Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Am J Hum Genet ; 110(10): 1769-1786, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37729906

ABSTRACT

Defects in hydroxymethylbilane synthase (HMBS) can cause acute intermittent porphyria (AIP), an acute neurological disease. Although sequencing-based diagnosis can be definitive, ∼⅓ of clinical HMBS variants are missense variants, and most clinically reported HMBS missense variants are designated as "variants of uncertain significance" (VUSs). Using saturation mutagenesis, en masse selection, and sequencing, we applied a multiplexed validated assay to both the erythroid-specific and ubiquitous isoforms of HMBS, obtaining confident functional impact scores for >84% of all possible amino acid substitutions. The resulting variant effect maps generally agreed with biochemical expectations and provide further evidence that HMBS can function as a monomer. Additionally, the maps implicated specific residues as having roles in active site dynamics, which was further supported by molecular dynamics simulations. Most importantly, these maps can help discriminate pathogenic from benign HMBS variants, proactively providing evidence even for yet-to-be-observed clinical missense variants.


Subject(s)
Hydroxymethylbilane Synthase , Porphyria, Acute Intermittent , Humans , Hydroxymethylbilane Synthase/chemistry , Hydroxymethylbilane Synthase/genetics , Hydroxymethylbilane Synthase/metabolism , Mutation, Missense/genetics , Porphyria, Acute Intermittent/diagnosis , Porphyria, Acute Intermittent/genetics , Amino Acid Substitution , Molecular Dynamics Simulation
2.
bioRxiv ; 2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36798224

ABSTRACT

Defects in hydroxymethylbilane synthase (HMBS) can cause Acute Intermittent Porphyria (AIP), an acute neurological disease. Although sequencing-based diagnosis can be definitive, ~⅓ of clinical HMBS variants are missense variants, and most clinically-reported HMBS missense variants are designated as "variants of uncertain significance" (VUS). Using saturation mutagenesis, en masse selection, and sequencing, we applied a multiplexed validated assay to both the erythroid-specific and ubiquitous isoforms of HMBS, obtaining confident functional impact scores for >84% of all possible amino-acid substitutions. The resulting variant effect maps generally agreed with biochemical expectation. However, the maps showed variants at the dimerization interface to be unexpectedly well tolerated, and suggested residue roles in active site dynamics that were supported by molecular dynamics simulations. Most importantly, these HMBS variant effect maps can help discriminate pathogenic from benign variants, proactively providing evidence even for yet-to-be-observed clinical missense variants.

3.
Life (Basel) ; 12(12)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36556425

ABSTRACT

High mortality rates have been reported in historical cohorts of acute intermittent porphyria (AIP) patients. The mortality associated with (hydroxymethylbilane synthase) HMBS variant heterozygosity is unknown. This study estimates all-cause mortality in pedigrees with HMBS gene variants that cause AIP. We collected data on the lifespan of individuals in Dutch AIP pedigrees and performed analyses using the family tree mortality ratio method. This gave us standardized mortality ratios for these pedigrees compared to the Dutch general population as a primary outcome. Between 1810 and 2017, the overall mortality in these pedigrees was identical to that of the general Dutch population: (SMR 1.01, p = 0.441). However, compared with the general population the SMR was significantly higher in women aged 45−64 years (SMR 1.99, p = 0.00003), which was based on excess mortality between 1915 and 1964 (SMR 1.94, p < 0.00002). In men aged 70−74 years, the SMR was 1.55 (p = 0.0021), based on excess mortality that occurred between 1925 and 1964 (SMR 1.92, p = 0000000003). Overall, mortality from HMBS variant heterozygosity was not increased compared with the general population. Severe excess mortality occurred in young women and old men between 1915 and 1964. Heterozygotes reached a normal lifespan during the past half-century, in parallel with disease awareness and the prevention of new attacks through family counselling.

4.
Int J Sport Nutr Exerc Metab ; 30(6): 386-395, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32998111

ABSTRACT

Fasting enhances the beneficial metabolic outcomes of exercise; however, it is unknown whether body composition is favorably modified on the short term. A baseline-follow-up study was carried out to assess the effect of an established protocol involving short-term combined exercise with fasting on body composition. One hundred seven recreationally exercising males underwent a 10-day intervention across 15 fitness centers in the Netherlands involving a 3-day gradual decrease of food intake, a 3-day period with extremely low caloric intake, and a gradual 4-day increase to initial caloric intake, with daily 30-min submaximal cycling. Using dual-energy X-ray absorptiometry analysis, all subjects substantially lost total body mass (-3.9 ± 1.9 kg; p < .001) and fat mass (-3.3 ± 1.3 kg; p < .001). Average lean mass was lost (-0.6 ± 1.5 kg; p < .001), but lean mass as a percentage of total body mass was not reduced. The authors observed a loss of -3.9 ± 1.9% android fat over total fat mass (p < .001), a loss of -2.2 ± 1.9% gynoid over total fat mass (p < .001), and reduced android/gynoid ratios (-0.05 ± 0.1; p < .001). Analyzing 15 preselected single-nucleotide polymorphisms in 13 metabolism-related genes revealed trending associations for thyroid state-related single-nucleotide polymorphisms rs225014 (deiodinase 2) and rs35767 (insulin-like growth factor1), and rs1053049 (PPARD). In conclusion, a short period of combined fasting and exercise leads to a substantial loss of body and fat mass without a loss of lean mass as a percentage of total mass.


Subject(s)
Body Composition , Exercise , Fasting , Absorptiometry, Photon , Adult , Energy Intake , Follow-Up Studies , Humans , Male , Middle Aged , Netherlands , Polymorphism, Single Nucleotide , Young Adult
5.
JAMA Dermatol ; 156(5): 570-575, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32186677

ABSTRACT

Importance: The effectiveness of afamelanotide treatment in patients with erythropoietic protoporphyria (EPP) in clinical practice who experience pain after light exposure that substantially impairs quality of life is unknown. Objective: To evaluate the association of afamelanotide treatment with outcomes in patients with EPP in regular practice during longer-term follow-up. Design, Setting, and Participants: This single-center, prospective postauthorization safety and efficacy cohort study was directed and approved by the European Medicines Agency. Data were collected from patients with EPP treated with afamelanotide at Erasmus MC between June 2016 and September 2018. Analysis began October 2018. Main Outcomes and Measures: Time spent outside during treatment, number of phototoxic reactions, disease-specific quality of life, usage of protective clothing, and adverse events. Results: A total of 117 patients with EPP (59 women [50.4%]; mean [SD] age, 43.0 [15.5] years) were treated with afamelanotide. Nearly all patients continued treatment (115 [98%]) with a median (interquartile range) follow-up of 2.0 (1.3-2.1) years. Compared with baseline, mean time spent outside during treatment increased significantly by an added 6.1 hours per week (95% CI, 3.62-8.67; P < .001). Mean quality of life score improved significantly by 14.01% (95% CI, 4.53%-23.50%; P < .001). Phototoxic reactions were less painful (ß, -0.85; 95% CI, -1.43 to -0.26; P < .001), but there was no significant difference in number or duration of reactions. Minor self-limiting adverse events occurred, such as nausea, fatigue, and headache. Conclusions and Relevance: This cohort study found that afamelanotide treatment was associated with improved clinical outcomes and a good safety profile for patients with EPP. The treatment has clinically significant, sustained positive associations with quality of life, is associated with increased duration of sun exposure, and is associated with less severe phototoxic reactions.


Subject(s)
Dermatologic Agents/administration & dosage , Protoporphyria, Erythropoietic/drug therapy , Quality of Life , alpha-MSH/analogs & derivatives , Adolescent , Adult , Aged , Cohort Studies , Dermatologic Agents/adverse effects , Female , Humans , Male , Middle Aged , Prospective Studies , Protoporphyria, Erythropoietic/physiopathology , Sunlight/adverse effects , Treatment Outcome , Young Adult , alpha-MSH/administration & dosage , alpha-MSH/adverse effects
6.
Genet Med ; 21(11): 2605-2613, 2019 11.
Article in English | MEDLINE | ID: mdl-31073229

ABSTRACT

With the advent of precision and genomic medicine, a critical issue is whether a disease gene variant is pathogenic or benign. Such is the case for the three autosomal dominant acute hepatic porphyrias (AHPs), including acute intermittent porphyria, hereditary coproporphyria, and variegate porphyria, each resulting from the half-normal enzymatic activities of hydroxymethylbilane synthase, coproporphyrinogen oxidase, and protoporphyrinogen oxidase, respectively. To date, there is no public database that documents the likely pathogenicity of variants causing the porphyrias, and more specifically, the AHPs with biochemically and clinically verified information. Therefore, an international collaborative with the European Porphyria Network and the National Institutes of Health/National Center for Advancing Translational Sciences/National Institute of Diabetes and Digestive and Kidney Diseases (NIH/NCATS/NIDDK)-sponsored Porphyrias Consortium of porphyria diagnostic experts is establishing an online database that will collate biochemical and clinical evidence verifying the pathogenicity of the published and newly identified variants in the AHP-causing genes. The overall goal of the International Porphyria Molecular Diagnostic Collaborative is to determine the pathogenic and benign variants for all eight porphyrias. Here we describe the overall objectives and the initial efforts to validate pathogenic and benign variants in the respective heme biosynthetic genes causing the AHPs.


Subject(s)
Porphyrias/genetics , Porphyrias/physiopathology , Virulence/genetics , Data Curation/methods , Databases, Factual , Female , Humans , Male , Pathology, Molecular , Porphobilinogen Synthase/deficiency , Porphobilinogen Synthase/genetics , Porphyria, Acute Intermittent/genetics , Porphyria, Acute Intermittent/physiopathology , Porphyrias, Hepatic/genetics , Porphyrias, Hepatic/physiopathology , United States
7.
J Card Surg ; 34(4): 226-228, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30847947

ABSTRACT

We report implantation of a left ventricular assist device (LVAD) in a patient with Parkinson's disease. Postoperative fluid overload together with insufficient LVAD output in the setting of vasodilation through levodopa likely caused renal hypoperfusion and acute kidney injury. A patient like ours, therefore, requires the highest possible increase of HM3 RPM and LVAD flow early after surgery.


Subject(s)
Heart Failure/surgery , Heart Ventricles/surgery , Heart-Assist Devices , Levodopa/adverse effects , Parkinson Disease/drug therapy , Vasodilation/drug effects , Acute Kidney Injury/etiology , Acute Kidney Injury/prevention & control , Heart Failure/complications , Heart-Assist Devices/adverse effects , Humans , Ischemia/etiology , Ischemia/prevention & control , Kidney/blood supply , Levodopa/administration & dosage , Male , Middle Aged , Parkinson Disease/complications , Postoperative Complications/etiology , Postoperative Complications/prevention & control
9.
Diabetologia ; 61(11): 2386-2397, 2018 11.
Article in English | MEDLINE | ID: mdl-30145664

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to evaluate the effect of sitagliptin on glucose tolerance, plasma lipids, energy expenditure and metabolism of brown adipose tissue (BAT), white adipose tissue (WAT) and skeletal muscle in overweight individuals with prediabetes (impaired glucose tolerance and/or impaired fasting glucose). METHODS: We performed a randomised, double-blinded, placebo-controlled trial in 30 overweight, Europid men (age 45.9 ± 6.2 years; BMI 28.8 ± 2.3 kg/m2) with prediabetes in the Leiden University Medical Center and the Alrijne Hospital between March 2015 and September 2016. Participants were initially randomly allocated to receive sitagliptin (100 mg/day) (n = 15) or placebo (n = 15) for 12 weeks, using a randomisation list that was set up by an unblinded pharmacist. All people involved in the study as well as participants were blinded to group assignment. Two participants withdrew from the study prior to completion (both in the sitagliptin group) and were subsequently replaced with two new participants that were allocated to the same treatment. Before and after treatment, fasting venous blood samples and skeletal muscle biopsies were obtained, OGTT was performed and body composition, resting energy expenditure and [18F] fluorodeoxyglucose ([18F]FDG) uptake by metabolic tissues were assessed. The primary study endpoint was the effect of sitagliptin on BAT volume and activity. RESULTS: One participant from the sitagliptin group was excluded from analysis, due to a distribution error, leaving 29 participants for further analysis. Sitagliptin, but not placebo, lowered glucose excursion (-40%; p < 0.003) during OGTT, accompanied by an improved insulinogenic index (+38%; p < 0.003) and oral disposition index (+44%; p < 0.003). In addition, sitagliptin lowered serum concentrations of triacylglycerol (-29%) and very large (-46%), large (-35%) and medium-sized (-24%) VLDL particles (all p < 0.05). Body weight, body composition and energy expenditure did not change. In skeletal muscle, sitagliptin increased mRNA expression of PGC1ß (also known as PPARGC1B) (+117%; p < 0.05), a main controller of mitochondrial oxidative energy metabolism. Although the primary endpoint of change in BAT volume and activity was not met, sitagliptin increased [18F] FDG uptake in subcutaneous WAT (sWAT; +53%; p < 0.05). Reported side effects were mild and transient and not necessarily related to the treatment. CONCLUSIONS/INTERPRETATION: Twelve weeks of sitagliptin in overweight, Europid men with prediabetes improves glucose tolerance and lipid metabolism, as related to increased [18F] FDG uptake by sWAT, rather than BAT, and upregulation of the mitochondrial gene PGC1ß in skeletal muscle. Studies on the effect of sitagliptin on preventing or delaying the progression of prediabetes into type 2 diabetes are warranted. TRIAL REGISTRATION: ClinicalTrials.gov NCT02294084. FUNDING: This study was funded by Merck Sharp & Dohme Corp, Dutch Heart Foundation, Dutch Diabetes Research Foundation, Ministry of Economic Affairs and the University of Granada.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Overweight/drug therapy , Overweight/metabolism , Prediabetic State/drug therapy , Sitagliptin Phosphate/therapeutic use , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adult , Blood Glucose/drug effects , Body Weight/drug effects , Carrier Proteins/genetics , Double-Blind Method , Energy Metabolism/drug effects , Humans , Male , Middle Aged , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Prediabetic State/metabolism , RNA-Binding Proteins
10.
11.
J Endocrinol ; 238(2): 91-106, 2018 08.
Article in English | MEDLINE | ID: mdl-29743343

ABSTRACT

Cold exposure of mice is a common method to stimulate brown adipose tissue (BAT) activity and induce browning of white adipose tissue (WAT) that has beneficial effects on whole-body lipid metabolism, including reduced plasma triglyceride (TG) concentrations. The liver is a key regulatory organ in lipid metabolism as it can take up as well as oxidize fatty acids. The liver can also synthesize, store and secrete TGs in VLDL particles. The effects of cold exposure on murine hepatic lipid metabolism have not been addressed. Here, we report the effects of 24-h exposure to 4°C on parameters of hepatic lipid metabolism of male C57BL/6J mice. Cold exposure increased hepatic TG concentrations by 2-fold (P < 0.05) but reduced hepatic lipogenic gene expression. Hepatic expression of genes encoding proteins involved in cholesterol synthesis and uptake such as the LDL receptor (LDLR) was significantly increased upon cold exposure. Hepatic expression of Cyp7a1 encoding the rate-limiting enzyme in the classical bile acid (BA) synthesis pathway was increased by 4.3-fold (P < 0.05). Hepatic BA concentrations and fecal BA excretion were increased by 2.8- and 1.3-fold, respectively (P < 0.05 for both). VLDL-TG secretion was reduced by approximately 50% after 24 h of cold exposure (P < 0.05). In conclusion, cold exposure has various, likely intertwined effects on the liver that should be taken into account when studying the effects of cold exposure on whole-body metabolism.


Subject(s)
Cold Temperature , Liver/metabolism , Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Animals , Cell Transdifferentiation/genetics , Down-Regulation/genetics , Gene Expression Regulation , Glycogen/metabolism , Lipid Metabolism/physiology , Lipogenesis/genetics , Lipoproteins, VLDL/blood , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Thermogenesis/physiology , Triglycerides/blood
12.
J Inherit Metab Dis ; 41(5): 809-817, 2018 09.
Article in English | MEDLINE | ID: mdl-29675607

ABSTRACT

INTRODUCTION: A small proportion of patients with acute intermittent porphyria (AIP) suffer from recurrent porphyric attacks, with a severely diminished quality of life. In this retrospective case-control study, the burden of disease is quantified and compared among three AIP patient subgroups: cases with recurrent attacks, cases with one or occasional attacks and asymptomatic carriers. METHODS: Data from patient records and questionnaires were collected in patients between 1960 and 2016 at the Erasmus Medical Center, Rotterdam, the Netherlands. We collected symptoms related to porphyria, porphyria related complications, attack frequency, hospitalisation frequency, hospitalisation days related to acute porphyric attacks, frequency of heme infusions and medical healthcare costs based on hospitalisations and heme therapy. RESULTS: In total 11 recurrent AIP cases, 24 symptomatic AIP cases and 53 AIP carriers as controls were included. All recurrent patients reported porphyria related symptoms, such as pain, neurological and/or psychiatric disorders, and nearly all developed complications, such as hypertension and chronic kidney disease. In the recurrent cases group, the median lifelong number of hospitalisation days related to porphyric attacks was 82 days per patient (range 10-374), and they spent a median of 346 days (range 34-945) at a day-care facility for prophylactic heme therapy; total follow-up time was 243 person-years (PYRS). In the symptomatic non-recurrent group the median lifelong number of hospitalisation days related to porphyric attacks was 7 days per patient (range 1-78), total follow-up time was 528 PYRS. The calculated total medical healthcare cost for recurrent cases group was €5.8 million versus €0.3 million for the symptomatic cases group.


Subject(s)
Heme/therapeutic use , Porphyria, Acute Intermittent/complications , Porphyria, Acute Intermittent/drug therapy , Porphyria, Acute Intermittent/economics , Adolescent , Adult , Carcinoma, Hepatocellular/etiology , Case-Control Studies , Cost of Illness , Female , Health Care Costs , Hospitalization/economics , Humans , Hypertension/etiology , Liver Neoplasms/etiology , Longitudinal Studies , Male , Middle Aged , Netherlands , Quality of Life , Renal Insufficiency, Chronic/etiology , Retrospective Studies , Surveys and Questionnaires , Young Adult
13.
Hypertension ; 69(6): 1145-1155, 2017 06.
Article in English | MEDLINE | ID: mdl-28396539

ABSTRACT

Renin, a key component in the regulation of blood pressure in mammals, is produced by the rare and highly specialized juxtaglomerular cells of the kidney. Chronic stimulation of renin release results in a recruitment of new juxtaglomerular cells by the apparent conversion of adjacent smooth muscle cells along the afferent arterioles. Because juxtaglomerular cells rapidly dedifferentiate when removed from the kidney, their developmental origin and the mechanism that explains their phenotypic plasticity remain unclear. To overcome this limitation, we have performed RNA expression analysis on 4 human renin-producing tumors. The most highly expressed genes that were common between the reninomas were subsequently used for in situ hybridization in kidneys of 5-day-old mice, adult mice, and adult mice treated with captopril. From the top 100 genes, 10 encoding for ligands were selected for further analysis. Medium of human embryonic kidney 293 cells transfected with the mouse cDNA encoding these ligands was applied to (pro)renin-synthesizing As4.1 cells. Among the ligands, only platelet-derived growth factor B reduced the medium and cellular (pro)renin levels, as well as As4.1 renin gene expression. In addition, platelet-derived growth factor B-exposed As4.1 cells displayed a more elongated and aligned shape with no alteration in viability. This was accompanied by a downregulated expression of α-smooth muscle actin and an upregulated expression of interleukin-6, suggesting a phenotypic shift from myoendocrine to inflammatory. Our results add 36 new genes to the list that characterize renin-producing cells and reveal a novel role for platelet-derived growth factor B as a regulator of renin-synthesizing cells.


Subject(s)
Gene Expression Profiling , Juxtaglomerular Apparatus/cytology , Kidney Diseases/genetics , Platelet-Derived Growth Factor/metabolism , Renin/biosynthesis , Analysis of Variance , Animals , Cells, Cultured , Disease Models, Animal , Gene Expression , Humans , In Situ Hybridization , Juxtaglomerular Apparatus/metabolism , Kidney Diseases/metabolism , Mice , Mice, Inbred C57BL , Random Allocation , Renin/genetics , Signal Transduction
14.
FASEB J ; 31(1): 14-28, 2017 01.
Article in English | MEDLINE | ID: mdl-27729415

ABSTRACT

Obesity and type 2 diabetes are associated disorders that involve a multiplicity of tissues. Both fasting and physical exercise are known to counteract dyslipidemia/hyperglycemia. Skeletal muscle plays a key role in the control of blood glucose levels, and the metabolic changes and related signaling pathways in skeletal muscle induced by fasting overlap with those induced by exercise. The reduction of fat disposal has been shown to extend to the liver and to white and brown adipose tissue and to involve an increase in their metabolic activities. In recent years signal transduction pathways related to exercise and fasting/food withdrawal in muscle have been intensively studied, both in animals and in humans. Combining fasting/food withdrawal with exercise in animals as well as in humans causes changes unlike those seen during fasting/food withdrawal or exercise alone, which favor repair of muscle over autophagy. In addition, compounds that mimic exercise have been studied in combination with exercise or fasting/food withdrawal. This review addresses our current knowledge of the mechanisms that underlie the individual and combined effects of fasting/food withdrawal, endurance or resistance exercise, and their mimetics, in muscle vs other organs in rodents and humans, and highlights which combinations may improve metabolic disorders.-Jaspers, R. T., Zillikens, M. C., Friesema, E. C. H., delli Paoli, G., Bloch, W., Uitterlinden, A. G., Goglia, F., Lanni, A., de Lange, P. Exercise, fasting, and mimetics: toward beneficial combinations.


Subject(s)
Exercise/physiology , Food Deprivation/physiology , Animals , Blood Glucose , Diabetes Mellitus, Type 2/metabolism , Humans , Lipids/blood , Obesity/metabolism
15.
Neurology ; 87(12): 1258-65, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27558376

ABSTRACT

OBJECTIVE: To identify the genetic etiology of a distinct leukoencephalopathy with autosomal recessive inheritance in a single family. METHODS: We analyzed available MRIs and retrospectively reviewed clinical information and laboratory investigations. We performed whole-exome sequencing to find the causal gene variants. RESULTS: We identified 3 family members with a similar MRI pattern characterized by symmetrical signal abnormalities in the periventricular and deep cerebral white matter, thalami, and central part of the pons. Cerebellar atrophy was noted in advanced disease stages. Clinical features were childhood-onset slowly progressive spastic paraparesis, cerebellar ataxia, peripheral neuropathy, and in 2 patients, optic atrophy as well as vertical gaze and convergence palsies and nystagmus. Whole-exome sequencing revealed compound heterozygous missense variants in the HMBS gene, both associated with the autosomal dominant disorder acute intermittent porphyria. Sanger sequencing of 6 healthy siblings confirmed the bi-allelic location of the variants and segregation with the disease. Patients had a slight and moderate increase in urinary and plasma porphobilinogen and 5'-aminolevulinic acid, respectively, and a 50% to 66% decrease in hydroxymethylbilane synthase enzyme activity compared to normal. CONCLUSIONS: Bi-allelic HMBS variants have been reported before as cause of severe encephalopathy with early childhood fatality in acute intermittent porphyria. Our cases demonstrate childhood onset, but milder and slower disease progression in middle-aged patients. With this, a novel phenotype can be added to the disease spectrum associated with bi-allelic HMBS variants: a leukoencephalopathy with early onset, slowly progressive neurologic symptomatology, and long life expectancy.


Subject(s)
Brain/diagnostic imaging , Hydroxymethylbilane Synthase/genetics , Leukoencephalopathies/complications , Porphyria, Acute Intermittent/complications , Family , Female , Genetic Variation , Humans , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/genetics , Magnetic Resonance Imaging , Male , Middle Aged , Porphyria, Acute Intermittent/diagnostic imaging , Porphyria, Acute Intermittent/genetics , Retrospective Studies
16.
Clin Sci (Lond) ; 130(14): 1209-20, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27129187

ABSTRACT

ARNI [dual AT1 (angiotensin II type 1) receptor-neprilysin inhibition] exerts beneficial effects on blood pressure and kidney function in heart failure, compared with ARB (AT1 receptor blockade) alone. We hypothesized that ARNI improves cardiac and kidney parameters in diabetic TGR(mREN2)27 rats, an angiotensin II-dependent hypertension model. Rats were made diabetic with streptozotocin for 5 or 12 weeks. In the final 3 weeks, rats were treated with vehicle, irbesartan (ARB) or irbesartan+thiorphan (ARNI). Blood pressure, measured by telemetry in the 5-week group, was lowered identically by ARB and ARNI. The heart weight/tibia length ratio in 12-week diabetic animals was lower after ARNI compared with after ARB. Proteinuria and albuminuria were observed from 8 weeks of diabetes onwards. ARNI reduced proteinuria more strongly than ARB, and a similar trend was seen for albuminuria. Kidneys of ARNI-treated animals showed less severe segmental glomerulosclerosis than those of ARB-treated animals. After 12 weeks, no differences between ARNI- and ARB-treated animals were found regarding diuresis, natriuresis, plasma endothelin-1, vascular reactivity (acetylcholine response) or kidney sodium transporters. Only ARNI-treated rats displayed endothelin type B receptor-mediated vasodilation. In conclusion, ARNI reduces proteinuria, glomerulosclerosis and heart weight in diabetic TGR(mREN2)27 rats more strongly than does ARB, and this occurs independently of blood pressure.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Blood Pressure/physiology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/prevention & control , Neprilysin/antagonists & inhibitors , Aminobutyrates/therapeutic use , Animals , Atrial Natriuretic Factor/blood , Biphenyl Compounds , Blood Pressure/drug effects , Drug Combinations , Rats , Rats, Sprague-Dawley , Streptozocin , Tetrazoles/therapeutic use , Valsartan
17.
Kidney Int ; 88(1): 109-20, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25830765

ABSTRACT

Neprilysin inhibitors prevent the breakdown of bradykinin and natriuretic peptides, promoting vasodilation and natriuresis. However, they also increase angiotensin II and endothelin-1. Here we studied the effects of a low and a high dose of the neprilysin inhibitor thiorphan on top of AT1 receptor blockade with irbesartan versus vehicle in TGR(mREN2)27 rats with high renin hypertension. Mean arterial blood pressure was unaffected by vehicle or thiorphan alone. Irbesartan lowered blood pressure, but after 7 days pressure started to increase again. Low- but not high-dose thiorphan prevented this rise. Only during exposure to low-dose thiorphan plus irbesartan did heart weight/body weight ratio, cardiac atrial natriuretic peptide expression, and myocyte size decrease significantly. Circulating endothelin-1 was not affected by low-dose thiorphan with or without irbesartan, but increased after treatment with high-dose thiorphan plus irbesartan. This endothelin-1 rise was accompanied by an increase in renal sodium-hydrogen exchanger 3 protein abundance, and an upregulation of constrictor vascular endothelin type B receptors. Consequently, the endothelin type B receptor antagonist BQ788 no longer enhanced endothelin-1-induced vasoconstriction (indicative of endothelin type B receptor-mediated vasodilation), but prevented it. Thus, optimal neprilysin inhibitor dosing reveals additional cardioprotective effects on top of AT1 receptor blockade in renin-dependent hypertension.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Biphenyl Compounds/pharmacology , Kidney/metabolism , Myocardium/pathology , Neprilysin/antagonists & inhibitors , Protease Inhibitors/pharmacology , Receptor, Angiotensin, Type 1/drug effects , Tetrazoles/pharmacology , Thiorphan/pharmacology , Animals , Arterial Pressure/drug effects , Atrial Natriuretic Factor/metabolism , Body Weight , Endothelin B Receptor Antagonists/pharmacology , Endothelin-1/blood , Irbesartan , Kidney/pathology , Myocytes, Cardiac/pathology , Oligopeptides/pharmacology , Organ Size , Piperidines/pharmacology , Protease Inhibitors/administration & dosage , Rats , Receptor, Endothelin B/metabolism , Renin-Angiotensin System/drug effects , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/metabolism , Thiorphan/administration & dosage , Up-Regulation , Vasoconstriction/drug effects , Vasodilation/drug effects
18.
Hypertension ; 65(6): 1316-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25870197

ABSTRACT

Women with preeclampsia display low renin-angiotensin-aldosterone system activity and a high antiangiogenic state, the latter characterized by high levels of soluble Fms-like tyrosine kinase (sFlt)-1 and reduced placental growth factor levels. To investigate whether renin-angiotensin-aldosterone system suppression in preeclampsia is because of this disturbed angiogenic balance, we measured mean arterial pressure, creatinine, endothelin-1 (ET-1), and renin-angiotensin-aldosterone system components in pregnant women with a high (≥85; n=38) or low (<85; n=65) soluble Fms-like tyrosine kinase-1/placental growth factor ratio. Plasma ET-1 levels were increased in women with a high ratio, whereas their plasma renin activity and plasma concentrations of renin, angiotensinogen, and aldosterone were decreased. Plasma renin activity-aldosterone relationships were identical in both the groups. Multiple regression analysis revealed that plasma renin concentration correlated independently with mean arterial pressure and plasma ET-1. Plasma ET-1 correlated positively with soluble Fms-like tyrosine kinase-1 and negatively with plasma renin concentration, and urinary protein correlated with plasma ET-1 and mean arterial pressure. Despite the lower plasma levels of renin and angiotensinogen in the high-ratio group, their urinary levels of these components were elevated. Correction for albumin revealed that this was because of increased glomerular filtration. Subcutaneous arteries obtained from patients with preeclampsia displayed an enhanced, AT2 receptor-mediated response to angiotensin II. In conclusion, a high antiangiogenic state associates with ET-1 activation, which together with the increased mean arterial pressure may underlie the parallel reductions in renin and aldosterone in preeclampsia. Because ET-1 also was a major determinant of urinary protein, our data reveal a key role for ET-1 in the pathogenesis of preeclampsia. Finally, the enhanced angiotensin responsiveness in preeclampsia involves constrictor AT2 receptors.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Endothelin-1/blood , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy Outcome , Renin-Angiotensin System/physiology , Adult , Case-Control Studies , Creatinine/blood , Endothelin-1/metabolism , Female , Humans , Placental Circulation/physiology , Pregnancy , Reference Values , Renin/blood , Renin/metabolism , Renin-Angiotensin System/drug effects , Risk Assessment
19.
Mol Endocrinol ; 28(12): 1961-70, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25389909

ABSTRACT

Monocarboxylate transporter 8 (MCT8) transports thyroid hormone (TH) across the plasma membrane. Mutations in MCT8 result in the Allan-Herndon-Dudley syndrome, comprising severe psychomotor retardation and elevated serum T3 levels. Because the neurological symptoms are most likely caused by a lack of TH transport into the central nervous system, the administration of a TH analog that does not require MCT8 for cellular uptake may represent a therapeutic strategy. Here, we investigated the therapeutic potential of the biologically active T3 metabolite Triac (TA3) by studying TA3 transport, metabolism, and action both in vitro and in vivo. Incubation of SH-SY5Y neuroblastoma cells and MO3.13 oligodendrocytes with labeled substrates showed a time-dependent uptake of T3 and TA3. In intact SH-SY5Y cells, both T3 and TA3 were degraded by endogenous type 3 deiodinase, and they influenced gene expression to a similar extent. Fibroblasts from MCT8 patients showed an impaired T3 uptake compared with controls, whereas TA3 uptake was similar in patient and control fibroblasts. In transfected cells, TA3 did not show significant transport by MCT8. Most importantly, treatment of athyroid Pax8-knockout mice and Mct8/Oatp1c1-double knockout mice between postnatal days 1 and 12 with TA3 restored T3-dependent neural differentiation in the cerebral and cerebellar cortex, indicating that TA3 can replace T3 in promoting brain development. In conclusion, we demonstrated uptake of TA3 in neuronal cells and in fibroblasts of MCT8 patients and similar gene responses to T3 and TA3. This indicates that TA3 bypasses MCT8 and may be used to improve the neural status of MCT8 patients.


Subject(s)
Mental Retardation, X-Linked/drug therapy , Mental Retardation, X-Linked/metabolism , Muscle Hypotonia/drug therapy , Muscle Hypotonia/metabolism , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Triiodothyronine/analogs & derivatives , Animals , Biological Transport/drug effects , COS Cells , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Humans , In Vitro Techniques , Membrane Transport Proteins , Mice , Mice, Knockout , Monocarboxylic Acid Transporters , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Symporters , Triiodothyronine/genetics , Triiodothyronine/metabolism , Triiodothyronine/therapeutic use
20.
Endocrinology ; 155(2): 618-26, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24265446

ABSTRACT

Monocarboxylate transporter 8 (MCT8) facilitates cellular influx and efflux of the thyroid hormones (THs) T(4) and T(3). Mutations in MCT8 lead to severe psychomotor retardation. Here, we studied the importance of 2 highly conserved residues (Arg445 in transmembrane domain 8 and Asp498 in transmembrane domain 10) for substrate recognition and helix interactions. We introduced single and double mutations (R445A, R445C, R445D, R445K, D498A, D498E, D498N, D498R, R445A+D498A, R445D+D498R, and R445K+D498E) in human MCT8 cDNA and studied the effects on MCT8-mediated TH uptake and metabolism in transfected cells. The impact of these mutations on MCT8 protein expression, dimerization capacity, and subcellular localization was studied by Western blotting and confocal microscopy. We found that mutations in Arg445 or Asp498 that alter the local charge resulted in a near-complete loss of TH uptake capacity, whereas the expression, stability, and subcellular localization of these mutant proteins was similar to those for wild-type MCT8. Given the impaired TH uptake, TH efflux could not be adequately studied. The importance of opposite charges at Arg445 and Asp498 was studied by exchanging these residues (R445D+D498R). In particular, T(4) uptake was less severely reduced by the exchange mutation than by the single mutations. Mutations of Arg445 and Asp498 to equally charged residues (R445K and/or D498E) resulted in TH uptake levels similar to wild-type MCT8. The presence of 2 oppositely charged residues at positions Arg445 and Asp498 that are predicted in close structural proximity is crucial for efficient TH uptake, which may indicate the presence of an, at least transient, charge pair between these residues.


Subject(s)
Biological Transport/genetics , Monocarboxylic Acid Transporters/genetics , Thyroid Gland/metabolism , Thyroid Hormones/metabolism , Cell Line , Humans , Monocarboxylic Acid Transporters/metabolism , Mutagenesis, Site-Directed , Mutation , Symporters
SELECTION OF CITATIONS
SEARCH DETAIL
...