Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
2.
Front Physiol ; 14: 1208105, 2023.
Article in English | MEDLINE | ID: mdl-37435301

ABSTRACT

Introduction: We previously reported that malignant hypertension is associated with impaired capillary density of target organs. Here, we tested the hypothesis that stabilization of hypoxia-inducible factor (HIF) in a modified "preconditioning" approach prevents the development of malignant hypertension. To stabilize HIF, we employed pharmacological inhibition of HIF prolyl hydroxylases (PHD), that profoundly affect HIF metabolism. Methods: Two-kidney, one-clip renovascular hypertension (2K1C) was induced in rats; controls were sham operated. 2K1C rats received either intermittent injections of the PHD inhibitor ICA (2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate) or placebo. Thirty-five days after clipping, the frequency of malignant hypertension was assessed (based on weight loss and the occurrence of characteristic vascular lesions). In addition, kidney injury was compared between all ICA treated versus all placebo treated 2K1C, regardless of the occurrence of malignant hypertension. HIF stabilization was evaluated by immunohistochemistry, and HIF target gene expression by RT-PCR. Results: Blood pressure was elevated to the same degree in ICA- and placebo-treated 2K1C compared to control rats. ICA treatment did not affect the frequency of malignant hypertension or the extent of kidney tissue fibrosis, inflammation, or capillary density. There was a trend towards higher mortality and worse kidney function in ICA-treated 2K1C rats. ICA increased the number of HIF-1α-positive renal tubular cell nuclei and induced several HIF-1 target genes. In contrast, expression of HIF-2α protein as well as HIF-2 target genes were markedly enhanced by 2K1C hypertension, irrespective of ICA treatment. Discussion: We conclude that intermittent PHD inhibition did not ameliorate severe renovascular hypertension in rats. We speculate that the unexpected strong renal accumulation of HIF-2α in renovascular hypertension, which could not be further augmented by ICA, may contribute to the lack of a benefit from PHD inhibition.

3.
Dalton Trans ; 51(17): 6839-6845, 2022 May 03.
Article in English | MEDLINE | ID: mdl-35438696

ABSTRACT

Bis(4-carboxylpyrazol-1-yl)acetic acid (H3bcpza) (2), obtained in a one-pot synthesis, contains carboxyl groups that differ in their pKa values. The ligand exhibits heteroscorpionate κ3-N,N,O-coordination whereupon the peripheral carboxyl groups are not involved in metal binding. The corresponding carbonyl complexes [Mn(H2bcpza)(CO)3] (4), [Re(H2bcpza)(CO)3] (5) and [Ru(H2bcpza)Cl(CO)2] (6a/6b) are partially soluble in water but readily soluble in PBS buffer.


Subject(s)
Coordination Complexes , Acetic Acid , Coordination Complexes/chemistry , Crystallography, X-Ray , Ligands , Solubility , Water
4.
Dalton Trans ; 51(10): 3902-3912, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35168247

ABSTRACT

The hetereoscorpionate ligands bis(pyrazol-1-yl)acetic acid (Hbpza) and bis(3,5-dimethylpyrazol-1-yl)acetic acid (Hbdmpza) are reacted with [Sn(OAc)2] or [Sn(acac)2] to yield the corresponding Sn(II) complexes. A single crystal X-ray determination for the in solution monomeric complex [Sn(bpza)2] (1a) revealed a dinuclear molecular structure [Sn2(bpza)4] (1b), with κ3-N,N,O-coordinated bpza ligands at each of the Sn(II) and two bpza ligands µ-bridging between the Sn(II) centres. The molecular structure of [Sn(bdmpza)2] (2) exhibits a homoleptic bisligand complex with both ligands displaced by the free electron pair, which is proven by DFT calculations. Oxidation of complex 2 in an attempt to synthesize a homoleptic Sn(IV) complex leads to the formation of [Sn(bdmpza)F3] (3). In addition, homoleptic bisligand gallium complexes [Ga(bdmpza)2][ClO4] (4) and [Ga(bpza)2][GaCl4] (5) were prepared and characterized by 71Ga NMR and IR spectroscopy as well as by X-ray crystallography.

5.
Kidney Int ; 100(1): 122-137, 2021 07.
Article in English | MEDLINE | ID: mdl-33705825

ABSTRACT

Kidney fibrosis is characterized by the development of myofibroblasts originating from resident kidney and immigrating cells. Myofibroblast formation and extracellular matrix production during kidney damage are triggered by various cytokines. Among these, transforming growth factor ß1 (TGFß1) is considered a central trigger for kidney fibrosis. We found a highly upregulated expression of TGFß1 and TGFß receptor 2 (TGFß-R2) mRNAs in kidney interstitial cells in experimental fibrosis. Here, we investigated the contribution of TGFß1 signaling in resident kidney interstitial cells to organ fibrosis using the models of adenine induced nephropathy and unilateral ureteral occlusion in mice. For this purpose TGFß1 signaling was interrupted by inducible deletion of the TGFß-R2 gene in interstitial cells expressing the fibroblast marker platelet derived growth factor receptor-ß. Expression of profibrotic genes was attenuated up to 50% in kidneys lacking TGFß-R2 in cells positive for platelet derived growth factor receptor-ß. Additionally, deletion of TGFß-R2 prevented the decline of erythropoietin production in ureter ligated kidneys. Notably, fibrosis associated expression of α-smooth muscle actin as a myofibroblast marker and deposits of extracellular collagens were not altered in mice with targeted deletion of TGFß-R2. Thus, our findings suggest an enhancing effect of TGFß1 signaling in resident interstitial cells that contributes to profibrotic gene expression and the downregulation of erythropoietin production, but not to the development of myofibroblasts during kidney fibrosis.


Subject(s)
Erythropoietin , Transforming Growth Factor beta1 , Animals , Fibroblasts , Fibrosis , Gene Expression , Kidney/pathology , Mice , Myofibroblasts/pathology , Transforming Growth Factor beta , Transforming Growth Factor beta1/genetics
6.
Dalton Trans ; 49(37): 13134-13141, 2020 Sep 29.
Article in English | MEDLINE | ID: mdl-32930692

ABSTRACT

Carbon-rich ruthenium allenylidene complexes bearing either a hexaarylbenzene (HAB) or a hexa-peri-hexabenzocoronene (HBC) substituent were synthesised. This was achieved via the corresponding propargyl alcohols with HAB and HBC substituents, which were accessible via 3 or 4 step reaction cascades. Reaction of the propargyl alcohols HC[triple bond, length as m-dash]C(OH)Ph(HAB) and HC[triple bond, length as m-dash]C(OH)Ph(HBC) with [RuCl(η5-C5H5)(PPh3)2] yielded the complexes [Ru(η5-C5H5)([double bond, length as m-dash]C[double bond, length as m-dash]C[double bond, length as m-dash]C(HAB)(Ph))(PPh3)2]PF6 and [Ru(η5-C5H5)([double bond, length as m-dash]C[double bond, length as m-dash]C[double bond, length as m-dash]C(HBC)(Ph))(PPh3)2]PF6. The latter of which shows interesting π-π-stacking behaviour in the solid state as well as aggregation in solution.

7.
Kidney Int ; 96(2): 378-396, 2019 08.
Article in English | MEDLINE | ID: mdl-31146971

ABSTRACT

Prolyl hydroxylase domain enzyme inhibitors (PHDIs) stabilize hypoxia-inducible factors (HIFs), and are protective in models of acute ischemic and inflammatory kidney disease. Whether PHDIs also confer protection in chronic inflammatory kidney disease models remains unknown. Here we investigated long-term effects of PHDI treatment in adenine-induced nephropathy as a model for chronic tubulointerstitial nephritis. After three weeks, renal dysfunction and tubulointerstitial damage, including proximal and distal tubular injury, tubular dilation and renal crystal deposition were significantly attenuated in PHDI-treated (the isoquinoline derivative ICA and Roxadustat) compared to vehicle-treated mice with adenine-induced nephropathy. Crystal-induced renal fibrosis was only partially diminished by treatment with ICA. Renoprotective effects of ICA treatment could not be attributed to changes in adenine metabolism or urinary excretion of the metabolite 2,8-dihydroxyadenine. ICA treatment reduced inflammatory infiltrates of F4/80+ mononuclear phagocytes in the kidneys and supported a regulatory, anti-inflammatory immune response. Furthermore, interstitial deposition of complement C1q was decreased in ICA-treated mice fed an adenine-enriched diet. Tubular cell-specific HIF-1α and myeloid cell-specific HIF-1α and HIF-2α expression were not required for the renoprotective effects of ICA. In contrast, depletion of mononuclear phagocytes with clodronate largely abolished the nephroprotective effects of PHD inhibition. Thus, our findings indicate novel and potent systemic anti-inflammatory properties of PHDIs that confer preservation of kidney function and structure in chronic tubulointerstitial inflammation and might counteract kidney disease progression.


Subject(s)
Nephritis, Interstitial/drug therapy , Phagocytes/drug effects , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Renal Insufficiency, Chronic/prevention & control , Adenine/metabolism , Adenine/toxicity , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Clodronic Acid/pharmacology , Complement C1q/immunology , Complement C1q/metabolism , Disease Models, Animal , Glycine/analogs & derivatives , Glycine/pharmacology , Glycine/therapeutic use , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Kidney Tubules/cytology , Kidney Tubules/drug effects , Kidney Tubules/immunology , Kidney Tubules/pathology , Male , Mice , Mice, Transgenic , Nephritis, Interstitial/blood , Nephritis, Interstitial/chemically induced , Nephritis, Interstitial/immunology , Phagocytes/immunology , Prolyl Hydroxylases/immunology , Prolyl-Hydroxylase Inhibitors/therapeutic use , Protective Agents/pharmacology , Protective Agents/therapeutic use , Renal Insufficiency, Chronic/immunology
8.
ChemMedChem ; 14(1): 94-99, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30380199

ABSTRACT

Prolyl hydroxylation domain (PHD) enzymes catalyze the hydroxylation of the transcription factor hypoxia-inducible factor (HIF) and serve as cellular oxygen sensors. HIF and the PHD enzymes regulate numerous potentially tissue-protective target genes which can adapt cells to metabolic and ischemic stress. We describe a fluorescent PHD inhibitor (1-chloro-4-hydroxybenzo[g]isoquinoline-3-carbonyl)glycine which is suited to fluorescence-based detection assays and for monitoring PHD inhibitors in biological systems. In cell-based assays, application of the fluorescent PHD inhibitor allowed co-localization with a cellular PHD enzyme and led to live cell imaging of processes involved in cellular oxygen sensing.


Subject(s)
Benzylisoquinolines/pharmacology , Fluorescent Dyes/pharmacology , Molecular Imaging/methods , Optical Imaging/methods , Prolyl Hydroxylases/metabolism , Prolyl-Hydroxylase Inhibitors/pharmacology , Benzylisoquinolines/chemical synthesis , Benzylisoquinolines/chemistry , Biocatalysis/drug effects , Dose-Response Relationship, Drug , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Molecular Structure , Prolyl-Hydroxylase Inhibitors/chemical synthesis , Prolyl-Hydroxylase Inhibitors/chemistry , Structure-Activity Relationship
9.
Kidney Int ; 93(3): 656-669, 2018 03.
Article in English | MEDLINE | ID: mdl-29241624

ABSTRACT

Primary/secondary hyperoxalurias involve nephrocalcinosis-related chronic kidney disease (CKD) leading to end-stage kidney disease. Mechanistically, intrarenal calcium oxalate crystal deposition is thought to elicit inflammation, tubular injury and atrophy, involving the NLRP3 inflammasome. Here, we found that mice deficient in NLRP3 and ASC adaptor protein failed to develop nephrocalcinosis, compromising conclusions on nephrocalcinosis-related CKD. In contrast, hyperoxaluric wild-type mice developed profound nephrocalcinosis. NLRP3 inhibition using the ß-hydroxybutyrate precursor 1,3-butanediol protected such mice from nephrocalcinosis-related CKD. Interestingly, the IL-1 inhibitor anakinra had no such effect, suggesting IL-1-independent functions of NLRP3. NLRP3 inhibition using 1,3-butanediol treatment induced a shift of infiltrating renal macrophages from pro-inflammatory (CD45+F4/80+CD11b+CX3CR1+CD206-) and pro-fibrotic (CD45+F4/80+CD11b+CX3CR1+CD206+TGFß+) to an anti-inflammatory (CD45+F4/80+CD11b+CD206+TGFß-) phenotype, and prevented renal fibrosis. Finally, in vitro studies with primary murine fibroblasts confirmed the non-redundant role of NLRP3 in the TGF-ß signaling pathway for fibroblast activation and proliferation independent of the NLRP3 inflammasome complex formation. Thus, nephrocalcinosis-related CKD involves NLRP3 but not necessarily via intrarenal IL-1 release but rather via other biological functions including TGFR signaling and macrophage polarization. Hence, NLRP3 may be a promising therapeutic target in hyperoxaluria and nephrocalcinosis.


Subject(s)
Cell Plasticity , Hyperoxaluria/metabolism , Inflammasomes/metabolism , Interleukin-1/metabolism , Kidney/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephrocalcinosis/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Butylene Glycols/pharmacology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Cell Plasticity/drug effects , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Hyperoxaluria/drug therapy , Hyperoxaluria/immunology , Hyperoxaluria/pathology , Inflammasomes/drug effects , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-1/immunology , Kidney/immunology , Kidney/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nephrocalcinosis/immunology , Nephrocalcinosis/pathology , Nephrocalcinosis/prevention & control , Phenotype , Receptors, Transforming Growth Factor beta/metabolism , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/prevention & control , Signal Transduction
10.
Chemistry ; 23(52): 12815-12824, 2017 Sep 18.
Article in English | MEDLINE | ID: mdl-28703303

ABSTRACT

Isopenicillin N synthase (IPNS) catalyses the four-electron oxidation of a tripeptide, l-δ-(α-aminoadipoyl)-l-cysteinyl-d-valine (ACV), to give isopenicillin N (IPN), the first-formed ß-lactam in penicillin and cephalosporin biosynthesis. IPNS catalysis is dependent upon an iron(II) cofactor and oxygen as a co-substrate. In the absence of substrate, the carbonyl oxygen of the side-chain amide of the penultimate residue, Gln330, co-ordinates to the active-site metal iron. Substrate binding ablates the interaction between Gln330 and the metal, triggering rearrangement of seven C-terminal residues, which move to take up a conformation that extends the final α-helix and encloses ACV in the active site. Mutagenesis studies are reported, which probe the role of the C-terminal and other aspects of the substrate binding pocket in IPNS. The hydrophobic nature of amino acid side-chains around the ACV binding pocket is important in catalysis. Deletion of seven C-terminal residues exposes the active site and leads to formation of a new type of thiol oxidation product. The isolated product is shown by LC-MS and NMR analyses to be the ene-thiol tautomer of a dithioester, made up from two molecules of ACV linked between the thiol sulfur of one tripeptide and the oxidised cysteinyl ß-carbon of the other. A mechanism for its formation is proposed, supported by an X-ray crystal structure, which shows the substrate ACV bound at the active site, its cysteinyl ß-carbon exposed to attack by a second molecule of substrate, adjacent. Formation of this product constitutes a new mode of reaction for IPNS and non-heme iron oxidases in general.


Subject(s)
Aldehydes/metabolism , Esters/metabolism , Oxidoreductases/metabolism , Sulfhydryl Compounds/chemistry , Aldehydes/chemistry , Binding Sites , Biocatalysis , Catalytic Domain , Cephalosporins/biosynthesis , Cephalosporins/chemistry , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Esters/chemistry , Iron/chemistry , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Conformation , Mutagenesis , Oxidation-Reduction , Oxidoreductases/genetics , Oxygen/chemistry , Oxygen/metabolism , Penicillins/biosynthesis , Penicillins/chemistry , Substrate Specificity
11.
Chem Commun (Camb) ; 53(25): 3579-3582, 2017 Mar 23.
Article in English | MEDLINE | ID: mdl-28289745

ABSTRACT

Artificial enzymes are required to catalyse non-natural reactions. Here, a hybrid catalyst was developed by embedding a novel Ru complex in the transport protein NikA. The protein scaffold activates the bound Ru complex to produce a catalyst with high regio- and stereo-selectivity. The hybrid efficiently and stably produced α-hydroxy-ß-chloro chlorohydrins from alkenes (up to 180 TON with a TOF of 1050 h-1).


Subject(s)
Alkenes/chemistry , Chlorohydrins/chemistry , Ruthenium/chemistry , Catalysis , Coordination Complexes/chemistry , Molecular Conformation , Spectroscopy, Fourier Transform Infrared , Stereoisomerism
12.
Kidney Int ; 91(3): 616-627, 2017 03.
Article in English | MEDLINE | ID: mdl-27927598

ABSTRACT

Chronic kidney disease (CKD) is associated with increased risk and worse prognosis of cardiovascular disease, including peripheral artery disease. An impaired angiogenic response to ischemia may contribute to poor outcomes of peripheral artery disease in patients with CKD. Hypoxia inducible factors (HIF) are master regulators of angiogenesis and therefore represent a promising target for therapeutic intervention. To test this we induced hind-limb ischemia in rats with CKD caused by 5/6 nephrectomy and administered two different treatments known to stabilize HIF protein in vivo: carbon monoxide and a pharmacological inhibitor of prolyl hydroxylation 2-(1-chloro-4- hydroxyisoquinoline-3-carboxamido) acetate (ICA). Expression levels of pro-angiogenic HIF target genes (Vegf, Vegf-r1, Vegf-r2, Ho-1) were measured by qRT-PCR. Capillary density was measured by CD31 immunofluorescence staining and HIF expression was evaluated by immunohistochemistry. Capillary density in ischemic skeletal muscle was significantly lower in CKD animals compared to sham controls. Rats with CKD showed significantly lower expression of HIF and all measured pro-angiogenic HIF target genes, including VEGF. Both HIF stabilizing treatments rescued HIF target gene expression in animals with CKD and led to significantly higher ischemia-induced capillary sprouting compared to untreated controls. ICA was effective regardless of whether it was administered before or after induction of ischemia and led to a HIF expression in skeletal muscle. Thus, impaired ischemia-induced angiogenesis in rats with CKD can be improved by HIF stabilization, even if started after onset of ischemia.


Subject(s)
Capillaries/drug effects , Carbon Monoxide/pharmacology , Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/drug therapy , Isoquinolines/pharmacology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Renal Insufficiency, Chronic/metabolism , Signal Transduction/drug effects , Animals , Capillaries/metabolism , Capillaries/physiopathology , Cell Line , Disease Models, Animal , Gene Expression Regulation , Glycine/pharmacology , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Hindlimb , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Ischemia/physiopathology , Male , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Protein Stability , Rats, Sprague-Dawley , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
J Am Soc Nephrol ; 28(3): 761-768, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27612997

ABSTRACT

Intrarenal crystals trigger inflammation and renal cell necroptosis, processes that involve TNF receptor (TNFR) signaling. Here, we tested the hypothesis that TNFRs also have a direct role in tubular crystal deposition and progression of hyperoxaluria-related CKD. Immunohistochemical analysis revealed upregulated tubular expression of TNFR1 and TNFR2 in human and murine kidneys with calcium oxalate (CaOx) nephrocalcinosis-related CKD compared with controls. Western blot and mRNA expression analyses in mice yielded consistent data. When fed an oxalate-rich diet, wild-type mice developed progressive CKD, whereas Tnfr1-, Tnfr2-, and Tnfr1/2-deficient mice did not. Despite identical levels of hyperoxaluria, Tnfr1-, Tnfr2-, and Tnfr1/2-deficient mice also lacked the intrarenal CaOx deposition and tubular damage observed in wild-type mice. Inhibition of TNFR signaling prevented the induced expression of the crystal adhesion molecules, CD44 and annexin II, in tubular epithelial cells in vitro and in vivo, and treatment with the small molecule TNFR inhibitor R-7050 partially protected hyperoxaluric mice from nephrocalcinosis and CKD. We conclude that TNFR signaling is essential for CaOx crystal adhesion to the luminal membrane of renal tubules as a fundamental initiating mechanism of oxalate nephropathy. Furthermore, therapeutic blockade of TNFR might delay progressive forms of nephrocalcinosis in oxalate nephropathy, such as primary hyperoxaluria.


Subject(s)
Hyperoxaluria/complications , Kidney Calculi/etiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Animals , Crystallization , Humans , Hyperoxaluria/metabolism , Mice , Mice, Inbred C57BL
14.
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
15.
Interact Cardiovasc Thorac Surg ; 22(5): 561-70, 2016 05.
Article in English | MEDLINE | ID: mdl-26819270

ABSTRACT

OBJECTIVES: The development of transplant arteriosclerosis, the hallmark feature of heart transplant rejection, is associated with a chronic immune response and also influenced by an initial injury to the graft through ischaemia and reperfusion. Hypoxia-inducible transcription factor (HIF)-1 pathway signalling has a protective effect against ischaemia-reperfusion injury and has already been demonstrated to ameliorate allograft nephropathy in previous animal studies. Therefore, the aim of this study was to investigate the effect of stabilization of hypoxia-inducible transcription factors with a prolyl-hydroxylase domain (PHD) inhibitor on transplant arteriosclerosis in an experimental aortic allograft model. METHODS: MHC-class I mismatched C.B10-H2(b)/LilMcdJ donor thoracic aortas were heterotopically transplanted into the abdominal aorta of BALB/c mice. Donor animals received a single dose of the PHD inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate (ICA) (40 mg/kg) or vehicle i.p. 4 h before transplantation. Intragraft HIF accumulation after ICA treatment was detected by immunohistochemistry before and after cold ischaemia (n = 5). Grafts were harvested 30 days after transplantation and analysed by histology (n = 7) and immunofluorescence (n = 7). In addition, intragraft mRNA expression for cytokines, adhesion molecules and growth factors was determined on Day 14 (n = 7). RESULTS: Donor preconditioning with ICA resulted in HIF accumulation in the aorta and induction of the HIF target genes vascular endothelial growth factor and transforming growth factor-beta. Vascular lesions were present in both experimental groups. However, there was significantly reduced intimal proliferation in preconditioned grafts when compared with vehicle controls [intimal proliferation 31.3 ± 8% (ICA) vs 55.3 ± 20% (control), P < 0.01]. In addition, experimental groups revealed a down-regulation of E-selectin (-57%) and MCP1 (-33%) expression after ICA pretreatment compared with controls, going along with decreased T-cell [1.4% CD4+ T-cell infiltration vs 8.4% (control) and 4.9% CD8+ T-cell infiltration vs 10.7% (control)], dendritic cell (0.6% dendritic cells infiltration vs 1.9% infiltration(control)] and macrophage infiltration [4.8% macrophages (ICA) vs 10.9% (control)] within vascular grafts. CONCLUSIONS: These data of an animal transplant model show that the pharmaceutical activation of HIF with endogenous up-regulation of protective target genes leads to adaptation of the graft to low oxygen-saturation and hereby attenuates the development of transplant arteriosclerosis and allograft injury. Pharmaceutical inhibition of PHDs appears to be a very attractive strategy for organ preservation that deserves further clinical evaluation.


Subject(s)
Aorta, Abdominal/transplantation , Arteriosclerosis/drug therapy , Gene Expression Regulation , Hypoxia-Inducible Factor 1/genetics , Prolyl-Hydroxylase Inhibitors/pharmacology , Allografts , Animals , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Disease Models, Animal , Hypoxia-Inducible Factor 1/metabolism , Immunohistochemistry , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors
16.
Kidney Int ; 88(6): 1283-1292, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26200943

ABSTRACT

Reduced nephron number predisposes to hypertension and kidney disease. Interaction of the branching ureteric bud and surrounding mesenchymal cells determines nephron number. Since oxygen supply may be critical for intrauterine development, we tested whether hypoxia and hypoxia-inducible factor-1α (HIF-1α) influence nephrogenesis. We found that HIF-1α is required for branching of MDCK cells. In addition, culture of metanephric mouse kidneys with ureteric bud cell-specific stabilization or knockout of HIF-1α revealed a positive impact of HIF-1α on nephrogenesis. In contrast, widespread stabilization of HIF-1α in metanephric kidneys through hypoxia or HIF stabilizers impaired nephrogenesis, and pharmacological HIF inhibition enhanced nephrogenesis. Several lines of evidence suggest an inhibitory effect through the hypoxia response of mesenchymal cells. HIF-1α was expressed in mesenchymal cells during nephrogenesis. Expression of the anti-branching factors Bmp4 and Vegfa, secreted by mesenchymal cells, was increased upon HIF stabilization. The conditioned medium from hypoxic metanephric kidneys inhibited MDCK branching, which was partially rescued by Vegfa antibodies. Thus, the effect of HIF-1α on nephrogenesis appears context dependent. While HIF-1α in the ureteric bud is of importance for proper branching morphogenesis, the net effect of hypoxia-induced HIF activation in the embryonic kidney appears to be mesenchymal cell-dependent inhibition of ureter branching.

17.
Pflugers Arch ; 467(10): 2141-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25578858

ABSTRACT

Several genetically modified mouse models implicated that prolyl-4-hydroxylase domain (PHD) enzymes are critical mediators for protecting tissues from an ischemic insult including myocardial infarction by affecting the stability and activation of hypoxia-inducible factor (HIF)-1 and HIF-2. Thus, the current efforts to develop small-molecule PHD inhibitors open a new therapeutic option for myocardial tissue protection during ischemia. Therefore, we aimed to investigate the applicability and efficacy of pharmacological HIFα stabilization by a small-molecule PHD inhibitor in the heart. We tested for protective effects in the acute phase of myocardial infarction after pre- or post-conditional application of the inhibitor. Application of the specific PHD inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate (ICA) resulted in HIF-1α and HIF-2α accumulation in heart muscle cells in vitro and in vivo. The rapid and robust responsiveness of cardiac tissue towards ICA was further confirmed by induction of the known HIF target genes heme oxygenase-1 and PHD3. Pre- and post-conditional treatment of mice undergoing myocardial infarction resulted in a significantly smaller infarct size. Tissue protection from ischemia after pre- or post-conditional ICA treatment demonstrates that there is a therapeutic time window for the application of the PHD inhibitor (PHI) post-myocardial infarction, which might be exploited for acute medical interventions.


Subject(s)
Cardiotonic Agents/therapeutic use , Glycine/analogs & derivatives , Isoquinolines/pharmacology , Myocardial Infarction/drug therapy , Prolyl-Hydroxylase Inhibitors/therapeutic use , Animals , Glycine/pharmacology , Glycine/therapeutic use , Hypoxia-Inducible Factor 1/metabolism , Ischemic Postconditioning , Ischemic Preconditioning, Myocardial , Isoquinolines/therapeutic use , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Transcription Factors/metabolism
18.
Inorg Chem ; 53(23): 12305-14, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25393757

ABSTRACT

The synthesis and structural characterization of new coordination polymers with the N,N-donor ligand trans-1,2-bis(N-methylimidazol-2-yl)ethylene (trans-bie) are reported. It was found that the acetate-bridged paddlewheel metal(II) complexes [M2(O2CCH3)4(trans-bie)]n with M = Rh, Ru, Mo, and Cr are linked by the trans-bie ligand to give a one-dimensional alternating chain. The metal-metal multiple bonds were analyzed with density functional theory and CASSCF/CASPT2 calculations (bond orders: Rh, 0.8; Ru, 1.7; Mo, 3.3).

19.
Chemistry ; 20(37): 11863-9, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25077463

ABSTRACT

Novel κ(3) -N,N,O ligands tend to form 1D coordination polymer strands. Deposition of 1D structures on highly oriented pyrolytic graphite (HOPG) was achieved from diluted solutions and polymer strands have been studied on HOPG by AFM/STM. Single strands were mapped by STM and their electronic properties were subsequently characterized by current imaging tunneling spectroscopy (CITS). Periodic density functional calculations simulating a polymer strand deposited on a HOPG surface are in agreement with the zig-zag structure indicated by experimental findings. Both the observed periodicity and the Zn-Zn distances can be reproduced in the simulations. Van der Waals interactions were found to play a major role for the geometry of the isolated polymer strand, for the adsorption geometry on HOPG, as well as for the adsorption energy.


Subject(s)
Graphite/chemistry , Imidazoles/chemistry , Microscopy, Scanning Tunneling/methods , Ligands , Surface Properties
20.
J Am Soc Nephrol ; 25(3): 465-74, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24203996

ABSTRACT

Polycystic kidney diseases are characterized by numerous bilateral renal cysts that continuously enlarge and, through compression of intact nephrons, lead to a decline in kidney function over time. We previously showed that cyst enlargement is accompanied by regional hypoxia, which results in the stabilization of hypoxia-inducible transcription factor-1α (HIF-1α) in the cyst epithelium. Here we demonstrate a correlation between cyst size and the expression of the HIF-1α-target gene, glucose transporter 1, and report that HIF-1α promotes renal cyst growth in two in vitro cyst models-principal-like MDCK cells (plMDCKs) within a collagen matrix and cultured embryonic mouse kidneys stimulated with forskolin. In both models, augmenting HIF-1α levels with the prolyl hydroxylase inhibitor 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetate enhanced cyst growth. In addition, inhibition of HIF-1α degradation through tubule-specific knockdown of the von Hippel-Lindau tumor suppressor increased cyst size in the embryonic kidney cyst model. In contrast, inhibition of HIF-1α by chetomin and knockdown of HIF-1α both decreased cyst growth in these models. Consistent with previous reports, plMDCK cyst enlargement was driven largely by transepithelial chloride secretion, which consists, in part, of a calcium-activated chloride conductance. plMDCKs deficient for HIF-1α almost completely lacked calcium-activated chloride secretion. We conclude that regional hypoxia in renal cysts contributes to cyst growth, primarily due to HIF-1α-dependent calcium-activated chloride secretion. These findings identify the HIF system as a novel target for inhibition of cyst growth.


Subject(s)
Chlorides/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Polycystic Kidney Diseases/etiology , Animals , Chloride Channels/metabolism , Dogs , Female , Gene Expression Regulation , Glucose Transport Proteins, Facilitative/metabolism , Hypoxia/physiopathology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred C57BL , Polycystic Kidney Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...