Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Adv Sci (Weinh) ; 10(29): e2301756, 2023 10.
Article in English | MEDLINE | ID: mdl-37485646

ABSTRACT

Astrocytes are diverse brain cells that form large networks communicating via gap junctions and chemical transmitters. Despite recent advances, the functions of astrocytic networks in information processing in the brain are not fully understood. In culture, brain slices, and in vivo, astrocytes, and neurons grow in tight association, making it challenging to establish whether signals that spread within astrocytic networks communicate with neuronal groups at distant sites, or whether astrocytes solely respond to their local environments. A multi-electrode array (MEA)-based device called AstroMEA is designed to separate neuronal and astrocytic networks, thus allowing to study the transfer of chemical and/or electrical signals transmitted via astrocytic networks capable of changing neuronal electrical behavior. AstroMEA demonstrates that cortical astrocytic networks can induce a significant upregulation in the firing frequency of neurons in response to a theta-burst charge-balanced biphasic current stimulation (5 pulses of 100 Hz × 10 with 200 ms intervals, 2 s total duration) of a separate neuronal-astrocytic group in the absence of direct neuronal contact. This result corroborates the view of astrocytic networks as a parallel mechanism of signal transmission in the brain that is separate from the neuronal connectome. Translationally, it highlights the importance of astrocytic network protection as a treatment target.


Subject(s)
Astrocytes , Gap Junctions , Gap Junctions/physiology , Neurons , Brain
2.
Vet Rec ; 185(14): 446-448, 2019 10 12.
Article in English | MEDLINE | ID: mdl-31604868
3.
Brain Behav ; 5(11): e00401, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26664787

ABSTRACT

AIMS: Synapses represent a major pathological target across a broad range of neurodegenerative conditions. Recent studies addressing molecular mechanisms regulating synaptic vulnerability and degeneration have relied heavily on invertebrate and mouse models. Whether similar molecular neuropathological changes underpin synaptic breakdown in large animal models and in human patients with neurodegenerative disease remains unclear. We therefore investigated whether molecular regulators of synaptic pathophysiology, previously identified in Drosophila and mouse models, are similarly present and modified in the brain of sheep with CLN5 Batten disease. METHODS: Gross neuropathological analysis of CLN5 Batten disease sheep and controls was used alongside postmortem MRI imaging to identify affected brain regions. Synaptosome preparations were then generated and quantitative fluorescent Western blotting used to determine and compare levels of synaptic proteins. RESULTS: The cortex was particularly affected by regional neurodegeneration and synaptic loss in CLN5 sheep, whilst the cerebellum was relatively spared. Quantitative assessment of the protein content of synaptosome preparations revealed significant changes in levels of seven out of eight synaptic neurodegeneration proteins investigated in the motor cortex, but not cerebellum, of CLN5 sheep (α-synuclein, CSP-α, neurofascin, ROCK2, calretinin, SIRT2, and UBR4). CONCLUSIONS: Synaptic pathology is a robust correlate of region-specific neurodegeneration in the brain of CLN5 sheep, driven by molecular pathways similar to those reported in Drosophila and rodent models. Thus, large animal models, such as sheep, represent ideal translational systems to develop and test therapeutics aimed at delaying or halting synaptic pathology for a range of human neurodegenerative conditions.


Subject(s)
Disease Models, Animal , Neuronal Ceroid-Lipofuscinoses/pathology , Sheep Diseases/pathology , Synapses/pathology , Animals , Cerebellum/pathology , Cerebral Cortex/pathology , Female , Male , Sheep
4.
Int J Exp Pathol ; 94(1): 1-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23110747

ABSTRACT

Connective tissue growth factor (CTGF, CCN2) is a member of the CCN family of matricellular proteins. It interacts with many other proteins, including plasma membrane proteins, modulating cell function. It is expressed at low levels in normal adult kidney cells but is increased in kidney diseases, playing important roles in inflammation and in the development of glomerular and interstitial fibrosis in chronic disease. This review reports the evidence for its expression in human and animal models of chronic kidney disease and summarizes data showing that anti-CTGF therapy can successfully attenuate fibrotic changes in several such models, suggesting that therapies targeting CTGF and events downstream of it in renal cells may be useful for the treatment of human kidney fibrosis. Connective tissue growth factor stimulates the development of fibrosis in the kidney in many ways including activating cells to increase extracellular matrix synthesis, inducing cell cycle arrest and hypertrophy, and prolonging survival of activated cells. The relationship between CTGF and the pro-fibrotic factor TGFß is examined and mechanisms by which CTGF promotes signalling by the latter are discussed. No specific cellular receptors for CTGF have been discovered but it interacts with and activates several plasma membrane proteins including low-density lipoprotein receptor-related protein (LRP)-1, LRP-6, tropomyosin-related kinase A, integrins and heparan sulphate proteoglycans. Intracellular signalling and downstream events triggered by such interactions are reviewed. Finally, the relationships between CTGF and several anti-fibrotic factors, such as bone morphogenetic factor-4 (BMP4), BMP7, hepatocyte growth factor, CCN3 and Oncostatin M, are discussed. These may determine whether injured tissue heals or progresses to fibrosis.


Subject(s)
Connective Tissue Growth Factor/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Animals , Connective Tissue Growth Factor/genetics , Cytokines/metabolism , Disease Progression , Fibrosis , Humans , Inflammation Mediators/metabolism , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/therapy , Prognosis , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
Diabetes ; 61(9): 2280-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22586581

ABSTRACT

CCN2, a secreted profibrotic protein, is highly expressed in diabetic nephropathy (DN) and implicated in its pathogenesis; however, the actions of CCN2 in DN remain elusive. We previously demonstrated that CCN2 triggers signaling via tropomyosin receptor kinase A (TrkA). Trace expression of TrkA is found in normal kidneys, but its expression is elevated in several nephropathies; yet its role in DN is unexplored. In this study we show de novo expression of TrkA in human and murine DN. We go on to study the molecular mechanisms leading to TrkA activation and show that it involves hypoxia, as demonstrated by ischemia-reperfusion injury and in vitro experiments mimicking hypoxia, implicating hypoxia as a common pathway leading to disease. We also expose renal cells to hyperglycemia, which led to TrkA phosphorylation in mesangial cells, tubular epithelial cells, and podocytes but not in glomerular endothelial cells and renal fibroblasts. In addition, we report that hyperglycemia caused an induction of phosphorylated extracellular signal-related kinase 1/2 and Snail1 that was abrogated by silencing of TrkA or CCN2 using small interfering RNA. In conclusion, we provide novel evidence that TrkA is activated in diabetic kidneys and suggest that anti-TrkA therapy may prove beneficial in DN.


Subject(s)
Connective Tissue Growth Factor/physiology , Diabetic Nephropathies/etiology , Hyperglycemia/complications , Animals , Connective Tissue Growth Factor/genetics , Diabetic Nephropathies/physiopathology , Humans , Hyperglycemia/physiopathology , Hypoxia/complications , Hypoxia/physiopathology , Kidney/pathology , MAP Kinase Signaling System/physiology , Mesangial Cells/metabolism , Mice , Phosphorylation , RNA, Small Interfering/pharmacology , Receptor, trkA/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology , Snail Family Transcription Factors , Transcription Factors/biosynthesis
6.
Matrix Biol ; 30(7-8): 396-403, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21864682

ABSTRACT

Accumulation of type I collagen is a key event in renal interstitial fibrosis. As there is no effective treatment, understanding the site where collagen is transcribed and the factors driving it in response to disease in vivo is critical for designing future therapies. The present research investigated the transcriptional activity of the COL1A2 gene in a mouse model of progressive fibrosis induced by aristolochic acid (aristolochic acid nephropathy, AAN). To achieve this we genetically modified mice to express a reporter gene (LacZ) and CCN2 (connective tissue growth factor) under the transcriptional control of the COL1A2 promoter /enhancer sequences. Using these mice we asked where is collagen actively transcribed and secondly, what is the role of CCN2 in AAN. Here, we report that de-novo transcription of the COL1A2 gene occurred predominantly in damaged tubular epithelial cells during progressive interstitial fibrosis in vivo. The activation of COL1A2 was studied by detection of the reporter gene LacZ and COL1A2 mRNA in interstitial, glomerular, vascular, and tubular epithelial tissue from laser capture microscopy. We also demonstrated that LacZ-positive cells co-express E-Cadherin a marker of epithelial origin which is consistent with an epithelial phenotype which is capable of collagen expression during injury. There was no evidence of detachment of these cells from tubules to become myofibroblasts. Moreover, we showed that the transgenic mice show a modest enhancement of CCN2 expression; however fibrosis induced by AA is the same in transgenics and controls suggesting that CCN2, at this level of expression, is not sufficient to enhance fibrogenesis. Overall our study provides a better understanding into the expression patterns and roles of two major extracellular matrix proteins: type I collagen and CCN2.


Subject(s)
Aristolochic Acids/adverse effects , Collagen Type I/metabolism , Epithelial Cells/metabolism , Fibrosis/pathology , Kidney/pathology , Animals , Aristolochic Acids/administration & dosage , Aristolochic Acids/pharmacology , Biomarkers/analysis , Blood Urea Nitrogen , Cadherins/metabolism , Collagen Type I/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Disease Progression , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Fibrosis/etiology , Fibrosis/metabolism , Kidney Tubules/cytology , Laser Capture Microdissection , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Transcriptional Activation , Transgenes
7.
Int J Exp Pathol ; 92(3): 143-50, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21554437

ABSTRACT

Epithelial to mesenchymal transition (EMT) is a well established biological process in metazoan embryological development. Over the past 15 years, investigators have sought to establish whether EMT also occurs in renal epithelial cells, following kidney injury, and to show that the mesenchymal cells formed could give rise to myofibroblasts which populate the renal interstitium, causing fibrosis within it. There is no doubt that proximal tubular epithelial cells (PTECs) can undergo EMT in vitro in response to TGFß-1 and other inflammatory stimuli. Moreover, the results of experiments with animal models of renal fibrosis and examination of biopsies from patients with chronic kidney disease have lent support to the hypothesis that EMT occurs in vivo. This review discusses some of the key evidence underlying that idea and summarises recent advances in understanding the molecular mechanism underlying the process. Early experiments using mice which were genetically engineered to mark PTECs with the LacZ gene to trace their fate following kidney injury provided evidence supporting the occurrence of EMT. Recently, however, cell lineage tracking experiments using the red fluorescent protein (RFP) as a high-resolution marker for cells of renal epithelial origin did not replicate this result; the interstitial space following kidney injury was devoid of RFP expressing cells, leading the investigators to reject the renal EMT hypothesis.


Subject(s)
Cell Differentiation , Epithelial Cells/pathology , Kidney/pathology , Mesoderm/pathology , Animals , Cell Adhesion , Disease Models, Animal , Disease Progression , Fibrosis , Humans , Mice
8.
Mol Biol Cell ; 22(11): 1836-44, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21471005

ABSTRACT

Transforming growth factor-ß (TGF-ß) is an inducer of type I collagen, and uncontrolled collagen production leads to tissue scarring and organ failure. Here we hypothesize that uncovering a molecular mechanism that enables us to switch off type I collagen may prove beneficial in treating fibrosis. For the first time, to our knowledge, we provide evidence that CUX1 acts as a negative regulator of TGF-ß and potent inhibitor of type I collagen transcription. We show that CUX1, a CCAAT displacement protein, is associated with reduced expression of type I collagen both in vivo and in vitro. We show that enhancing the expression of CUX1 results in effective suppression of type I collagen. We demonstrate that the mechanism by which CUX1 suppresses type I collagen is through interfering with gene transcription. In addition, using an in vivo murine model of aristolochic acid (AA)-induced interstitial fibrosis and human AA nephropathy, we observe that CUX1 expression was significantly reduced in fibrotic tissue when compared to control samples. Moreover, silencing of CUX1 in fibroblasts from kidneys of patients with renal fibrosis resulted in increased type I collagen expression. Furthermore, the abnormal CUX1 expression was restored by addition of TGF-ß via the p38 mitogen-activated protein kinase pathway. Collectively, our study demonstrates that modifications of CUX1 expression lead to aberrant expression of type I collagen, which may provide a molecular basis for fibrogenesis.


Subject(s)
Collagen Type I/antagonists & inhibitors , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Aristolochic Acids , Cells, Cultured , Collagen Type I/genetics , Dose-Response Relationship, Drug , Feedback, Physiological , Fibrosis/drug therapy , Fibrosis/prevention & control , Gene Expression Regulation/drug effects , Humans , Kidney/pathology , Kidney Diseases/chemically induced , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Smad3 Protein/metabolism , Smad7 Protein , Transcription Factors , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism
9.
AIDS Patient Care STDS ; 24(12): 781-5, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21091231

ABSTRACT

Performing rapid HIV testing in nontraditional clinical settings such as dental clinics is a potential method for targeting high-risk individuals who may not otherwise access health care settings that offer HIV testing. In March 2008, Harlem Hospital Center, located in New York City, launched a counselor-based rapid HIV testing initiative in its on-site dental clinic. A full-time, trained counselor consented and tested patients as they waited for their appointments. HIV screening was performed using a whole-blood, finger-stick rapid HIV test. Through this initiative, 3864 HIV tests were performed from March 1, 2008 to December 31, 2009, representing 3565 unique individuals and 97.6% of dental patients approached for testing. Of those tested, the mean age was 38.5 years, with 47.1% female, 75.5% black, and 20.6% Hispanic. Self-reported HIV risk behaviors included 73.5% with recent unprotected heterosexual intercourse, 4.6% with recent or past injection drug use, and 2.6% who identified as men who have sex with men. Nineteen previously undiagnosed individuals (0.53%) were confirmed HIV positive. Of these individuals, mean age was 38.3 years with males representing 84.2%. Fifteen newly diagnosed patients (78.9%) were linked to care. Of those linked to care, median initial CD4 cell count was 317 cells/mm(3); 6 of these individuals (40%) had CD4 cell counts below 200 cells/mm(3). Our results demonstrate that a counselor-based rapid HIV testing program with linkage to specialized HIV care can be successfully integrated into the dental clinic setting.


Subject(s)
AIDS Serodiagnosis/methods , Delivery of Health Care, Integrated , Dental Clinics , HIV Infections/diagnosis , Hospitals, Urban , Adult , Counseling , Female , HIV Infections/prevention & control , HIV-1 , Humans , Male , New York City , Patient Acceptance of Health Care , Time Factors
10.
J Cell Commun Signal ; 3(2): 95-104, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19214781

ABSTRACT

Connective tissue growth factor (CTGF/CCN2) is a member of the CCN family of matricellular proteins. Its expression is induced by a number of factors including TGF-beta. It has been associated with fibrosis in various tissues including the kidney. Diabetic nephropathy (DN) develops in about 30% of patients with diabetes and is characterized by thickening of renal basement membranes, fibrosis in the glomerulus (glomerulosclerosis), tubular atrophy and interstitial fibrosis, all of which compromise kidney function. This review examines changes in CTGF expression in the kidney in DN, the effects they have on glomerular mesangial and podocyte cells and the tubulointerstitium, and how these contribute to driving fibrotic changes in the disease. CTGF can bind to several other growth factors modifying their function. CTGF is also able to interact with receptors on cells, including integrins, tyrosine receptor kinase A (TrkA), low density lipoprotein receptor-related protein (LRP) and heparan sulphate proteoglycans. These interactions, the intracellular signalling pathways they activate, and the cellular responses evoked are reviewed. CTGF also induces the expression of chemokines which themselves have pharmacological actions on cells. CTGF may prompt some responses by acting through several different mechanisms, possibly simultaneously. For example, CTGF is often described as an effector of TGF-beta. It can promote TGF-beta signalling by binding directly to the growth factor, promoting its interaction with the TGF-beta receptor; by triggering intracellular signalling on binding the TrkA receptor, which leads to the transcriptional repression of Smad7, an inhibitor of the TGF-beta signalling pathway; and by binding to BMP-7 whose own signalling pathway opposing TGF-beta is inhibited, leading to enhanced TGF-beta signalling.

11.
Biochem J ; 406(1): 131-8, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17489738

ABSTRACT

Activated mesangial cells are thought to play a pivotal role in the development of kidney fibrosis under chronic pathological conditions, including DN (diabetic nephropathy). Their prolonged survival may enhance the development of the disease since they express increased amounts of growth factors and extracellular matrix proteins. CTGF (connective tissue growth factor) is one of the growth factors produced by activated mesangial cells and is reported to play a key role in the pathogenesis of DN. Previous studies have shown that addition of exogenous CTGF to HMCs (human mesangial cells) rapidly activates ERK1/2 (extracellular-signal-regulated kinase 1/2) MAPK (mitogen-activated protein kinase) and JNK (c-Jun N-terminal kinase) MAPK, but not the p38 MAPK, despite the activation of the upstream kinases, MKK3/6 (MAPK kinase 3/6). The aim of the present study was to investigate whether the lack of phosphorylated p38 MAPK by CTGF has an anti-apoptotic effect on activated HMCs. We show that in HMC CTGF induces the rapid transcriptional activation and synthesis of MKP-1 (MAPK phosphatase-1), a dual specificity phosphatase that dephosphorylates p38 MAPK. This in turn prevents the anti-apoptotic protein, Bcl-2, from being phosphorylated and losing its function, leading to the survival of the cells. Knockout of MKP-1 protein in mesangial cells treated with CTGF, using siRNA (small interfering RNA) or antisense oligonucleotides, allows p38 MAPK activation and induces mesangial cell death.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Mesangial Cells/cytology , Mesangial Cells/enzymology , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , Adult , Cell Cycle Proteins/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Connective Tissue Growth Factor , Dual Specificity Phosphatase 1 , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Immediate-Early Proteins/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mesangial Cells/drug effects , Models, Biological , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Nephron Exp Nephrol ; 104(4): e129-34, 2006.
Article in English | MEDLINE | ID: mdl-16902316

ABSTRACT

In the adult kidney, the cellular phenotypes are maintained by a strict balance of growth factors. Epithelial-to-mesenchymal transition (EMT) is a program whereby injured epithelial cells that function as ion and fluid transporters become matrix remodelling mesenchymal cells. This process requires either transcriptional repression of genes that maintain the epithelial phenotype and transcriptional activation, or relieved repression of genes needed for functional myofibroblasts. The transcriptional regulators are controlled by several integrated signalling pathways which are triggered by growth factors. Emerging evidence indicates that the growth factors TGFbeta/CTGF and BMP-7/HGF are the main determinants that maintain the two cellular phenotypes. Both TGFbeta and BMP-7 counteract the activity of each other by cross-inducing their respective inhibitory Smads. Both growth factors may also induce the expression of other factors that can change the cellular environment and enhance their function. Chronic kidney diseases (regardless of the aetiology of the disease) are associated with increased TGFbeta and CTGF expression levels which, in turn, have an inverse effect on the activity level of BMP-7 and HGF, leading to an EMT of injured tubular epithelial cells and a progression of the disease. A detailed understanding of the complex interrelationship between these growth factors may lead to the development of novel drugs.


Subject(s)
Epithelial Cells/physiology , Intercellular Signaling Peptides and Proteins/physiology , Mesoderm/physiology , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/physiology , Cell Differentiation/drug effects , Connective Tissue Growth Factor , Hepatocyte Growth Factor/physiology , Immediate-Early Proteins/physiology , Smad6 Protein/physiology , Smad7 Protein/physiology , Transforming Growth Factor beta/physiology
13.
Am J Nephrol ; 26(4): 381-7, 2006.
Article in English | MEDLINE | ID: mdl-16873994

ABSTRACT

BACKGROUND/AIM: Recent studies have suggested that statins may play a role in the protection against renal failure which is independent of cholesterol reduction. Activation of RhoGTPases is a key step in renal tubular cells' epithelial-to-mesenchymal transition (EMT) which contributes to renal interstitial fibrosis. We hypothesized that statins could act by inhibiting the synthesis of the isoprenoids, such as geranylgeranyl pyrophosphate, which is essential for membrane attachment and biological activity of RhoGTPases, RhoA and Rac1. METHODS: Human proximal tubular epithelial cells (HK2) were used to examine the inhibitory effect of statins on EMT induced with medium conditioned by activated peripheral blood mononuclear cells. RESULTS: Our study demonstrates that the statins lovastatin, simvastatin, and pravastatin inhibit HK2 cells to undergo EMT. Inhibition of EMT in HK2 cells with these statins resulted in a reduction of RhoA and Rac1 activation in both the cytoplasmic and membrane-bound forms, in preservation of the expression of the epithelial cell markers E-cadherin and cytokeratin-19, and in a decrease in Fn-EDA expression, a marker for the myofibroblast phenotype. The decreased levels of activated RhoA and Rac1 in both the cytoplasmic and membrane fractions of the cells were reversed by geranylgeranyl pyrophosphate and mevalonate, and thus attributable to the inhibition of isoprenylation of RhoGTPases by statins. CONCLUSION: This phenomenon could explain the beneficial effect of statins on EMT and on renal fibrosis prevention.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Line , Dose-Response Relationship, Drug , Humans
14.
Arthritis Rheum ; 52(11): 3479-91, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16255039

ABSTRACT

OBJECTIVE: To use human cartilage samples and a mouse model of osteoarthritis (OA) to determine whether extracellular superoxide dismutase (EC-SOD) is a constituent of cartilage and to evaluate whether there is a relationship between EC-SOD deficiency and OA. METHODS: Samples of human cartilage were obtained from femoral heads at the time of joint replacement surgery for OA or femoral neck fracture. Samples of mouse tibial cartilage obtained from STR/ort mice and CBA control mice were compared at 5, 15, and 35 weeks of age. EC-SOD was measured by enzyme-linked immunosorbent assay, Western blotting, and immunohistochemistry techniques. Real-time quantitative reverse transcription-polymerase chain reaction was used to measure messenger RNA for EC-SOD and for endothelial cell, neuronal, and inducible nitric oxide synthases. Nitrotyrosine formation was assayed by Western blotting in mouse cartilage and by fluorescence immunohistochemistry in human cartilage. RESULTS: Human articular cartilage contained large amounts of EC-SOD (mean +/- SEM 18.8 +/- 3.8 ng/gm wet weight of cartilage). Cartilage from patients with OA had an approximately 4-fold lower level of EC-SOD compared with cartilage from patients with hip fracture. Young STR/ort mice had decreased levels of EC-SOD in tibial cartilage before histologic evidence of disease occurred, as well as significantly more nitrotyrosine formation at all ages studied. CONCLUSION: EC-SOD, the major scavenger of reactive oxygen species in extracellular spaces, is decreased in humans with OA and in an animal model of OA. Our findings suggest that inadequate control of reactive oxygen species plays a role in the pathophysiology of OA.


Subject(s)
Cartilage, Articular/enzymology , Free Radical Scavengers/metabolism , Osteoarthritis, Hip/enzymology , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Adult , Aged , Animals , Cartilage, Articular/pathology , Disease Models, Animal , Female , Femur Head , Hindlimb , Humans , Male , Mice , Mice, Inbred CBA , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Osteoarthritis, Hip/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/genetics , Tibia , Tyrosine/analogs & derivatives , Tyrosine/metabolism
15.
Respir Res ; 6: 56, 2005 Jun 09.
Article in English | MEDLINE | ID: mdl-15946381

ABSTRACT

BACKGROUND: Fibroblastic foci are characteristic features in lung parenchyma of patients with idiopathic pulmonary fibrosis (IPF). They comprise aggregates of mesenchymal cells which underlie sites of unresolved epithelial injury and are associated with progression of fibrosis. However, the cellular origins of these mesenchymal phenotypes remain unclear. We examined whether the potent fibrogenic cytokine TGF-beta1 could induce epithelial mesenchymal transition (EMT) in the human alveolar epithelial cell line, A549, and investigated the signaling pathway of TGF-beta1-mediated EMT. METHODS: A549 cells were examined for evidence of EMT after treatment with TGF-beta1. EMT was assessed by: morphology under phase-contrast microscopy; Western analysis of cell lysates for expression of mesenchymal phenotypic markers including fibronectin EDA (Fn-EDA), and expression of epithelial phenotypic markers including E-cadherin (E-cad). Markers of fibrogenesis, including collagens and connective tissue growth factor (CTGF) were also evaluated by measuring mRNA level using RT-PCR, and protein by immunofluorescence or Western blotting. Signaling pathways for EMT were characterized by Western analysis of cell lysates using monoclonal antibodies to detect phosphorylated Erk1/2 and Smad2 after TGF-beta1 treatment in the presence or absence of MEK inhibitors. The role of Smad2 in TGF-beta1-mediated EMT was investigated using siRNA. RESULTS: The data showed that TGF-beta1, but not TNF-alpha or IL-1beta, induced A549 cells with an alveolar epithelial type II cell phenotype to undergo EMT in a time-and concentration-dependent manner. The process of EMT was accompanied by morphological alteration and expression of the fibroblast phenotypic markers Fn-EDA and vimentin, concomitant with a downregulation of the epithelial phenotype marker E-cad. Furthermore, cells that had undergone EMT showed enhanced expression of markers of fibrogenesis including collagens type I and III and CTGF. MMP-2 expression was also evidenced. TGF-beta1-induced EMT occurred through phosphorylation of Smad2 and was inhibited by Smad2 gene silencing; MEK inhibitors failed to attenuate either EMT-associated Smad2 phosphorylation or the observed phenotypic changes. CONCLUSION: Our study shows that TGF-beta1 induces A549 alveolar epithelial cells to undergo EMT via Smad2 activation. Our data support the concept of EMT in lung epithelial cells, and suggest the need for further studies to investigate the phenomenon.


Subject(s)
Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Transforming Growth Factor beta/administration & dosage , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibronectins/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Pulmonary Alveoli/drug effects , Respiratory Mucosa/drug effects , Transforming Growth Factor beta1
16.
Exp Cell Res ; 307(2): 305-14, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15950619

ABSTRACT

Transforming growth factor-beta (TGFbeta) drives fibrosis in diseases such as diabetic nephropathy (DN). Connective tissue growth factor (CTGF; CCN2) has also been implicated in this, but the molecular mechanism is unknown. We show that CTGF enhances the TGFbeta/Smad signaling pathway by transcriptional suppression of Smad 7 following rapid and sustained induction of the transcription factor TIEG-1. Smad 7 is a known antagonist of TGFbeta signaling and TIEG-1 is a known repressor of Smad 7 transcription. CTGF enhanced TGFbeta-induced phosphorylation and nuclear translocation of Smad 2 and Smad 3 in mesangial cells. Antisense oligonucleotides directed against TIEG-1 prevented CTGF-induced downregulation of Smad 7. CTGF enhanced TGFbeta-stimulated transcription of the SBE4-Luc reporter gene and this was markedly reduced by TIEG-1 antisense oligonucleotides. Expression of the TGFbeta-responsive genes PAI-1 and Col III over 48 h was maximally stimulated by TGFbeta+CTGF compared to TGFbeta alone, while CTGF alone had no significant effect. TGFbeta-stimulated expression of these genes was markedly reduced by both CTGF and TIEG-1 antisense oligonucleotides, consistent with the endogenous induction of CTGF by TGFbeta. We propose that under pathological conditions, where CTGF expression is elevated, CTGF blocks the negative feedback loop provided by Smad 7, allowing continued activation of the TGFbeta signaling pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Glomerular Mesangium/physiology , Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Signal Transduction/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/physiology , Cell Cycle Proteins/genetics , Cells, Cultured , Collagen Type III/genetics , Connective Tissue Growth Factor , Cyclin-Dependent Kinase Inhibitor p15 , DNA-Binding Proteins/genetics , Early Growth Response Transcription Factors , Gene Expression/drug effects , Glomerular Mesangium/cytology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Kruppel-Like Transcription Factors , Oligoribonucleotides, Antisense/pharmacology , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/genetics , Recombinant Proteins/pharmacology , Smad Proteins , Smad2 Protein , Smad3 Protein , Smad7 Protein , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins/genetics
17.
J Am Soc Nephrol ; 16(7): 1977-84, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15901767

ABSTRACT

ESRD is characterized by an interstitial infiltrate of inflammatory cells in association with tubular atrophy, epithelial mesenchymal transdifferentiation (EMT), and interstitial fibrosis. Human proximal tubular epithelial cells (HK2 cells) undergo EMT in response to activated PBMC conditioned medium (aPBMC-CM), showing acquisition of a fibroblastoid morphology, increased fibronectin-EDA (EDA) expression, loss of junctional E-cadherin localization, and cytokeratin 19 (K19) expression. The signaling pathway(s) that regulates EMT in response to aPBMC-CM is not well understood. This study shows that aPBMC-CM induces a rapid activation of RhoA, Rac1, and Cdc42 activity in HK2 cells from 15 min to 48 h. Moreover, infection with adenovirus expressing constitutively active RhoA, Rac1, and Cdc42 significantly increased the expression of EDA and downregulated expression of E-cadherin and K19. Dominant negative RhoA expression suppressed aPBMC-CM-induced upregulation of EDA but did not restore the expression of E-cadherin and K19. Constitutively active RhoA activated the Rho kinase and its downstream effectors, whereas constitutively active Rac1 and Cdc42 activated the P21-activated protein kinase in epithelial cells. In further experiments, HK2 cells were treated with toxin B, exoenzyme C3, Y-27632, and HA1077. These strategies, inhibiting the Rho/Rho kinase pathway, as well as the Rac1/Cdc42/P21-activated protein kinase pathway, blocked transdifferentiation of HK2 cells in response to aPBMC-CM. To conclude, these results indicate that aPBMC-CM activates RhoA, Rac1, and Cdc42 and their downstream effectors, leading to HK2 cells undergoing transdifferentiation. Therefore, activation of small RhoGTPases is a key step in the mechanism of EMT and likely to be a contributor to tubulointerstitial fibrosis.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/physiology , Kidney Tubules, Proximal/physiology , Mesoderm/physiology , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Cells, Cultured , Culture Media, Conditioned , Fibrosis , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/pathology , Leukocytes, Mononuclear , Signal Transduction , cdc42 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/genetics
18.
J Am Soc Nephrol ; 16(2): 340-51, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15601748

ABSTRACT

Connective tissue growth factor (CTGF) is implicated as a factor promoting tissue fibrosis in several disorders, including diabetic nephropathy. However, the molecular mechanism(s) by which it functions is not known. CTGF rapidly activates several intracellular signaling molecules in human mesangial cells (HMC), including extracellular signal-related kinase 1/2, Jun NH(2)-terminal kinase, protein kinase B, CaMK II, protein kinase Calpha, and protein kinase Cdelta, suggesting that it functions via a signaling receptor. Treating HMC with CTGF stimulated tyrosine phosphorylation of proteins 75 to 80 and 140 to 180 kD within 10 min, and Western blot analysis of anti-phosphotyrosine immunoprecipitates identified the neurotrophin receptor TrkA (molecular weight approximately 140 kD). Cross-linking rCTGF to cell surface proteins with 3,3'-dithiobis(sulfosuccinimidylpropionate) revealed that complexes formed with TrkA and with the general neurotrophin co-receptor p75(NTR). rCTGF stimulated phosphorylation of TrkA (tyr 490, 674/675). K252a, a known selective inhibitor of Trk, blocked this phosphorylation, CTGF-induced activation of signaling proteins, and CTGF-dependent induction of the transcription factor TGF-beta-inducible early gene in HMC. It is concluded that TrkA serves as a tyrosine kinase receptor for CTGF.


Subject(s)
Glomerular Mesangium/cytology , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Receptor, trkA/drug effects , Receptor, trkA/physiology , Signal Transduction/physiology , Adult , Base Sequence , Binding Sites , Blotting, Western , Cells, Cultured , Connective Tissue Growth Factor , Cross-Linking Reagents , Fluorescent Antibody Technique , Humans , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
19.
Curr Opin Nephrol Hypertens ; 13(1): 53-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15090860

ABSTRACT

PURPOSE OF REVIEW: Connective tissue growth factor (CCN2) has recently received much attention as a possible key determinant of progressive renal fibrosis. However, the mechanism(s) by which this growth factor functions is not known. The purpose of this review is to summarize and discuss the recent findings regarding the possible mechanisms involved. RECENT FINDINGS: Emerging evidence from in-vitro studies of renal cells indicates that connective tissue growth factor is a crucial mediator for transforming growth factor-beta-induced cellular dysfunction, manifest by increased cellular hypertrophy, synthesis of extracellular matrix proteins and their deposition and assembly around the cells. Indeed, recent evidence suggests that the interrelationship between connective tissue growth factor and transforming growth factor-beta is stronger than first thought. While transforming growth factor-beta induces the expression of connective tissue growth factor, the latter plays a key role in both bioactivation of latent transforming growth factor-beta and the promotion of its Smad signalling activity. SUMMARY: Connective tissue growth factor is clearly implicated in the pathogenesis of progressive renal disease. Although there is much to learn about the production, function, and mechanism of action of connective tissue growth factor, some progress has been made in understanding the molecular basis of its relationship with transforming growth factor-beta. Elucidating the signal transduction pathways activated by connective tissue growth factor will also definitely help to clarify other actions of connective tissue growth factor which may be independent of transforming growth factor-beta. Because of the inflammatory and immunosuppressive properties of transforming growth factor-beta, connective tissue growth factor seems to be an attractive alternative therapeutic target for combating renal fibrosis.


Subject(s)
Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Kidney Diseases/physiopathology , Animals , Connective Tissue Growth Factor , Humans , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Kidney/metabolism , Transforming Growth Factor beta/metabolism
20.
J Am Soc Nephrol ; 15(1): 21-32, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14694154

ABSTRACT

Interactions between inflammatory infiltrates and resident tubular epithelial cells may play important roles in the development of tubulointerstitial fibrosis, by promoting epithelial cell-myofibroblast transdifferentiation (EMT). Human proximal tubular epithelial cells transdifferentiated to myofibroblasts after treatment with activated PBMC conditioned medium. mRNA and protein levels for alpha-smooth muscle actin, collagen I, and fibronectin EDA(+) (markers for the myofibroblastic phenotype) were increased, whereas those for E-cadherin and cytokeratin 19 (markers for the epithelial phenotype) were decreased. cDNA microarray analysis was used to identify other changes in gene expression that might point to novel molecular mechanisms driving EMT. Of 1176 array genes, 61 demonstrated at least a twofold change at at least two consecutive time points, of the five time points examined (0.5, 4, 8, 16, and 48 h). Of these genes, 59% were upregulated and 41% were downregulated. The array indicated upregulation of expression of the oncostatin M (OSM)-specific receptor beta subunit from 4 to 48 h after exposure of kidney epithelial cells to activated PBMC conditioned medium, which contained high levels of OSM. In additional experiments, it was demonstrated that OSM induced EMT. OSM activated the Jak/Stat signaling pathway in epithelial cells, and a specific inhibitor of Jak2 blocked both its phosphorylation after exposure to OSM and the induction of alpha-actin and loss of cytokeratin 19 expression. Therefore, OSM is a novel inducer of EMT and is likely to be one of several cytokines produced by inflammatory infiltrates that contribute to this and subsequent tubulointerstitial fibrosis.


Subject(s)
Cell Differentiation , Cytokines/physiology , Epithelial Cells/cytology , Fibroblasts/cytology , Leukocytes, Mononuclear/metabolism , Myoblasts/cytology , Peptides/physiology , Signal Transduction , Cell Differentiation/genetics , Cells, Cultured , Gene Expression , Humans , Kidney Tubules/cytology , Oligonucleotide Array Sequence Analysis , Oncostatin M , Urothelium/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...