Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Eur Urol Focus ; 9(6): 983-991, 2023 11.
Article in English | MEDLINE | ID: mdl-37105783

ABSTRACT

BACKGROUND: Molecular signatures in prostate cancer (PCa) tissue can provide useful prognostic information to improve the understanding of a patient's risk of harbouring aggressive disease. OBJECTIVE: To develop and validate a gene signature that adds independent prognostic information to clinical parameters for better treatment decisions and patient management. DESIGN, SETTING, AND PARTICIPANTS: Expression of 14 genes was evaluated in radical prostatectomy (RP) tissue from an Irish cohort of PCa patients (n = 426). A six-gene molecular risk score (MRS) was identified with strong prognostic performance to predict adverse pathology (AP) at RP or biochemical recurrence (BCR). The MRS was combined with the Cancer of the Prostate Risk Assessment (CAPRA) score, to create a molecular and clinical risk score (MCRS), and validated in a Swedish cohort (n = 203). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The primary AP outcome was assessed by the likelihood ratio statistics and area under the receiver operating characteristics curves (AUC) from logistic regression models. The secondary time to BCR outcome was assessed by likelihood ratio statistics and C-indexes from Cox proportional hazard regression models. RESULTS AND LIMITATIONS: The six-gene signature was significantly (p < 0.0001) prognostic and added significant prognostic value to clinicopathological features for AP and BCR outcomes. For both outcomes, both the MRS and the MCRS increased the AUC/C-index when added to European Association of Urology (EAU) and CAPRA scores. Limitations include the retrospective nature of this study. CONCLUSIONS: The six-gene signature has strong performance for the prediction of AP and BCR in an independent clinical validation study. MCRS improves prognostic evaluation and can optimise patient management after RP. PATIENT SUMMARY: We found that the expression panel of six genes can help predict whether a patient is likely to have a disease recurrence after radical prostatectomy surgery.


Subject(s)
Neoplasm Recurrence, Local , Prostatic Neoplasms , Male , Humans , Retrospective Studies , Risk Assessment/methods , Neoplasm Recurrence, Local/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/surgery , Prostatic Neoplasms/pathology , Prostate/pathology
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3095-3104, 2017 12.
Article in English | MEDLINE | ID: mdl-28923783

ABSTRACT

Bone morphogenetic protein-7 (BMP-7) counteracts pro-fibrotic effects of TGFß1 in cultured renal cells and protects from fibrosis in acute and chronic renal injury models. Using the unilateral ureteral obstruction (UUO) model of chronic renal fibrosis, we investigated the effect of exogenous-rhBMP-7 on pro-fibrotic signaling pathways mediated by TGFß1 and hypoxia. Mice undergoing UUO were treated with vehicle or rhBMP-7 (300µg/kg i.p.) every other day for eight days and kidneys analysed for markers of fibrosis and SMAD, MAPK, and PI3K signaling. In the kidney, collecting duct and tubular epithelial cells respond to BMP-7 via activation of SMAD1/5/8. Phosphorylation of SMAD1/5/8 was reduced in UUO kidneys from vehicle-treated animals yet maintained in UUO kidneys from BMP-7-treated animals, confirming renal bioactivity of exogenous rhBMP-7. BMP-7 inhibited Collagen Iα1 and Collagen IIIα1 gene expression and Collagen I protein accumulation, while increasing expression of Collagen IVα1 in UUO kidneys. Activation of SMAD2, SMAD3, ERK, p38 and PI3K/Akt signaling occurred during fibrogenesis and BMP-7 significantly attenuated SMAD3 and Akt signaling in vivo. Analysis of renal collecting duct (mIMCD) and tubular epithelial (HK-2) cells stimulated with TGFß1 or hypoxia (1% oxygen) to activate Akt provided further evidence that BMP-7 specifically inhibited PI3K/Akt signaling. PTEN is a negative regulator of PI3K and BMP-7 increased PTEN expression in vivo and in vitro. These data demonstrate an important mechanism by which BMP-7 orchestrates renal protection through Akt inhibition and highlights Akt inhibitors as anti-fibrotic therapeutics.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology , Fibrosis/prevention & control , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Cell Hypoxia/physiology , Cell Line , Collagen/metabolism , Disease Models, Animal , Fibrosis/enzymology , Fibrosis/pathology , Kidney Diseases/enzymology , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Kidney Tubules/drug effects , Kidney Tubules/pathology , Mice , PTEN Phosphohydrolase/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
3.
Development ; 143(10): 1742-52, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27190038

ABSTRACT

Hypoxia is encountered in either pathological or physiological conditions, the latter of which is seen in amniote embryos prior to the commencement of a functional blood circulation. During the hypoxic stage, a large number of neural crest cells arise from the head neural tube by epithelial-to-mesenchymal transition (EMT). As EMT-like cancer dissemination can be promoted by hypoxia, we investigated whether hypoxia contributes to embryonic EMT. Using chick embryos, we show that the hypoxic cellular response, mediated by hypoxia-inducible factor (HIF)-1α, is required to produce a sufficient number of neural crest cells. Among the genes that are involved in neural crest cell development, some genes are more sensitive to hypoxia than others, demonstrating that the effect of hypoxia is gene specific. Once blood circulation becomes fully functional, the embryonic head no longer produces neural crest cells in vivo, despite the capability to do so in a hypoxia-mimicking condition in vitro, suggesting that the oxygen supply helps to stop emigration of neural crest cells in the head. These results highlight the importance of hypoxia in normal embryonic development.


Subject(s)
Head/embryology , Neural Crest/cytology , Amino Acids, Dicarboxylic/pharmacology , Animals , Avian Proteins/metabolism , Biomarkers/metabolism , Cell Hypoxia/drug effects , Cell Lineage/drug effects , Cell Movement/drug effects , Cell Plasticity/drug effects , Cells, Cultured , Chick Embryo , Epithelial-Mesenchymal Transition/drug effects , Hyperoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neural Tube/drug effects , Neural Tube/metabolism , Staining and Labeling
4.
Am J Physiol Renal Physiol ; 311(1): F35-45, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27122540

ABSTRACT

Diabetic nephropathy is the most common microvascular complication of diabetes mellitus, manifesting as mesangial expansion, glomerular basement membrane thickening, glomerular sclerosis, and progressive tubulointerstitial fibrosis leading to end-stage renal disease. Here we describe the functional characterization of Wnt6, whose expression is progressively lost in diabetic nephropathy and animal models of acute tubular injury and renal fibrosis. We have shown prominent Wnt6 and frizzled 7 (FzD7) expression in the mesonephros of the developing mouse kidney, suggesting a role for Wnt6 in epithelialization. Importantly, TCF/Lef reporter activity is also prominent in the mesonephros. Analysis of Wnt family members in human renal biopsies identified differential expression of Wnt6, correlating with severity of the disease. In animal models of tubular injury and fibrosis, loss of Wnt6 was evident. Wnt6 signals through the canonical pathway in renal epithelial cells as evidenced by increased phosphorylation of GSK3ß (Ser9), nuclear accumulation of ß-catenin and increased TCF/Lef transcriptional activity. FzD7 was identified as a putative receptor of Wnt6. In vitro Wnt6 expression leads to de novo tubulogenesis in renal epithelial cells grown in three-dimensional culture. Importantly, Wnt6 rescued epithelial cell dedifferentiation in response to transforming growth factor-ß (TGF-ß); Wnt6 reversed TGF-ß-mediated increases in vimentin and loss of epithelial phenotype. Wnt6 inhibited TGF-ß-mediated p65-NF-κB nuclear translocation, highlighting cross talk between the two pathways. The critical role of NF-κB in the regulation of vimentin expression was confirmed in both p65(-/-) and IKKα/ß(-/-) embryonic fibroblasts. We propose that Wnt6 is involved in epithelialization and loss of Wnt6 expression contributes to the pathogenesis of renal fibrosis.


Subject(s)
Cell Differentiation/genetics , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Wnt Proteins/genetics , Wnt Proteins/physiology , Animals , Epithelial Cells/pathology , Female , Fibrosis , Frizzled Receptors , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , I-kappa B Proteins/genetics , Kidney/embryology , Kidney Diseases/chemically induced , Kidney Tubules/growth & development , Mice , Mice, Knockout , Phosphorylation , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vimentin/biosynthesis
6.
Biochim Biophys Acta ; 1833(8): 1969-78, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23567938

ABSTRACT

TGF-ß1 is a prototypic profibrotic cytokine and major driver of fibrosis in the kidney and other organs. Induced in high glucose-1 (IHG-1) is a mitochondrial protein which we have recently reported to be associated with renal disease. IHG-1 amplifies responses to TGF-ß1 and regulates mitochondrial biogenesis by stabilising the transcriptional co-activator peroxisome proliferator-activated receptor gamma coactivator-1-alpha. Here we report that the mitochondrial localisation of IHG-1 is pivotal in the amplification of TGF-ß1 signalling. We demonstrate that IHG-1 expression is associated with repression of the endogenous TGF-ß1 inhibitor Smad7. Intriguingly, expression of a non-mitochondrial deletion mutant of IHG-1 (Δmts-IHG-1) repressed TGF-ß1 fibrotic signalling in renal epithelial cells. In cells expressing Δmts-IHG-1 fibrotic responses including CCN2/connective tissue growth factor, fibronectin and jagged-1 expression were reduced following stimulation with TGF-ß1. Δmts-IHG-1 modulation of TGF-ß1 signalling was associated with increased Smad7 protein expression. Δmts-IHG-1 modulated TGF-ß1 activity by increasing Smad7 protein expression as it failed to inhibit TGF-ß1 transcriptional responses when endogenous Smad7 expression was knocked down. These data indicate that mitochondria modulate TGF-ß1 signal transduction and that IHG-1 is a key player in this modulation.


Subject(s)
Fibrosis/metabolism , Mitochondria/genetics , Proteins/metabolism , Smad7 Protein/biosynthesis , Transforming Growth Factor beta1/metabolism , Amino Acid Sequence , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Epithelial Cells/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Fibrosis/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Molecular Sequence Data , Phosphorylation , Proteins/genetics , Serrate-Jagged Proteins , Signal Transduction , Smad7 Protein/genetics , Smad7 Protein/metabolism , Transforming Growth Factor beta1/genetics
7.
J Am Soc Nephrol ; 24(4): 627-37, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23520204

ABSTRACT

Lipoxins, which are endogenously produced lipid mediators, promote the resolution of inflammation, and may inhibit fibrosis, suggesting a possible role in modulating renal disease. Here, lipoxin A4 (LXA4) attenuated TGF-ß1-induced expression of fibronectin, N-cadherin, thrombospondin, and the notch ligand jagged-1 in cultured human proximal tubular epithelial (HK-2) cells through a mechanism involving upregulation of the microRNA let-7c. Conversely, TGF-ß1 suppressed expression of let-7c. In cells pretreated with LXA4, upregulation of let-7c persisted despite subsequent stimulation with TGF-ß1. In the unilateral ureteral obstruction model of renal fibrosis, let-7c upregulation was induced by administering an LXA4 analog. Bioinformatic analysis suggested that targets of let-7c include several members of the TGF-ß1 signaling pathway, including the TGF-ß receptor type 1. Consistent with this, LXA4-induced upregulation of let-7c inhibited both the expression of TGF-ß receptor type 1 and the response to TGF-ß1. Overexpression of let-7c mimicked the antifibrotic effects of LXA4 in renal epithelia; conversely, anti-miR directed against let-7c attenuated the effects of LXA4. Finally, we observed that several let-7c target genes were upregulated in fibrotic human renal biopsies compared with controls. In conclusion, these results suggest that LXA4-mediated upregulation of let-7c suppresses TGF-ß1-induced fibrosis and that expression of let-7c targets is dysregulated in human renal fibrosis.


Subject(s)
Kidney/drug effects , Kidney/pathology , Lipoxins/pharmacology , MicroRNAs/metabolism , Transforming Growth Factor beta1/metabolism , Cadherins/drug effects , Cadherins/metabolism , Cells, Cultured , Fibronectins/drug effects , Fibronectins/metabolism , Fibrosis , Humans , Kidney/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , MicroRNAs/drug effects , Receptor, Notch1/drug effects , Receptor, Notch1/metabolism , Signal Transduction , Thrombospondins/drug effects , Thrombospondins/metabolism , Transforming Growth Factor beta1/drug effects
8.
FASEB J ; 26(10): 4287-94, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22700871

ABSTRACT

Aging and adiposity are associated with chronic low-grade inflammation, which underlies the development of obesity-associated complications, including type 2 diabetes mellitus (T2DM). The mechanisms underlying adipose inflammation may include macrophage infiltration and activation, which, in turn, affect insulin sensitivity of adipocytes. There is a growing appreciation that specific lipid mediators (including lipoxins, resolvins, and protectins) can promote the resolution of inflammation. Here, we investigated the effect of lipoxin A4 (LXA4), the predominant endogenously generated lipoxin, on adipose tissue inflammation. Using adipose tissue explants from perigonadal depots of aging female C57BL/6J mice (Animalia, Chordata, Mus musculus) as a model of age-associated adipose inflammation, we report that LXA4 (1 nM) attenuates adipose inflammation, decreasing IL-6 and increasing IL-10 expression (P<0.05). The altered cytokine milieu correlated with increased GLUT-4 and IRS-1 expression, suggesting improved insulin sensitivity. Further investigations revealed the ability of LXA4 to rescue macrophage-induced desensitization to insulin-stimulated signaling and glucose uptake in cultured adipocytes, using vehicle-stimulated cells as controls. This was associated with preservation of Akt activation and reduced secretion of proinflammatory cytokines, including TNF-α. We therefore propose that LXA4 may represent a potentially useful and novel therapeutic strategy to subvert adipose inflammation and insulin resistance, key components of T2DM.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/immunology , Inflammation/drug therapy , Lipoxins/therapeutic use , Adipose Tissue/metabolism , Animals , Female , Glucose Transporter Type 4/metabolism , Inflammation/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Interleukin-10/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
FASEB J ; 25(9): 2967-79, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21628447

ABSTRACT

Unresolved inflammation underlies the development of fibrosis and organ failure. Here, we investigate the potential of the proresolving eicosanoid lipoxinA4 (LXA4) and its synthetic analog benzo-LXA4 to prophylactically modulate fibrotic and inflammatory responses in a model of early renal fibrosis, unilateral ureteric obstruction (UUO). Male Wistar rats (Animalia, Chordata, Rattus norvegicus) were injected intravenously with vehicle (0.1% ethanol), LXA4 (45 µg/250-g rat), or benzo-LXA4 (15 µg/250-g rat) 15 min prior to surgery and sacrificed 3 d postligation. Renal gene and protein expression, collagen deposition, macrophage infiltration, and apoptosis were analyzed using manipulated kidneys from sham operations as control. Lipoxins (LXs) attenuated collagen deposition and renal apoptosis (P<0.05) and shifted the inflammatory milieu toward resolution, inhibiting TNF-α and IFN-γ expression, while stimulating proresolving IL-10. LXs attenuated UUO-induced activation of MAP kinases, Akt, and Smads (P<0.05) in injured kidneys. We explored whether the underlying mechanism reflected LX-induced modulation of fibroblast activation. Using cultured rat renal NRK-49F fibroblasts, we report that LXA4 (1 nM) inhibits TGF-ß1 (10 ng/ml)-induced activation of Smad2 and MAP-kinases (P<0.05), and furthermore, LXA4 reduced TGF-ß1-stimulated PAI-1 luciferase activation (P<0.05) relative to vehicle-stimulated cells. We propose that LXs may represent a potentially useful and novel therapeutic strategy for consideration in the context of renal fibrosis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Kidney Diseases/drug therapy , Kidney/pathology , Lipoxins/therapeutic use , Animals , Apoptosis/drug effects , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Fibrosis/drug therapy , Fibrosis/prevention & control , Gene Expression Regulation , Kidney/drug effects , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Ligation , Lipoxins/chemistry , Male , Rats , Rats, Wistar , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
10.
Am J Physiol Renal Physiol ; 295(4): F1023-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18667485

ABSTRACT

Chronic hypoxia accelerates renal fibrosis. The chief mediator of the hypoxic response is hypoxia-inducible factor 1 (HIF-1) and its oxygen-sensitive component HIF-1alpha. HIF-1 regulates a wide variety of genes, some of which are closely associated with tissue fibrosis. To determine the specific role of HIF-1 in renal fibrosis, we generated a knockout mouse in which tubular epithelial expression of von Hippel-Lindau tumor suppressor (VHL), which acts as a ubiquitin ligase to promote proteolysis of HIF-1alpha, was targeted. We investigated the effect of VHL deletion (i.e., stable expression of HIF-1alpha) histologically and used the anti-HIF-1alpha agent [3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole] (YC-1) to test whether inhibition of HIF-1alpha could represent a novel approach to treating renal fibrosis. The area of renal fibrosis was significantly increased in a 5/6 renal ablation model of VHL-/- mice and in all VHL-/- mice at least 60 wk of age. Injection of YC-1 inhibited the progression of renal fibrosis in unilateral ureteral obstruction model mice. In conclusion, HIF-1alpha appears to be a critical contributor to the progression of renal fibrosis and could be a useful target for its treatment.


Subject(s)
Epithelial Cells/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/physiopathology , Kidney Diseases/physiopathology , Kidney Tubules, Proximal/physiology , Aging/pathology , Aging/physiology , Animals , Disease Models, Animal , Enzyme Activators/pharmacology , Epithelial Cells/pathology , Female , Fibrosis , Gene Deletion , Gene Expression/physiology , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Indazoles/pharmacology , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Transfection , Up-Regulation/physiology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
11.
Cell Cycle ; 7(9): 1128-32, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18418042

ABSTRACT

Capillary rarefaction is a hallmark of fibrotic diseases and results in reduced blood perfusion and oxygen delivery. In the kidney, tubulointerstitial fibrosis, which leads to the destruction of renal tissue and the irreversible loss of kidney function, is associated with hypoxia and the activation of Hypoxia-Inducible-Factor (HIF) signaling. HIF-1 and HIF-2 are basic-helix-loop-helix transcription factors that allow cells to survive in a low oxygen environment by regulating energy metabolism, vascular remodeling, erythropoiesis, cellular proliferation and apoptosis. Recent studies suggest that HIF activation promotes epithelial to mesenchymal transition (EMT) and renal fibrogenesis. These findings raise the possibility that the spectrum of HIF activated biological responses to hypoxic stress may differ under conditions of acute and chronic hypoxia. Here we discuss the role of HIF signaling in the pathogenesis and progression of chronic kidney disease.


Subject(s)
Fibrosis/metabolism , Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , Nephritis, Interstitial/metabolism , Animals , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Fibrosis/genetics , Fibrosis/physiopathology , Humans , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1/genetics , Mesoderm/metabolism , Nephritis, Interstitial/physiopathology , Regional Blood Flow/physiology , Transforming Growth Factor beta1/metabolism
12.
J Clin Invest ; 117(12): 3810-20, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18037992

ABSTRACT

Hypoxia has been proposed as an important microenvironmental factor in the development of tissue fibrosis; however, the underlying mechanisms are not well defined. To examine the role of hypoxia-inducible factor-1 (HIF-1), a key mediator of cellular adaptation to hypoxia, in the development of fibrosis in mice, we inactivated Hif-1alpha in primary renal epithelial cells and in proximal tubules of kidneys subjected to unilateral ureteral obstruction (UUO) using Cre-loxP-mediated gene targeting. We found that Hif-1alpha enhanced epithelial-to-mesenchymal transition (EMT) in vitro and induced epithelial cell migration through upregulation of lysyl oxidase genes. Genetic ablation of epithelial Hif-1alpha inhibited the development of tubulointerstitial fibrosis in UUO kidneys, which was associated with decreased interstitial collagen deposition, decreased inflammatory cell infiltration, and a reduction in the number of fibroblast-specific protein-1-expressing (FSP-1-expressing) interstitial cells. Furthermore, we demonstrate that increased renal HIF-1alpha expression is associated with tubulointerstitial injury in patients with chronic kidney disease. Thus, we provide clinical and genetic evidence that activation of HIF-1 signaling in renal epithelial cells is associated with the development of chronic renal disease and may promote fibrogenesis by increasing expression of extracellular matrix-modifying factors and lysyl oxidase genes and by facilitating EMT.


Subject(s)
Cell Movement , Epithelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia/metabolism , Kidney Tubules, Proximal/metabolism , Ureteral Obstruction/metabolism , Animals , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Cell Hypoxia , Cell Movement/genetics , Chronic Disease , Collagen/biosynthesis , Collagen/genetics , Epithelial Cells/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Hypoxia/genetics , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Tubules, Proximal/pathology , Mice , Mice, Knockout , Protein-Lysine 6-Oxidase/biosynthesis , Protein-Lysine 6-Oxidase/genetics , S100 Calcium-Binding Protein A4 , S100 Proteins , Signal Transduction/genetics , Up-Regulation/genetics , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology
13.
Mol Cell Biol ; 25(8): 3163-72, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15798202

ABSTRACT

Patients with germ line mutations in the VHL tumor suppressor gene are predisposed to the development of highly vascularized tumors within multiple tissues. Loss of pVHL results in constitutive activation of the transcription factors HIF-1 and HIF-2, whose relative contributions to the pathogenesis of the VHL phenotype have yet to be defined. In order to examine the role of HIF in von Hippel-Lindau (VHL)-associated vascular tumorigenesis, we utilized Cre-loxP-mediated recombination to inactivate hypoxia-inducible factor-1alpha (Hif-1alpha) and arylhydrocarbon receptor nuclear translocator (Arnt) genes in a VHL mouse model of cavernous liver hemangiomas and polycythemia. Deletion of Hif-1alpha did not affect the development of vascular tumors and polycythemia, nor did it suppress the increased expression of vascular endothelial growth factor (Vegf) and erythropoietin (Epo). In contrast, phosphoglycerokinase (Pgk) expression was substantially decreased, providing evidence for target gene-dependent functional redundancy between different Hif transcription factors. Inactivation of Arnt completely suppressed the development of hemangiomas, polycythemia, and Hif-induced gene expression. Here, we demonstrate genetically that the development of VHL-associated vascular tumors in the liver depends on functional ARNT. Furthermore, we provide evidence that individual HIF transcription factors may play distinct roles in the development of specific VHL disease manifestations.


Subject(s)
DNA-Binding Proteins/physiology , Genes, Tumor Suppressor , Hemangioma, Cavernous/genetics , Liver Neoplasms/genetics , Receptors, Aryl Hydrocarbon/physiology , Transcription Factors/physiology , von Hippel-Lindau Disease/genetics , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , DNA-Binding Proteins/genetics , Erythropoietin/genetics , Erythropoietin/metabolism , Gene Deletion , Gene Expression , Gene Silencing , Hemangioma, Cavernous/metabolism , Hepatocytes/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit , Liver/blood supply , Liver/pathology , Liver Neoplasms/metabolism , Mice , Mice, Knockout , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Polycythemia/genetics , Polycythemia/metabolism , Receptors, Aryl Hydrocarbon/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/physiology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Von Hippel-Lindau Tumor Suppressor Protein , von Hippel-Lindau Disease/metabolism
14.
Am J Physiol Renal Physiol ; 287(6): F1223-32, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15315937

ABSTRACT

CTGF plays a significant role in the development of renal fibrosis by mediating the fibrotic effects of transforming growth factor (TGF)-beta(1) and has been shown to be hypoxia inducible in human breast cancer cells. It has been suggested that hypoxia is an important underlying cause for the development of renal fibrosis through the modulation of profibrotic genes. One of the key mediators of the cell's response to lowered oxygen environments is hypoxia-inducible-factor-1 (HIF-1), a basic helix-loop-helix transcription factor, which enables cells to adapt to hypoxia by regulating the expression of genes involved in increasing oxygen availability (VEGF, erythropoietin) and enhancing glucose uptake and metabolism (Glut-1, PGK). In this paper, we have used primary tubular epithelial cell cultures from a tetracycline-inducible-Hif-1alpha knockout murine model to further elucidate the role of Hif-1 in the hypoxic-induction of Ctgf expression. We show that hypoxia response elements present upstream of Ctgf enable direct interaction of Hif-1 transcription factor with the Ctgf promoter, resulting in increased transcription of Ctgf mRNA. Cells deficient in Hif-1alpha were incapable of inducing Ctgf mRNA in response to hypoxia, suggesting an absolute requirement of Hif-1. Furthermore, the observed Hif-1-mediated hypoxic stimulation of Ctgf expression was found to occur independently of TGF-beta(1) signaling. Our findings have important implications for a number of fibrotic disorders in which hypoxia, CTGF, and TGF-beta(1) are involved, including renal, dermal, hepatic, and pulmonary fibrosis.


Subject(s)
Cell Hypoxia , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Transcription Factors/physiology , Animals , Base Sequence , Binding Sites , Cells, Cultured , Connective Tissue Growth Factor , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit , Immediate-Early Proteins/genetics , Integrases/genetics , Intercellular Signaling Peptides and Proteins/genetics , Kidney Tubules/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Response Elements/genetics , Signal Transduction , Smad Proteins , Trans-Activators/physiology , Transcription Factors/deficiency , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1
15.
Kidney Int ; 64(6): 2079-91, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633130

ABSTRACT

BACKGROUND: Tubulointerstitial inflammation and fibrosis are pathologic hallmarks of end-stage renal disease (ESRD). Here we have used DNA microarray technology to monitor the transcriptomic responses to murine unilateral ureteral obstruction (UUO) with a view to identifying molecular modulators of tubulointerstitial fibrosis. METHODS: Using Affymetrix Mu74Av2 microarrays, gene expression 4 and 10 days postobstruction was investigated relative to control contralateral kidneys. Candidate profibrogenic genes were further investigated in epithelial cells undergoing epithelial to mesenchymal transition (EMT) in vitro. RESULTS: mRNA levels for 1091 gene/EST sequences, of a total of 12,488 displayed on the microarray, were altered twofold or greater by days 4 and 10 postobstruction compared to contralateral control kidneys. Genes were categorised into functional groups, including modulators of cytoskeletal and extracellular matrix metabolism, cell growth, signalling, and transcription/translational events. Among the potentially profibrogenic genes, whose mRNA levels were increased after UUO, were fibroblast-inducible secreted protein (fisp-12), the murine homologue of connective tissue growth factor (CTGF), collagen XVIIIalpha1, secreted protein acidic and rich in cysteine (SPARC), and src-suppressed C-kinase substrate (SSeCKS). A sustained increase in fisp-12 mRNA level was observed during EMT induced by transforming growth factor-beta1 (TGF-beta1) and epidermal growth factor (EGF). CONCLUSION: Altered gene expression in murine UUO has been demonstrated. Increased expression of fisp-12, SPARC, and SSeCKS has been shown in response to TGF-beta1 treatment and during EMT, suggesting that these genes may offer potential therapeutic targets against tubulointerstitial fibrosis.


Subject(s)
Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Ureteral Obstruction/metabolism , A Kinase Anchor Proteins , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Collagen Type XVIII/genetics , Connective Tissue Growth Factor , Disease Progression , Epidermal Growth Factor/pharmacology , Gene Expression , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mitogens/genetics , Osteonectin/genetics , Protein Isoforms/genetics , RNA, Messenger/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Ureteral Obstruction/genetics , Ureteral Obstruction/pathology , Ureteral Obstruction/physiopathology
16.
Kidney Int ; 64(2): 480-92, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12846743

ABSTRACT

BACKGROUND: Lipoxins are lipoxygenase-derived eicosanoids with anti-inflammatory and proresolution bioactivities in vitro and in vivo. We have previously demonstrated that the stable synthetic LXA4 analog 15-epi-16-(FPhO)-LXA4-Me is renoprotective in murine renal ischemia/reperfusion injury, as gauged by lower serum creatinine, attenuated leukocyte infiltration, and reduced morphologic tubule injury. METHODS: We employed complementary oligonucleotide microarray and bioinformatic analyses to probe the transcriptomic events that underpin lipoxin renoprotection in this setting. RESULTS: Microarray-based analysis identified three broad categories of genes whose mRNA levels are altered in response to ischemia/reperfusion injury, including known genes previously implicated in the pathogenesis of ischemia/reperfusion injury [e.g., intercellular adhesion molecule-1 (ICAM-1), p21, KIM-1], known genes not previously associated with ischemia/reperfusion injury, and cDNAs representing yet uncharacterized genes. Characterization of expressed sequence tags (ESTs) displayed on microarrays represents a major challenge in studies of global gene expression. A bioinformatic annotation pipeline successfully annotated a large proportion of ESTs modulated during ischemia/reperfusion injury. The differential expression of a representative group of these ischemia/reperfusion injury-modulated genes was confirmed by real-time polymerase chain reaction. Prominent among the up-regulated genes were claudin-1, -3, and -7, and ADAM8. Interestingly, the former response was claudin-specific and was not observed with other claudins expressed by the kidney (e.g., claudin-8 and -6) or indeed with other components of the renal tight junctions (e.g., occludin and junctional adhesion molecule). Noteworthy among the down-regulated genes was a cluster of transport proteins (e.g., aquaporin-1) and the zinc metalloendopeptidase meprin-1 beta implicated in renal remodeling. CONCLUSION: Treatment with the lipoxin analog 15-epi-16-(FPhO)-LXA4-Me prior to injury modified the expression of many differentially expressed pathogenic mediators, including cytokines, growth factors, adhesion molecules, and proteases, suggesting a renoprotective action at the core of the pathophysiology of acute renal failure (ARF). Importantly, this lipoxin-modulated transcriptomic response included many genes expressed by renal parenchymal cells and was not merely a reflection of a reduced renal mRNA load resulting from attenuated leukocyte recruitment. The data presented herein suggest a framework for understanding drivers of kidney injury in ischemia/reperfusion and the molecular basis for renoprotection by lipoxins in this setting.


Subject(s)
Kidney/physiopathology , Lipoxins/pharmacology , Reperfusion Injury/drug therapy , Reperfusion Injury/physiopathology , Transcription, Genetic/drug effects , ADAM Proteins , Acute Kidney Injury/drug therapy , Acute Kidney Injury/physiopathology , Animals , Antigens, CD/genetics , Claudin-1 , Claudin-3 , Claudins , DNA, Complementary , Epidermal Growth Factor/genetics , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...