Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Diabetologia ; 66(1): 213-222, 2023 01.
Article in English | MEDLINE | ID: mdl-36114428

ABSTRACT

AIMS/HYPOTHESIS: Serum sex hormone-binding globulin (SHBG) has been proposed to act as a hepatokine that contributes to the extrahepatic complications observed in non-alcoholic fatty liver disease (NAFLD). However, it remains uncertain whether serum SHBG mediates the association between intrahepatic lipids (IHL) and type 2 diabetes. Therefore, we studied whether, and to what extent, serum SHBG mediates the association between IHL content and type 2 diabetes. METHODS: We used cross-sectional data from the Maastricht Study (n=1554), a population-based cohort study with oversampling of individuals with type 2 diabetes. Type 2 diabetes status was assessed by oral glucose tolerance test, and IHL content was measured using 3T Dixon MRI. Mediation analyses were performed to assess the role of serum SHBG in mediating the association between IHL content and type 2 diabetes. RESULTS: IHL content was significantly associated with type 2 diabetes in women and men (OR 1.08 [95% CI 1.04, 1.14] and OR 1.12 [95% CI 1.08, 1.17], respectively). Serum SHBG significantly mediated the association between IHL content and type 2 diabetes. The contribution of serum SHBG was higher in women (OR 1.04 [95% CI 1.02, 1.07]; proportion mediated 50.9% [95% CI 26.7, 81.3]) than in men (OR 1.02 [95% CI 1.01, 1.03]; proportion mediated 17.2% [95% CI 9.6, 27.6]). Repeat analyses with proxies of type 2 diabetes and adjustment for covariates did not substantially affect the results. CONCLUSIONS/INTERPRETATION: In this large-scale population-based cohort study, serum SHBG was found to be a mediator of the association between IHL content and type 2 diabetes. These findings extend our understanding of the potential mechanisms by which NAFLD is a risk factor for type 2 diabetes, and further elaborate on the role of SHBG as a hepatokine.


Subject(s)
Diabetes Mellitus, Type 2 , Liver , Sex Hormone-Binding Globulin , Female , Humans , Cohort Studies , Cross-Sectional Studies , Lipids , Male , Liver/metabolism
2.
Nutrients ; 14(22)2022 Nov 21.
Article in English | MEDLINE | ID: mdl-36432614

ABSTRACT

α-Dicarbonyls and advanced glycation end products (AGEs) may contribute to the pathogenesis of insulin resistance by a variety of mechanisms. To investigate whether young insulin-resistant subjects present markers of increased dicarbonyl stress, we determined serum α-dicarbonyls-methylglyoxal, glyoxal, 3-deoxyglucosone; their derived free- and protein-bound, and urinary AGEs using the UPLC/MS-MS method; soluble receptors for AGEs (sRAGE), and cardiometabolic risk markers in 142 (49% females) insulin resistant (Quantitative Insulin Sensitivity Check Index (QUICKI) ≤ 0.319) and 167 (47% females) age-, and waist-to-height ratio-matched insulin-sensitive controls aged 16-to-22 years. The between-group comparison was performed using the two-factor (sex, presence/absence of insulin resistance) analysis of variance; multiple regression via the orthogonal projection to latent structures model. In comparison with their insulin-sensitive peers, young healthy insulin-resistant individuals without diabetes manifest alterations throughout the α-dicarbonyls-AGEs-sRAGE axis, dominated by higher 3-deoxyglucosone levels. Variables of α-dicarbonyls-AGEs-sRAGE axis were associated with insulin sensitivity independently from cardiometabolic risk markers, and sex-specifically. Cleaved RAGE associates with QUICKI only in males; while multiple α-dicarbonyls and AGEs independently associate with QUICKI particularly in females, who displayed a more advantageous cardiometabolic profile compared with males. Further studies are needed to elucidate whether interventions alleviating dicarbonyl stress ameliorate insulin resistance.


Subject(s)
Cardiovascular Diseases , Insulin Resistance , Male , Female , Humans , Glycation End Products, Advanced , Case-Control Studies , Insulin
3.
Sci Rep ; 11(1): 22675, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34811435

ABSTRACT

There has been increasing interest in finding non-invasive biomarkers for neurodegenerative diseases such as Alzheimer's disease (AD). This observational study investigated AD-specific biomarkers in tear fluid. Tear fluid was collected from a total of 65 subjects, including 23 patients with subjective cognitive decline (SCD), 22 patients with mild cognitive impairment (MCI), 11 dementia patients and 9 healthy controls (HC). Levels of amyloid-beta peptides (AB38, AB40, AB42), total-tau (t-tau) and phosphorylated-tau (p-tau) were determined using multiplex immunoassays. Levels of AB40 and t-tau were detectable in the vast majority (> 94%) of tear fluid samples. Cerebrospinal fluid (CSF) was available from a subset of patients. In this group, tear t-tau levels were significantly higher in people with dementia compared to SCD patients. Tear t-tau levels were elevated in patients with neurodegeneration (classified according to the A/T/N system) compared to patients without neurodegeneration. Negative correlations were found between CSF AB42 and CSF t-tau, and between CSF AB42 and tear t-tau. In summary, this study shows the potential of tau proteins in tear fluid to be associated with disease severity and neurodegeneration.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Cognitive Dysfunction/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Severity of Illness Index , Tears/chemistry , tau Proteins/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Alzheimer Disease/epidemiology , Biomarkers/cerebrospinal fluid , Case-Control Studies , Cognitive Dysfunction/epidemiology , Female , Humans , Male , Middle Aged , Netherlands/epidemiology
4.
Food Res Int ; 147: 110547, 2021 09.
Article in English | MEDLINE | ID: mdl-34399524

ABSTRACT

SCOPE: Dietary advanced glycation endproducts (AGEs) are associated with negative biological effects, possibly due to accumulation in plasma and tissues and through modulation of inflammation and gut microbiota. Whether these biological consequences are reversible by limiting dietary AGE intake is unknown. METHODS AND RESULTS: Young healthy C57BL/6 mice were fed a standard chow (n = 10) or a baked chow high AGE-diet (n = 10) (~1.8-6.9 fold increased protein-bound Nε-(carboxymethyl)lysine (CML), Nε-(1-carboxyethyl)lysine (CEL), and Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1)) for 10 weeks or a switch diet with baked chow for 5 weeks followed by 5 weeks of standard chow (n = 10). We assessed accumulation of AGEs in plasma, kidney, and liver and measured inflammatory markers and gut microbial composition. After 10 weeks of baked chow, a substantial panel of AGEs were increased in plasma, liver, and kidney. These increases were normalized after the switch diet. The inflammatory z-score increased after the baked chow diet. Gut microbial composition differed significantly between groups, with enriched Dubosiella spp. dominating these alterations. CONCLUSION: A high AGE-diet led to an increase of AGEs in plasma, kidney, and liver and to more inflammation and modification of the gut microbiota. These effects were reversed or discontinued by a diet lower in AGEs.


Subject(s)
Gastrointestinal Microbiome , Glycation End Products, Advanced , Animals , Diet , Inflammation , Mice , Mice, Inbred C57BL
5.
Diabetes Care ; 44(1): 157-163, 2021 01.
Article in English | MEDLINE | ID: mdl-33144352

ABSTRACT

OBJECTIVE: Diabetes is a risk factor for severe limb ischemia (SLI), a condition associated with high mortality, morbidity, and limb loss. The reactive glucose-derived dicarbonyl methylglyoxal (MGO) is a major precursor for advanced glycation end products (AGEs) and a potential driver of cardiovascular disease. We investigated whether plasma MGO levels are associated with poor outcomes in SLI. RESEARCH DESIGN AND METHODS: We measured plasma levels of MGO, free AGEs, and d-lactate, the detoxification end product of MGO, with ultraperformance liquid chromatography-tandem mass spectrometry at baseline in 160 patients (64.8 ± 13.3 years, 67.5% male, 37.5% with diabetes) with no-option SLI and recorded major adverse outcomes (n = 86, comprising n = 53 deaths and n = 49 amputations [first event counted]) over the 5-year follow-up. Data were analyzed with linear or Cox regression, after Ln-transformation of the independent variables, adjusted for sex, age, trial arm, diabetes, estimated glomerular filtration rate, systolic blood pressure, cholesterol levels, and BMI. Associations are reported per 1 SD plasma marker. RESULTS: Higher plasma MGO levels were associated with more adverse outcomes (relative risk 1.44; 95% CI 1.11-1.86) and amputations separately (1.55; 1.13-2.21). We observed a similar but weaker trend for mortality (1.28; 0.93-1.77). The MGO-derived AGE Nε-(carboxyethyl)lysine was also associated with more adverse outcomes (1.46; 1.00-2.15) and amputations (1.71; 1.04-2.79). d-Lactate was not associated with adverse incident outcomes. Higher plasma MGO levels were also associated with more inflammation and white blood cells and fewer progenitor cells. CONCLUSIONS: Plasma MGO levels are associated with adverse outcomes in SLI. Future studies should investigate whether MGO-targeting therapies improve outcomes in SLI.


Subject(s)
Cardiovascular Diseases , Diabetes Mellitus , Amputation, Surgical , Case-Control Studies , Female , Glycation End Products, Advanced , Humans , Ischemia , Male , Pyruvaldehyde
6.
J Diabetes Res ; 2019: 6289831, 2019.
Article in English | MEDLINE | ID: mdl-31218233

ABSTRACT

Advanced glycation end products (AGEs) accumulate in fatty livers and may contribute to low-grade inflammation (LGI), potentially via their receptor, RAGE. It is unknown if the AGE accumulation in fatty livers results in elevated circulating AGEs. In a cohort study, we investigated the association of liver fat and hepatocellular damage with circulating AGEs and soluble RAGE (sRAGE) and subsequently the association of circulating AGEs and sRAGE with LGI. Cross-sectional associations of liver fat percentage (eLF%; ln-transformed) and liver enzymes (LE score; standardized) with circulating AGEs (free CML, CEL, and MG-H1 in nM and protein-bound CML, CEL, and pentosidine in nmol/mmol lysine; ln-transformed) and sRAGE (pg/ml, ln-transformed) and additionally of AGEs and sRAGE with LGI (standardized) were determined by multiple linear regression. eLF% was positively associated with circulating free CEL (ß = 0.090; 95% CI 0.041; 0.139) but inversely with protein-bound CML (ß = -0.071; 95% CI -0.108; -0.034). Similarly, the LE score was positively associated with free CML (ß = 0.044; 95% CI 0.012; 0.076) and CEL (ß = 0.040; 95% CI 0.009; 0.072) but inversely with protein-bound CML (ß = -0.037; 95% CI -0.060; -0.013). Free CML (ß = 0.297; 95% CI 0.049; 0.545) was positively associated with LGI, while protein-bound CML (ß = -0.547; 95% CI -0.888; -0.207) was inversely associated, although this association was absent after adjustment for BMI. eLF% and LE score were not associated with sRAGE and sRAGE not with LGI after adjustment for BMI. Liver fat and enzymes were positively associated with circulating free AGEs, which were associated with LGI. In contrast, inverse relations were observed of liver fat and enzymes with circulating protein-bound AGEs and of protein-bound AGEs with LGI. These data suggest that hepatic steatosis and inflammation affect the formation and degradation of hepatic protein-bound AGEs resulting in elevated circulating free AGE levels. These alterations in AGE levels might influence LGI, but this is likely independent of RAGE.


Subject(s)
Glycation End Products, Advanced/metabolism , Inflammation , Liver/pathology , Aged , Arginine/analogs & derivatives , Arginine/metabolism , Biomarkers/metabolism , Body Mass Index , Cohort Studies , Cross-Sectional Studies , Fatty Liver/metabolism , Female , Humans , Liver/enzymology , Lysine/analogs & derivatives , Lysine/metabolism , Male , Middle Aged , Oxidative Stress , Receptor for Advanced Glycation End Products/metabolism , Regression Analysis , Treatment Outcome
7.
Sci Rep ; 8(1): 15256, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323247

ABSTRACT

Non-alcoholic fatty liver disease is a spectrum of liver diseases ranging from steatosis only to non-alcoholic steatohepatitis (NASH). The latter is characterized by hepatic inflammation, which increases the risk of cardiovascular disease. It is poorly understood which factors contribute to the onset of hepatic inflammation characterizing the progression from steatosis to NASH. Previously, we demonstrated increased advanced glycation endproducts (AGEs) in the livers of NASH patients. We hypothesise that AGEs play a key role in NASH development by activating their proinflammatory receptor, RAGE. RAGE-deficient mice and wildtype littermates, both on Ldlr-/- background, were fed a Western type diet (WTD) for 3 or 12 weeks. Flow cytometry, histology, gene expression and AGE measurements were performed to evaluate the effects of RAGE deficiency. RAGE-deficient mice displayed reduced weight gain and visceral fat expansion compared to control mice. No difference in adipose tissue inflammation was observed between groups. RAGE deficiency did not affect WTD-induced monocytosis, circulating lipids or hepatic steatosis. WTD-induced hepatic neutrophil and macrophage accumulation and atherosclerotic plaque development was comparable between control and RAGE-deficient mice. No difference in AGE levels was observed. RAGE does not seem to play a major role in the development of NASH or atherosclerosis in a hyperlipidemic mouse model.


Subject(s)
Atherosclerosis/genetics , Inflammation/genetics , Non-alcoholic Fatty Liver Disease/genetics , Receptor for Advanced Glycation End Products/genetics , Receptors, LDL/genetics , Animals , Atherosclerosis/etiology , Atherosclerosis/pathology , Diet, Western/adverse effects , Disease Models, Animal , Glycation End Products, Advanced/genetics , Humans , Inflammation/pathology , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/pathology , Risk Factors
8.
J Clin Endocrinol Metab ; 103(2): 759-767, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29211893

ABSTRACT

Context: Impaired insulin-mediated muscle microvascular recruitment (IMMR) may add to the development of insulin resistance and hypertension. Increased aldosterone levels have been linked to these obesity-related complications in severely to morbidly obese individuals and to impaired microvascular function in experimental studies. Objectives: To investigate whether aldosterone levels are associated with IMMR, insulin sensitivity, and blood pressure in lean and moderately abdominally obese men, and to study the effect of weight loss. Design, Setting, Participants, Intervention, Main Outcome Measures: In 25 lean and 53 abdominally obese men, 24-hour blood pressure measurement was performed, and aldosterone levels were measured using ultra-performance liquid chromatography tandem mass spectrometry. Insulin sensitivity was assessed by determining whole-body glucose disposal during a hyperinsulinemic clamp. IMMR in forearm skeletal muscle was measured with contrast-enhanced ultrasonography. These assessments were repeated in the abdominally obese men following an 8-week weight loss or weight stable period. Results: Sodium excretion and aldosterone levels were similar in lean and abdominally obese participants, but sodium excretion was inversely associated with aldosterone concentration only in the lean individuals [lean, ß/100 mmol sodium excretion (adjusted for age and urinary potassium excretion) = -0.481 (95% confidence interval, -0.949 to -0.013); abdominally obese, ß/100 mmol sodium excretion = -0.081 (95% confidence interval, -0.433 to 0.271); P for interaction = 0.02]. Aldosterone was not associated with IMMR, insulin sensitivity, or blood pressure and was unaffected by weight loss. Conclusion: In moderately abdominally obese men, the inverse relationship between sodium excretion and aldosterone concentration is less than that in lean men but does not translate into higher aldosterone levels. The absolute aldosterone level does not explain differences in microvascular and metabolic insulin sensitivity and blood pressure between lean and moderately abdominally obese men.


Subject(s)
Aldosterone/blood , Insulin Resistance/physiology , Microvessels/metabolism , Muscle, Skeletal/metabolism , Obesity, Abdominal/metabolism , Obesity, Abdominal/therapy , Weight Loss/physiology , Adolescent , Adult , Aged , Blood Glucose/metabolism , Blood Pressure/physiology , Female , Humans , Hypertension/complications , Hypertension/metabolism , Male , Middle Aged , Muscle, Skeletal/blood supply , Obesity, Abdominal/complications , Obesity, Abdominal/physiopathology , Thinness/blood , Thinness/metabolism , Thinness/physiopathology , Weight Reduction Programs , Young Adult
9.
Arterioscler Thromb Vasc Biol ; 35(12): 2707-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26449750

ABSTRACT

OBJECTIVE: Adipose tissue inflammation contributes to the development of complications, such as insulin resistance and type 2 diabetes mellitus. We previously reported that plasma levels of N(ε)-(carboxymethyl)lysine (CML) were decreased in obese subjects resulting from CML accumulation in adipose tissue and that this CML accumulation plays an important role in adipose tissue inflammation. The objective of this study is to investigate associations between obesity (body mass index, waist circumference, and trunk fat mass), plasma CML (as an inversely correlated marker of CML accumulation in adipose tissue), and low-grade inflammation (LGI) in a large sample of individuals whose weight status ranged from normal to morbid obesity. APPROACH AND RESULTS: We studied 1270 individuals of the Cohort on Diabetes and Atherosclerosis Maastricht Study and Hoorn Study, in whom protein-bound CML levels were measured by UPLC-Tandem MS (ultra performance liquid chromatography-tandem mass spectrometry), and 6 inflammatory markers were measured with multiarrays. These inflammatory markers were compiled into an LGI score. Multiple linear regression, adjusted for covariates, showed that (1) waist circumference was inversely associated with protein-bound CML plasma levels (standardized regression coefficient [ß]=-0.357 [95% confidence interval: -0.414; -0.301]); (2) protein-bound CML was inversely associated with LGI score (ß=-0.073 [-0.130;-0.015]); and (3) the association between waist circumference and LGI (ß=0.262 [0.203;0.321]) was attenuated after adjustment for protein-bound CML plasma levels and other potential mediators (to ß=0.202 [0.138;0.266]), with CML explaining the greatest portion of the attenuation (≈12%). Further analysis with dual-energy X-ray absorptiometry measures of body composition confirmed a strong inverse association of fat mass preferentially accumulated in the trunk with protein-bound CML plasma levels, significantly explaining ≈21% of the trunk fat-LGI association. CONCLUSIONS: Obesity, in particular central obesity, is characterized by greater levels of LGI but by lower levels of circulating CML; the latter significantly explaining a portion of the positive association between central obesity and inflammation.


Subject(s)
Inflammation Mediators/blood , Inflammation/blood , Lysine/analogs & derivatives , Obesity, Abdominal/blood , Obesity, Morbid/blood , Absorptiometry, Photon , Adiposity , Adult , Aged , Biomarkers/blood , Body Mass Index , Chromatography, Liquid , Cross-Sectional Studies , Disease Progression , Female , Humans , Inflammation/diagnosis , Inflammation/epidemiology , Linear Models , Lysine/blood , Male , Middle Aged , Multivariate Analysis , Netherlands/epidemiology , Obesity, Abdominal/diagnosis , Obesity, Abdominal/epidemiology , Obesity, Abdominal/physiopathology , Obesity, Morbid/diagnosis , Obesity, Morbid/epidemiology , Obesity, Morbid/physiopathology , Prospective Studies , Risk Factors , Tandem Mass Spectrometry , Waist Circumference
10.
PLoS One ; 8(3): e58576, 2013.
Article in English | MEDLINE | ID: mdl-23472208

ABSTRACT

BACKGROUND: In terms of time, effort and quality, multiplex technology is an attractive alternative for well-established single-biomarker measurements in clinical studies. However, limited data comparing these methods are available. METHODS: We measured, in a large ongoing cohort study (n = 574), by means of both a 4-plex multi-array biomarker assay developed by MesoScaleDiscovery (MSD) and single-biomarker techniques (ELISA or immunoturbidimetric assay), the following biomarkers of low-grade inflammation: C-reactive protein (CRP), serum amyloid A (SAA), soluble intercellular adhesion molecule 1 (sICAM-1) and soluble vascular cell adhesion molecule 1 (sVCAM-1). These measures were realigned by weighted Deming regression and compared across a wide spectrum of subjects' cardiovascular risk factors by ANOVA. RESULTS: Despite that both methods ranked individuals' levels of biomarkers very similarly (Pearson's r all≥0.755) absolute concentrations of all biomarkers differed significantly between methods. Equations retrieved by the Deming regression enabled proper realignment of the data to overcome these differences, such that intra-class correlation coefficients were then 0.996 (CRP), 0.711 (SAA), 0.895 (sICAM-1) and 0.858 (sVCAM-1). Additionally, individual biomarkers differed across categories of glucose metabolism, weight, metabolic syndrome and smoking status to a similar extent by either method. CONCLUSIONS: Multiple low-grade inflammatory biomarker data obtained by the 4-plex multi-array platform of MSD or by well-established single-biomarker methods are comparable after proper realignment of differences in absolute concentrations, and are equally associated with cardiovascular risk factors, regardless of such differences. Given its greater efficiency, the MSD platform is a potential tool for the quantification of multiple biomarkers of low-grade inflammation in large ongoing and future clinical studies.


Subject(s)
Biomarkers/metabolism , Electrochemistry/methods , Luminescence , Aged , Blood Glucose/metabolism , Body Mass Index , C-Reactive Protein/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Inflammation , Intercellular Adhesion Molecule-1/blood , Male , Middle Aged , Nephelometry and Turbidimetry/methods , Oligonucleotide Array Sequence Analysis , Prospective Studies , Serum Amyloid A Protein/metabolism , Smoking , Vascular Cell Adhesion Molecule-1/blood
11.
Exp Diabetes Res ; 2012: 234812, 2012.
Article in English | MEDLINE | ID: mdl-22474418

ABSTRACT

BACKGROUND: Plasma and urinary levels of D-lactate have been linked to the presence of diabetes. Previously developed techniques have shown several limitations to further evaluate D-lactate as a biomarker for this condition. METHODS: D- and L-lactate were quantified using ultraperformance liquid chromatography tandem mass spectrometry with labelled internal standard. Samples were derivatized with diacetyl-L-tartaric anhydride and separated on a C(18)-reversed phase column. D- and L-lactate were analysed in plasma and urine of controls, patients with inflammatory bowel disease (IBD), and patients with type 2 diabetes (T2DM). RESULTS: Quantitative analysis of D- and L-lactate was achieved successfully. Calibration curves were linear (r(2) > 0.99) over the physiological and pathophysiological ranges. Recoveries for urine and plasma were between 96% and 113%. Inter- and intra-assay variations were between 2% and 9%. The limits of detection of D-lactate and L-lactate in plasma were 0.7 µmol/L and 0.2 µmol/L, respectively. The limits of detection of D-lactate and L-lactate in urine were 8.1 nmol/mmol creatinine and 4.4 nmol/mmol creatinine, respectively. Plasma and urinary levels of D- and L-lactate were increased in patients with IBD and T2DM as compared with controls. CONCLUSION: The presented method proved to be suitable for the quantification of D- and L-lactate and opens the possibility to explore the use of D-lactate as a biomarker.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Lactic Acid/metabolism , Adult , Aged , Chromatography, Reverse-Phase , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/urine , Female , Humans , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/urine , Lactic Acid/blood , Lactic Acid/urine , Male , Middle Aged , Tandem Mass Spectrometry
12.
Article in English | MEDLINE | ID: mdl-19188098

ABSTRACT

A rapid and sensitive single-column high-performance liquid chromatography method and application for the detection of protein bound pentosidine is described. Pentosidine, a cross-link between arginine and lysine, is a well-characterized advanced glycation endproduct. In order to detect protein-bound pentosidine, plasma proteins were hydrolysed in 6N HCl. Detection of pentosidine is done based on its own fluorescence characteristics using fluorimetric detection (E(x)=325 nm, E(m)=385 nm). Separation is done, with a run-to-run time of 30 min, on a C(18) Allspehere ODS-II column with a citric acid acetonitrile buffer. This detection enables sensitive and specific determination of protein bound pentosidine in plasma with a detection limit of 2.2 nmol/l or 0.02 pmol/mg protein (signal-to-noise: 6). The intra-assay coefficient variation is 6.5% at a plasma pentosidine concentration of 0.47 pmol/mg protein and 2.0% at a concentration of 1.27 pmol/mg protein. The inter-assay coefficient variation is 3.1% at a plasma pentosidine concentration of 0.43 pmol/mg protein and 1.6% at a concentration of 1.40 pmol/mg protein. Linearity is tested in 4 different plasma samples and showed linearity (0-200 nmol/l, r(2)>0.99). Recovery of pentosidine in 4 different plasma samples at different concentration levels is 102+/-10% (mean+/-SD). Using this method protein bound pentosidine concentration is investigated in healthy controls (n=24, age 67+/-9 years) and patients with end stage renal disease (n=24, age 65+/-10 years). Higher plasma concentrations of protein bound pentosidine are measured in the patient group as compared with the control group 3.05 (2.03-3.92)pmol/mg protein and 0.21 (0.19-0.33)pmol/mg protein, respectively (median (interquartile range), p<0.00001). These results are consistent with previously reported results.


Subject(s)
Arginine/analogs & derivatives , Chromatography, High Pressure Liquid/methods , Lysine/analogs & derivatives , Aged , Arginine/blood , Female , Humans , Lysine/blood , Male , Middle Aged , Spectrometry, Fluorescence/instrumentation
13.
J Physiol ; 552(Pt 1): 163-75, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12909684

ABSTRACT

The aim of this study was to determine the effects of a clinically relevant single course of prenatal betamethasone in the rat on growth parameters with particular reference to brain cell proliferation and apoptosis. We report that administration of 170 microg kg-1 betamethasone twice within 4 h to E20 pregnant rats conveys moderate somatic growth retardation. Further, using a measure of brain cell proliferation independent of blood-brain barrier (BBB) permeability, we demonstrate for the first time that betamethasone is chronically anti-proliferative to brain cells without inducing caspase-3-mediated apoptosis. More importantly we show that there is a significant and sexually divergent rebound of neural proliferation which occurs earlier in males than in females and continues until at least 21 days of postnatal life. BBB permeability to [3H]thymidine was significantly increased by steroid treatment re-iterating the fact that tracer studies not correcting for BBB permeability, such as bromodeoxyuridine (BrdU), may be questionable in this type of study. Further, prenatal steroid treatment did not alter postnatal corticosterone levels. In summary we show that prenatal betamethasone conveys significant and long-lasting side effects and that its human clinical application in preterm labour needs more careful consideration as compared to the relative ease with which it is prescribed today.


Subject(s)
Betamethasone/pharmacology , Brain/cytology , Brain/drug effects , Glucocorticoids/pharmacology , Prenatal Exposure Delayed Effects , Animals , Apoptosis/drug effects , Birth Weight , Blood-Brain Barrier/physiology , Brain/growth & development , Caspase 3 , Caspases/metabolism , Cell Division/drug effects , Corticosterone/blood , Female , Head/anatomy & histology , Head/growth & development , Hyperglycemia/chemically induced , Litter Size , Organ Size , Pregnancy , Rats , Rats, Inbred F344 , Survival Rate
14.
Am J Ophthalmol ; 134(3): 367-75, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12208248

ABSTRACT

PURPOSE: To investigate the presence of basic fibroblast growth factor (bFGF), glutamine synthetase (GS), and interleukin-6 (IL-6) in vitreous fluid from eyes with retinal detachment complicated by proliferative vitreoretinopathy (PVR). DESIGN: Comparative case series; experimental study. METHODS: In a prospective study, we measured bFGF, GS, IL-6, and total protein in vitreous fluid samples from 53 eyes from 53 consecutive patients with PVR operated on in our hospital. As controls, vitreous fluid samples from eyes with a macular hole (n = 9) or pucker (n = 11) were used. MAIN OUTCOME MEASURES: Laboratory data of the patient group were compared with the control group and correlated with various clinical data, especially with visual recovery and redetachment. RESULTS: For IL-6, bFGF, and total protein we found significantly higher levels in PVR patients' eyes than in control eyes (P =.03, P =.046, and P <.0001, respectively). Within the PVR group, no significant correlation was found for IL-6, bFGF, GS, or total protein with the various tested clinical variables. CONCLUSIONS: We found increased levels of IL-6, bFGF, and total protein in vitreous fluid from patients' eyes with PVR. Whether the increased levels of IL-6, bFGF, and total protein are the result of an injury-induced upregulation of these proteins as part of a self-protective mechanism of the retina to minimize photoreceptor damage after the mechanical injury induced by retinal detachment is, at present, not known.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Glutamate-Ammonia Ligase/metabolism , Interleukin-6/metabolism , Retinal Detachment/metabolism , Vitreoretinopathy, Proliferative/metabolism , Vitreous Body/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Enzyme-Linked Immunosorbent Assay , Eye Proteins/metabolism , Female , Humans , Male , Middle Aged , Prospective Studies , Retinal Detachment/etiology , Retinal Detachment/surgery , Vitrectomy , Vitreoretinopathy, Proliferative/complications , Vitreoretinopathy, Proliferative/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...