Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Sci Rep ; 10(1): 15608, 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32958810

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Sci Rep ; 7(1): 14451, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089512

ABSTRACT

The Baltic Sea population of the common eider (Somateria mollissima) has declined dramatically during the last two decades. Recently, widespread episodic thiamine (vitamin B1) deficiency has been demonstrated in feral birds and suggested to contribute significantly to declining populations. Here we show that the decline of the common eider population in the Baltic Sea is paralleled by high mortality of the pulli a few days after hatch, owing to thiamine deficiency and probably also thereby associated abnormal behaviour resulting in high gull predation. An experiment with artificially incubated common eider eggs collected in the field revealed that thiamine treatment of pulli had a therapeutic effect on the thiamine status of the brain and prevented death. The mortality was 53% in untreated specimens, whereas it was only 7% in thiamine treated specimens. Inability to dive was also linked to brain damage typical for thiamine deficiency. Our results demonstrate how thiamine deficiency causes a range of symptoms in the common eider pulli, as well as massive die-offs a few days after hatch, which probably are the major explanation of the recent dramatic population declines.


Subject(s)
Ducks/metabolism , Thiamine Deficiency/metabolism , Thiamine/metabolism , Animals , Baltic States , Birds , Eggs , Reproduction/drug effects
3.
Sci Rep ; 6: 38821, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27958327

ABSTRACT

Many wildlife populations are declining at rates higher than can be explained by known threats to biodiversity. Recently, thiamine (vitamin B1) deficiency has emerged as a possible contributing cause. Here, thiamine status was systematically investigated in three animal classes: bivalves, ray-finned fishes, and birds. Thiamine diphosphate is required as a cofactor in at least five life-sustaining enzymes that are required for basic cellular metabolism. Analysis of different phosphorylated forms of thiamine, as well as of activities and amount of holoenzyme and apoenzyme forms of thiamine-dependent enzymes, revealed episodically occurring thiamine deficiency in all three animal classes. These biochemical effects were also linked to secondary effects on growth, condition, liver size, blood chemistry and composition, histopathology, swimming behaviour and endurance, parasite infestation, and reproduction. It is unlikely that the thiamine deficiency is caused by impaired phosphorylation within the cells. Rather, the results point towards insufficient amounts of thiamine in the food. By investigating a large geographic area, by extending the focus from lethal to sublethal thiamine deficiency, and by linking biochemical alterations to secondary effects, we demonstrate that the problem of thiamine deficiency is considerably more widespread and severe than previously reported.


Subject(s)
Birds/metabolism , Bivalvia/metabolism , Skates, Fish/metabolism , Thiamine Deficiency , Anguilla/metabolism , Animals , Animals, Wild/metabolism , Chickens/metabolism , Female , Mytilus/metabolism , Salmon/metabolism
4.
Sci Rep ; 6: 27589, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27270328

ABSTRACT

Melanocortin therapy by using adrenocorticotropic hormone (ACTH) or non-steroidogenic melanocortin peptides attenuates proteinuria and glomerular injury in experimental glomerular diseases and induces remission of nephrotic syndrome in patients with diverse glomerulopathies, even those resistant to steroids. The underlying mechanism remains elusive, but the role of melanocortin 1 receptor (MC1R) has been implicated and was examined here. Four patients with congenital red hair color and nephrotic syndrome caused by idiopathic membranous nephropathy or focal segmental glomerulosclerosis were confirmed by gene sequencing to bear dominant-negative MC1R mutations. Despite prior corticosteroid resistance, all patients responded to ACTH monotherapy and ultimately achieved clinical remission, inferring a steroidogenic-independent and MC1R-dispensable anti-proteinuric effect of melanocortin signaling. In confirmatory animal studies, the protective effect of [Nle(4), D-Phe(7)]-α-melanocyte stimulating hormone (NDP-MSH), a potent non-steroidogenic pan-melanocortin receptor agonist, on the lipopolysaccharide elicited podocytopathy was completely preserved in MC1R-null mice, marked by reduced albuminuria and diminished histologic signs of podocyte injury. Moreover, in complementary in vitro studies, NDP-MSH attenuated the lipopolysaccharide elicited apoptosis, hypermotility and impairment of filtration barrier function equally in primary podocytes derived from MC1R-null and wild-type mice. Collectively, our findings suggest that melanocortin therapy confers a proteinuria reducing and podoprotective effect in proteinuric glomerulopathies via MC1R-independent mechanisms.


Subject(s)
Kidney Glomerulus/drug effects , Melanocortins/administration & dosage , Proteinuria/drug therapy , Receptor, Melanocortin, Type 1/genetics , Adrenocorticotropic Hormone/administration & dosage , Adrenocorticotropic Hormone/metabolism , Animals , Glomerulosclerosis, Focal Segmental/drug therapy , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/pathology , Humans , Kidney Glomerulus/injuries , Kidney Glomerulus/pathology , Melanocortins/genetics , Mice , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Podocytes/drug effects , Proteinuria/genetics , Proteinuria/metabolism , Proteinuria/pathology , Signal Transduction/drug effects , alpha-MSH/administration & dosage
5.
Basic Clin Pharmacol Toxicol ; 117(6): 375-82, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26046936

ABSTRACT

During clinical development of analgesics, it is important to have access to pharmacologically specific human pain models. o-Chlorobenzylidene malononitrile (CS) is a selective and potent agonist of the transient receptor potential ankyrin repeat 1 (TRPA1), which is a transducer molecule in nociceptors sensing reactive chemical species. While CS has been subject to extensive toxicological investigations in animals and human beings, its effects on intradermal or subcutaneous injection have not previously been reported. We have investigated the potential of CS to be used as an agonist on TRPA1 in human experimental pain studies. A calcium influx assay was used to confirm the capacity of CS to activate TRPA1 with >100,000 times the selectivity over the transient receptor potential vanilloid receptor 1. CS dose-dependently (EC50 0.9 µM) released calcitonin gene-related peptide in rat dorsal root ganglion cultures, supporting involvement in pain signalling. In a local tolerance study, injection of a single intradermal dose of 20 mM CS to rats resulted in superficial, circular crusts at the injection sites after approximately 4 days. The histopathology evaluation revealed a mild, acute inflammatory reaction in the epidermis and dermis at the intradermal CS injection site 1 day after administration. After 14 days, the epidermal epithelium was fully restored. The symptoms were not considered to be adverse, and it is suggested that doses up to 20 µL of 20 mM CS can be safely administered to human beings. In conclusion, our data support development of a CS human dermal pain model.


Subject(s)
Nerve Tissue Proteins/agonists , Nociceptive Pain/chemically induced , Skin/innervation , TRPC Cation Channels/agonists , Transient Receptor Potential Channels/agonists , o-Chlorobenzylidenemalonitrile/toxicity , Animals , CHO Cells , Calcitonin Gene-Related Peptide/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling/drug effects , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Injections, Intradermal , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nociception/drug effects , Nociceptive Pain/metabolism , Nociceptive Pain/physiopathology , Pain Threshold/drug effects , Rats, Wistar , TRPA1 Cation Channel , TRPC Cation Channels/metabolism , Time Factors , Transfection , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , o-Chlorobenzylidenemalonitrile/administration & dosage
6.
Arch Toxicol ; 89(3): 423-36, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24798087

ABSTRACT

The environmental neurotoxin ß-N-methylamino-L-alanine (BMAA) has been implicated in the etiology of neurodegenerative disease, and recent studies indicate that BMAA can be misincorporated into proteins. BMAA is a developmental neurotoxicant that can induce long-term learning and memory deficits, as well as regionally restricted neuronal degeneration and mineralization in the hippocampal CA1. The aim of the study was to characterize long-term changes (2 weeks to 6 months) further in the brain of adult rats treated neonatally (postnatal days 9-10) with BMAA (460 mg/kg) using immunohistochemistry (IHC), transmission electron microscopy, and laser capture microdissection followed by LC-MS/MS for proteomic analysis. The histological examination demonstrated progressive neurodegenerative changes, astrogliosis, microglial activation, and calcification in the hippocampal CA1 3-6 months after exposure. The IHC showed an increased staining for α-synuclein and ubiquitin in the area. The ultrastructural examination revealed intracellular deposition of abundant bundles of closely packed parallel fibrils in neurons, axons, and astrocytes of the CA1. Proteomic analysis of the affected site demonstrated an enrichment of chaperones (e.g., clusterin, GRP-78), cytoskeletal and intermediate filament proteins, and proteins involved in the antioxidant defense system. Several of the most enriched proteins (plectin, glial fibrillar acidic protein, vimentin, Hsp 27, and ubiquitin) are known to form complex astrocytic inclusions, so-called Rosenthal fibers, in the neurodegenerative disorder Alexander disease. In addition, TDP-43 and the negative regulator of autophagy, GLIPR-2, were exclusively detected. The present study demonstrates that neonatal exposure to BMAA may offer a novel model for the study of hippocampal fibril formation in vivo.


Subject(s)
Amino Acids, Diamino/toxicity , CA1 Region, Hippocampal/drug effects , Calcinosis/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeleton/drug effects , Molecular Chaperones/metabolism , Animals , Animals, Newborn , CA1 Region, Hippocampal/growth & development , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/ultrastructure , Calcinosis/chemically induced , Chromatography, Liquid , Cyanobacteria Toxins , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Immunohistochemistry , Microscopy, Electron, Transmission , Protein Folding , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Ubiquitin/metabolism , alpha-Synuclein/metabolism
7.
BMC Nephrol ; 15: 74, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24886340

ABSTRACT

BACKGROUND: Treatment of idiopathic membranous nephropathy with nephrotic syndrome is still controversial. There is currently little known about the clinical use of renal biomarkers which may explain contradictory results obtained from clinical trials. In order to assess whether IgG-uria can predict the outcome in membranous nephropathy, we examined the value of baseline EF-IgG in predicting remission and progression of nephrotic syndrome. METHODS: In a prospective cohort of 84 (34 female) idiopathic membranous nephropathy patients with nephrotic syndrome we validated the ability of the clinically available urine biomarker, IgG, to predict the risk of kidney disease progression and the beneficial effect of immunosuppression with steroids and cyclophosphamide. The fractional excretion of IgG (FE-IgG) and α1-microglobulin (FE-α1m), urine albumin/creatinine ratio, and eGFR were measured at the time of kidney biopsy. Primary outcome was progression to end stage kidney failure or kidney function (eGFR) decline ≥ 50% of baseline. Patients were followed up for 7.2 ± 4.1 years (range 1-16.8). RESULTS: High FE-IgG (≥ 0.02) predicted an increased risk of kidney failure (Hazard Ratio, (HR) 8.2, 95%CI 1.0-66.3, p=0.048) and lower chance of remission (HR 0.18, 95%CI 0.09-0.38, p<0.001). The ten-year cumulative risk of kidney failure was 51.7% for patients with high FE-IgG compared to only 6.2% for patients with low FE-IgG. During the study, only 24% of patients with high FE-IgG entered remission compared to 90% of patients with low FE-IgG. Combined treatment with steroids and cyclophosphamide decreased the progression rate (-40%) and increased the remission rate (+36%) only in patients with high FE-IgG. CONCLUSION: In idiopathic membranous nephropathy patients with nephrotic syndrome, FE-IgG could be useful for predicting kidney disease progression, remission, and response to treatment.


Subject(s)
Glomerulonephritis, Membranous/drug therapy , Glomerulonephritis, Membranous/urine , Immunoglobulin G/urine , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/urine , Renal Agents/therapeutic use , Biomarkers/urine , Female , Glomerulonephritis, Membranous/diagnosis , Humans , Male , Middle Aged , Nephrotic Syndrome/diagnosis , Prospective Studies , Reproducibility of Results , Risk Factors , Sensitivity and Specificity , Treatment Outcome
8.
J Toxicol Pathol ; 27(1): 31-42, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24791065

ABSTRACT

AZD3783, a cationic amphiphilic drug and a potent inhibitor of the 5-hydroxytryptamine (5-HT1B) receptor, was explored as a potential treatment for depression. To support clinical trials, repeat dose toxicity studies in rats and dogs were conducted. Here we report toxicity findings in dogs after dosing from 1 to 3 months. In the 1-month study, there were minimal neuronal vacuolation in the brain, a marked increase in liver enzymes accompanied by hepatocellular degeneration/necrosis and phospholipidosis (PLD), and PLD/cholecystitis in the gallbladder of animals dosed at 47 mg/kg/day. In the 3-month study, neurotoxicity resulted in euthanasia of one animal dosed at 30 mg/kg/day after 86 days. Extensive pathologic changes were seen in all animals in retina epithelium (inclusion bodies), brain (neuronal vacuolation, degeneration, or necrosis and nerve fiber degeneration), spinal ganglia (vacuolation, degeneration, or necrosis), as well as sciatic and optic nerves (degeneration). Pigment-laden macrophages were observed in the lung, kidney, liver, gallbladder, bone marrow, gastrointestinal tract, and lymphoid tissues. Also seen were vitrel and retinal hemorrhage in the eyes. A brain concentration and pathology study showed that the concentration of AZD3783 in the brain was approximately 4 times higher than in the plasma after 4 weeks of dosing, however, they were similar in all regions examined, and did not correlate with areas with pathologic findings. Our findings with AZD3783 in dogs have not been reported previously with other CNS compounds that effect through serotonergic pharmacology.

9.
Proc Natl Acad Sci U S A ; 111(14): 5409-14, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706865

ABSTRACT

One hallmark of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) is infiltration of leukocytes into the CNS, where chemokines and their receptors play a major mediatory role. CX3CR1 is a chemokine receptor involved in leukocyte adhesion and migration and hence a mediator of immune defense reactions. The role of CX3CR1 in MS and EAE pathogenesis however remains to be fully assessed. Here, we demonstrate CX3CR1 mRNA expression on inflammatory cells within active plaque areas in MS brain autopsies. To test whether blocking CNS infiltration of peripheral leukocytes expressing CX3CR1 would be a suitable treatment strategy for MS, we developed a selective, high-affinity inhibitor of CX3CR1 (AZD8797). The compound is active outside the CNS and AZD8797 treatment in Dark Agouti rats with myelin oligodendrocyte glycoprotein-induced EAE resulted in reduced paralysis, CNS pathology, and incidence of relapses. The compound is effective when starting treatment before onset, as well as after the acute phase. This treatment strategy is mechanistically similar to, but more restricted than, current very late antigen-4-directed approaches that have significant side effects. We suggest that blocking CX3CR1 on leukocytes outside the CNS could be an alternative approach to treat MS.


Subject(s)
Brain/metabolism , Disease Models, Animal , Multiple Sclerosis/pathology , Receptors, Chemokine/antagonists & inhibitors , Animals , CX3C Chemokine Receptor 1 , Chronic Disease , Rats , Receptors, Chemokine/metabolism , Recurrence
10.
J Neurosci ; 33(24): 10075-84, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23761903

ABSTRACT

Aß, the product of APP (amyloid precursor protein), has been implicated in the pathophysiology of Alzheimer's disease (AD). ß-Site APP cleaving enzyme1 (BACE1) is the enzyme initiating the processing of the APP to Aß peptides. Small molecule BACE1 inhibitors are expected to decrease Aß-peptide generation and thereby reduce amyloid plaque formation in the brain, a neuropathological hallmark of AD. BACE1 inhibition thus addresses a key mechanism in AD and its potential as a therapeutic target is currently being addressed in clinical studies. Here, we report the discovery and the pharmacokinetic and pharmacodynamic properties of BACE1 inhibitor AZ-4217, a high potency compound (IC50 160 pM in human SH-SY5Y cells) with an excellent in vivo efficacy. Central efficacy of BACE1 inhibition was observed after a single dose in C57BL/6 mice, guinea pigs, and in an APP transgenic mouse model of cerebral amyloidosis (Tg2576). Furthermore, we demonstrate that in a 1 month treatment paradigm BACE1 inhibition of Aß production does lower amyloid deposition in 12-month-old Tg2576 mice. These results strongly support BACE1 inhibition as concretely impacting amyloid deposition and therefore potentially an important approach for therapeutic intervention in AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid/metabolism , Enzyme Inhibitors/pharmacology , Neurons/drug effects , Neurons/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/therapeutic use , Female , Guinea Pigs , Humans , Isoindoles/pharmacology , Isoindoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Peptide Fragments/metabolism , Pyridones/pharmacology , Pyridones/therapeutic use , Time Factors
11.
J Neurosci ; 32(48): 17297-305, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197721

ABSTRACT

γ-Secretase inhibition represents a major therapeutic strategy for lowering amyloid ß (Aß) peptide production in Alzheimer's disease (AD). Progress toward clinical use of γ-secretase inhibitors has, however, been hampered due to mechanism-based adverse events, primarily related to impairment of Notch signaling. The γ-secretase inhibitor MRK-560 represents an exception as it is largely tolerable in vivo despite displaying only a small selectivity between Aß production and Notch signaling in vitro. In exploring the molecular basis for the observed tolerability, we show that MRK-560 displays a strong preference for the presenilin 1 (PS1) over PS2 subclass of γ-secretases and is tolerable in wild-type mice but causes dose-dependent Notch-related side effect in PS2-deficient mice at drug exposure levels resulting in a substantial decrease in brain Aß levels. This demonstrates that PS2 plays an important role in mediating essential Notch signaling in several peripheral organs during pharmacological inhibition of PS1 and provide preclinical in vivo proof of concept for PS2-sparing inhibition as a novel, tolerable and efficacious γ-secretase targeting strategy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Brain/drug effects , Presenilin-2/metabolism , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Mice , Presenilin-2/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use
12.
Toxicol Sci ; 130(2): 391-404, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22872059

ABSTRACT

The cyanobacterial toxin ß-N-methylamino-L-alanine (BMAA) has been proposed to contribute to neurodegenerative disease. We have previously reported a selective uptake of BMAA in the mouse neonatal hippocampus and that exposure during the neonatal period causes learning and memory impairments in adult rats. The aim of this study was to characterize effects in the brain of 6-month-old rats treated neonatally (postnatal days 9-10) with the glutamatergic BMAA. Protein changes were examined using the novel technique Matrix-Assisted Laser Desorption Ionization (MALDI) imaging mass spectrometry (IMS) for direct imaging of proteins in brain cryosections, and histological changes were examined using immunohistochemistry and histopathology. The results showed long-term changes including a decreased expression of proteins involved in energy metabolism and intracellular signaling in the adult hippocampus at a dose (150 mg/kg) that gave no histopathological lesions in this brain region. Developmental exposure to a higher dose (460 mg/kg) also induced changes in the expression of S100ß, histones, calcium- and calmodulin-binding proteins, and guanine nucleotide-binding proteins. At this dose, severe lesions in the adult hippocampus including neuronal degeneration, cell loss, calcium deposits, and astrogliosis were evident. The data demonstrate subtle, sometimes dose-dependent, but permanent effects of a lower neonatal dose of BMAA in the adult hippocampus suggesting that BMAA could potentially disturb many processes during the development. The detection of BMAA in seafood stresses the importance of evaluating the magnitude of human exposure to this neurotoxin.


Subject(s)
Amino Acids, Diamino/toxicity , Bacterial Toxins/toxicity , Hippocampus/drug effects , Marine Toxins/toxicity , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/chemically induced , Neurotoxicity Syndromes/etiology , Age Factors , Animals , Animals, Newborn , Cyanobacteria Toxins , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Food Contamination , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Rats , Rats, Wistar , Risk Assessment , Signal Transduction/drug effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors
13.
Toxicol Pathol ; 40(1): 18-32, 2012.
Article in English | MEDLINE | ID: mdl-22131108

ABSTRACT

Tesaglitazar was developed as a dual peroxisome proliferator-activated receptor (PPARα/γ). To support the clinical program, a hamster carcinogenicity study was performed. The only neoplastic findings possibly related to treatment with tesaglitazar were low incidences of hemangioma and hemangiosarcoma in the liver of male animals. A high-power, two-year investigative study with interim necropsies was performed to further elucidate these findings. Treatment with tesaglitazar resulted in changes typical for exaggerated PPARα pharmacology in rodents, such as hepatocellular hypertrophy and hepatocellular carcinoma, but not an increased frequency of hemangiosarcomas. At the highest dose level, there was an increased incidence of sinusoidal dilatation and hemangiomas. No increased endothelial cell (EC) proliferation was detected in vivo, which was confirmed by in vitro administration to ECs. Immunohistochemistry and gene expression analyses indicated increased cellular stress and vascular endothelial growth factor (VEGF) expression in the liver, which may have contributed to the sinusoidal dilatation. A two-fold increase in the level of circulating VEGF was detected in the hamster at all dose levels, whereas no effect on VEGF was observed in patients treated with tesaglitazar. In conclusion, investigations have demonstrated that tesaglitazar does not produce hemangiosarcomas in hamster despite a slight effect on vascular morphology in the liver.


Subject(s)
Alkanesulfonates/toxicity , Liver Neoplasms, Experimental/chemically induced , PPAR alpha/agonists , PPAR gamma/agonists , Phenylpropionates/toxicity , Animals , Area Under Curve , Carcinogenicity Tests , Cell Proliferation/drug effects , Cricetinae , Female , Gene Expression Profiling , Hemangioma/chemically induced , Hemangiosarcoma/chemically induced , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunohistochemistry , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Statistics, Nonparametric , Vascular Endothelial Growth Factor A/metabolism
14.
Drug Metab Dispos ; 39(7): 1131-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21436404

ABSTRACT

Reliable and stable in vitro cellular systems maintaining specific liver functions important for drug metabolism and disposition are urgently needed in preclinical drug discovery and development research. The cell line HepaRG exhibits promising properties such as expression and function of drug-metabolizing enzymes and transporter proteins, which resemble those found in freshly isolated human hepatocytes. In this study, HepaRG cells were cultured up to 68 days in a three-dimensional multicompartment capillary membrane bioreactor, which enables high-density cell culture under dynamic conditions. The activity of drug-metabolizing cytochrome P450 (P450) enzymes was investigated by a cocktail of substrates for CYP1A1/2 (phenacetin), CYP2C9 (diclofenac), CYP2B6 (bupropion), and CYP3A4 (midazolam). The model P450 substrates, which were introduced to the bioreactor system mimicking in vivo bolus doses, showed stable metabolism over the entire experimental period of several weeks with the exception of bupropion hydroxylase, which increased over time. Ketoconazole treatment decreased the CYP3A4 activity by 69%, and rifampicin induced the CYP3A4- and CYP2B6-dependent activity 6-fold, which predicts well the magnitude of changes observed in vivo. Moreover, polarity of transporter expression and formation of tissue-like structures including bile canaliculi were demonstrated by immune histochemistry. The long-lasting bioreactor system using HepaRG cells thus provides a promising and stable liver-like in vitro model for continuous investigations of the hepatic kinetics of drugs and of drug-drug interactions, which well predict the situation in vivo in humans.


Subject(s)
Bioreactors , Cytochrome P-450 Enzyme System/metabolism , Cell Line , Humans , Substrate Specificity
15.
Vet Immunol Immunopathol ; 141(1-2): 162-7, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21419498

ABSTRACT

Borna disease virus (BDV) is a neurotropic, negative-stranded RNA virus, which causes a non-suppurative meningoencephalomyelitis in a wide range of animals. In cats, BDV infection leads to staggering disease. In spite of a vigorous immune response the virus persists in the central nervous system (CNS) in both experimentally and naturally infected animals. Since the CNS is vulnerable to cytotoxic effects mediated via NK-cells and cytotoxic T-cells, other non-cytolytic mechanisms such as the interferon (IFN) system is favourable for viral clearance. In this study, IFN-γ expression in the brain of cats with clinical signs of staggering disease (N=12) was compared to the expression in cats with no signs of this disease (N=7) by quantitative RT-PCR. The IFN-γ expression was normalised against the expression of three reference genes (HPRT, RPS7, YWHAZ). Cats with staggering disease had significantly higher expression of IFN-γ compared to the control cats (p-value ≤ 0.001). There was no significant difference of the IFN-γ expression in BDV-positive (N=7) and -negative (N=5) cats having clinical signs of staggering disease. However, as BDV-RNA still could be detected, despite an intense IFN-γ expression, BDV needs to have mechanisms to evade this antiviral immune response of the host, to be able to persist.


Subject(s)
Borna Disease/immunology , Borna disease virus , Brain/immunology , Cat Diseases/virology , Interferon-gamma/biosynthesis , Animals , Brain/metabolism , Brain/virology , Cat Diseases/immunology , Cat Diseases/metabolism , Cats , Female , Male , Polymerase Chain Reaction/veterinary
16.
Behav Brain Res ; 219(2): 310-20, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21315110

ABSTRACT

We have reported previously that exposure to the cyanobacterial neurotoxin ß-N-methylamino-L-alanine (BMAA) during the neonatal period causes cognitive impairments in adult rats. The aim of this study was to investigate the long-term effects of neonatal BMAA exposure on learning and memory mechanisms and to identify early morphological changes in the neonatal brain. BMAA was injected subcutaneously in rat pups on postnatal days 9-10. BMAA (50 and 200 mg/kg) caused distinct deficits in spatial learning and memory in adult animals but no morphological changes. No impairment of recognition memory was detected, suggesting that neonatal exposure to BMAA preferentially affects neuronal systems that are important for spatial tasks. Histopathological examination revealed early neuronal cell death as determined by TUNEL staining in the hippocampus 24 h after a high dose (600 mg/kg) of BMAA whereas no changes were observed at lower doses (50 and 200 mg/kg). In addition, there was a low degree of neuronal cell death in the retrosplenial and cingulate cortices, areas that are also important for cognitive function. Taken together, these results indicate that BMAA is a developmental neurotoxin inducing long-term changes in cognitive function. The risk posed by BMAA as a potential human neurotoxin merits further consideration, particularly if the proposed biomagnifications in the food chain are confirmed.


Subject(s)
Amino Acids, Diamino/toxicity , Amino Acids, Dicarboxylic/toxicity , Cell Death/physiology , Hippocampus/physiology , Learning Disabilities/chemically induced , Learning Disabilities/psychology , Memory Disorders/chemically induced , Memory Disorders/psychology , Neurons/pathology , Neurotoxins/toxicity , Animals , Animals, Newborn , Anxiety/psychology , Behavior, Animal/drug effects , Body Weight/drug effects , Cognition/drug effects , Cyanobacteria/chemistry , Dose-Response Relationship, Drug , Emotions/drug effects , Exploratory Behavior/drug effects , Female , Hippocampus/pathology , Learning Disabilities/pathology , Maze Learning/drug effects , Memory Disorders/pathology , Motor Activity/drug effects , Pregnancy , Rats , Rats, Wistar , Recognition, Psychology/drug effects
17.
Toxicol Pathol ; 39(2): 325-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21270424

ABSTRACT

The dual peroxisome-proliferator-activated receptor (PPAR) α/γ agonist tesaglitazar has been shown to produce fibrosarcomas in rats. Here, the authors studied morphology, proliferation, differentiation, and inflammation markers in adipose tissue from rats exposed to 1, 3, or 10 µmol/kg tesaglitazar for 2 or 12 weeks, including recovery groups (12 weeks treatment followed by 12 weeks recovery), and 3 or 10 µmol/kg tesaglitazar for 24 weeks. Subcutaneous white and brown fat revealed reversible dose-related histopathological alterations and after 12 and 24 weeks developed areas of thickened skin (fatty lumps). There was a dose-dependent increase in proliferation of interstitial cells in white and brown fat as shown by increased mitotic index in all dose groups after 2 weeks. This was limited to the high dose after 12 and 24 weeks in white fat. Gene expression analyses showed that while tesaglitazar induced differentiation of adipose tissue characterized with a switch in cyclin D1 and D3 mRNA by 12 weeks, longer exposure at high doses reversed this differentiation concurrent with a reappearance of early adipocyte and inflammatory markers. These data suggest that sustained increased turnover of mesenchymal cells in adipose tissues, concomitant with onset of inflammation and fibrosis, drives development of fibrosarcomas in rats treated with tesaglitazar.


Subject(s)
Adipose Tissue/drug effects , Fibrosarcoma/chemically induced , PPAR alpha/agonists , PPAR gamma/agonists , Adipocytes , Adipose Tissue/metabolism , Adipose Tissue/pathology , Alkanesulfonates/blood , Alkanesulfonates/metabolism , Analysis of Variance , Animals , Biomarkers , Cell Proliferation , Fibrosarcoma/pathology , Gene Expression , Inflammation/chemically induced , Male , Phenylpropionates/blood , Phenylpropionates/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar
18.
J Am Soc Nephrol ; 21(8): 1290-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20507942

ABSTRACT

Membranous nephropathy is one of the most common causes of nephrotic syndrome in adults. Recent reports suggest that treatment with adrenocorticotropic hormone (ACTH) reduces proteinuria, but the mechanism of action is unknown. Here, we identified gene expression of the melanocortin receptor MC1R in podocytes, glomerular endothelial cells, mesangial cells, and tubular epithelial cells. Podocytes expressed most MC1R protein, which colocalized with synaptopodin but not with an endothelial-specific lectin. We treated rats with passive Heymann nephritis (PHN) with MS05, a specific MC1R agonist, which significantly reduced proteinuria compared with untreated PHN rats (P < 0.01). Furthermore, treatment with MC1R agonists improved podocyte morphology and reduced oxidative stress. In summary, podocytes express MC1R, and MC1R agonism reduces proteinuria, improves glomerular morphology, and reduces oxidative stress in nephrotic rats with PHN. These data may explain the proteinuria-reducing effects of ACTH observed in patients with membranous nephropathy, and MC1R agonists may provide a new therapeutic option for these patients.


Subject(s)
Adrenocorticotropic Hormone/therapeutic use , Hormones/therapeutic use , Proteinuria/prevention & control , Receptor, Melanocortin, Type 1/agonists , Adult , Aged , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Female , Glomerulonephritis, Membranous/complications , Humans , Male , Mesangial Cells/metabolism , Middle Aged , Podocytes/metabolism , Proteinuria/etiology , Rats , Receptor, Melanocortin, Type 1/biosynthesis , Urothelium/cytology , Urothelium/metabolism
19.
Int J Mol Med ; 23(5): 621-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19360321

ABSTRACT

Serum alanine aminotransferase (ALT) is used as a clinical marker of hepatotoxicity. Three forms of human ALT have been identified, ALT1 and 2 and an alternative splice variant of ALT2 (herein called ALT2_2). The standard ALT activity assay does not discriminate between ALT from different organs, or the isoforms measured in the plasma. Here, we show that ALT1 and 2 possess similar enzymatic activity for alanine and pyruvate but with different Km and kcat values, while recombinant ALT2_2 protein does not possess any enzymatic activity. Isolation of organelles from cultured human skeletal muscle cells, showed localisation of ALT2 to the mitochondrial fraction and endoplasmatic reticulum (ER), but not to the cytosol. In human hepatocytes, on the other hand, ALT1 was only localised to the cytosol and ER, with no detection in mitochondria. ALT2 was not detected in cultured human hepatocytes, liver extract or tissue using Western blotting or immunohistochemistry. The islet of Langerhans and cardiomyocytes were other examples of cells with high expression of catalytic ALT2. A clinical method for selective measurement of ALT1 and 2 in human plasma is described, and both ALT1 and 2 were immunoprecipitated from human plasma and structurally detected using Western blotting techniques.


Subject(s)
Alanine Transaminase/analysis , Alanine Transaminase/blood , Liver/metabolism , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Alanine Transaminase/metabolism , Cells, Cultured , Female , Hepatocytes/metabolism , Humans , Male , Middle Aged , Plasma/chemistry , Plasma/metabolism , Recombinant Proteins/metabolism , Serum/metabolism , Substrate Specificity , Young Adult
20.
APMIS Suppl ; (124): 46-9, 2008.
Article in English | MEDLINE | ID: mdl-18771097

ABSTRACT

In the early 1970s a fatal neurological disorder in cats was reported in the areas around Lake Mälaren in central Sweden. The major signs were hind-leg ataxia, as well as absence or marked decrease in postural reactions and in some cases behavioural changes. The pathology of the disorder was characterized as a non-suppurative meningoencephalomyelitis, but the etiology was not determined. Almost twenty years later, the disorder now known as staggering disease (SD), was further characterized both clinically and pathologically. The same histopathological picture was seen as in the previous study, with inflammatory nodules, neuronal degeneration and perivascular cuffs mainly consisting of lymphocytes. The most severe inflammatory changes were seen in the grey matter of the brain stem, basal ganglia and hippocampus. Clinically the same major neurological signs were seen. Although the cats were examined for several known infectious agents causing central nervous system (CNS) disturbances, no etiological cause of SD was determined.


Subject(s)
Borna Disease , Borna disease virus , Animals , Borna Disease/epidemiology , Borna Disease/pathology , Borna Disease/virology , Borna disease virus/genetics , Cat Diseases/epidemiology , Cat Diseases/pathology , Cat Diseases/virology , Cats , Central Nervous System/pathology , Inflammation/pathology , Meningoencephalitis/epidemiology , Meningoencephalitis/pathology , Meningoencephalitis/veterinary , Meningoencephalitis/virology , Molecular Epidemiology , Nerve Degeneration/pathology , Phylogeny , Sweden/epidemiology , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...