas Represoras/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA