Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Rep ; 7(1): 812, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28400560

ABSTRACT

Fundamental cell signaling mechanisms that regulate dynamic remodeling of the extracellular matrix (ECM) in mechanically loaded tissues are not yet clearly understood. Trabecular meshwork (TM) tissue in the eye is under constant mechanical stress and continuous remodeling of ECM is crucial to maintain normal aqueous humor drainage and intraocular pressure (IOP). However, excessive ECM remodeling can cause fibrosis of the TM as in primary open-angle glaucoma (POAG) patients, and is characterized by increased resistance to aqueous humor drainage, elevated IOP, optic nerve degeneration and blindness. Increased levels of active transforming growth factor-ß2 (TGF-ß2) in the aqueous humor is the main cause of fibrosis of TM in POAG patients. Herein, we report a novel finding that, in TM cells, TGF-ß-induced increase in collagen expression is associated with phosphorylation of phosphatase and tensin homolog (PTEN) at residues Ser380/Thr382/383. Exogenous overexpression of a mutated form of PTEN with enhanced phosphatase activity prevented the TGF-ß-induced collagen expression by TM cells. We propose that rapid alteration of PTEN activity through changes in its phosphorylation status could uniquely regulate the continuous remodeling of ECM in the normal TM. Modulating PTEN activity may have high therapeutic potential to alleviating the fibrosis of TM in POAG patients.


Subject(s)
Glaucoma, Open-Angle/metabolism , PTEN Phosphohydrolase/metabolism , Protein Processing, Post-Translational , Trabecular Meshwork/pathology , Transforming Growth Factor beta/pharmacology , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibrosis , Glaucoma, Open-Angle/pathology , Humans , Phosphorylation , Trabecular Meshwork/metabolism , Transforming Growth Factor beta/metabolism
2.
Matrix Biol ; 43: 35-41, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644104

ABSTRACT

Elevated adhesive signaling promotes fibrosis. Protein phosphatase and tensin homologue (PTEN) dephosphorylates focal adhesion kinase and suppresses the activation of Akt and hence suppresses adhesive signaling. Loss of PTEN expression is associated with lung fibrosis, but whether PTEN expression by type I collagen-expressing cells controls lung fibrosis is unclear. Here, we use mice expressing tamoxifen-dependent cre recombinase expressed under the control of a COL1A2 promoter/enhancer and mice harboring floxed-PTEN and/or floxed-CCN2 alleles to assess whether loss of PTEN expression by type I collagen producing cells results in lung fibrosis in a CCN2-dependent fashion. In vivo, loss of PTEN expression resulted in the overexpression of both collagen type I and the pro-adhesive matricellular protein connective tissue growth factor (CTGF/CCN2). However, α-smooth muscle actin expression was unaffected. Loss of CCN2 expression by lung fibroblasts rescues this phenotype; i.e.., mice deficient in both PTEN and CCN2 in collagen type I-expressing cells do not develop significant collagen deposition in the lung. PTEN expression by collagen type I-expressing cells controls collagen deposition; therapeutic strategies blocking CCN2 may be of benefit in blocking excessive collagen deposition in fibrosis.


Subject(s)
Connective Tissue Growth Factor/metabolism , Fibroblasts/metabolism , PTEN Phosphohydrolase/genetics , Pulmonary Fibrosis/pathology , Animals , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Connective Tissue Growth Factor/genetics , Fibroblasts/pathology , Gene Expression Regulation , Lung/cytology , Lung/metabolism , Lung/pathology , Mice , PTEN Phosphohydrolase/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism
3.
Clin Exp Ophthalmol ; 43(5): 443-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25495256

ABSTRACT

BACKGROUND: To assess the effects of amniotic membrane extract (AMX) on cellular activity of primary human corneal epithelial (HCE) cells under mechanical and oxidative stress, and on human limbal cells under oxidative stress. METHODS: Corneal mechanical stress was simulated with a linear scratch in confluent HCE cell plates, then incubated with 0.1% AMX for 48 and 72 h. Subjecting HCE cultures to 0.5 mmol/L tertiary-butylhydroperoxide for 1 h simulated an oxidative stress. 0.1% AMX-treated cultures were compared with controls at 24 and 48 h using cellular viability assay, along with 12-h AMX pretreatment and human limbal cell comparisons. RESULTS: Mechanical stress on HCE cultures revealed a statistically significant distance ratio at 48 and 72 h in favour of 0.1% AMX-treated cultures (P = 0.021 and 0.035, respectively). Oxidative stress did not reveal any significant difference in cellular viability of AMX-treated versus control cultures. Twelve hour AMX pre-treatment prior to oxidative stress revealed a significant difference after 24 h from oxidative injury (73.3% AMX vs. 66.0% control, P = 0.035), but not after 48 h. Human limbal cells demonstrated significantly improved oxidative viability compared with HCE cells, with (91.0% vs. 82.0% control, P = 0.017) and without 0.1% AMX pre-treatment (91.2% vs. 83.7% control, P = 0.019). CONCLUSIONS: HCE cells treated with AMX healed faster after mechanical insult, suggesting a potential benefit in acute corneal injuries. Under oxidative stress, human limbal cells, a more proliferative cell type, showed superior viability compared with HCE cells.


Subject(s)
Amnion/chemistry , Epithelium, Corneal/drug effects , Limbus Corneae/drug effects , Tissue Extracts/pharmacology , Cell Proliferation/drug effects , Cell Survival , Epithelium, Corneal/cytology , Humans , Limbus Corneae/cytology , Oxidative Stress , Stress, Mechanical , Wound Healing/physiology , tert-Butylhydroperoxide/toxicity
4.
J Cell Commun Signal ; 8(2): 113-24, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24604397

ABSTRACT

Several studies have established the role of activated corneal keratocytes in the fibrosis of the cornea. However, the role of keratocytes in maintaining the structural integrity of a normal cornea is less appreciated. We focus on the probable functions of integrins in the eye and of the importance of integrin-mediated keratocyte interactions with stromal matrix in the maintenance of corneal integrity. We point out that further understanding of how keratocytes interact with their matrix could establish a novel direction in preventing corneal pathology including loss of structural integrity as in keratoconus or as in fibrosis of the corneal stroma.

5.
Arthritis Rheum ; 65(11): 2940-4, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23983074

ABSTRACT

OBJECTIVE: Protein phosphatase and tensin homolog (PTEN) expression is reduced in dermal fibroblasts isolated from patients with diffuse cutaneous systemic sclerosis, a fibrotic autoimmune disease. In support of this finding, deletion of the PTEN gene in the dermal fibroblasts of mice has been shown to result in skin fibrosis and in vivo overexpression of connective tissue growth factor (CTGF; CCN2), a proadhesive matricellular protein; however, whether CCN2 is required for the fibrosis caused by loss of PTEN is unclear. This study was undertaken to investigate the role of CCN2 in fibrosis caused by reduced PTEN expression. METHODS: We generated conditional knockout mice in which PTEN was deleted in fibroblasts, either alone or in combination with CCN2. Skin samples were collected for histologic examination, immunohistochemical analysis, and collagen assay. RESULTS: Loss of CCN2 resulted in resistance to the increases in collagen production and myofibroblast recruitment that are caused by loss of PTEN. CCN2 deficiency did not impair Akt phosphorylation or the increases in the intensity of proliferating cell nuclear antigen staining that were caused by loss of PTEN. CONCLUSION: These data are consistent with the notion that CCN2 is required for particular aspects of the fibroproliferative response; therapeutic strategies blocking CCN2 may be of clinical benefit in combating fibrotic disease.


Subject(s)
Connective Tissue Growth Factor/genetics , Dermis/pathology , Fibroblasts/pathology , PTEN Phosphohydrolase/genetics , Scleroderma, Diffuse/genetics , Scleroderma, Diffuse/pathology , Animals , Collagen/metabolism , Connective Tissue Growth Factor/metabolism , Dermis/metabolism , Fibroblasts/metabolism , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Mice , Mice, Knockout , PTEN Phosphohydrolase/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Scleroderma, Diffuse/metabolism
6.
Invest Ophthalmol Vis Sci ; 52(11): 7799-806, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21873663

ABSTRACT

PURPOSE: The precise role of a normal keratocyte in maintaining corneal structural integrity is unclear; it is generally considered to remain quiescent at the end of cell division. Given that integrins are essential for cell/extracellular matrix interactions, the authors tested the hypothesis that integrin expression by keratocytes is essential for corneal structure and function. METHODS: Using a tamoxifen-dependent cre recombinase expressed under the control of a fibroblast-specific promoter/enhancer, the authors conditionally deleted the integrin ß1 (Itgb1) gene in mouse keratocytes during the postnatal matrix maturation phase of the cornea. The effects of this deletion were monitored histologically and by macroscopic observation of the cornea. RESULTS: The resultant cornea shows an initial thinning of the stroma, reduced space between collagen fibrils, loss of epithelial layers and subsequent edema, thickening of Descemet's membrane, and degenerative changes in the endothelial cell layer, with eventual scarring. These pathologic changes have some similarities to human corneal disease keratoconus. The phenotype did not develop when Itgb1 was deleted after complete corneal maturation. CONCLUSIONS: Loss of integrin ß1 expression in keratocytes during the phase of stromal maturation results in corneal thinning and edema. Keratocyte-ECM interaction is essential for matrix maturation and thus in the maintenance of corneal structural integrity. This model has relevance in understanding corneal diseases such as keratoconus.


Subject(s)
Corneal Edema/pathology , Corneal Stroma/cytology , Fibroblasts/pathology , Integrin beta1/physiology , Keratoconus/pathology , Animals , Cells, Cultured , Corneal Edema/metabolism , Corneal Stroma/metabolism , Cyclin D1/metabolism , Endothelium, Corneal/pathology , Epithelium, Corneal/metabolism , Epithelium, Corneal/pathology , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Gene Deletion , Integrases/genetics , Keratoconus/metabolism , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Polymerase Chain Reaction
7.
J Invest Dermatol ; 131(10): 1996-2003, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21654839

ABSTRACT

Fibrosis represents a common pathway leading to organ failure and death in many diseases and has no effective therapy. Dysregulated repair and excessive tissue scarring provides a unifying mechanism for pathological fibrosis. The protein phosphatase and tensin homolog (PTEN) acts to dephosphorylate proteins, which promotes tissue repair and thus could be a key fibrogenic mediator. To test this hypothesis, we first showed that PTEN expression was reduced in skin fibroblasts from patients with the fibrotic autoimmune disease diffuse systemic sclerosis (dSSc). To evaluate whether this deficiency could be sufficient for fibrogenesis in vivo, we deleted PTEN in adult mouse fibroblasts. Compared with littermate control mice, loss of PTEN resulted in a 3-fold increase in dermal thickness due to excess deposition of collagen. PTEN-deleted fibroblasts showed elevated Akt phosphorylation and increased expression of connective tissue growth factor (CTGF/CCN2). Selective inhibition of the phosphatidylinositol 3-kinase/Akt pathway reduced the overexpression of collagen and CCN2 by PTEN-deficient fibroblasts. Overexpression of PTEN reduced the overexpression of type I collagen and CCN2 by dSSc fibroblasts. Thus, PTEN appears to be a potential in vivo master regulator of fibrogenesis; PTEN agonists may represent anti-fibrotic treatments.


Subject(s)
Fibroblasts/cytology , Fibrosis/pathology , Gene Expression Regulation , PTEN Phosphohydrolase/genetics , Scleroderma, Diffuse/pathology , Skin/pathology , Animals , Cell Line , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Female , Fibrosis/genetics , Humans , Immunohistochemistry/methods , Mice , Phosphorylation
8.
Arthritis Res Ther ; 13(1): R6, 2011.
Article in English | MEDLINE | ID: mdl-21266028

ABSTRACT

INTRODUCTION: Microsomal prostaglandin E2 synthase-1 (mPGES-1) is an inducible enzyme that acts downstream of cyclooxygenase (COX) to specifically catalyze the conversion of prostaglandin (PG) H2 to PGE2. mPGES-1 plays a key role in inflammation, pain and arthritis; however, the role of mPGES-1 in fibrogenesis is largely unknown. Herein, we examine the role of mPGES-1 in a mouse model of skin scleroderma using mice deficient in mPGES-1. METHODS: Wild type (WT) and mPGES-1 null mice were subjected to the bleomycin model of cutaneous skin scleroderma. mPGES-1 expressions in scleroderma fibroblasts and in fibroblasts derived from bleomycin-exposed mice were assessed by Western blot analysis. Degree of fibrosis, dermal thickness, inflammation, collagen content and the number of α-smooth muscle actin (α-SMA)-positive cells were determined by histological analyses. The quantity of the collagen-specific amino acid hydroxyproline was also measured. RESULTS: Compared to normal skin fibroblasts, mPGES-1 protein expression was elevated in systemic sclerosis (SSc) fibroblasts and in bleomycin-exposed mice. Compared to WT mice, mPGES-1-null mice were resistant to bleomycin-induced inflammation, cutaneous thickening, collagen production and myofibroblast formation. CONCLUSIONS: mPGES-1 expression is required for bleomycin-induced skin fibrogenesis. Inhibition of mPGES-1 may be a viable method to alleviate the development of cutaneous sclerosis and is a potential therapeutic target to control the onset of fibrogenesis.


Subject(s)
Intramolecular Oxidoreductases/metabolism , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Blotting, Western , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibrosis/chemically induced , Fibrosis/metabolism , Fibrosis/pathology , Fluorescent Antibody Technique , Immunohistochemistry , Intramolecular Oxidoreductases/deficiency , Male , Mice , Mice, Knockout , Prostaglandin-E Synthases , Scleroderma, Systemic/chemically induced
9.
PLoS One ; 5(11): e13885, 2010 Nov 08.
Article in English | MEDLINE | ID: mdl-21079736

ABSTRACT

BACKGROUND: Advanced age contributes to clinical manifestations of many retinopathies and represents a major risk factor for age-related macular degeneration, a leading cause of visual impairment and blindness in the elderly. Rod photoreceptors are especially vulnerable to genetic defects and changes in microenvironment, and are among the first neurons to die in normal aging and in many retinal degenerative diseases. The molecular mechanisms underlying rod photoreceptor vulnerability and potential biomarkers of the aging process in this highly specialized cell type are unknown. METHODOLOGY/PRINCIPAL FINDINGS: To discover aging-associated adaptations that may influence rod function, we have generated gene expression profiles of purified rod photoreceptors from mouse retina at young adult to early stages of aging (1.5, 5, and 12 month old mice). We identified 375 genes that showed differential expression in rods from 5 and 12 month old mouse retina compared to that of 1.5 month old retina. Quantitative RT-PCR experiments validated expression change for a majority of the 25 genes that were examined. Macroanalysis of differentially expressed genes using gene class testing and protein interaction networks revealed overrepresentation of cellular pathways that are potentially photoreceptor-specific (angiogenesis and lipid/retinoid metabolism), in addition to age-related pathways previously described in several tissue types (oxidative phosphorylation, stress and immune response). CONCLUSIONS/SIGNIFICANCE: Our study suggests a progressive shift in cellular homeostasis that may underlie aging-associated functional decline in rod photoreceptors and contribute to a more permissive state for pathological processes involved in retinal diseases.


Subject(s)
Aging , Gene Expression Profiling , Homeostasis/genetics , Retinal Rod Photoreceptor Cells/metabolism , Animals , Cluster Analysis , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Retina/growth & development , Retina/metabolism , Retinal Diseases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
10.
Invest Ophthalmol Vis Sci ; 50(12): 5965-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19578024

ABSTRACT

PURPOSE: In proliferative vitreoretinopathy retinal pigment epithelial (RPE) cells undergo epithelial-mesenchymal transformation (EMT). Vitreous and transforming growth factor-beta (TGFbeta) have been implicated in this EMT. The role of TGFbeta in the vitreous-mediated transformation of low-passage human RPE cells was investigated. METHODS: Cells were treated with vitreous or TGFbeta2. SB431542 was used to inhibit TGFbeta signaling. Morphology was investigated using phase-contrast or confocal microscopy. Motility was measured using a monolayer-wounding assay. Invasion was determined using basement membrane matrix-based assays. Gene expression was measured by quantitative PCR, immunohistochemistry, or immunoblotting. RESULTS: Changes in phosphorylation or cellular localization of Smad -2, -3, or -4 indicated a TGFbeta-like activity in vitreous. Cortical actin filaments in untreated cells were replaced by stress fibers after TGFbeta treatment, but peripheral actin aggregates were seen in vitreous-treated cells. SB431542 did not block the morphologic change induced by vitreous. Vitreous-treated cells exhibited increased motility and invasion, whereas TGFbeta-treated cells did not. However, SB431542 decreased vitreous-meditated changes in motility and invasion. The levels of mRNA for genes indicative of myofibroblast differentiation (alpha-SMA and CTGF) were increased by treatment with TGFbeta but suppressed by vitreous. TGFbeta or vitreous caused increased expression of Snail1. CONCLUSIONS: Vitreous or TGFbeta caused a fibroblast-like morphology and induced Snail1, a marker of EMT. TGFbeta activity in vitreous was necessary but not sufficient for the vitreous-induced motile, invasive phenotype. However, differences in the cytoskeletal organization and in the expression of CTGF and alpha-SMA suggested that TGFbeta-treatment caused differentiation along a myofibroblast pathway, whereas vitreous treatment suppressed myofibroblast formation.


Subject(s)
Retinal Pigment Epithelium/cytology , Transforming Growth Factor beta2/pharmacology , Vitreous Body/physiology , Actins/genetics , Benzamides/pharmacology , Cell Differentiation/drug effects , Cell Line, Transformed , Cell Movement/drug effects , Cells, Cultured , Connective Tissue Growth Factor/genetics , Dioxoles/pharmacology , Gene Expression Regulation/drug effects , Humans , Immunoblotting , Immunohistochemistry , Microscopy, Confocal , Microscopy, Phase-Contrast , Phosphorylation , RNA, Messenger/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Retinal Pigment Epithelium/physiology , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/genetics
11.
Arthritis Rheum ; 60(1): 234-41, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19116914

ABSTRACT

OBJECTIVE: Fibrosis is believed to occur through normal tissue remodeling failing to terminate. Tissue repair intimately involves the ability of fibroblasts to contract extracellular matrix (ECM), and enhanced ECM contraction is a hallmark of fibrotic cells in various conditions, including scleroderma. Some fibrogenic transcriptional responses to transforming growth factor beta (TGFbeta), including alpha-smooth muscle actin (alpha-SMA) expression and ECM contraction, require focal adhesion kinase/Src (FAK/Src). The present study was undertaken to assess whether TGFbeta-activated kinase 1 (TAK1) acts downstream of FAK/Src to mediate fibrogenic responses in fibroblasts. METHODS: We used microarray, real-time polymerase chain reaction, Western blot, and collagen gel contraction assays to assess the ability of wild-type and TAK1-knockout fibroblasts to respond to TGFbeta1. RESULTS: The ability of TGF to induce TAK1 was blocked by the FAK/Src inhibitor PP2. JNK phosphorylation in response to TGFbeta1 was impaired in the absence of TAK1. TGFbeta could not induce matrix contraction or expression of a group of fibrotic genes, including alpha-SMA, in the absence of TAK1. CONCLUSION: These results suggest that TAK1 operates downstream of FAK/Src in mediating fibrogenic responses and that targeting of TAK1 may be a viable antifibrotic strategy in the treatment of certain disorders, including scleroderma.


Subject(s)
Actins/genetics , Extracellular Matrix/physiology , Fibroblasts/physiology , Actins/metabolism , Animals , Cell Line, Transformed , Fibroblasts/cytology , Fibrosis , Focal Adhesion Kinase 1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphorylation/physiology , Reverse Transcriptase Polymerase Chain Reaction , src-Family Kinases/metabolism
12.
J Cell Commun Signal ; 3(2): 89-94, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19156539

ABSTRACT

The CCN (cyr61, ctgf, nov) family of modular proteins regulate diverse biological affects including cell adhesion, matrix production, tissue remodelling, proliferation and differentiation. Recent targeted gene disruption studies have demonstrated the CCN family to be developmentally essential for chondrogenesis, osteogenesis and angiogenesis. CCN2 is induced by agents such as angiotensin II, endothelin-1, glucocorticoids, HGF, TGFbeta, and VEGF, and by hypoxia and biomechanical and shear stress. Dysregulated expression of CCN2 has also been widely documented in many fibroproliferative diseases. This mini-review will focus on CCN2, and the recent progress in understanding CCN2 gene regulation in health and disease. That CCN2 should be considered a novel and informative surrogate clinical bio-marker for fibroproliferative disease is discussed.

13.
Fibrogenesis Tissue Repair ; 1(1): 3, 2008 Oct 13.
Article in English | MEDLINE | ID: mdl-19014648

ABSTRACT

In skin, connective tissue growth factor (CTGF/CCN2) is induced during tissue repair. However, what the exact cell types are that express CTGF in normal and wounded skin remain controversial. In this report, we use transgenic knock-in mice in which the Pacific jellyfish Aequorea victoria enhanced green fluorescent protein (E-GFP) gene has been inserted between the endogenous CTGF promoter and gene. Unwounded (day 0) and wounded (days 3 and 7) skin was examined for GFP to detect cells in which the CTGF promoter was active, alpha-smooth muscle actin (alpha-SMA) to detect myofibroblasts, and NG2 expression to detect pericytes. In unwounded mice, CTGF expression was absent in epidermis and was present in a few cells in the dermis. Upon wounding, CTGF expression was induced in the dermis. Double immunolabeling revealed that CTGF-expressing cells also expressed alpha-SMA, indicating the CTGF was expressed in myofibroblasts. A subset (approximately 30%) of myofibroblasts were also NG2 positive, indicating that pericytes significantly contributed to the number of myofibroblasts in the wound. Pericytes also expressed CTGF. Collectively, these results indicate that CTGF expression in skin correlates with myofibroblast induction, and that CTGF-expressing pericytes are significant contributors to myofibroblast activity during cutaneous tissue repair.

14.
J Cell Commun Signal ; 2(1-2): 19-23, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18649016

ABSTRACT

Connective tissue growth factor (CTGF, CCN2) is induced in response to TGFbeta in fibroblasts. In this report, we show that C2 ceramide reduced the ability of TGFbeta to induce CCN2 protein, mRNA and promoter activity in fibroblasts. C2 ceramide reduced the ability of TGFbeta to induce the generic Smad responsive promoter/reporter construct SBE-luciferase. These results suggest that C2 ceramide reduces the action of TGFbeta in fibroblasts via Smad antagonism.

15.
Vision Res ; 48(3): 366-76, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17904189

ABSTRACT

Mutations in the cilia-centrosomal protein Retinitis Pigmentosa GTPase Regulator (RPGR) are a frequent cause of retinal degeneration. The RPGR gene undergoes complex alternative splicing and encodes multiple protein isoforms. To elucidate the function of major RPGR isoforms (RPGR 1-19 and RPGR ORF15), we have generated isoform-specific antibodies and examined their expression and localization in the retina. Using sucrose-gradient centrifugation, immunofluorescence and co-immunoprecipitation methods, we show that RPGR isoforms localize to distinct sub-cellular compartments in mammalian photoreceptors and associate with a number of cilia-centrosomal proteins. The RCC1-like domain of RPGR, which is present in all major RPGR isoforms, is sufficient to target it to the cilia and centrosomes in cultured cells. Our findings indicate that multiple isotypes of RPGR may perform overlapping yet somewhat distinct transport-related functions in photoreceptors.


Subject(s)
Carrier Proteins/physiology , Eye Diseases, Hereditary/genetics , Eye Proteins/physiology , Genetic Diseases, X-Linked/genetics , Retinitis Pigmentosa/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cattle , Cell Cycle/physiology , Cells, Cultured , Centrosome/metabolism , Eye Diseases, Hereditary/metabolism , Eye Proteins/genetics , Eye Proteins/immunology , Eye Proteins/metabolism , Genetic Diseases, X-Linked/metabolism , Humans , Mice , Photoreceptor Cells, Vertebrate/metabolism , Protein Isoforms/metabolism , Protein Isoforms/physiology , Retina/metabolism , Retinitis Pigmentosa/metabolism , Species Specificity
16.
Am J Hum Genet ; 81(5): 1098-103, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17924349

ABSTRACT

We report mutations in the gene for topoisomerase I-binding RS protein (TOPORS) in patients with autosomal dominant retinitis pigmentosa (adRP) linked to chromosome 9p21.1 (locus RP31). A positional-cloning approach, together with the use of bioinformatics, identified TOPORS (comprising three exons and encoding a protein of 1,045 aa) as the gene responsible for adRP. Mutations that include an insertion and a deletion have been identified in two adRP-affected families--one French Canadian and one German family, respectively. Interestingly, a distinct phenotype is noted at the earlier stages of the disease, with an unusual perivascular cuff of retinal pigment epithelium atrophy, which was found surrounding the superior and inferior arcades in the retina. TOPORS is a RING domain-containing E3 ubiquitin ligase and localizes in the nucleus in speckled loci that are associated with promyelocytic leukemia bodies. The ubiquitous nature of TOPORS expression and a lack of mutant protein in patients are highly suggestive of haploinsufficiency, rather than a dominant negative effect, as the molecular mechanism of the disease and make rescue of the clinical phenotype amenable to somatic gene therapy.


Subject(s)
Genes, Dominant , Mutation/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Pigment Epithelium of Eye/blood supply , Pigment Epithelium of Eye/pathology , Retinitis Pigmentosa/genetics , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Base Sequence , Child , Chromosomes, Human , DNA Mutational Analysis , Exons/genetics , Female , Humans , Male , Middle Aged , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Pedigree , Ubiquitin-Protein Ligases/metabolism
17.
Invest Ophthalmol Vis Sci ; 48(4): 1853-63, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17389521

ABSTRACT

PURPOSE: In proliferative vitreoretinopathy (PVR), retinal pigment epithelial (RPE) cells enter the vitreous and proliferate. They become fibroblast-like and participate in the formation of contractile membranes, which can lead to retinal detachment. Vitreous treatment of RPE cells in vitro results in similar morphologic changes. This study was conducted to examine vitreous-induced modulation of gene expression in RPE cells. METHODS: Low-passage human RPE cell lines derived from three donors were each treated for 6, 12, 24, or 48 hours with complete medium or complete medium containing 25% vitreous. Changes in mRNA levels were examined by using microarrays. Real-time quantitative PCR (qPCR) was used to measure mRNA expression of a subset of genes in cells from three additional donors. Immunohistochemistry and immunoblot analysis were used to examine protein expression. RESULTS: Vitreous treatment caused a progressive reprogramming of gene expression. qPCR confirmed vitreous modulation of mRNA levels of 10 of 10 genes. Changes consistent with a transition from an epithelial to a mesenchymal phenotype were observed. Downregulated genes included genes associated with differentiated RPE cells. Upregulated genes included genes associated with stress and inflammation. Pathway analysis indicated that the transforming growth factor-beta/bone morphogenetic protein (BMP) pathway and the focal adhesion pathway may play a role in this process. BMP-2 protein and mRNA were increased. CONCLUSIONS: Despite the biological variation in vitreous and RPE donors, vitreous reproducibly modulated a limited number of mRNAs. Many of these changes were consistent with the more fibroblast-like appearance of vitreous-treated cells and with the pathobiology of PVR. TGF-beta and BMP-2 may be important modulators of vitreous-induced changes in gene expression.


Subject(s)
Eye Proteins/genetics , Gene Expression/physiology , Pigment Epithelium of Eye/metabolism , Vitreous Body/physiology , Cells, Cultured , Gene Expression Profiling , Humans , Immunoblotting , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Pigment Epithelium of Eye/cytology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
18.
Mol Vis ; 13: 66-78, 2007 Jan 24.
Article in English | MEDLINE | ID: mdl-17277740

ABSTRACT

PURPOSE: When human retinal pigment epithelial (RPE) cells come in contact with vitreous, they undergo changes in gene expression that include inflammatory and anti-oxidant responses. The effects of vitreous on expression of heme oxygenase-1 (HO-1), metallothionein (MT) -1a and -2a, and c-fos were investigated. Activator protein-1 (AP-1) binding sites are located in the promoter region of HO-1 and MT genes and the effects of vitreous on c-fos activity were investigated. METHODS: Low passage cultures of human RPE cells were grown in the presence or absence of vitreous or transforming growth factor-beta (TGF-beta). The expression of HO-1 and MTs was measured by real time PCR and, in the case of HO-1, by immunoblotting and immunofluorescence microscopy. Specific inhibitors were used to investigate possible signaling pathways. The effect of vitreous on activation of AP-1 transcription factor was determined by immunoblotting, electrophoretic mobility shift assays, or immunofluorescence microscopy. RESULTS: Incubation of RPE cells with vitreous resulted in increased expression of HO-1, MT-1a and MT-2a. TGF-beta caused an increase in HO-1 expression, although not to the extent mediated by vitreous, but had little effect on MT expression. Addition of inhibitors of TGF-beta signaling (SB431542 or TGF-beta-neutralizing antibodies) decreased the vitreous induction of HO-1. Several reactive oxygen species (ROS) quenchers inhibited the TGF-beta-induced or vitreous-induced elevation of HO-1 mRNA but had no effect on vitreous-mediated induction of MT expression. Inhibitors of the mitogen-activated protein kinase (p38MAPK; SB203580) and Jun N-terminal kinase (JNK; SP600125) pathways inhibited vitreous-induction of HO-1. C-fos, a component of AP-1 transcription factor complexes, exhibited increased expression and activation in the presence of vitreous. CONCLUSIONS: TGF-beta, a known component of vitreous, can account for some but not all of the regulation of the anti-oxidant, anti-inflammatory HO-1 gene in human RPE cells, but it does not participate in the vitreous-mediated upregulation of MTs. Both vitreous and TGF-beta signals increased HO-1 expression via ROS but the latter were not involved in vitreous-mediated MT expression. Increased p38, JNK, and c-fos activation may be implicated in vitreous modulation of HO-1.


Subject(s)
Heme Oxygenase-1/biosynthesis , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/physiology , Vitreous Body/physiology , Activin Receptors, Type I/antagonists & inhibitors , Benzamides/pharmacology , Biological Transport/physiology , Cell Nucleus/metabolism , Cells, Cultured , Dioxoles/pharmacology , Enzyme Activation/physiology , Heme Oxygenase-1/genetics , Humans , Metallothionein/genetics , Metallothionein/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor AP-1/metabolism , Vitreous Body/cytology
19.
Hum Mol Genet ; 15(11): 1847-57, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16632484

ABSTRACT

Centrosome- and cilia-associated proteins play crucial roles in establishing polarity and regulating intracellular transport in post-mitotic cells. Using genetic mapping and positional candidate strategy, we have identified an in-frame deletion in a novel centrosomal protein CEP290 (also called NPHP6), leading to early-onset retinal degeneration in a newly identified mouse mutant, rd16. We demonstrate that CEP290 localizes primarily to centrosomes of dividing cells and to the connecting cilium of retinal photoreceptors. We show that, in the retina, CEP290 associates with several microtubule-based transport proteins including RPGR, which is mutated in approximately 15% of patients with retinitis pigmentosa. A truncated CEP290 protein (DeltaCEP290) is detected in the rd16 retina, but in considerably reduced amounts; however, the mutant protein exhibits stronger association with specific RPGR isoform(s). Immunogold labeling studies demonstrate the redistribution of RPGR and of phototransduction proteins in the photoreceptors of rd16 retina. Our findings suggest a critical function for CEP290 in ciliary transport and provide insights into the mechanism of early-onset photoreceptor degeneration.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Carrier Proteins/metabolism , Centrosome/ultrastructure , Eye Proteins/genetics , Gene Deletion , Nuclear Proteins/genetics , Retinal Degeneration/genetics , Animals , Base Sequence , Carrier Proteins/genetics , Cell Cycle Proteins , Centrosome/metabolism , Cytoskeletal Proteins , Disease Models, Animal , Eye Proteins/metabolism , Humans , Mice , Models, Genetic , Molecular Sequence Data , Mutation , Nuclear Proteins/physiology , Protein Binding
20.
J Biol Chem ; 280(39): 33580-7, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16043481

ABSTRACT

Mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene account for almost 20% of patients with retinitis pigmentosa. Most mutations are detected in alternatively spliced RPGR-ORF15 isoform(s), which are primarily but not exclusively expressed in the retina. We show that, in addition to the axoneme, the RPGR-ORF15 protein is localized to the basal bodies of photoreceptor connecting cilium and to the tip and axoneme of sperm flagella. Mass spectrometric analysis of proteins that were immunoprecipitated from the retinal axoneme-enriched fraction using an anti-ORF15 antibody identified two chromosome-associated proteins, structural maintenance of chromosomes (SMC) 1 and SMC3. Using pulldown assays, we demonstrate that the interaction of RPGR with SMC1 and SMC3 is mediated, at least in part, by the RCC1-like domain of RPGR. This interaction was not observed with phosphorylation-deficient mutants of SMC1. Both SMC1 and SMC3 localized to the cilia of retinal photoreceptors and Madin-Darby canine kidney cells, suggesting a broader physiological relevance of this interaction. Additional immunoprecipitation studies revealed the association of RPGR-ORF15 isoform(s) with the intraflagellar transport polypeptide IFT88 as well as microtubule motor proteins, including KIF3A, p150Glued, and p50-dynamitin. Inhibition of dynein function by overexpressing p50 abrogated the localization of RPGR-ORF15 to basal bodies. Taken together, these results provide novel evidence for the possible involvement of RPGR-ORF15 in microtubule organization and regulation of transport in primary cilia.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Eye Proteins/metabolism , Retinitis Pigmentosa/prevention & control , Animals , Carrier Proteins/genetics , Carrier Proteins/ultrastructure , Cell Cycle Proteins/ultrastructure , Chondroitin Sulfate Proteoglycans/ultrastructure , Chromosomal Proteins, Non-Histone/ultrastructure , Eye Proteins/genetics , Eye Proteins/ultrastructure , Humans , Mice , Mice, Knockout , Microtubules/ultrastructure , Mutation , Open Reading Frames , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...