Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Pediatr Orthop ; 41(1): e7-e13, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32804861

ABSTRACT

BACKGROUND: Treatment of symptomatic hip dysplasia in skeletally mature patients with spastic cerebral palsy (CP) can be challenging. This study examines our technical experience with the Bernese periacetabular osteotomy (PAO) in combination with adjunctive procedures in the treatment of this complex hip deformity. METHODS: Sixteen consecutive patients (18 hips) with symptomatic CP hip dysplasia were treated with a PAO and variable adjunctive procedures and retrospectively reviewed. Two patient (2 hips) were excluded due to insufficient follow-up. The average age at the time of surgery was 17.7 years (range: 13 to 28 y). We compared the preoperative to postoperative changes in radiographic parameters as well as early outcomes as measured by patient assessment of hip pain and function using the modified Harris Hip Score (mHHS). RESULTS: The average time of follow-up was 3.3 years (range: 2.0 to 6.3 y). Tönnis angles decreased from a median of 30 degrees (range: 18 to 45 degrees) preoperatively to a median of 6 degrees (range: -9 to 21 degrees) postoperatively. Lateral center-edge angles increased from a median of -8 degrees (range: -28 to 15 degrees) to a median of 32 degrees (range: 19 to 38 degrees). Anterior center-edge angles increased from a median of 2 degrees (range: -22 to 39 degrees) to a median of 35 degrees (range: 22 to 47 degrees). The extrusion index decreased from a median of 57% preoperatively (range: 35% to 73%) to a median of 21% (range: 11% to 36%) postoperatively.The median mHHS was 62 (range: 37 to 81) preoperatively and 85 (range: 65 to 100) postoperatively. Notably, the pain component of the mHHS improved from 20 (range: 0 to 44) to 42 (range: 30 to 44). Tönnis osteoarthritis grade preoperatively was either 0 (11 hips) or 1 (5 hips) and remained unchanged in 11 hips and increased by 1 grade in 5 hips. CONCLUSIONS: It has been our experience that the Bernese PAO in combination with appropriate adjunctive treatments has provided a very satisfactory surgical approach in the treatment of CP hip dysplasia. In the adolescent and young adult with spastic CP, utilizing the Bernese PAO technique makes it possible to obtain redirection of often a very severe acetabular dysplasia. Adjunctive soft tissue procedures and a proximal femoral osteotomy are frequently necessary to maintain postoperative stability. A notable improvement in the quality of life and function directly attributable to our surgical treatment of their pre-existing problematic hip dysplasia has been consistently noted in early follow-up for our patients. LEVEL OF EVIDENCE: Level IV-therapeutic.


Subject(s)
Cerebral Palsy/complications , Femur/surgery , Hip Dislocation/surgery , Osteotomy/methods , Acetabulum/surgery , Adolescent , Adult , Arthralgia , Female , Hip Dislocation/etiology , Humans , Male , Osteotomy/adverse effects , Quality of Life , Radiography , Retrospective Studies , Treatment Outcome , Young Adult
2.
Diabetologia ; 63(5): 1043-1054, 2020 05.
Article in English | MEDLINE | ID: mdl-31974732

ABSTRACT

AIMS/HYPOTHESIS: Evidence that glucose-dependent insulinotropic peptide (GIP) and/or the GIP receptor (GIPR) are involved in cardiovascular biology is emerging. We hypothesised that GIP has untoward effects on cardiovascular biology, in contrast to glucagon-like peptide 1 (GLP-1), and therefore investigated the effects of GIP and GLP-1 concentrations on cardiovascular disease (CVD) and mortality risk. METHODS: GIP concentrations were successfully measured during OGTTs in two independent populations (Malmö Diet Cancer-Cardiovascular Cohort [MDC-CC] and Prevalence, Prediction and Prevention of Diabetes in Botnia [PPP-Botnia]) in a total of 8044 subjects. GLP-1 (n = 3625) was measured in MDC-CC. The incidence of CVD and mortality was assessed via national/regional registers or questionnaires. Further, a two-sample Mendelian randomisation (2SMR) analysis between the GIP pathway and outcomes (coronary artery disease [CAD] and myocardial infarction) was carried out using a GIP-associated genetic variant, rs1800437, as instrumental variable. An additional reverse 2SMR was performed with CAD as exposure variable and GIP as outcome variable, with the instrumental variables constructed from 114 known genetic risk variants for CAD. RESULTS: In meta-analyses, higher fasting levels of GIP were associated with risk of higher total mortality (HR[95% CI] = 1.22 [1.11, 1.35]; p = 4.5 × 10-5) and death from CVD (HR[95% CI] 1.30 [1.11, 1.52]; p = 0.001). In accordance, 2SMR analysis revealed that increasing GIP concentrations were associated with CAD and myocardial infarction, and an additional reverse 2SMR revealed no significant effect of CAD on GIP levels, thus confirming a possible effect solely of GIP on CAD. CONCLUSIONS/INTERPRETATION: In two prospective, community-based studies, elevated levels of GIP were associated with greater risk of all-cause and cardiovascular mortality within 5-9 years of follow-up, whereas GLP-1 levels were not associated with excess risk. Further studies are warranted to determine the cardiovascular effects of GIP per se.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular Diseases/mortality , Gastric Inhibitory Polypeptide/metabolism , Glucose/metabolism , Adult , Aged , Female , Genotype , Glucagon-Like Peptide 1/metabolism , Humans , Male , Middle Aged , Prospective Studies , Receptors, Gastrointestinal Hormone/metabolism
3.
Diabetes ; 69(3): 424-435, 2020 03.
Article in English | MEDLINE | ID: mdl-31806622

ABSTRACT

Central to the development of diabetic macro- and microvascular disease is endothelial dysfunction, which appears well before any clinical sign but, importantly, is potentially reversible. We previously demonstrated that hyperglycemia activates nuclear factor of activated T cells (NFAT) in conduit and medium-sized resistance arteries and that NFAT blockade abolishes diabetes-driven aggravation of atherosclerosis. In this study, we test whether NFAT plays a role in the development of endothelial dysfunction in diabetes. NFAT-dependent transcriptional activity was elevated in skin microvessels of diabetic Akita (Ins2 +/- ) mice when compared with nondiabetic littermates. Treatment of diabetic mice with the NFAT blocker A-285222 reduced NFATc3 nuclear accumulation and NFAT-luciferase transcriptional activity in skin microvessels, resulting in improved microvascular function, as assessed by laser Doppler imaging and iontophoresis of acetylcholine and localized heating. This improvement was abolished by pretreatment with the nitric oxide (NO) synthase inhibitor l-N G-nitro-l-arginine methyl ester, while iontophoresis of the NO donor sodium nitroprusside eliminated the observed differences. A-285222 treatment enhanced dermis endothelial NO synthase expression and plasma NO levels of diabetic mice. It also prevented induction of inflammatory cytokines interleukin-6 and osteopontin, lowered plasma endothelin-1 and blood pressure, and improved mouse survival without affecting blood glucose. In vivo inhibition of NFAT may represent a novel therapeutic modality to preserve endothelial function in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Endothelium, Vascular/drug effects , Microvessels/drug effects , NFATC Transcription Factors/antagonists & inhibitors , Pyrazoles/pharmacology , Acetylcholine/pharmacology , Animals , Blood Pressure/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Endothelin-1/drug effects , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Insulin/genetics , Interleukin-6/metabolism , Iontophoresis , Mice , Microvessels/metabolism , Microvessels/physiopathology , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/metabolism , Nitroprusside/pharmacology , Osteopontin/drug effects , Osteopontin/metabolism , Skin/blood supply , Survival Rate , Ultrasonography, Doppler , Vasodilator Agents/pharmacology
4.
Diab Vasc Dis Res ; 15(4): 302-313, 2018 07.
Article in English | MEDLINE | ID: mdl-29499628

ABSTRACT

AIMS: Despite vast clinical experience linking diabetes and atherosclerosis, the molecular mechanisms leading to accelerated vascular damage are still unclear. Here, we investigated the effects of nuclear factor of activated T-cells inhibition on plaque burden in a novel mouse model of type 2 diabetes that better replicates human disease. METHODS & RESULTS: IGF-II/LDLR-/-ApoB100/100 mice were generated by crossbreeding low-density lipoprotein receptor-deficient mice that synthesize only apolipoprotein B100 (LDLR-/-ApoB100/100) with transgenic mice overexpressing insulin-like growth factor-II in pancreatic ß cells. Mice have mild hyperglycaemia and hyperinsulinaemia and develop complex atherosclerotic lesions. In vivo treatment with the nuclear factor of activated T-cells blocker A-285222 for 4 weeks reduced atherosclerotic plaque area and degree of stenosis in the brachiocephalic artery of IGF-II/LDLR-/-ApoB100/100 mice, as assessed non-invasively using ultrasound biomicroscopy prior and after treatment, and histologically after termination. Treatment had no impact on plaque composition (i.e. muscle, collagen, macrophages). The reduced plaque area could not be explained by effects of A-285222 on plasma glucose, insulin or lipids. Inhibition of nuclear factor of activated T-cells was associated with increased expression of atheroprotective NOX4 and of the anti-oxidant enzyme catalase in aortic vascular smooth muscle cells. CONCLUSION: Targeting the nuclear factor of activated T-cells signalling pathway may be an attractive approach for the treatment of diabetic macrovascular complications.


Subject(s)
Apolipoproteins B/deficiency , Atherosclerosis/prevention & control , Brachiocephalic Trunk/drug effects , Insulin-Like Growth Factor II/deficiency , NFATC Transcription Factors/antagonists & inhibitors , Plaque, Atherosclerotic , Pyrazoles/pharmacology , Receptors, LDL/deficiency , Animals , Apolipoprotein B-100 , Apolipoproteins B/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Brachiocephalic Trunk/metabolism , Brachiocephalic Trunk/pathology , Catalase/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Female , Genetic Predisposition to Disease , Insulin-Like Growth Factor II/genetics , Male , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 4/metabolism , NFATC Transcription Factors/metabolism , Oxidative Stress/drug effects , Phenotype , Receptors, LDL/genetics , Signal Transduction
5.
Nat Commun ; 8(1): 1920, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203863

ABSTRACT

Impaired albumin reabsorption by proximal tubular epithelial cells (PTECs) has been highlighted in diabetic nephropathy (DN), but little is known about the underlying molecular mechanisms. Here we find that ORAI1-3, are preferentially expressed in PTECs and downregulated in patients with DN. Hyperglycemia or blockade of insulin signaling reduces the expression of ORAI1-3. Inhibition of ORAI channels by BTP2 and diethylstilbestrol or silencing of ORAI expression impairs albumin uptake. Transgenic mice expressing a dominant-negative Orai1 mutant (E108Q) increases albuminuria, and in vivo injection of BTP2 exacerbates albuminuria in streptozotocin-induced and Akita diabetic mice. The albumin endocytosis is Ca2+-dependent and accompanied by ORAI1 internalization. Amnionless (AMN) associates with ORAIs and forms STIM/ORAI/AMN complexes after Ca2+ store depletion. STIM1/ORAI1 colocalizes with clathrin, but not with caveolin, at the apical membrane of PTECs, which determines clathrin-mediated endocytosis. These findings provide insights into the mechanisms of protein reabsorption and potential targets for treating diabetic proteinuria.


Subject(s)
Albumins/metabolism , Albuminuria/genetics , Calcium Channels/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Nephropathies/metabolism , Epithelial Cells/metabolism , Kidney Tubules, Proximal/metabolism , ORAI1 Protein/genetics , ORAI2 Protein/genetics , Albumins/drug effects , Albuminuria/metabolism , Anilides/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers , Calcium Channels/metabolism , Case-Control Studies , Caveolins/metabolism , Cell Line , Clathrin/metabolism , Diethylstilbestrol/pharmacology , Down-Regulation , Endocytosis , Epithelial Cells/drug effects , Estrogens, Non-Steroidal/pharmacology , Female , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Kidney/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Middle Aged , Mutation , ORAI1 Protein/antagonists & inhibitors , ORAI1 Protein/metabolism , ORAI2 Protein/antagonists & inhibitors , ORAI2 Protein/metabolism , Renal Reabsorption/drug effects , Renal Reabsorption/genetics , Stromal Interaction Molecule 1/metabolism , Thiadiazoles/pharmacology
6.
Diabetes ; 65(1): 239-54, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26395740

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone with extrapancreatic effects beyond glycemic control. Here we demonstrate unexpected effects of GIP signaling in the vasculature. GIP induces the expression of the proatherogenic cytokine osteopontin (OPN) in mouse arteries via local release of endothelin-1 and activation of CREB. Infusion of GIP increases plasma OPN concentrations in healthy individuals. Plasma endothelin-1 and OPN concentrations are positively correlated in patients with critical limb ischemia. Fasting GIP concentrations are higher in individuals with a history of cardiovascular disease (myocardial infarction, stroke) when compared with control subjects. GIP receptor (GIPR) and OPN mRNA levels are higher in carotid endarterectomies from patients with symptoms (stroke, transient ischemic attacks, amaurosis fugax) than in asymptomatic patients, and expression associates with parameters that are characteristic of unstable and inflammatory plaques (increased lipid accumulation, macrophage infiltration, and reduced smooth muscle cell content). While GIPR expression is predominantly endothelial in healthy arteries from humans, mice, rats, and pigs, remarkable upregulation is observed in endothelial and smooth muscle cells upon culture conditions, yielding a "vascular disease-like" phenotype. Moreover, the common variant rs10423928 in the GIPR gene is associated with increased risk of stroke in patients with type 2 diabetes.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Endothelial Cells/metabolism , Endothelin-1/genetics , Gastric Inhibitory Polypeptide/metabolism , Myocytes, Smooth Muscle/metabolism , Osteopontin/genetics , RNA, Messenger/metabolism , Receptors, Gastrointestinal Hormone/genetics , Aged , Aged, 80 and over , Animals , Aorta/cytology , Blotting, Western , Cardiovascular Diseases/genetics , Carotid Arteries/cytology , Case-Control Studies , Coronary Vessels/cytology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelin-1/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Mice , Mice, Knockout , Microscopy, Confocal , Microvessels/cytology , Middle Aged , Osteopontin/metabolism , Peripheral Arterial Disease/metabolism , Plaque, Atherosclerotic/metabolism , Polymorphism, Single Nucleotide , Rats , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction , Stroke/complications , Stroke/genetics , Stroke/metabolism , Sus scrofa , Swine
7.
Proc Natl Acad Sci U S A ; 113(2): 434-9, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26712011

ABSTRACT

Cyclooxygenase-2 (COX-2) is an inducible enzyme that drives inflammation and is the therapeutic target for widely used nonsteroidal antiinflammatory drugs (NSAIDs). However, COX-2 is also constitutively expressed, in the absence of overt inflammation, with a specific tissue distribution that includes the kidney, gastrointestinal tract, brain, and thymus. Constitutive COX-2 expression is therapeutically important because NSAIDs cause cardiovascular and renal side effects in otherwise healthy individuals. These side effects are now of major concern globally. However, the pathways driving constitutive COX-2 expression remain poorly understood. Here we show that in the kidney and other sites, constitutive COX-2 expression is a sterile response, independent of commensal microorganisms and not associated with activity of the inflammatory transcription factor NF-κB. Instead, COX-2 expression in the kidney but not other regions colocalized with nuclear factor of activated T cells (NFAT) transcription factor activity and was sensitive to inhibition of calcineurin-dependent NFAT activation. However, calcineurin/NFAT regulation did not contribute to constitutive expression elsewhere or to inflammatory COX-2 induction at any site. These data address the mechanisms driving constitutive COX-2 and suggest that by targeting transcription it may be possible to develop antiinflammatory therapies that spare the constitutive expression necessary for normal homeostatic functions, including those important to the cardiovascular-renal system.


Subject(s)
Cyclooxygenase 2/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , Signal Transduction , Transcription, Genetic , Animals , Cyclooxygenase 2/metabolism , Cyclosporine/pharmacology , Cytokines/metabolism , Female , Gene Expression Regulation, Enzymologic/drug effects , Germ-Free Life , Kidney/drug effects , Kidney/metabolism , Lipopolysaccharides/pharmacology , Luciferases/metabolism , Male , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tissue Distribution/drug effects , Transcription, Genetic/drug effects
8.
J Diabetes Res ; 2015: 428473, 2015.
Article in English | MEDLINE | ID: mdl-25918731

ABSTRACT

The pathogenesis of diabetic retinopathy (DR) remains unclear but hyperglycemia is an established risk factor. Endothelial dysfunction and changes in Ca2+ signaling have been shown to precede the onset of DR. We recently demonstrated that high extracellular glucose activates the Ca(2+)/calcineurin-dependent transcription factor NFAT in cerebral arteries and aorta, promoting the expression of inflammatory markers. Here we show, using confocal immunofluorescence, that NFAT is expressed in the endothelium of retinal microvessels and is readily activated by high glucose. This was inhibited by the NFAT blocker A-285222 as well as by the ectonucleotidase apyrase, suggesting a mechanism involving the release of extracellular nucleotides. Acute hyperglycemia induced by an IP-GTT (intraperitoneal glucose tolerance test) resulted in increased NFATc3 nuclear accumulation and NFAT-dependent transcriptional activity in retinal vessels of NFAT-luciferase reporter mice. In both Akita (Ins2(+/-) ) and streptozotocin- (STZ-) induced diabetic mice, NFAT transcriptional activity was elevated in retinal vessels. In vivo inhibition of NFAT with A-285222 decreased the expression of OPN and ICAM-1 mRNA in retinal vessels, prevented a diabetes driven downregulation of anti-inflammatory IL-10 in retina, and abrogated the increased vascular permeability observed in diabetic mice. Results identify NFAT signaling as a putative target for treatment of microvascular complications in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Microvessels/metabolism , NFATC Transcription Factors/metabolism , Retinal Vein/metabolism , Animals , Aorta/metabolism , Calcium/metabolism , Diabetes Complications , Glucose Tolerance Test , Hyperglycemia/metabolism , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microcirculation , Osteopontin/metabolism , Permeability , Pyrazoles/chemistry , Retinal Vessels/pathology , Signal Transduction
9.
J Mol Med (Berl) ; 93(5): 511-21, 2015 May.
Article in English | MEDLINE | ID: mdl-25471481

ABSTRACT

UNLABELLED: ORAI and stromal interaction molecule (STIM) are store-operated channel molecules that play essential roles in human physiology through a coupling mechanism of internal Ca(2+) store to Ca(2+) influx. However, the roles of ORAI and STIM in vascular endothelial cells under diabetic conditions remain unknown. Here, we investigated expression and signalling pathways of ORAI and STIM regulated by high glucose or hyperglycaemia using in vitro cell models, in vivo diabetic mice and tissues from patients. We found that ORAI1-3 and STIM1-2 were ubiquitously expressed in human vasculatures. Their expression was upregulated by chronic treatment with high glucose (HG, 25 mM D-glucose), which was accompanied by enhanced store-operated Ca(2+) influx in vascular endothelial cells. The increased expression was also observed in the aortae from genetically modified Akita diabetic mice (C57BL/6-Ins2(Akita)/J) and streptozocin-induced diabetic mice, and aortae from diabetic patients. HG-induced upregulation of ORAI and STIM genes was prevented by the calcineurin inhibitor cyclosporin A and NFATc3 siRNA. Additionally, in vivo treatment with the nuclear factor of activated T cells (NFAT) inhibitor A-285222 prevented the gene upregulation in Akita mice. However, HG had no direct effects on ORAI1-3 currents and the channel activation process through cytosolic STIM1 movement in the cells co-expressing STIM1-EYFP/ORAIs. We concluded that upregulation of STIM/ORAI through Ca(2+)-calcineurin-NFAT pathway is a novel mechanism causing abnormal Ca(2+) homeostasis and endothelial dysfunction under hyperglycaemia. KEY MESSAGE: ORAI1-3 and STIM1-2 are ubiquitously expressed in vasculatures and upregulated by high glucose. Increased expression is confirmed in Akita (Ins2(Akita)/J) and STZ diabetic mice and patients. Upregulation mechanism is mediated by Ca(2+)/calcineurin/NFATc3 signalling. High glucose has no direct effects on ORAI1-3 channel activity and channel activation process.


Subject(s)
Calcineurin/metabolism , Calcium Channels/metabolism , Calcium/metabolism , Glucose/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction , Animals , Calcium Channels/genetics , Cell Line , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Male , Mice , Mice, Transgenic , Middle Aged , RNA, Messenger , Up-Regulation
10.
PLoS One ; 8(6): e65020, 2014.
Article in English | MEDLINE | ID: mdl-23755169

ABSTRACT

OBJECTIVE OF THE STUDY: Diabetic patients have a much more widespread and aggressive form of atherosclerosis and therefore, higher risk for myocardial infarction, peripheral vascular disease and stroke, but the molecular mechanisms leading to accelerated damage are still unclear. Recently, we showed that hyperglycemia activates the transcription factor NFAT in the arterial wall, inducing the expression of the pro-atherosclerotic protein osteopontin. Here we investigate whether NFAT activation may be a link between diabetes and atherogenesis. METHODOLOGY AND PRINCIPAL FINDINGS: Streptozotocin (STZ)-induced diabetes in apolipoprotein E(-/-) mice resulted in 2.2 fold increased aortic atherosclerosis and enhanced pro-inflammatory burden, as evidenced by elevated blood monocytes, endothelial activation- and inflammatory markers in aorta, and pro-inflammatory cytokines in plasma. In vivo treatment with the NFAT blocker A-285222 for 4 weeks completely inhibited the diabetes-induced aggravation of atherosclerosis, having no effect in non-diabetic mice. STZ-treated mice exhibited hyperglycemia and higher plasma cholesterol and triglycerides, but these were unaffected by A-285222. NFAT-dependent transcriptional activity was examined in aorta, spleen, thymus, brain, heart, liver and kidney, but only augmented in the aorta of diabetic mice. A-285222 completely blocked this diabetes-driven NFAT activation, but had no impact on the other organs or on splenocyte proliferation or cytokine secretion, ruling out systemic immunosuppression as the mechanism behind reduced atherosclerosis. Instead, NFAT inhibition effectively reduced IL-6, osteopontin, monocyte chemotactic protein 1, intercellular adhesion molecule 1, CD68 and tissue factor expression in the arterial wall and lowered plasma IL-6 in diabetic mice. CONCLUSIONS: Targeting NFAT signaling may be a novel and attractive approach for the treatment of diabetic macrovascular complications.


Subject(s)
Atherosclerosis/complications , Atherosclerosis/pathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Disease Progression , NFATC Transcription Factors/antagonists & inhibitors , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/blood , Biomarkers/metabolism , Blood Glucose/metabolism , Body Weight/drug effects , Cholesterol/blood , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Inflammation/pathology , Interleukin-6/blood , Mice, Inbred C57BL , Monocytes/metabolism , NFATC Transcription Factors/metabolism , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Signal Transduction/drug effects , Transcription, Genetic/drug effects
11.
PLoS One ; 8(8): e70465, 2013.
Article in English | MEDLINE | ID: mdl-23936435

ABSTRACT

AIMS: Sight-threatening diabetic retinopathy has been treated with photocoagulation for decades but the mechanisms behind the beneficial clinical effects are poorly understood. One target of irradiation and a potential player in this process is the retinal pigment epithelium (RPE). Here we establish an in vitro model for photocoagulation of human RPE cells. METHODS: ARPE-19 cells were exposed to photocoagulation and studied at various time points up to 168h. Lesion morphology, necrosis and apoptosis were investigated by light microscopy; LIVE/DEAD staining and measurements of lactate dehydrogenase activity; and TUNEL- and ELISA-based quantification of DNA fragments, respectively. Cell migration and proliferation were explored using docetaxel and mitomycin C; temporal and spatial changes in proliferation were assessed by confocal immunofluorescence of proliferating cell nuclear antigen. Gene expression was measured by qPCR. RESULTS: Photocoagulation of ARPE-19 resulted in denaturation of proteins and reproducible lesion formation. A transient peak in necrosis, followed by a peak in apoptosis was observed in cells within the lesions at 6h and 24h, respectively after photocoagulation. Cell proliferation was depressed during the first hours after photocoagulation, back to control levels at 24h and augmented in the following days. These effects were not limited to cells in the lesions, but also evident in neighbouring cells. Changes in cell proliferation during lesion repair were preceded by changes in cell migration. Altered mRNA expression of genes previously implicated in the regulation of cell proliferation (FOS, IL-1ß, IL-8, HMGA2), migration and tissue repairing (TGFBR2, ADAMTS6, TIMP3, CTGF) was observed, as well as increased expression of the alarmin IL33 and the cytoprotective gene HSPA6. CONCLUSIONS: Using a laser system and experimental settings that comply with standards used in clinical practice, we have established a suitable model for in vitro photocoagulation of human RPE cells to isolate their contribution to the beneficial effects of laser treatment.


Subject(s)
Apoptosis/radiation effects , Cell Movement/radiation effects , Cytoprotection/genetics , Gene Expression Regulation/radiation effects , Laser Coagulation , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/surgery , Cell Line , Cell Proliferation/radiation effects , Cytoprotection/radiation effects , Humans , Necrosis , Retinal Pigment Epithelium/metabolism , Time Factors
12.
Arterioscler Thromb Vasc Biol ; 33(10): 2316-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950143

ABSTRACT

OBJECTIVE: Interleukin-19 (IL-19) is a putative Th2, anti-inflammatory interleukin. Its expression and potential role in atherogenesis are unknown. IL-19 is not detected in normal artery and is expressed to a greater degree in plaque from symptomatic versus asymptomatic patients, suggesting a compensatory counter-regulatory function. We tested whether IL-19 could reduce atherosclerosis in susceptible mice and identified plausible mechanisms. APPROACH AND RESULTS: LDLR(-/-) mice fed an atherogenic diet and injected with either 1.0 or 10.0 ng/g per day recombinant mouse IL-19 had significantly less plaque area in the aortic arch compared with controls (P<0.0001). Weight gain, cholesterol, and triglyceride levels were not significantly different. Gene expression in splenocytes from IL-19-treated mice demonstrated immune cell Th2 polarization, with decreased expression of T-bet, interferon-γ, interleukin-1ß, and interleukin-12ß and increased expression of GATA3 and FoxP3 mRNA. A greater percentage of lymphocytes were Th2 polarized in IL-19-treated mice. Cellular characterization of plaque by immunohistochemistry demonstrated that IL-19-treated mice have significantly less macrophage infiltrate compared with controls (P<0.001). Intravital microscopy revealed significantly less leukocyte adhesion in wild-type mice injected with IL-19 and fed an atherogenic diet compared with controls. Treatment of cultured endothelial cells, vascular smooth muscle cells, and bone marrow-derived macrophages with IL-19 resulted in a significant decrease in chemokine mRNA and mRNA stability protein human antigen R. CONCLUSIONS: These data suggest that IL-19 is a potent inhibitor of experimental atherosclerosis, with diverse mechanisms including immune cell polarization, decrease in macrophage adhesion, and decrease in gene expression. This may identify IL-19 as a novel therapeutic to limit vascular inflammation.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Interleukin-10/pharmacology , Aged , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers/blood , Carotid Artery Diseases/immunology , Carotid Artery Diseases/pathology , Cells, Cultured , Cholesterol/blood , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Interleukins/metabolism , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Proteins/pharmacology , Spleen/drug effects , Spleen/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Time Factors , Triglycerides/blood
13.
Gastroenterology ; 143(5): 1352-1360.e7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22841788

ABSTRACT

BACKGROUND & AIMS: The signaling mechanisms that regulate trypsinogen activation and inflammation in acute pancreatitis (AP) are unclear. We explored the involvement of the calcium- and calcineurin-dependent transcription factor nuclear factor of activated T cells (NFAT) in development of AP in mice. METHODS: We measured levels of myeloperoxidase and macrophage inflammatory protein 2 (CXCL2), trypsinogen activation, and tissue damage in the pancreas 24 hours after induction of AP by retrograde infusion of taurocholate into the pancreatic ducts of wild-type, NFAT luciferase reporter (NFAT-luc), and NFATc3-deficient mice. We isolated acinar cells and measured NFAT nuclear accumulation, trypsin activity, and expression of NFAT-regulated genes. RESULTS: Infusion of taurocholate increased the transcriptional activity of NFAT in the pancreas, aorta, lung, and spleen of NFAT-luc mice. Inhibition of NFAT with A-285222 blocked taurocholate-induced activation of NFAT in all organs. A-285222 also reduced taurocholate-induced increases in levels of amylase, myeloperoxidase, and CXCL2; activation of trypsinogen; necrosis of acinar cells; edema; leukocyte infiltration; and hemorrhage in the pancreas. NFATc3-deficient mice were protected from these effects of taurocholate. Similar results were obtained using an l-arginine-induced model of AP. Reverse-transcription polymerase chain reaction and confocal immunofluorescence analyses showed that NFATc3 is expressed by acinar cells. NFATc3 expression was activated by stimuli that increase intracellular calcium levels, and activation was prevented by the calcineurin blocker cyclosporin A or A-285222. Activation of trypsinogen by secretagogues in acinar cells was prevented by pharmacologic inhibition of NFAT signaling or lack of NFATc3. A-285222 also reduced expression of inflammatory cytokines such as CXCL2 in acinar cells. CONCLUSIONS: NFATc3 regulates trypsinogen activation, inflammation, and pancreatic tissue damage during development of AP in mice and might be a therapeutic target.


Subject(s)
Acinar Cells/metabolism , NFATC Transcription Factors/metabolism , Neutrophils/physiology , Pancreatitis/metabolism , Trypsinogen/metabolism , Acinar Cells/drug effects , Amylases/blood , Amylases/drug effects , Animals , Aorta/metabolism , Cell Nucleus/metabolism , Chemokine CXCL2/drug effects , Chemokine CXCL2/metabolism , Lung/metabolism , Mice , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/genetics , Neutrophils/drug effects , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/pathology , Peroxidase/drug effects , Peroxidase/metabolism , Pyrazoles/pharmacology , Signal Transduction , Spleen/metabolism , Statistics, Nonparametric , Taurocholic Acid , Trypsinogen/drug effects
14.
Cardiovasc Res ; 93(3): 414-23, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22116621

ABSTRACT

AIMS: Alternative transcription and splicing of the allograft inflammatory factor-1 (AIF-1) gene results in the expression of two different proteins: AIF-1 and interferon responsive transcript-1 (IRT-1).  Here, we explore the impact of AIF-1 and IRT-1 on vascular smooth muscle cell (VSMC) activation and neointima formation, the mechanisms underlying their alternative splicing, and associations of AIF-1 and IRT-1 mRNA with parameters defining human atherosclerotic plaque phenotype. METHODS AND RESULTS: Translation of AIF-1 and IRT-1 results in different products with contrasting cellular distribution and functions. Overexpression of AIF-1 stimulates migration and proliferation of human VSMCs, whereas IRT-1 exerts opposite effects. Adenoviral infection of angioplasty-injured rat carotid arteries with AdAIF-1 exacerbates intima hyperplasia, whereas infection with AdIRT-1 reduces neointima. Expression of these variants is modulated by changes in nuclear factor of activated T-cells (NFAT) activity.  Pharmacological inhibition of NFAT or targeting of NFATc3 with small interfering RNA (siRNA) lowers the AIF-1/IRT-1 ratio and favours an anti-proliferative outcome.  NFAT acts as a repressor on the IRT-1 transcriptional start site, which is also sensitive to interferon-γ stimulation. Expression of AIF-1 mRNA in human carotid plaques associates with less extracellular matrix and a more pro-inflammatory plaque and plasma profile, features that may predispose to plaque rupture. In contrast, expression of IRT-1 mRNA associates with a less aggressive phenotype and less VSMCs at the most stenotic region of the plaque. CONCLUSION: Inhibition of NFAT signalling, by shifting the AIF-1/IRT-1 ratio, may be an attractive target to regulate the VSMC response to injury and manipulate plaque stability in atherosclerosis.


Subject(s)
Alternative Splicing/physiology , Carotid Artery Diseases , Coronary Artery Disease , DNA-Binding Proteins/genetics , NFATC Transcription Factors/metabolism , Neointima , Angioplasty, Balloon, Coronary/adverse effects , Animals , Calcium-Binding Proteins , Carotid Arteries/pathology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Coronary Restenosis/metabolism , Coronary Restenosis/pathology , Coronary Vessels/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Microfilament Proteins , Muscle, Smooth, Vascular/pathology , Myometrium/blood supply , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
15.
Diabetes ; 60(9): 2424-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21810601

ABSTRACT

OBJECTIVE: The incretin hormone GIP (glucose-dependent insulinotropic polypeptide) promotes pancreatic ß-cell function by potentiating insulin secretion and ß-cell proliferation. Recently, a combined analysis of several genome-wide association studies (Meta-analysis of Glucose and Insulin-Related Traits Consortium [MAGIC]) showed association to postprandial insulin at the GIP receptor (GIPR) locus. Here we explored mechanisms that could explain the protective effects of GIP on islet function. RESEARCH DESIGN AND METHODS: Associations of GIPR rs10423928 with metabolic and anthropometric phenotypes in both nondiabetic (N = 53,730) and type 2 diabetic individuals (N = 2,731) were explored by combining data from 11 studies. Insulin secretion was measured both in vivo in nondiabetic subjects and in vitro in islets from cadaver donors. Insulin secretion was also measured in response to exogenous GIP. The in vitro measurements included protein and gene expression as well as measurements of ß-cell viability and proliferation. RESULTS: The A allele of GIPR rs10423928 was associated with impaired glucose- and GIP-stimulated insulin secretion and a decrease in BMI, lean body mass, and waist circumference. The decrease in BMI almost completely neutralized the effect of impaired insulin secretion on risk of type 2 diabetes. Expression of GIPR mRNA was decreased in human islets from carriers of the A allele or patients with type 2 diabetes. GIP stimulated osteopontin (OPN) mRNA and protein expression. OPN expression was lower in carriers of the A allele. Both GIP and OPN prevented cytokine-induced reduction in cell viability (apoptosis). In addition, OPN stimulated cell proliferation in insulin-secreting cells. CONCLUSIONS: These findings support ß-cell proliferative and antiapoptotic roles for GIP in addition to its action as an incretin hormone. Identification of a link between GIP and OPN may shed new light on the role of GIP in preservation of functional ß-cell mass in humans.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/genetics , Islets of Langerhans/metabolism , Osteopontin/genetics , Alleles , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Genome-Wide Association Study , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Male , Osteopontin/metabolism
16.
J Bone Miner Res ; 25(3): 520-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19821773

ABSTRACT

Children with limited or no ability to ambulate frequently sustain fragility fractures. Joint contractures, scoliosis, hip dysplasia, and metallic implants often prevent reliable measures of bone mineral density (BMD) in the proximal femur and lumbar spine, where BMD is commonly measured. Further, the relevance of lumbar spine BMD to fracture risk in this population is questionable. In an effort to obtain bone density measures that are both technically feasible and clinically relevant, a technique was developed involving dual-energy X-ray absorptiometry (DXA) measures of the distal femur projected in the lateral plane. The purpose of this study is to test the hypothesis that these new measures of BMD correlate with fractures in children with limited or no ability to ambulate. The relationship between distal femur BMD Z-scores and fracture history was assessed in a cross-sectional study of 619 children aged 6 to 18 years with muscular dystrophy or moderate to severe cerebral palsy compiled from eight centers. There was a strong correlation between fracture history and BMD Z-scores in the distal femur; 35% to 42% of those with BMD Z-scores less than -5 had fractured compared with 13% to 15% of those with BMD Z-scores greater than -1. Risk ratios were 1.06 to 1.15 (95% confidence interval 1.04-1.22), meaning a 6% to 15% increased risk of fracture with each 1.0 decrease in BMD Z-score. In clinical practice, DXA measure of BMD in the distal femur is the technique of choice for the assessment of children with impaired mobility.


Subject(s)
Bone Density , Cerebral Palsy/diagnostic imaging , Femur/diagnostic imaging , Fractures, Bone/diagnostic imaging , Muscular Dystrophies/diagnostic imaging , Absorptiometry, Photon , Adolescent , Cerebral Palsy/complications , Child , Disabled Children , Female , Fractures, Bone/complications , Fractures, Bone/etiology , Humans , Male , Muscular Dystrophies/complications
17.
Arterioscler Thromb Vasc Biol ; 30(2): 218-24, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19965778

ABSTRACT

OBJECTIVE: Hyperglycemia is a recognized risk factor for cardiovascular disease in diabetes. Recently, we reported that high glucose activates the Ca(2+)/calcineurin-dependent transcription factor nuclear factor of activated T cells (NFAT) in arteries ex vivo. Here, we sought to determine whether hyperglycemia activates NFAT in vivo and whether this leads to vascular complications. METHODS AND RESULTS: An intraperitoneal glucose-tolerance test in mice increased NFATc3 nuclear accumulation in vascular smooth muscle. Streptozotocin-induced diabetes resulted in increased NFATc3 transcriptional activity in arteries of NFAT-luciferase transgenic mice. Two NFAT-responsive sequences in the osteopontin (OPN) promoter were identified. This proinflammatory cytokine has been shown to exacerbate atherosclerosis and restenosis. Activation of NFAT resulted in increased OPN mRNA and protein in native arteries. Glucose-induced OPN expression was prevented by the ectonucleotidase apyrase, suggesting a mechanism involving the release of extracellular nucleotides. The calcineurin inhibitor cyclosporin A or the novel NFAT blocker A-285222 prevented glucose-induced OPN expression. Furthermore, diabetes resulted in higher OPN expression, which was significantly decreased by in vivo treatment with A-285222 for 4 weeks or prevented in arteries from NFATc3(-/-) mice. CONCLUSIONS: These results identify a glucose-sensitive transcription pathway in vivo, revealing a novel molecular mechanism that may underlie vascular complications of diabetes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/etiology , Hyperglycemia/metabolism , Muscle, Smooth, Vascular/metabolism , NFATC Transcription Factors/metabolism , Osteopontin/metabolism , Animals , Apyrase/pharmacology , Arteries/metabolism , Binding Sites , Calcineurin/metabolism , Calcineurin Inhibitors , Cyclosporine/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/prevention & control , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Glucose Tolerance Test , Humans , Hyperglycemia/complications , Hyperglycemia/drug therapy , Jurkat Cells , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/deficiency , NFATC Transcription Factors/genetics , Osteopontin/deficiency , Osteopontin/genetics , Promoter Regions, Genetic , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Uridine Triphosphate/metabolism
18.
Arterioscler Thromb Vasc Biol ; 29(10): 1465-70, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19755528

ABSTRACT

OBJECTIVE: Vascular inflammation is a key feature of both micro- and macrovascular complications in diabetes. Several lines of evidence have implicated the cytokine tumor necrosis factor (TNF) alpha as an important mediator of inflammation in diabetes. In the present study we evaluated the role of TNF alpha in streptozotocin (STZ)-induced diabetes on vascular inflammation in C57BL/6 wild-type and apoE-/- mice. METHODS AND RESULTS: Diabetes increased the expression of vascular cell adhesion molecule (VCAM)-1 in cerebral arteries 150 m in diameter as well as the macrophage accumulation in aortic root atherosclerotic plaques in apoE-/- mice. A more pronounced vascular inflammatory response was observed in diabetic TNF alpha-deficient apoE-/- mice. These mice were also characterized by increased accumulation of IgG and IgM autoantibodies in atherosclerotic lesions. Diabetes also increased VCAM-1 expression and plaque formation in apoE-competent TNF alpha -/- mice, whereas no such effects were observed in C57BL/6 wild-type mice. CONCLUSIONS: The present findings suggest that TNF alpha does not mediate diabetic-induced vascular inflammation in mice and reveal an unexpected protective role for TNF alpha. These effects are partly attributable to a direct antiinflammatory role of TNF alpha, but may also reflect a defective development of the immune system in these mice.


Subject(s)
Atherosclerosis/etiology , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Inflammation/etiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apolipoproteins E/physiology , Autoantibodies/analysis , Blood Glucose/analysis , Cerebral Arteries/chemistry , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Streptozocin , Vascular Cell Adhesion Molecule-1/blood
19.
J Am Acad Orthop Surg ; 17(6): 369-77, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19474446

ABSTRACT

Volar locking plate fixation via open reduction and internal fixation is an increasingly accepted method for managing displaced distal radius fractures. Volar plating offers biomechanically stable fixation, allows early rehabilitation, and enables fixation of comminuted or osteopenic bone. The literature reporting complications of volar plate fixation is limited primarily to case reports and small case series. The surgeon must be mindful of potential soft-tissue, neurovascular, and osseous complications, such as extensor tendon and flexor tendon injury, flexor pollicis rupture, carpal tunnel syndrome, complex regional pain syndrome, and loss of reduction, as well as hardware failure. Increased awareness of potential complications may lead to more prompt recognition and treatment when they do arise.


Subject(s)
Bone Plates , Fracture Fixation, Internal/instrumentation , Radius Fractures/surgery , Fracture Fixation, Internal/adverse effects , Humans , Prosthesis Failure
SELECTION OF CITATIONS
SEARCH DETAIL
...