Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Clin Apher ; 34(4): 423-433, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30817043

ABSTRACT

INTRODUCTION: Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition with monoclonal antibodies has complemented the armamentarium of lipid-lowering therapy (LLT) before the final step of commencing chronic lipoprotein apheresis (LA). Data are scarce on patients who, after escalation of LLT with PCSK9 antibodies, have commenced chronic LA or PCSK9 antibody treatment during ongoing long-term LA. PATIENTS AND METHODS: In this study, a cohort of 110 patients with established atherosclerotic cardiovascular disease (ASCVD) due to hypercholesterolemia or concomitant lipoprotein(a)-hyperlipoproteinemia, who received PCSK9 antibodies for the first time during routine care, were consecutively identified. RESULTS: Mean LDL-C concentration prior to initiation of LA or PCSK9 antibody treatment was 5.3 ± 2.6 mmol/L (205 ± 102 mg/dL). Due to established ASCVD, the risk-adjusted LDL-C target value was <1.8 mmol/L (<70 mg/dL) in all patients. Use of PCSK9 antibodies increased the proportion of patients attaining the LDL-C target concentration by 41.8% overall. Treatment emergent adverse events (TEAE) associated with PCSK9 antibody medication were reported in 35 patients (31.8%). Discontinuation of PCSK9 antibody therapy due to TEAEs occurred in 25 patients (22.7%). CONCLUSION: Finally, 55.5% of patients received a combination of PCSK9 antibody therapy and LA at individually optimized treatment frequencies resulting in an increase of target attainment in 54.1% of patients. About 18.1% of chronic LA patients terminated LA treatment in this real-world study. The termination of long-term LA therapy, which has hitherto prevented the progression of ASCVD, requires careful individual risk assessment and cannot be recommended by the general criteria of LDL-C reduction.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Blood Component Removal/methods , Combined Modality Therapy/methods , Lipoproteins/isolation & purification , PCSK9 Inhibitors , Atherosclerosis/therapy , Cholesterol, LDL/isolation & purification , Enzyme Inhibitors/therapeutic use , Female , Humans , Hypercholesterolemia/therapy , Lipids/isolation & purification , Lipoprotein(a)/isolation & purification , Male , Middle Aged , Proprotein Convertase 9/immunology
2.
Transpl Immunol ; 39: 66-73, 2016 11.
Article in English | MEDLINE | ID: mdl-27590486

ABSTRACT

BACKGROUND: Obliterative bronchiolitis (OB) is the major limiting factor for long-term survival after lung transplantation. As previously shown, donor treatment with a PHD-inhibitor activating hypoxia-inducible transcription factors (HIFs) prevents graft injury both in an allogenic kidney and aortic allograft transplant model. The aim of this study was to investigate the effect of HIF activation with a PHD-inhibitor on the development of OB. METHODS: Fully MHC-mismatched C57BL/6 (H-2b) donor tracheas were orthotopically transplanted into CBA/J (H-2k) recipients. Donor animals received a single dose of PHD-inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate (ICA) (40mg/kg i.p.) or vehicle control 4h before transplantation. Transplanted tracheas were harvested 14 or 30days after transplantation and were analyzed by histology, by immunofluorescence and by rtPCR for mRNA expression. RESULTS: Donor pre-conditioning with ICA resulted in HIF accumulation and induction of HIF target genes: HO-1, VEGF, MIF, TGFß, and EpoR, which persisted during different times of ischemia. Grafts of vehicle treated controls showed substantially more luminal obliteration on postoperative day 30 in contrast to groups pre-treated with ICA [luminal obliteration 29.2±5% (ICA) vs. 36.7±8% (control), p<0.01]. We found significantly lower expression of TNFα, PDGFß, MCP-1, E-selectin, and ICAM-1 14days after ICA premedication. In addition ICA pre-treated groups revealed decreased T-cell and macrophage infiltration in tracheal grafts on days 30 after transplantation (p<0.05). CONCLUSION: Pre-treatment with ICA effectively reduced obliterative bronchiolitis. Our data suggest that activation of hypoxia-inducible transcription factors (HIFs) and thereby adaptation to low oxygen prevents the development of OB and allograft injury. Pharmaceutical inhibition of PHDs appears to be an attractive strategy for organ preservation that deserves further clinical evaluation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Bronchiolitis Obliterans/drug therapy , Glycine/analogs & derivatives , Graft Rejection/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoquinolines/therapeutic use , Lung Transplantation , Macrophages/immunology , Prolyl-Hydroxylase Inhibitors/therapeutic use , T-Lymphocytes/immunology , Transplantation Conditioning/methods , Animals , Cell Movement , Gene Expression Regulation , Glycine/therapeutic use , Heme Oxygenase-1/genetics , Humans , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Models, Animal , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/genetics
3.
Nephrol Dial Transplant ; 27(3): 929-36, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21742784

ABSTRACT

BACKGROUND: Inhibition of the HIF regulating prolyl hydroxylation domain (PHDs) proteins prior to renal injury (preconditioning) has been shown to protect the kidney via activation of hypoxia-inducible transcription factors (HIF). Application of erythropoietin (EPO), one of the HIF target genes, has also been shown to be nephroprotective, and it remains unclear to what extent the effect of HIF induction is mediated by EPO. It is also unknown whether HIF activation after the onset of ischaemia (postconditioning) is still able to protect the kidney. METHODS: Using a rat model of renal ischaemia-reperfusion injury, animals were treated with the PHD inhibitor (PHD-I) 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate (ICA), vehicle (Veh) or recombinant human EPO (300 IU/kg) 6 h (ICA or Veh) or 30 min (EPO) prior to ischaemia (preconditioning) or with ICA prior to reperfusion (postconditioning). Renal function was assessed at baseline, 24 h and 72 h. After 72 h, kidneys were processed for histology and morphometric analysis. HIF immunohistochemistry and real-time polymerase chain reaction for HIF target genes, including EPO, were performed to evaluate ICA effects. RESULTS: ICA treatment resulted in stabilization of HIF-1α and -2α and up-regulation of HIF target genes in a dose-dependent manner. Preconditional activation of HIF by ICA significantly improved serum creatinine levels and renal morphology in comparison to Veh (P < 0.05), while postconditional ICA treatment was ineffective. EPO therapy improved tissue morphology but had no impact on the course of serum creatinine. CONCLUSION: These findings are in line with the concept that PHD-Is exert their protective effects through accumulation of HIF target gene products, with time requirements for increased transcription and translation of HIF-dependent genes, and suggest that their renoprotective effect is not predominately mediated by EPO.


Subject(s)
Acute Kidney Injury/prevention & control , Erythropoietin/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Reperfusion Injury/prevention & control , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Enzyme Inhibitors/pharmacology , Epoetin Alfa , Hypoxia/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Recombinant Proteins/therapeutic use , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
4.
Nephrol Dial Transplant ; 26(11): 3458-65, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21804086

ABSTRACT

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure. Aberrant epithelial cell proliferation is a major cause of progressive cyst enlargement in ADPKD. Since activation of the Ras/Raf signaling system has been detected in cyst-lining epithelia, inhibition of Raf kinase has been proposed as an approach to retard the progression of ADPKD. Methods and results. PLX5568, a novel selective small molecule inhibitor of Raf kinases, attenuated proliferation of human ADPKD cyst epithelial cells. It reduced in vitro cyst growth of Madin-Darby Canine Kidney cells and of human ADPKD cells within a collagen gel. In male cy/+ rats with polycystic kidneys, PLX5568 inhibited renal cyst growth along with a significant reduction in the number of proliferating cell nuclear antigen- and phosphorylated extracellular signal-regulated kinase-positive cyst-lining epithelial cells. Furthermore, treated animals showed increased capacity to concentrate urine. However, PLX5568 did not lead to a consistent improvement of renal function. Moreover, although relative cyst volume was decreased, total kidney-to-body weight ratio was not significantly reduced by PLX5568. Further analyses revealed a 2-fold increase of renal and hepatic fibrosis in animals treated with PLX5568. CONCLUSIONS: PLX5568 attenuated cyst enlargement in vitro and in a rat model of ADPKD without improving kidney function, presumably due to increased renal fibrosis. These data suggest that effective therapies for the treatment of ADPKD will need to target fibrosis as well as the growth of cysts.


Subject(s)
Cell Proliferation/drug effects , Cysts/pathology , Kidney/physiopathology , Liver Cirrhosis/chemically induced , Polycystic Kidney Diseases/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Animals , Cells, Cultured , Cysts/drug therapy , Dogs , Epithelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases , Humans , Immunoblotting , Immunoenzyme Techniques , Kidney/drug effects , Male , Mitogen-Activated Protein Kinases , Phosphorylation/drug effects , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/pathology , Protein Kinase Inhibitors/toxicity , Rats , Rats, Sprague-Dawley
5.
J Am Soc Nephrol ; 21(12): 2151-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21115615

ABSTRACT

The reasons for inadequate production of erythropoietin (EPO) in patients with ESRD are poorly understood. A better understanding of EPO regulation, namely oxygen-dependent hydroxylation of the hypoxia-inducible transcription factor (HIF), may enable targeted pharmacological intervention. Here, we tested the ability of fibrotic kidneys and extrarenal tissues to produce EPO. In this phase 1 study, we used an orally active prolyl-hydroxylase inhibitor, FG-2216, to stabilize HIF independent of oxygen availability in 12 hemodialysis (HD) patients, six of whom were anephric, and in six healthy volunteers. FG-2216 increased plasma EPO levels 30.8-fold in HD patients with kidneys, 14.5-fold in anephric HD patients, and 12.7-fold in healthy volunteers. These data demonstrate that pharmacologic manipulation of the HIF system can stimulate endogenous EPO production. Furthermore, the data indicate that deranged oxygen sensing--not a loss of EPO production capacity--causes renal anemia.


Subject(s)
Erythropoietin/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Anemia/prevention & control , Erythropoietin/blood , Female , Follow-Up Studies , Humans , Male , Middle Aged , Nephrectomy , Procollagen-Proline Dioxygenase/administration & dosage , Procollagen-Proline Dioxygenase/pharmacokinetics , Reference Values , Renal Dialysis , Risk Assessment , Treatment Outcome
6.
Mol Cancer Res ; 7(1): 88-98, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19147540

ABSTRACT

The mammalian target of rapamycin (mTOR) regulates cellular growth and proliferation, mainly by controlling cellular translation. Most tumors show constitutive activation of the mTOR pathway. In hypoxia, mTOR is inactivated, which is believed to be part of the program of the cell to maintain energy homeostasis. However, certain proteins are believed to be preferentially translated during hypoxia via 5' terminal oligopyrimidine tract mechanisms with controversial discussion about the involvement of the mTOR-dependent ribosomal protein S6 (rpS6). The hypoxia-inducible transcription factor (HIF) is the master regulator of hypoxic adaptation and itself strongly implicated in tumor growth. HIF is translationally regulated by mTOR. The regulatory features and the involvement of molecular oxygen itself in this regulation of HIF by mTOR are poorly understood. mTOR inhibition leads to profound attenuation of HIFalpha protein in the majority of primary and cancer cells studied. Under severe hypoxia, no influence of mTOR inhibitors was observed; thus, stimulation of HIFalpha by mTOR may only be relevant under mild hypoxia or even normoxia. HIF expression and phosphorylated rpS6 negatively correlate in experimental tumors. In cell culture, prolonged hypoxia abolishes rpS6 phosphorylation, which seems to be partly independent of the upstream p70S6 kinase. We show that hypoxic repression of rpS6 is largely dependent on HIF, implicating a negative feedback loop, which may influence cellular translational rates and metabolic homeostasis. These data implicate that the hypoxic microenvironment renders tumor cells resistant to mTOR inhibition, at least concerning hypoxic gene activation, which would add to the difficulties of other established therapeutic strategies in hypoxic cancer tissues.


Subject(s)
Cell Hypoxia/genetics , Hypoxia-Inducible Factor 1/biosynthesis , Protein Kinases/genetics , Cell Line, Tumor , HeLa Cells , Homeostasis , Humans , Immunohistochemistry , Luciferases/genetics , Oxygen Consumption , Protein Biosynthesis , Ribonucleases , TOR Serine-Threonine Kinases , Transfection
7.
Curr Opin Crit Care ; 14(6): 621-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19005302

ABSTRACT

PURPOSE OF REVIEW: Acute kidney injury (AKI) frequently occurs in critically ill patients and is an independent risk factor for poor outcome. The prevention of kidney injury in intensive care remains a great challenge as specific nephroprotective therapies are still lacking. The present review summarizes recent evidence for the use of erythropoietin as a promising candidate to provide protection from AKI. RECENT FINDINGS: Beyond the known hematopoietic actions of erythropoietin, a number of preclinical studies demonstrated that erythropoietin possesses pleiotropic, organ-protecting properties. Preconditional and postconditional erythropoietin treatment was shown to protect from ischemic, toxic and septic AKI. Despite heterogeneities in study design and dose, erythropoietin consistently ameliorated renal injury. The mechanisms of protection remain largely unclear but may involve reduction of apoptosis, induction of cellular proliferation and tissue repair as well as mobilization of stem cells. SUMMARY: Animal studies revealed a physiological basis for the use of erythropoietin in AKI, which may be clinically applicable to prevent AKI in critically ill patients, but clinical studies are still lacking.


Subject(s)
Acute Kidney Injury/prevention & control , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Critical Illness , Erythropoietin/therapeutic use , Acute Kidney Injury/etiology , Hematopoiesis , Humans , Intracellular Signaling Peptides and Proteins , Recombinant Proteins , Ribonucleoproteins, Small Nuclear , Risk Factors , Tumor Suppressor Proteins/metabolism
8.
J Am Soc Nephrol ; 19(3): 486-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18256363

ABSTRACT

The contribution of hypoxia to cisplatin-induced renal tubular injury is controversial. Because the hypoxia-inducible factor (HIF) pathway is a master regulator of adaptation to hypoxia, we measured the effects of cisplatin on HIF accumulation in vitro and in vivo, and tested whether hypoxic preconditioning is protective against cisplatin-induced injury. We found that cisplatin did not stabilize HIF-1alpha protein in vitro or in vivo under normoxic conditions. However, hypoxic preconditioning of cisplatin-treated proximal tubular cells in culture reduced apoptosis in an HIF-1alpha-dependent fashion and increased cell proliferation as measured by BrdU incorporation. In vivo, rats preconditioned with carbon monoxide before cisplatin administration had significantly better renal function than rats kept in normoxic conditions throughout. Moreover, the histomorphological extent of renal damage and tubular apoptosis was reduced by the preconditional treatment. Therefore, development of pharmacologic agents to induce renal HIF might provide a new approach to ameliorate cisplatin-induced nephrotoxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia , Kidney/drug effects , Acute Kidney Injury/chemically induced , Animals , Apoptosis/drug effects , Carbon Monoxide/pharmacology , Cell Line , DNA/biosynthesis , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
9.
Methods Enzymol ; 435: 221-45, 2007.
Article in English | MEDLINE | ID: mdl-17998057

ABSTRACT

Since the first description of a protective effect of hypoxic preconditioning in the heart, the principle of reducing tissue injury in response to ischemia by prior exposure to hypoxia was confirmed in a number of cells and organs. However, despite impressive preclinical results, hypoxic preconditioning has so far failed to reach clinical application. Nevertheless, it remains of significant interest to induce genes that are normally activated during hypoxia and ischemia as part of an endogenous escape mechanism prior to or during the early phase of an ischemic insult. This approach has recently been greatly facilitated by the identification of hypoxia-inducible factors (HIFs), transcription factors that operate as a master switch in the cellular response to hypoxia. Far more than 100 target genes are regulated by HIF, including genes such as erythropoietin and hemoxygenase-1, which have been shown to be tissue-protective. The identification of small molecule inhibitors of the oxygen-sensing HIF-prolyl hydroxlases now offers the possibility to mimic the hypoxic response by pharmacological stabilization of HIF in order to achieve organ protection. Oxygen-independent activation of HIF is therefore a promising therapeutic strategy for the prevention of organ injury and failure.


Subject(s)
Ischemia/prevention & control , Ischemic Preconditioning/methods , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Carbon Monoxide/pharmacology , Cell Hypoxia , Gene Expression , Gene Targeting , Humans , Mice , Myocardium/enzymology , Procollagen-Proline Dioxygenase/metabolism , Rats , Transcription Factors/genetics
10.
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
11.
J Am Soc Nephrol ; 17(7): 1970-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16762988

ABSTRACT

Activation of hypoxia-inducible transcription factor (HIF) has been identified as an important mechanism of cellular adaptation to low oxygen. Normoxic degradation of HIF is mediated by oxygen-dependent hydroxylation of specific prolyl residues of the regulative alpha-subunits by HIF prolyl hydroxylases (PHD). It was hypothesized that inhibition of HIF degradation by either hypoxia or pharmacologic inhibition of PHD would confer protection against subsequent ischemic injury. For testing this hypothesis ischemic acute renal failure was induced in rats by 40 min of clamping of the left renal artery after right-sided nephrectomy. Before surgery, pretreatment with either carbon monoxide, leading to tissue hypoxia, or the novel PHD inhibitor FG-4487 was applied. No toxic effects of FG-4487 were observed. Both pretreatments strongly induced the accumulation of HIF-1alpha and HIF-2alpha in tubular and peritubular cells, respectively, as well as HIF target gene expression. The course of subsequent ischemic injury was significantly ameliorated by both strategies of preconditioning, as evident from a significant improvement of serum creatinine and serum urea after 24 and 72 h. Furthermore, tissue injury and apoptosis were less severe, which were quantified by application of a standardized histologic scoring system in a blinded manner. In conclusion, the data provide proof of principle that preconditional activation of the HIF system protects against ischemic injury. Inhibiting the activity of HIF hydroxylases therefore seems to have considerable clinical perspectives.


Subject(s)
Acute Kidney Injury/metabolism , Carbon Monoxide/administration & dosage , Hypoxia-Inducible Factor 1/metabolism , Hypoxia , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Acute Kidney Injury/prevention & control , Animals , Apoptosis/drug effects , Cell Line , Creatinine/blood , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/drug effects , Male , Premedication , Procollagen-Proline Dioxygenase/adverse effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Urea/blood
12.
Eur J Heart Fail ; 8(4): 347-54, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16513418

ABSTRACT

UNLABELLED: Prolyl hydroxylase domain-containing enzymes (PHD) hydroxylate a proline residue that controls the degradation of hypoxia inducible factor (HIF). Hypoxia inhibits this hydroxylation thus increasing HIF levels. HIF is upregulated in ischemic tissues, growing tumors and in nonischemic, mechanically stressed myocardium. Pharmacological inhibition of prolyl 4-hydroxylase (P4-H) stabilizes HIF-protein in vitro and may modulate collagen turnover. The aims of this study were to investigate whether inhibition of P4-H protects myocardium against ischemia, and whether the observed effects are related to modulation of collagen metabolism or due to the stabilization of HIF. METHODS: Rats were treated with a specific P4-H inhibitor (P4-HI) or vehicle starting 2 days before induction of myocardial infarction (MI). Rats were investigated 7 or 30 days after MI. Induction of HIF-1alpha and -2alpha was visualized by immunohistochemistry. Expression of growth factors (connective tissue growth factor, Osteopontin) and mRNA expression and protein levels of Collagen I and III as well as HIF-2alpha were measured. RESULTS: P4-HI augments HIF in the myocardium as early as 24 h after treatment. P4-HI did not alter the MI-induced enhanced expression of growth factors and collagen. Treatment with P4-HI significantly reduced heart and lung weight, improved left ventricular contractility, prevented left ventricular enlargement and improved left ventricular ejection fraction without affecting infarct size after 30 days. CONCLUSIONS: Specific inhibition of the P4-H improved cardiac function without affecting the infarct size after experimental myocardial infarction in rats. Stabilization of HIF rather than inhibition of collagen maturation by P4-HI may prevent cardiac remodeling after MI.


Subject(s)
Collagen/metabolism , Hypoxia-Inducible Factor 1/metabolism , Myocardial Infarction/metabolism , Animals , Base Sequence , Collagen/genetics , DNA Primers , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Male , Myocardial Infarction/physiopathology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , RNA, Messenger/genetics , Rats , Rats, Wistar
13.
Kidney Int Suppl ; (99): S46-51, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16336576

ABSTRACT

The kidney shows a remarkable discrepancy between blood supply and oxygenation. Despite high blood flow and oxygen delivery, oxygen tensions in the kidney are comparatively low, in particular in the renal medulla. The reason for this lies in the parallel arrangement of arterial and venous preglomerular and postglomerular vessels, which allows oxygen to pass from arterioles into the postcapillary venous system via shunt diffusion. The limitation in renal tissue oxygen supply renders the kidney susceptible to hypoxia and has long been recognized as an important factor in the pathogenesis of acute renal injury. In recent years, evidence has accumulated that hypoxia does also play a significant role in the pathogenesis and progression of chronic renal disease, because different types of kidney disease are usually associated with a rarefication of postglomerular capillaries. In both acute and chronic diseases, tissue hypoxia does not only imply the risk of energy deprivation but also induces regulatory mechanisms and has a profound influence on gene expression. In particular, the transcription factor hypoxia inducible factor (HIF) is involved in cellular regulation of angiogenesis, vasotone, glucose metabolism, and cell death and survival decisions. HIF has been shown to be activated in renal disease and presumably plays a major role in protective responses to oxygen deprivation. Recent insights into the regulation of HIF increase our understanding of the role of hypoxia in disease progression and open new options to improve hypoxia tolerance and to induce nephroprotection.


Subject(s)
Kidney Diseases/physiopathology , Oxygen/physiology , Cell Hypoxia , Chronic Disease , Disease Progression , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Kidney Diseases/pathology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/pathology , Kidney Failure, Chronic/physiopathology , Kidney Glomerulus/pathology , Kidney Glomerulus/physiology , Kidney Glomerulus/physiopathology , Kidney Tubules/blood supply , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Neovascularization, Physiologic
14.
Biol Chem ; 386(8): 785-90, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16201874

ABSTRACT

The kidney is a target organ for thyroid hormone action and a variety of renal transport processes are altered in response to impaired thyroid functions. To investigate the effect of thyroid hormone on the expression of the renal proximal tubular high-affinity-type H(+)-peptide cotransporter (PEPT2) in rats, hypothyroidism was induced in animals by administration of methimazole (0.05%) via drinking water. After 7 weeks of treatment, hypothyroidism was confirmed by determining serum free T(3) and free T(4) concentrations. Northern blotting was used to examine the expression of PEPT2 mRNA in kidney tissues from hypothyroid rats compared to control rats. Hypothyroidism resulted in an increased level of total renal PEPT2 mRNA (121.1+/-3.3% vs. control 100+/-2.8%; p=0.008). The mRNA results were confirmed by immuno-blotting, which demonstrated significantly increased protein levels (162% vs. control 100%; p<0.01). Immunohistochemistry also revealed increased PEPT2 protein levels in the proximal tubules of treated compared to non-treated rats. In summary, PEPT2 is the first proximal tubule transporter protein that shows increased expression in states of hypothyreosis. As PEPT2 reabsorbs filtered di- and tripeptides and peptide-like drugs, the present findings may have important implications in nutritional amino acid homeostasis and for drug dynamics in states of altered thyroid function.


Subject(s)
Hypothyroidism/metabolism , Symporters/metabolism , Animals , Antithyroid Agents/pharmacology , Biological Transport , Blotting, Northern , Gene Expression , Hypothyroidism/blood , Hypothyroidism/chemically induced , Immunohistochemistry , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Methimazole , Peptides/metabolism , RNA, Messenger/metabolism , Rats , Symporters/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...