Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
Exp Eye Res ; 200: 108206, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32882212

RESUMO

Keratins are the forming units of intermediate filaments (IF) that provide mechanical support, and formation of desmosomes between cells and hemi desmosomes with basement membranes for epithelium integrity. Keratin IF are polymers of obligate heterodimer consisting one type I keratin and one type II keratin molecules. There are 54 functional keratin genes in human genome, which are classified into three major groups, i.e., epithelial keratins, hair follicle cell-specific epithelial keratins and hair keratins. Their expression is cell type-specific and developmentally regulated. Corneal epithelium expresses a subgroup of keratins similar to those of epidermal epithelium. Limbal basal stem cells express K5/K14, and K8/K18 and K8/K19 IF suggesting that there probably are two populations of limbal stem cells (LSCs). In human, LSCs at limbal basal layer can directly stratify and differentiate to limbal suprabasal cells that express K3/K12 IF, or centripetally migrate then differentiate to corneal basal transient amplifying cells (TAC) that co-express both K3/K12 and K5/K14 prior to moving upward and assuming suprabasal cells phenotype of only K3/K12 expression that signifies corneal type epithelium differentiation. In rodent, the differentiated cornea epithelial cells express K5/K12 in lieu of K3/K12, because K3 allele exists as a pseudogene and does not encode a functional K3 protein. The basal corneal cells of new-born mice originate from surface ectoderm during embryonic development slowly commit to differentiation of becoming TAC co-expressing K5/K12 and K5/K14 IF. However, the centripetal migration may still occur at a slower rate in young mice, which is accelerated during wound healing. In this review, we will discuss and compare the cornea-specific keratins expression patterns between corneal and epidermal epithelial cells during mouse development, and between human and mouse during development and homeostasis in adult, and pathology caused by a mutation of keratins.


Assuntos
Córnea/metabolismo , Queratinas/biossíntese , Animais , Diferenciação Celular , Células Cultivadas , Córnea/crescimento & desenvolvimento , Humanos , Limbo da Córnea/crescimento & desenvolvimento , Limbo da Córnea/metabolismo , Células-Tronco/citologia
2.
Nature ; 511(7509): 353-7, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25030174

RESUMO

Corneal epithelial homeostasis and regeneration are sustained by limbal stem cells (LSCs), and LSC deficiency is a major cause of blindness worldwide. Transplantation is often the only therapeutic option available to patients with LSC deficiency. However, while transplant success depends foremost on LSC frequency within grafts, a gene allowing for prospective LSC enrichment has not been identified so far. Here we show that ATP-binding cassette, sub-family B, member 5 (ABCB5) marks LSCs and is required for LSC maintenance, corneal development and repair. Furthermore, we demonstrate that prospectively isolated human or murine ABCB5-positive LSCs possess the exclusive capacity to fully restore the cornea upon grafting to LSC-deficient mice in xenogeneic or syngeneic transplantation models. ABCB5 is preferentially expressed on label-retaining LSCs in mice and p63α-positive LSCs in humans. Consistent with these findings, ABCB5-positive LSC frequency is reduced in LSC-deficient patients. Abcb5 loss of function in Abcb5 knockout mice causes depletion of quiescent LSCs due to enhanced proliferation and apoptosis, and results in defective corneal differentiation and wound healing. Our results from gene knockout studies, LSC tracing and transplantation models, as well as phenotypic and functional analyses of human biopsy specimens, provide converging lines of evidence that ABCB5 identifies mammalian LSCs. Identification and prospective isolation of molecularly defined LSCs with essential functions in corneal development and repair has important implications for the treatment of corneal disease, particularly corneal blindness due to LSC deficiency.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Limbo da Córnea/citologia , Limbo da Córnea/fisiologia , Regeneração , Células-Tronco/metabolismo , Cicatrização , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/deficiência , Transportadores de Cassetes de Ligação de ATP/deficiência , Animais , Apoptose , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Transplante de Células-Tronco , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
3.
Hum Mol Genet ; 26(19): 3776-3791, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28934388

RESUMO

Recently, we identified biallelic mutations of SLC25A46 in patients with multiple neuropathies. Functional studies revealed that SLC25A46 may play an important role in mitochondrial dynamics by mediating mitochondrial fission. However, the cellular basis and pathogenic mechanism of the SLC25A46-related neuropathies are not fully understood. Thus, we generated a Slc25a46 knock-out mouse model. Mice lacking SLC25A46 displayed severe ataxia, mainly caused by degeneration of Purkinje cells. Increased numbers of small, unmyelinated and degenerated optic nerves as well as loss of retinal ganglion cells indicated optic atrophy. Compound muscle action potentials in peripheral nerves showed peripheral neuropathy associated with degeneration and demyelination in axons. Mutant cerebellar neurons have large mitochondria, which exhibit abnormal distribution and transport. Biochemically mutant mice showed impaired electron transport chain activity and accumulated autophagy markers. Our results suggest that loss of SLC25A46 causes degeneration in neurons by affecting mitochondrial dynamics and energy production.


Assuntos
Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas de Transporte de Fosfato/genética , Proteínas de Transporte de Fosfato/metabolismo , Animais , Ataxia/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Dinâmica Mitocondrial/fisiologia , Mutação , Células Ganglionares da Retina/patologia
4.
Development ; 142(19): 3383-93, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443636

RESUMO

The development of organs with an epithelial parenchyma relies on reciprocal mesenchymal-epithelial communication. Mouse corneal epithelium stratification is the consequence of a coordinated developmental process based on mesenchymal-epithelial interactions. The molecular mechanism underlying these interactions remains unclear. The Wnt/ß-catenin signaling pathway is involved in fundamental aspects of development through the regulation of various growth factors. Here, we show that conditional ablation of either ß-catenin (Ctnnb1(cKO)) or co-receptors Lrp5/6 (Lrp5/6(cKO)) in corneal stromal cells results in precocious stratification of the corneal epithelium. By contrast, ectopic expression of a murine Ctnnb1 gain-of-function mutant (Ctnnb1(cGOF)) retards corneal epithelium stratification. We also discovered that Bmp4 is upregulated in the absence of ß-catenin in keratocytes, which further triggers ERK1/2 (Mapk3/1) and Smad1/5 phosphorylation and enhances transcription factor p63 (Trp63) expression in mouse corneal basal epithelial cells and in a human corneal epithelial cell line (HTCE). Interestingly, mouse neonates given a subconjunctival BMP4 injection displayed a phenotype resembling that of Ctnnb1(cKO). Conditional ablation of Bmp4 eradicates the phenotype produced in Ctnnb1(cKO) mice. Furthermore, ChIP and promoter-luciferase assays show that ß-catenin binds to and suppresses Bmp4 promoter activity. These data support the concept that cross-talk between the Wnt/ß-catenin/Bmp4 axis (in the stromal mesenchyme) and Bmp4/p63 signaling (in the epithelium) plays a pivotal role in epithelial stratification during corneal morphogenesis.


Assuntos
Proteína Morfogenética Óssea 4/antagonistas & inibidores , Epitélio Corneano/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Morfogênese/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Western Blotting , Proteína Morfogenética Óssea 4/administração & dosagem , Imunoprecipitação da Cromatina , Doxiciclina , Fluorescência , Galactosídeos , Técnicas Histológicas , Imuno-Histoquímica , Indóis , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Luciferases , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Fosfoproteínas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transativadores/metabolismo
5.
Eur J Immunol ; 46(12): 2852-2861, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27682997

RESUMO

TH17 cells play an essential role in the development of both human multiple sclerosis and animal experimental autoimmune encephalomyelitis (EAE). Nevertheless, it is not well understood how the pathogenicity of TH17 cells is controlled in the autoimmune neuroinflammation. In vitro, we found Lumican (Lum), an extracellular matrix (ECM) protein, is selectively expressed by TH17 cells among tested murine TH subsets. Lum deficiency leads to earlier onset and enhanced severity of EAE. This enhanced disease in Lum-deficient mice is associated with increased production of IL-17 and IL-21 and decreased TH17 cell apoptosis. Dysregulation in cytokine production appears to be specific to TH17 cells as TH1 and TH2 cell polarization and/or cytokine production were unaltered. Furthermore, adoptive transfer of myelin oligodendrocyte glycoprotein specific TH17 cells derived from Lum-deficient mice led to earlier onset and increased severity of disease compared to controls highlighting a TH17-cell-intrinsic effect of Lum. Taken together, our results suggest that Lum negatively regulates encephalitic TH17 cells, implicating a potential therapeutic pathway in TH17 cell mediated autoimmune and inflammatory diseases.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Lumicana/metabolismo , Esclerose Múltipla/imunologia , Células Th17/imunologia , Animais , Células Cultivadas , Feminino , Humanos , Lumicana/genética , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/imunologia , Equilíbrio Th1-Th2
6.
Biochem Biophys Res Commun ; 482(4): 1304-1311, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27939890

RESUMO

With aging and stress, the myocardium undergoes structural remodeling, often leading to fibrosis. The purpose of this study is to examine whether lumican, one of the class II small leucine-rich proteoglycans, has a protective role in cardiac remodeling and fibrosis. In attempts to elucidate the hypothesis that lumican may have a protective role in cardiac remodeling and fibrosis, we compared the cardiac phenotypes of young (3-month-old) and elder (6-month- and 12-month-old) lumican-null (Lum-/-) mice. Extra-cellular matrix remodeling and apoptosis are examined to determine the roles of lumican on age-dependent cardiac fibrosis induced by isoproterenol. Compared to wild type littermates, Lum-/- mice exhibited higher mortality due to significantly impaired systolic function, which was associated with an increase of atrial natriuretic peptide (ANP) secreted by the ventricles in response to excessive stretching of myocytes. Masson's Trichrome and silver stains showed significantly more severe ventricle fibrosis in Lum-/- mice. Interestingly, rate of cell death mediated via apoptosis illustrated by the expression of caspase 3 and TUNEL assay was lower in Lum-/- mice after isoproterenol infusion. In addition, Lum-/- mice exhibited higher levels of TGF-ß, collagen I/III, and membrane-type matrix metalloproteinase-1 (MT1-MMP/MMP-14) during cardiac remodeling. This study shows that alternations of lumican might be implicated in the pathogenesis of cardiac fibrosis and suggests lumican as novel targets for cardiac fibrosis therapy. Further studies are required to define the mechanism by which lumican modulates cardiac remodeling.


Assuntos
Coração/fisiologia , Isoproterenol/química , Lumicana/genética , Miocárdio/patologia , Animais , Apoptose , Fator Natriurético Atrial/química , Caspase 3/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Matriz Extracelular/metabolismo , Fibrose , Homozigoto , Sulfato de Queratano/química , Lumicana/metabolismo , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Reação em Cadeia da Polimerase , Fator de Crescimento Transformador beta/metabolismo
7.
Exp Eye Res ; 163: 58-63, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28950938

RESUMO

The epidermal growth factor receptor (EGFR) signaling has a pivotal role in the regulation of morphogenesis during development and maintenance of homeostasis in adult eyelid and its adnexa. Studies have demonstrated that during eyelid morphogenesis the EGFR signaling pathway is responsible for keratinocyte and mesenchymal cell proliferation and migration at the eyelid tip. For meibomian gland morphogenesis, EGFR signaling activation stimulates meibomian gland epithelial cell proliferation. EGFR signaling pathway functions through multiple downstream signals such as ERK, Rho/ROCK and integrin and is regulated by a variety of upstream signals including Adam17, GPR48 and FGFR signaling. Herein we review the literature that describe the role of EGFR and its related signaling pathways in eyelid and meibomian gland morphogenesis.


Assuntos
Receptores ErbB/fisiologia , Pálpebras/embriologia , Glândulas Tarsais/embriologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Epiteliais/fisiologia , Pálpebras/fisiologia , Humanos , Glândulas Tarsais/fisiologia , Transdução de Sinais/fisiologia
8.
Dev Biol ; 406(2): 147-57, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26363126

RESUMO

Transforming growth factor alpha (TGFα) belongs to the epidermal growth factor (EGF) family and is known to play an important role during eyelid morphogenesis. In this study, we showed that ectopic expression of TGFα in the stroma of Kera-rtTA/tet-O-TGFα bitransgenic mice results in precocious eye opening, abnormal morphogenesis of the meibomian gland, tendon and tarsal plate malformation and epithelium hyperplasia. TGFα did not change proliferation and differentiation of meibocytes, but promoted proliferation and inhibited differentiation of the tarsal plate tenocytes. These results suggest that proper formation of the tendon and tarsal plate in the mouse eyelid is required for normal morphogenesis of the meibomian gland.


Assuntos
Pálpebras/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glândulas Tarsais/anormalidades , Morfogênese/fisiologia , Fator de Crescimento Transformador alfa/metabolismo , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Primers do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas Histológicas , Imuno-Histoquímica , Glândulas Tarsais/embriologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Tendões/citologia , Tendões/embriologia , Fator de Crescimento Transformador alfa/genética
9.
J Biol Chem ; 290(32): 19770-9, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26109068

RESUMO

Gene-environment interactions determine the biological outcomes through mechanisms that are poorly understood. Mouse embryonic eyelid closure is a well defined model to study the genetic control of developmental programs. Using this model, we investigated how exposure to dioxin-like environmental pollutants modifies the genetic risk of developmental abnormalities. Our studies reveal that mitogen-activated protein 3 kinase 1 (MAP3K1) signaling is a focal point of gene-environment cross-talk. Dioxin exposure, acting through the aryl hydrocarbon receptor (AHR), blocked eyelid closure in genetic mutants in which MAP3K1 signaling was attenuated but did not disturb this developmental program in either wild type or mutant mice with attenuated epidermal growth factor receptor or WNT signaling. Exposure also markedly inhibited c-Jun phosphorylation in Map3k1(+/-) embryonic eyelid epithelium, suggesting that dioxin-induced AHR pathways can synergize with gene mutations to inhibit MAP3K1 signaling. Our studies uncover a novel mechanism through which the dioxin-AHR axis interacts with the MAP3K1 signaling pathways during fetal development and provide strong empirical evidence that specific gene alterations can increase the risk of developmental abnormalities driven by environmental pollutant exposure.


Assuntos
Dioxinas/toxicidade , Poluentes Ambientais/toxicidade , Pálpebras/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , MAP Quinase Quinase Quinase 1/genética , Receptores de Hidrocarboneto Arílico/genética , Animais , Embrião de Mamíferos , Epitélio/anormalidades , Epitélio/efeitos dos fármacos , Epitélio/embriologia , Epitélio/metabolismo , Pálpebras/anormalidades , Pálpebras/efeitos dos fármacos , Pálpebras/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Interação Gene-Ambiente , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinase 1/metabolismo , Camundongos , Morfogênese/efeitos dos fármacos , Morfogênese/genética , Fosforilação , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais
10.
Development ; 140(3): 594-605, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23293291

RESUMO

Conjunctival goblet cells primarily synthesize mucins to lubricate the ocular surface, which is essential for normal vision. Notch signaling has been known to associate with goblet cell differentiation in intestinal and respiratory tracts, but its function in ocular surface has yet to be fully characterized. Herein, we demonstrate that conditional inhibition of canonical Notch signaling by expressing dominant negative mastermind-like 1 (dnMaml1) in ocular surface epithelia resulted in complete suppression of goblet cell differentiation during and subsequent to development. When compared with the ocular surface of wild-type mice (OS(Wt)), expression of dnMaml1 at the ocular surface (OS(dnMaml1)) caused conjunctival epithelial hyperplasia, aberrant desquamation, failure of Mucin 5ac (Muc5ac) synthesis, subconjunctival inflammation and epidermal metaplasia in cornea. In addition, conditional deletion of Notch1 from the ocular surface epithelia partially recapitulated OS(dnMaml1) phenotypes. We have demonstrated that N1-ICD (Notch1 intracellular domain) transactivated the mouse Krüppel-like factor 4 (Klf) promoter and that Klf4 directly bound to and significantly potentiated the Muc5ac promoter. By contrast, OS(dnMaml1) dampened Klf4 and Klf5 expression, and diminished Muc5ac synthesis. Collectively, these findings indicated that Maml-mediated Notch signaling plays a pivotal role in the initiation and maintenance of goblet cell differentiation for normal ocular surface morphogenesis and homeostasis through regulation of Klf4 and Klf5.


Assuntos
Túnica Conjuntiva/metabolismo , Epitélio Corneano/patologia , Receptor Notch1/metabolismo , Transdução de Sinais , Ativação Transcricional , Animais , Diferenciação Celular , Proliferação de Células , Túnica Conjuntiva/embriologia , Túnica Conjuntiva/patologia , Córnea/embriologia , Córnea/metabolismo , Córnea/patologia , Epitélio Corneano/embriologia , Epitélio Corneano/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Hiperplasia/genética , Hiperplasia/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Aparelho Lacrimal/metabolismo , Aparelho Lacrimal/patologia , Metaplasia/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Mucina-5AC/genética , Mucina-5AC/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Receptor Notch1/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Mol Vis ; 22: 168-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26957900

RESUMO

PURPOSE: To identify the lineage that contributes to the morphogenesis of the meibomian gland. METHODS: To examine which cell lineage gives rise to the meibomian gland, the expression of Pax6 as well as that of various cytokeratin markers, including keratin 14 (Krt14), Krt15, Krt4, and Krt10, was examined with immunofluorescent staining of C57BL/6J mouse eyelids from P2 to P11 pups and adult mice. RESULTS: Pax6 was localized to the cytoplasm within the acinar region of the meibomian glands during morphogenesis but was absent in the fully developed gland. Keratin 14 was expressed throughout the gland at all stages whereas keratin 15 was absent at all stages. Keratin 4, a marker of mucosal lineage, was present throughout the gland and was colocalized with keratin 10 (epidermal lineage marker) in the developing duct at P4. This colocalization region decreased as the gland developed becoming restricted to the central duct near the opening to the acini in the fully developed gland. CONCLUSIONS: We identified a unique cell lineage that expresses markers characteristic of mucosal and epidermal epithelia during meibomian gland morphogenesis. This unique group of cells was located in the central duct with a concentration near the ductule orifice. The expression of these cells reduced during meibomian gland morphogenesis and may play a role in the development and homeostasis of the gland.


Assuntos
Linhagem da Célula/fisiologia , Pálpebras/crescimento & desenvolvimento , Glândulas Tarsais/crescimento & desenvolvimento , Morfogênese/fisiologia , Animais , Biomarcadores/metabolismo , Proteínas do Olho/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Homeodomínio/metabolismo , Queratina-10/metabolismo , Queratina-4/metabolismo , Glândulas Tarsais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Proteínas Repressoras/metabolismo
12.
Mol Vis ; 21: 793-803, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26283861

RESUMO

PURPOSE: To examine the developmental pathobiology of the eyelid and the cornea caused by epithelial ß-catenin gain-of-function (gof) during mouse embryogenesis. METHODS: Compound mutant mice (Ctnnb1(GOFOSE) , gof of ß-catenin in the epidermis and the ocular surface epithelium) were generated by time-mating keratin 5-promoter-Cre recombinase (Krt5-Cre) and Ctnnb1(fE3/WT) (floxed exon 3 of Ctnnb1) mice. Eyes obtained from wild-type (WT) and mutant embryos at various gestation stages until E18.5 were examined with histology and immunohistochemistry. The ultrastructure of the ocular tissues of the E18.5 embryos was also examined. RESULTS: Expression of the gof-ß-catenin mutant protein in the epidermis severely impaired eyelid morphogenesis at E15.5, E17.5, and E18.5. The mutant stroma exhibited impaired keratocyte differentiation with accelerated cell proliferation and reduction in the accumulation of collagen type I. The mutant embryos also showed hyperproliferative nodules in the ocular surface epithelia with anomaly of cornea-type epithelial differentiation and the absence of the epithelial basement membrane. CONCLUSIONS: Expression of the gof-ß-catenin mutant protein in basal epithelial cells disrupts eyelid and cornea morphogenesis during mouse embryonic development due to the perturbation of cell proliferation and differentiation of the epithelium and the neural crest-derived mesenchyme.


Assuntos
Córnea/embriologia , Córnea/metabolismo , Pálpebras/embriologia , Pálpebras/metabolismo , Mutação , beta Catenina/genética , beta Catenina/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Córnea/citologia , Transição Epitelial-Mesenquimal , Epitélio/embriologia , Epitélio/metabolismo , Pálpebras/citologia , Feminino , Idade Gestacional , Queratina-5/genética , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Morfogênese/genética , Gravidez , Regiões Promotoras Genéticas , Transdução de Sinais
13.
BMC Ophthalmol ; 15 Suppl 1: 155, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26818606

RESUMO

Mesenchymal stem cells (MSC) have become a promising tool for cell therapy in regenerative medicine. They are readily available, demonstrate powerful differentiation capabilities and present immunosuppressive properties that aid them in surviving from host immune rejection for its great potential use in allograft. Currently clinical trials are underway using MSC, both culture-expanded allogeneic and autologous, for the treatment of a range of diseases not treatable by conventional therapies. A vast array of studies has dedicated towards the use of MSC for treating corneal diseases with very promising outcomes. MSC have successfully differentiated into keratocytes both in vitro and in vivo, and corneal epithelial cells in vitro, but it is uncertain if MSC can assume corneal epithelial cells in vivo. However, to date few studies have unequivocally established the efficacy of MSC for treating corneal endothelial defects. Currently, the diversity in protocols of the isolation and expansion of MSC are hindering to the assessment of cell treatment ability and the further development of treatment regimens. Therefore, future studies should develop international standards for MSC isolation and characterization. In this review, we discuss recent advances in MSC for treating ocular surface diseases.


Assuntos
Doenças da Córnea/cirurgia , Epitélio Corneano/citologia , Transplante de Células-Tronco Mesenquimais/métodos , Animais , Diferenciação Celular , Doenças da Córnea/patologia , Modelos Animais de Doenças , Células Epiteliais/citologia , Humanos , Camundongos , Medicina Regenerativa
14.
Physiol Genomics ; 46(8): 277-89, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24550211

RESUMO

Increased angiogenesis, inflammation, and proliferation are hallmarks of diseased tissues, and in vivo models of these disease phenotypes can provide insight into disease pathology. Dstn(corn1) mice, deficient for the actin depolymerizing factor destrin (DSTN), display an increase of serum response factor (SRF) that results in epithelial hyperproliferation, inflammation, and neovascularization in the cornea. Previous work demonstrated that conditional ablation of Srf from the corneal epithelium of Dstn(corn1) mice returns the cornea to a wild-type (WT) like state. This result implicated SRF as a major regulator of genes that contributes to abnormal phenotypes in Dstn(corn1) cornea. The purpose of this study is to identify gene networks that are affected by increased expression of Srf in the Dstn(corn1) cornea. Microarray analysis led to characterization of gene expression changes that occur when conditional knockout of Srf rescues mutant phenotypes in the cornea of Dstn(corn1) mice. Comparison of gene expression values from WT, Dstn(corn1) mutant, and Dstn(corn1) rescued cornea identified >400 differentially expressed genes that are downstream from SRF. Srf ablation had a significant effect on genes associated with epithelial cell-cell junctions and regulation of actin dynamics. The majority of genes affected by SRF are downregulated in the Dstn(corn1) mutant cornea, suggesting that increased SRF negatively affects transcription of SRF gene targets. ChIP-seq analysis on Dstn(corn1) mutant and WT tissue revealed that, despite being present in higher abundance, SRF binding is significantly decreased in the Dstn(corn1) mutant cornea. This study uses a unique model combining genetic and genomic approaches to identify genes that are regulated by SRF. These findings expand current understanding of the role of SRF in both normal and abnormal tissue homeostasis.


Assuntos
Córnea/metabolismo , Destrina/genética , Células Epiteliais/metabolismo , Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Fator de Resposta Sérica/genética , Actinas/genética , Actinas/metabolismo , Animais , Destrina/metabolismo , Regulação para Baixo/genética , Junções Intercelulares/genética , Junções Intercelulares/metabolismo , Camundongos , Mutação/genética , Fenótipo , Fator de Resposta Sérica/metabolismo , Transcrição Gênica/genética
15.
Am J Pathol ; 183(1): 247-56, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23685109

RESUMO

Decorin, a small leucine-rich proteoglycan (SLRP), is involved in the pathophysiology of human congenital stromal corneal dystrophy (CSCD). This disease is characterized by corneal opacities and vision impairment. In reported cases, the human gene encoding decorin contains point mutations in exon 10, generating a truncated form of decorin lacking the C-terminal 33 amino acid residues. We have previously described a transgenic mouse model carrying a similar mutation in the decorin gene that leads to an ocular phenotype characterized by corneal opacities identical to CSCD in humans. We have also identified abnormal synthesis and secretion of various SLRPs in mutant mouse corneas. In the present study, we found that mutant C-terminal truncated decorin was retained in the cytoplasm of mouse keratocytes in vivo and of transfected human embryonic kidney cells. This resulted in endoplasmic reticulum stress and an unfolded protein response. Thus, we propose a novel cell-based mechanism underlying CSCD in which a truncated SLRP protein core is retained intracellularly, its accumulation triggering endoplasmic reticulum stress that results in abnormal SLRP synthesis and secretion, which ultimately affects stromal structure and corneal transparency.


Assuntos
Distrofias Hereditárias da Córnea/genética , Decorina/genética , Retículo Endoplasmático/fisiologia , Mutação Puntual , Animais , Distrofias Hereditárias da Córnea/metabolismo , Decorina/metabolismo , Imunofluorescência , Marcadores Genéticos , Células HEK293 , Humanos , Immunoblotting , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase , Transporte Proteico , Estresse Fisiológico , Sequências Repetidas Terminais
16.
Stem Cells ; 31(10): 2116-26, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23897660

RESUMO

Mucopolysaccharidosis (MPS) are a family of related disorders caused by a mutation in one of the lysosomal exoglycosidases which leads to the accumulation of glycosaminoglycans (GAGs). MPS VII, caused by a mutation in ß-glucuronidase, manifests hepatomegaly, skeletal dysplasia, short stature, corneal clouding, and developmental delay. Current treatment regimens for MPS are not effective for treating corneal clouding and impaired mental development. We hypothesized that human umbilical mesenchymal stem cells (UMSCs) transplanted into the corneal stroma could participate in the catabolism of GAGs providing a means of cell therapy for MPS. For such treatment, human UMSCs were intrastromally transplanted into corneas of MPS VII mice. UMSC transplantation restored the dendritic and hexagonal morphology of host keratocytes and endothelial cells, respectively, and in vivo confocal microscopy (HRT-II) revealed reduced corneal haze. Immunohistochemistry using antibodies against heparan sulfate and chondroitin sulfate chains as well as lysosomal-associated membrane protein 2 revealed a decrease in GAG content and both lysosomal number and size in the treated corneas. Labeling UMSC intracellular compartments prior to transplantation revealed the distribution of UMSC vesicles throughout the corneal stroma and endothelium. An in vitro coculture assay between skin fibroblasts isolated from MPS VII mice and UMSC demonstrated that neutral vesicles released by the UMSC are taken up by the fibroblasts and proceed to fuse with the acidic lysosomes. Therefore, transplanted UMSCs participate both in extracellular GAG turnover and enable host keratocytes to catabolize accumulated GAG products, suggesting that UMSC could be a novel alternative for treating corneal defects associated with MPS and other congenital metabolic disorders.


Assuntos
Doenças da Córnea/terapia , Transplante de Células-Tronco Mesenquimais , Mucopolissacaridose VII/terapia , Animais , Técnicas de Cocultura , Ceratócitos da Córnea/metabolismo , Substância Própria/metabolismo , Substância Própria/patologia , Feminino , Glicosaminoglicanos/metabolismo , Humanos , Lisossomos/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Faloidina/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
17.
Exp Cell Res ; 319(7): 967-81, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23399832

RESUMO

The stromal reaction surrounding tumors leads to the formation of a tumor-specific microenvironment, which may play either a restrictive role or a supportive role in the growth and progression of the tumors. Lumican, a small leucine-rich proteoglycan (SLRP) of the extracellular matrix (ECM), regulates collagen fibrillogenesis. Recently, lumican has also been shown to regulate cell behavior during embryonic development, tissue repair and tumor progression. The role of lumican in cancer varies according to the type of tumor. In this study we analyze the role of lumican in the pathogenesis of prostate cancer both in vivo and in vitro. Overall lumican up-regulation was observed in the primary tumors analyzed through both real-time PCR and immunostaining. The increase in lumican expression was observed in the reactive stroma surrounding prostate primary tumors with fibrotic deposition surrounding the acinar glands. In vitro analysis demonstrated that lumican inhibited both the migration and invasion of metastatic prostate cancer cells isolated from lymph node, bone and brain. Moreover, prostate cancer cells seeded on lumican presented a decrease in the formation of cellular projections, lamellipodia detected by a decreased rearrangement in ZO-1, keratin 8/18, integrin ß1 and MT1-MMP, and invadopodia detected by disruption of α-smooth muscle actin, cortactin and N-WASP. Moreover, a significant increase in prostate cancer cell invasion was observed through the peritoneum of lumican knockout mice, further demonstrating the restrictive role lumican present in the ECM has on prostate cancer invasion. In conclusion, lumican present in the reactive stroma surrounding prostate primary tumors plays a restrictive role on cancer progression, and we therefore postulate that lumican could be a valuable marker in prostate cancer staging.


Assuntos
Proteoglicanas de Sulfatos de Condroitina/biossíntese , Sulfato de Queratano/biossíntese , Neoplasias da Próstata/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Proteoglicanas de Sulfatos de Condroitina/deficiência , Humanos , Integrina beta1/metabolismo , Sulfato de Queratano/deficiência , Lumicana , Masculino , Camundongos , Camundongos Knockout , Neoplasias da Próstata/patologia , Regulação para Cima
18.
Ocul Surf ; 32: 39-47, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38218582

RESUMO

PURPOSE: To design a novel efficacious scAAV-Gusb viral vector for treating Mucopolysaccharidosis Type VII (MPS VII) caused by a mutation in the ß-Glu gene (Gusb allele). METHODS: ß-Glu expression of single-stranded AAV-Gusb (ssAAV-Gusb) and self-complementary AAV (scAAV-Gusb) vectors are tested with cultured murine Gusb fibroblasts. The scAAV-Gusb vector was chosen in further studies to prolong the life span and treat corneal pathology of Gusb mice via intrahepatic injection of neonates and intrastromal injection in adults, respectively. Corneal pathology was studied using HRT2 in vivo confocal microscope and histochemistry in mice corneas. RESULTS: Both ssAAV-Gusb and scAAV-Gusb vectors expressed murine ß-Glu in cultured Gusb fibroblasts. The scAAV-Gusb vector had higher transduction efficiency than the ssAAV-Gusb vector. To prolong the life span of Gusb mice, neonates (3 days old) were administered with scAAV-Gusb virus via intrahepatic injection. The treatment improves the survival rate of Gusb mice, prolonging the median survival rate from 22.5 weeks (untreated) to 50 weeks (treated). Thereafter, we determined the efficacy of the scAAV-Gusb virus in ameliorating corneal cloudiness observed in aged Gusb mice. Both corneal cloudiness and stroma thickness decreased, and there was the presence of ß-Glu enzyme activity in the Gusb corneas receiving scAAV-Gusb virus associated with morphology change of amoeboid stromal cells in untreated to characteristic dendritic keratocytes morphology after 4-12 weeks of scAAV-Gusb virus injection. CONCLUSION: Intrahepatic injection of scAAV-Gusb is efficacious in prolonging the life span of Gusb mice, and intrastromal injection can ameliorate corneal phenotypes. Both strategies can be adapted for treating other MPS.


Assuntos
Dependovirus , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos , Mucopolissacaridose VII , Animais , Camundongos , Terapia Genética/métodos , Dependovirus/genética , Mucopolissacaridose VII/terapia , Mucopolissacaridose VII/genética , Fibroblastos , Opacidade da Córnea/terapia , Células Cultivadas , Microscopia Confocal , Córnea/patologia , Camundongos Endogâmicos C57BL
19.
Lab Invest ; 93(2): 207-17, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23207449

RESUMO

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.


Assuntos
Lesões da Córnea , Substância Própria/fisiopatologia , Regulação da Expressão Gênica/genética , Tenascina/deficiência , Cicatrização/fisiologia , Análise de Variância , Animais , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Fibronectinas/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1/metabolismo
20.
J Cell Sci ; 124(Pt 15): 2561-72, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21730020

RESUMO

Notch signaling is pivotal for the morphogenesis and homeostasis of many tissues. We found that aberrant Notch activation in mouse neural-crest-derived periocular mesenchymal cells (POMCs), which contribute to the formation of corneal and eyelid stroma, results in blepharophimosis. Compound transgenic mice overexpressing the Notch1 intracellular domain (N1-ICD) in POMCs (POMC(N1-ICD)) showed relatively minor effects on the cornea, but increased cell apoptosis and decreased cell proliferation during eyelid morphogenesis. Eyelid closure at E15.5 and eyelid formation at birth were incomplete. In further analyses, overexpression of N1-ICD impaired eyelid levator smooth muscle formation by downregulating the transcription factor FoxL2. This is similar to the effect of haploinsufficiency of FOXL2 in humans, which results in type II BPES (blepharophimosis, ptosis and epicanthus inversus syndrome). In vitro studies showed that FoxL2 expression is augmented by a low dose of N1-ICD but was downregulated by a high dose, depending on the extent of Hes-1 and Hey-1 activation. Moreover, transfection of CMV-FoxL2 enhanced α-SMA promoter activity. These data strongly imply that a physiologically low level of Notch1 is crucial for proper FoxL2 expression in POMCs, which is, in turn, essential for Müeller muscle formation and normal eyelid development.


Assuntos
Blefarofimose/metabolismo , Blefarofimose/patologia , Pálpebras/metabolismo , Pálpebras/patologia , Fatores de Transcrição Forkhead/metabolismo , Músculo Liso/metabolismo , Receptor Notch1/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Blefarofimose/genética , Western Blotting , Proliferação de Células , Imunoprecipitação da Cromatina , Proteína Forkhead Box L2 , Fatores de Transcrição Forkhead/genética , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Células NIH 3T3 , Receptor Notch1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA