Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Neural Regen Res ; 19(7): 1437-1445, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38051885

ABSTRACT

ABSTRACT: Currently, there is a lack of effective medicines capable of halting or reversing the progression of neurodegenerative disorders, including amyotrophic lateral sclerosis, Parkinson's disease, multiple sclerosis, or Alzheimer's disease. Given the unmet medical need, it is necessary to reevaluate the existing paradigms of how to target these diseases. When considering neurodegenerative diseases from a systemic neurometabolic perspective, it becomes possible to explain the shared pathological features. This innovative approach presented in this paper draws upon extensive research conducted by the authors and researchers worldwide. In this review, we highlight the importance of metabolic mitochondrial dysfunction in the context of neurodegenerative diseases. We provide an overview of the risk factors associated with developing neurodegenerative disorders, including genetic, epigenetic, and environmental factors. Additionally, we examine pathological mechanisms implicated in these diseases such as oxidative stress, accumulation of misfolded proteins, inflammation, demyelination, death of neurons, insulin resistance, dysbiosis, and neurotransmitter disturbances. Finally, we outline a proposal for the restoration of mitochondrial metabolism, a crucial aspect that may hold the key to facilitating curative therapeutic interventions for neurodegenerative disorders in forthcoming advancements.

2.
J Pers Med ; 11(3)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809485

ABSTRACT

Depression is one of the most prevalent mental diseases worldwide. Patients with psychiatric diseases often have a history of childhood neglect, indicating that early-life experiences predispose to psychiatric diseases in adulthood. Two strong models were used in the present study: the maternal separation/early deprivation model (MS) and the chronic mild stress model (CMS). In both models, we found changes in the expression of a number of genes such as Creb and Npy. Strikingly, there was a clear regulation of expression of four genes involved in the AP-1 complex: c-Fos, c-Jun, FosB, and Jun-B. Interestingly, different expression levels were observed depending on the model, whereas the combination of the models resulted in a normal level of gene expression. The effects of MS and CMS on gene expression were associated with distinct histone methylation/acetylation patterns of all four genes. The epigenetic changes, like gene expression, were also dependent on the specific stressor or their combination. The obtained results suggest that single life events leave a mark on gene expression and the epigenetic signature of gene promoters, but a combination of different stressors at different life stages can further change gene expression through epigenetic factors, possibly causing the long-lasting adverse effects of stress.

3.
Mol Pain ; 16: 1744806920962925, 2020.
Article in English | MEDLINE | ID: mdl-33021154

ABSTRACT

BACKGROUND: Chronic postoperative pain affects approximately 20% of patients with knee osteoarthritis after total knee replacement. Circulating microRNAs can be found in serum and might act as biomarkers in a variety of diseases. The current study aimed to investigate the preoperative expression of circulating microRNAs as potential predictive biomarkers for the development of chronic postoperative pain in the year following total knee replacement. METHODS: Serum samples, collected preoperatively from 136 knee osteoarthritis patients, were analyzed for 21 circulatory microRNAs. Pain intensity was assessed using a visual analog scale before and one year after total knee replacement. Patients were divided into a low-pain relief group (pain relief percentage <30%) and a high-pain relief group (pain relief percentage >30%) based on their pain relief one year after total knee replacement, and differences in microRNAs expression were analyzed between the two groups. RESULTS: We found that three microRNAs were preoperatively dysregulated in serum in the low-pain relief group compared with the high-pain relief group. MicroRNAs hsa-miR-146a-5p, -145-5p, and -130 b-3p exhibited fold changes of 1.50, 1.55, and 1.61, respectively, between the groups (all P values < 0.05). Hsa-miR-146a-5p and preoperative pain intensity correlated positively with postoperative pain relief (respectively, R = 0.300, P = 0.006; R = 0.500, P < 0.001). DISCUSSION: This study showed that patients with a low postoperative pain relief present a dysregulation of circulating microRNAs. Altered circulatory microRNAs expression correlated with postoperative pain relief, indicating that microRNAs can serve as predictive biomarkers of pain outcome after surgery and hence may foster new strategies for preventing chronic postoperative pain after total knee replacement (TKR).


Subject(s)
MicroRNAs/blood , Osteoarthritis, Knee/complications , Pain, Postoperative/blood , Aged , Biomarkers/blood , Female , Gene Expression Regulation/genetics , Humans , Linear Models , Male , MicroRNAs/genetics , Middle Aged , Osteoarthritis, Knee/surgery , Pain Measurement , Pain, Postoperative/complications , Pain, Postoperative/genetics
4.
Sci Rep ; 10(1): 15583, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32973137

ABSTRACT

The etiology of CNS diseases including multiple sclerosis, Parkinson's disease and amyotrophic lateral sclerosis remains elusive despite decades of research resulting in treatments with only symptomatic effects. In this study, we provide evidence that a metabolic shift from glucose to lipid is a key mechanism in neurodegeneration. We show that, by downregulating the metabolism of lipids through the key molecule carnitine palmitoyl transferase 1 (CPT1), it is possible to reverse or slowdown disease progression in experimental models of autoimmune encephalomyelitis-, SOD1G93A and rotenone models, mimicking these CNS diseases in humans. The effect was seen both when applying a CPT1 blocker or by using a Cpt1a P479L mutant mouse strain. Furthermore, we show that diet, epigenetics, and microbiota are key elements in this metabolic shift. Finally, we present a systemic model for understanding the complex etiology of neurodegeneration and how different regulatory systems are interconnected through a central metabolic pathway that becomes deregulated under specific conditions.


Subject(s)
Brain/pathology , Carnitine O-Palmitoyltransferase/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gastrointestinal Microbiome , Metabolic Networks and Pathways , Parkinson Disease/pathology , Superoxide Dismutase-1/physiology , Animals , Brain/metabolism , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/genetics , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Male , Mice , Mutation , Parkinson Disease/etiology , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Rotenone/toxicity
5.
Mol Neurobiol ; 57(8): 3526-3539, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32542592

ABSTRACT

Ferroportin plays an essential role for iron transport through the blood-brain barrier (BBB), which is formed by brain capillary endothelial cells (BCECs). To maintain the integrity of the BBB, the BCECs gain support from pericytes and astrocytes, which together with neurons form the neurovascular unit (NVU). The objectives of the present study were to investigate ferroportin expression in primary cells of the NVU and to determine if ferroportin mRNA (Fpn) expression is epigenetically regulated. Primary rat BCECs, pericytes, astrocytes, and neurons all expressed ferroportin mRNA at varying levels, with BCECs exhibiting the highest expression of Fpn, peaking when co-cultured but examined separately from astrocytes. Conversely, Fpn expression was lowest in isolated astrocytes, which correlated with high DNA methylation in their Slc40a1 promoter. To provide further evidence for epigenetic regulation, mono-cultured BCECs, pericytes, and astrocytes were treated with the histone deacetylase inhibitors valproic acid (VPA) and sodium butyrate (SB), which significantly increased Fpn and ferroportin protein in BCECs and pericytes. Furthermore, 59Fe export from BCECs was elevated after treatment with VPA. In conclusion, we present first time evidence stating that Fpn expression is epigenetically regulated in BCECs, which may have implications for pharmacological induction of iron transport through the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Neurons/metabolism , Pericytes/metabolism , Animals , Astrocytes/metabolism , Biological Transport/physiology , Brain/metabolism , Capillaries/metabolism , Coculture Techniques/methods , Endothelium, Vascular/metabolism , Epigenesis, Genetic/physiology , Rats
6.
Neurosci Lett ; 704: 145-152, 2019 06 21.
Article in English | MEDLINE | ID: mdl-30974230

ABSTRACT

The CHRNA7 gene encoding the α7 nicotinic acetylcholine receptor (nAChR) has repeatedly been linked with schizophrenia and the P50 sensory gating deficit. The α7 nAChR is considered a promising drug target for treatment of cognitive dysfunction in schizophrenia and improves memory and executive functions in patients and healthy individuals. However, clinical trials with pro-cognitive drugs are challenged by large inter-individual response variations and these have been linked to genotypic variations reducing CHRNA7 expression and α7 nAChR function. Genetic variants as well as environmental conditions may cause epigenetic dysregulation and it has previously been found that DNA methylation of a region surrounding the transcription start site of CHRNA7 is important for tissue specific regulation and gene silencing. In the present study we identify two additional regions involved in epigenetic regulation of the CHRNA7 promoter. In human temporal cortex we find large variations in expression of CHRNA7 and establish evidence for a significant correlation with DNA methylation levels of one region. We then establish evidence that genotypic variations can influence methylation levels of the CHRNA7 promoter. Epigenetic dysregulation can be reversed by pharmacological intervention and in HeLa cells. Valproate, a commonly used mood stabiliser, caused demethylation and increased CHRNA7 expression in HeLA cells. Similar demethylation effect and increased CHRNA7 expression was obtained in SH-SY5Y cells stimulated concomitantly with valproate and nicotine. In summary, both genetic and epigenetic information could be useful to predict treatment outcomes in patients and epigenetic modulation may serve as a mechanism for potentiating the effects of α7 nAChR agonists.


Subject(s)
DNA Methylation , Histone Deacetylase Inhibitors/pharmacology , Valproic Acid/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Brain/metabolism , Brain/pathology , Cells, Cultured , Drug Interactions , Epigenesis, Genetic , Gene Expression/drug effects , Humans , Nicotine/pharmacology , Promoter Regions, Genetic , Transcription, Genetic , alpha7 Nicotinic Acetylcholine Receptor/genetics
7.
Mol Neurobiol ; 56(4): 2375-2378, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30128649

ABSTRACT

The original version of this article unfortunately contained mistakes on Figs. 1, 2, and 7 as some of the data were not visible. With this, the correct images are hereby published.

8.
Mol Neurobiol ; 56(4): 2362-2374, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30027341

ABSTRACT

Ferroportin is the only known iron exporter, and its regulation seems to be controlled at both transcriptional, post-transcriptional, and post-translational levels. The objective of the current work was to investigate how cellular iron status affects the expression of the ferroportin gene Fpn under the influence of hepcidin, known to post-translational lower the available ferroportin protein. Nerve growth factor-beta (NGF-ß)-differentiated PC12 cells, used as a model of neuronal cells, were evaluated in terms of their viability and expression of ferroportin after inducing cellular iron overload with ferric ammonium citrate (FAC) or hepcidin, iron deficiency with deferoxamine (DFO), or hepcidin in combination with FAC or DFO. Ferritin mRNA was significantly upregulated following treatment with 20 mM FAC. The viability of the differentiated PC12 cells was significantly reduced after treatment with 30 mM FAC or 1.0 µM hepcidin, but when combining FAC and hepcidin treatment, the cells remained unaffected. The expression of Fpn was concurrently upregulated after treatment with FAC in combination with hepcidin. Fifty millimolar DFO also increased Fpn. Together, these data point towards a transcriptional induction of Fpn in response to changes in cellular iron levels. Epigenetic regulation of Fpn may also occur as changes in genes associated with epigenetic regulation of Fpn were demonstrated.


Subject(s)
Cation Transport Proteins/genetics , Cell Differentiation , Hepcidins/pharmacology , Iron/pharmacology , Neurons/cytology , Neurons/metabolism , Transcription, Genetic , Animals , Cation Transport Proteins/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Deferoxamine/pharmacology , Epigenesis, Genetic/drug effects , Ferric Compounds/pharmacology , Models, Biological , Neurons/drug effects , PC12 Cells , Quaternary Ammonium Compounds/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcription, Genetic/drug effects
9.
PLoS One ; 12(1): e0170121, 2017.
Article in English | MEDLINE | ID: mdl-28095495

ABSTRACT

The bromodomain containing 1 gene, BRD1 is essential for embryogenesis and CNS development. It encodes a protein that participates in histone modifying complexes and thereby regulates the expression of a large number of genes. Genetic variants in the BRD1 locus show association with schizophrenia and bipolar disorder and risk alleles in the promoter region correlate with reduced BRD1 expression. Insights into the transcriptional regulation of BRD1 and the pathogenic mechanisms associated with BRD1 risk variants, however, remain sparse. By studying transcripts in human HeLa and SH-SY5Y cells we provide evidence for differences in relative expression of BRD1 transcripts with three alternative 5' UTRs (exon 1C, 1B, and 1A). We further show that expression of these transcript variants covaries negatively with DNA methylation proportions in their upstream promoter regions suggesting that promoter usage might be regulated by DNA methylation. In line with findings that the risk allele of the rs138880 SNP in the BRD1 promoter region correlates with reduced BRD1 expression, we find that it is also associated with moderate regional BRD1 promoter hypermethylation in both adipose tissue and blood. Importantly, we demonstrate by inspecting available DNA methylation and expression data that these regions undergo changes in methylation during fetal brain development and that differences in their methylation proportions in fetal compared to postnatal frontal cortex correlate significantly with BRD1 expression. These findings suggest that BRD1 may be dysregulated in both the developing and mature brain of risk allele carriers. Finally, we demonstrate that commonly used mood stabilizers Lithium, Valproate, and Carbamazepine affect the expression of BRD1 in SH-SY5Y cells. Altogether this study indicates a link between genetic risk and epigenetic dysregulation of BRD1 which raises interesting perspectives for targeting the mechanisms pharmacologically.


Subject(s)
Adenocarcinoma/genetics , DNA Methylation , Neuroblastoma/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic/genetics , Schizophrenia/genetics , Adenocarcinoma/pathology , Brain/metabolism , Brain/pathology , Female , Fetus/metabolism , Fetus/pathology , Gene Expression Regulation , Genetic Predisposition to Disease , HeLa Cells , Histone Acetyltransferases , Histone Chaperones , Humans , Neuroblastoma/pathology , Schizophrenia/pathology
10.
Fluids Barriers CNS ; 12: 19, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26246240

ABSTRACT

BACKGROUND: Primary brain capillary endothelial cells (BCECs) are a promising tool to study the blood-brain barrier (BBB) in vitro, as they maintain many important characteristics of the BBB in vivo, especially when co-cultured with pericytes and/or astrocytes. A novel strategy for drug delivery to the brain is to transform BCECs into protein factories by genetic modifications leading to secretion of otherwise BBB impermeable proteins into the central nervous system. However, a huge challenge underlying this strategy is to enable transfection of non-mitotic BCECs, taking a non-viral approach. We therefore aimed to study transfection in primary, non-mitotic BCECs cultured with defined BBB properties without disrupting the cells' integrity. METHODS: Primary cultures of BCECs, pericytes and astrocytes were generated from rat brains and used in three different in vitro BBB experimental arrangements, which were characterised based on a their expression of tight junction proteins and other BBB specific proteins, high trans-endothelial electrical resistance (TEER), and low passive permeability to radiolabeled mannitol. Recombinant gene expression and protein synthesis were examined in primary BCECs. The BCECs were transfected using a commercially available transfection agent Turbofect™ to express the red fluorescent protein HcRed1-C1. The BCECs were transfected at different time points to monitor transfection in relation to mitotic or non-mitotic cells, as indicated by fluorescence-activated cell sorting analysis after 5-and 6-carboxylfluorescein diacetate succinidyl ester incorporation. RESULTS: The cell cultures exhibited important BBB characteristics judged from their expression of BBB specific proteins, high TEER values, and low passive permeability. Among the three in vitro BBB models, co-culturing with BCECs and astrocytes was well suited for the transfection studies. Transfection was independent of cell division and with equal efficacy between the mitotic and non-mitotic BCECs. Importantly, transfection of BCECs exhibiting BBB characteristics did not alter the integrity of the BCECs cell layer. CONCLUSIONS: The data clearly indicate that non-viral gene therapy of BCECs is possible in primary culture conditions with an intact BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Cell Culture Techniques/methods , Endothelial Cells/metabolism , Transfection , Animals , Astrocytes/metabolism , Astrocytes/physiology , Blood-Brain Barrier/cytology , Capillary Permeability , Cell Division , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Pericytes/metabolism , Pericytes/physiology , Rats , Rats, Sprague-Dawley
11.
Mol Cell Neurosci ; 67: 137-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26141855

ABSTRACT

Electroconvulsive therapy (ECT) remains one of the most effective treatments of major depression. Unfortunately, some patients report side effects, of which the most prominent are memory deficits. The immediate early gene Arc plays a critical role in the maintenance phase of long-term potentiation and consolidation of memory in the rat brain. We recently observed increased methylation of the Arc promoter 24h after acute electroconvulsive stimulation (ECS) in rats, which could cause decreased Arc expression and provide an explanation for the observed memory deficits. In the present study we investigated the methylation and expression changes of Arc at 48h post-ECS and determined the role of de-novo methylation in that process. We initially measured expression of DNA methyltransferases (Dnmt1 and Dnmt3a) and Arc 1, 4, 8, 16, 24, and 48h after a single ECS. Arc expression increased approximately 10-fold at 1 and 4h after ECS, and subsequently decreased below sham levels. Four hours after ECS we also observed a significant increase in Dnmt3a expression, which was attenuated in a second experiment by the use of DNMT inhibitor decitabine (5-aza-2-deoxycytidine). We then investigated Arc gene expression and methylation changes at 48h post-ECS and we found a slightly reduced Arc expression in ECS-treated rats as compared to sham. In animals that received decitabine we observed a significant decrease in Dnmt3a expression and an increase of Arc expression in both ECS and sham groups. The same tendency for reduced Arc expression after ECS, as compared to sham was observed despite the blocking of DNA methylation with decitabine. The DNA methylation as measured by pyrosequencing is decreased 48h post-ECS both in the promoter and intragenic regions as a response to ECS regardless of the treatment with decitabine. Overall the results suggest that DNA methylation is involved in regulating Arc expression but is not the causal mechanism responsible for reducing Arc expression after ECS. We speculate that the decrease is caused by ECS-induced HDAC2 upregulation and decreased H3 acetylation at the Arc promoter.


Subject(s)
Cytoskeletal Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Nerve Tissue Proteins/genetics , Transcranial Direct Current Stimulation , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Brain/metabolism , Brain/physiology , Cytoskeletal Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , DNA Methyltransferase 3A , Decitabine , Enzyme Inhibitors/pharmacology , Male , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley
12.
Neurobiol Dis ; 81: 108-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25801802

ABSTRACT

Chronic inflammation in the substantia nigra (SN) accompanies conditions with progressive neurodegeneration. This inflammatory process contributes to gradual iron deposition that may catalyze formation of free-radical mediated damage, hence exacerbating the neurodegeneration. This study examined proteins related to iron-storage (ferritin) and iron-export (ferroportin) (aka metal transporter protein 1, MTP1) in a model of neurodegeneration. Ibotenic acid injected stereotactically into the striatum leads to loss of GABAergic neurons projecting to SN pars reticulata (SNpr), which subsequently leads to excitotoxicity in the SNpr as neurons here become vulnerable to their additional glutamatergic projections from the subthalamic nucleus. This imbalance between glutamate and GABA eventually led to progressive shrinkage of the SNpr and neuronal loss. Neuronal cell death was accompanied by chronic inflammation as revealed by the presence of cells expressing ED1 and CD11b in the SNpr and the adjacent white matter mainly denoted by the crus cerebri. The SNpr also exhibited changes in iron metabolism seen as a marked accumulation of inflammatory cells containing ferric iron and ferritin with morphology corresponding to macrophages and microglia. Ferritin was detected in neurons of the lesioned SNpr in contrast to the non-injected side. Compared to non-injected rats, surviving neurons of the SNpr expressed ferroportin at unchanged level. Analyses of dissected SNpr using RT-qPCR showed a rise in ferritin-H and -L transcripts with increasing age but no change was observed in the lesioned side compared to the non-lesioned side, indicating that the increased expression of ferritin in the lesioned side occurred at the post-transcriptional level. Hepcidin transcripts were higher in the lesioned side in contrast to ferroportin mRNA that remained unaltered. The continuous entry of iron-containing inflammatory cells into the degenerating SNpr and their subsequent demise is probably responsible for iron donation in neurodegeneration. This is accompanied by only a slight increase in neuronal ferritin and not ferroportin, which suggests that the iron-containing debris of dying inflammatory cells and degenerating neurons gets scavenged by invading macrophages and activated microglia to prevent an increase in neuronal iron.


Subject(s)
Brain/pathology , Encephalitis/complications , Ferritins/metabolism , Iron/metabolism , Microglia/metabolism , Neurodegenerative Diseases/complications , Analysis of Variance , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Brain/metabolism , Disease Models, Animal , Ectodysplasins/metabolism , Ferritins/genetics , Ibotenic Acid/toxicity , Male , Microglia/drug effects , Neural Pathways/pathology , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors
13.
Gene ; 539(1): 8-14, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24518690

ABSTRACT

Electroconvulsive therapy (ECT) remains one of the most effective treatments of major depression. It has been suggested that the mechanisms of action involve gene expression. In recent decades there have been several investigations of gene expression following both acute and chronic electroconvulsive stimulation (ECS). These studies have focused on several distinct gene targets but have generally included only few time points after ECS for measuring gene expression. Here we measured gene expression of three types of genes: Immediate early genes, synaptic proteins, and neuropeptides at six time points following an acute ECS. We find significant increases for c-Fos, Egr1, Neuritin 1 (Nrn 1), Bdnf, Snap29, Synaptotagmin III (Syt 3), Synapsin I (Syn 1), and Psd95 at differing time points after ECS. For some genes these changes are prolonged whereas for others they are transient. Npy expression significantly increases whereas the gene expression of its receptors Npy1r, Npy2r, and Npy5r initially decreases. These decreases are followed by a significant increase for Npy2r, suggesting anticonvulsive adaptations following seizures. In summary, we find distinct changes in mRNA quantities that are characteristic for each gene. Considering the observed transitory and inverse changes in expression patterns, these data underline the importance of conducting measurements at several time points post-ECS.


Subject(s)
Electroconvulsive Therapy/adverse effects , Genes, Immediate-Early/genetics , Hippocampus/metabolism , Synaptic Membranes/metabolism , Animals , Gene Expression Profiling , Male , Models, Animal , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/biosynthesis , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Seizures/genetics , Synaptic Membranes/genetics , Transcriptome
14.
Pharmaceutics ; 5(2): 246-60, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-24300449

ABSTRACT

The aim of the present study was to evaluate the transfection potential of chitosan-coated, green-fluorescent magnetic nanoparticles (MNPs) (chi-MNPs) after encapsulation inside polyethylglycol (PEG)ylated liposomes that produced lipid-encapsulated chitosan-coated MNPs (lip-MNPs), and also to evaluate how these particles would distribute in vivo after systemic injection. The transfection potential of both chi-MNPs and lip-MNPs was evaluated in vitro in rat brain endothelial 4 (RBE4) cells with and without applying a magnetic field. Subsequently, the MNPs were evaluated in vivo in young rats. The in vitro investigations revealed that the application of a magnetic field resulted in an increased cellular uptake of the particles. The lip-MNPs were able to transfect the RBE4 cells with an incidence of approximately 20% of a commercial transfection agent. The in vivo distribution studies revealed that lip-MNPs had superior pharmacokinetic properties due to evasion of the RES, including hepatic Kuppfer cells and macrophages in the spleen. In conclusion, we were able to design a novel lipid-encapsulated MNP with the ability to carry genetic material, with favorable pharmacokinetic properties, and under the influence of a magnetic field with the capability to mediate transfection in vitro.

15.
Behav Pharmacol ; 23(8): 735-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23075705

ABSTRACT

In the present study we report the finding that the quality of maternal care, in early life, increased the susceptibility to stress exposure in adulthood, when rats were exposed to the chronic mild stress paradigm. Our results indicate that high, as opposed to low maternal care, predisposed rats to a differential stress-coping ability. Thus rats fostered by low maternal care dams became more prone to adopt a stress-susceptible phenotype developing an anhedonic-like condition. Moreover, low maternal care offspring had lower weight gain and lower locomotion, with no additive effect of stress. Subchronic exposure to chronic mild stress induced an increase in faecal corticosterone metabolites, which was only significant in rats from low maternal care dams. Examination of glucocorticoid receptor exon 17 promoter methylation in unchallenged adult, maternally characterized rats, showed an insignificant tendency towards higher total cytosine methylation in rats from low maternal care dams. Assessment of methylation in the resilient versus anhedonic-like rat phenotypes, revealed only minor differences. Thus, maternal care status seems to be a strong predictor or trait marker for the behavioural phenotype.


Subject(s)
Depression/etiology , Maternal Behavior/physiology , Stress, Physiological , Animals , Behavior, Animal , Corticosterone/metabolism , DNA Methylation , Disease Models, Animal , Disease Susceptibility , Female , Locomotion/physiology , Male , Pregnancy , Promoter Regions, Genetic , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Weight Gain/physiology
16.
Curr Pharm Biotechnol ; 13(12): 2349-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23016641

ABSTRACT

The endothelial cells of the brain form the blood-brain barrier (BBB) that denotes a major restraint for drug entry to the brain. Traditional attempts to bypass the BBB have been by formulation of drugs with lipophilicity or low molecular weight designed to enable transport via solute nutrient transporters. The identification of many new targets in the brain cells form new ways of thinking drug design as modern therapeutics could be proteins and molecules of genetic origins like siRNA and cDNA that are prevented from entry into the brain unless encapsulated in drug carriers. In many chronic disorders affecting the central nervous system, the BBB is physically intact which further limits the entry of large molecules. The desirable entry of such molecules will be made by formulation of particular drug carriers that will enable their transport into the brain endothelium, or even through the endothelium and into the brain. This review discusses the potential of different principles for drug therapy to the brain with these main emphases on drug transport through the BBB: i) the effects of molecular lipidization, ii) the involvement of solute nutrient carriers, iii) targeted delivery using small peptides with high membrane penetrating properties, iv) treatment with magnetic nanoparticles. These different principles for therapy are also discussed with focus on possibilities of their improvement for targeted delivery to the brain.


Subject(s)
Brain/metabolism , Drug Delivery Systems , Adsorption , Animals , Cell-Penetrating Peptides/administration & dosage , Endocytosis , Humans , Magnetic Phenomena , Membrane Proteins/metabolism , Nanoparticles/administration & dosage , Pharmaceutical Preparations/administration & dosage
17.
Brain Res Bull ; 88(5): 507-13, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22613772

ABSTRACT

Electroconvulsive stimulation (ECS) remains one of the most effective treatments of major depression. However, the underlying molecular changes still remain to be elucidated. Since ECS causes rapid and significant changes in gene expression we have looked at epigenetic regulation of two important immediate early genes that are both induced after ECS: c-Fos and Arc. We examined Arc and c-Fos protein expression and found Arc present over 4 h, in contrast to c-Fos presence lasting only 1 h. Both genes had returned to baseline expression at 24 h post-ECS. Histone H4 acetylation (H4Ac) is one of the important epigenetic marks associated with gene activation. We show increased H4Ac at the c-Fos promoter at 1 h post-ECS. Surprisingly, we also observed a significant increase in DNA methylation of the Arc gene promoter at 24 h post-ECS. DNA methylation, which is responsible for gene silencing, is a rather stable covalent modification. This suggests that Arc expression has been repressed and may consequently remain inhibited for a prolonged period post-ECS. Arc plays a critical role in the maintenance phase of long-term potentiation (LTP) and consolidation of memory in the rat brain. Thus, this study is one of the first to demonstrate DNA methylation as a regulator of ECS-induced gene expression and it provides a molecular link to the memory deficits observed after ECS.


Subject(s)
Cytoskeletal Proteins/genetics , Electroshock , Epigenesis, Genetic/genetics , Hippocampus/physiology , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins c-fos/genetics , Animals , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/physiology , DNA Methylation/genetics , Electroshock/methods , Male , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/physiology , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/physiology , Rats , Rats, Sprague-Dawley , Time Factors
18.
J Control Release ; 151(1): 45-50, 2011 Apr 10.
Article in English | MEDLINE | ID: mdl-21251935

ABSTRACT

The blood-brain barrier (BBB) formed by brain capillary endothelial cells protects the brain against potentially harmful substances present in the circulation, but also restricts exogenous substances such as pharmacologically acting drugs or proteins from entering the brain. A novel and rather unchallenged approach to allow proteins to enter the brain is gene therapy based on delivery of genetic material into brain capillary endothelial cells. In theory in vivo transfection will allow protein expression and secretion from brain capillary endothelial cells and further into the brain. This would denote a new paradigm for therapy to transport proteins across the BBB. The aim of this study was to investigate the possibility to use brain capillary endothelial cells as factories for recombinant protein production. Non-viral gene carriers were prepared from pullulan, a polysaccharide, and spermine, a naturally occurring polyamine that were additionally conjugated with plasmid DNA. We were able to transfect rat brain endothelial cells (RBE4s) and human brain microvascular endothelial cells (HBMECs). Transfection of HBMECs with pullulan-spermine conjugated with plasmid DNA bearing cDNA encoding human growth hormone 1 (hGH1), led to secretion of hGH1 protein into the growth medium. Hence, the pullulan-spermine delivery system is a very promising method for delivering DNA to brain endothelial cells with potential for using these cells as factories for secretion of proteins.


Subject(s)
Blood-Brain Barrier/metabolism , DNA/administration & dosage , Endothelial Cells/metabolism , Glucans/chemistry , Growth Hormone/genetics , Transfection , Adult , Cells, Cultured , DNA/genetics , Female , Growth Hormone/metabolism , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spermine/chemistry
19.
Biometals ; 24(2): 357-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21213119

ABSTRACT

Iron is crucial for a variety of cellular functions in neuronal cells. Neuronal iron uptake is reflected in a robust and consistent expression of transferrin receptors and divalent metal transporter 1 (DMT 1). Conversely, the mechanisms by which neurons neutralize and possibly excrete iron are less clear. Studies indicate that neurons express ferroportin which could reflect a mechanism for iron export. We mapped the distribution of ferroportin in the adult mouse brain using an antibody prepared from a peptide representing amino acid sequences 223-303 of mouse ferroportin. The antibody specifically detected ferroportin in brain homogenates, whereas homogenates of cultured endothelial cells were devoid of immunoreactivity. In brain sections, ferroportin was confined to neuronal cell bodies and peripheral processes of cerebral cortex, hippocampus, thalamus, brain stem, and cerebellum. In brain stem ferroportin-labeling was particularly high in neurons of cranial nerve nuclei and reticular formation. Ferroportin was hardly detectable in striatum, pallidum, or hypothalamus. Among non-neuronal cells, ferroportin was detected in oligodendrocytes and choroid plexus epithelial cells. A comparison with previous studies on the distribution of transferrin receptors in neurons shows that many neuronal pools coincide with those expressing ferroportin. The data therefore indicate that neuronal iron homeostasis consists of a delicate balance between transferrin receptor-mediated uptake of iron-transferrin and ferroportin-related iron excretion. The findings also suggest a particular high turnover of iron in neuronal regions, such as habenula, hippocampus, reticular formation and cerebellum, as several neurons in these regions exhibit a prominent co-expression of transferrin receptors and ferroportin.


Subject(s)
Brain/cytology , Brain/metabolism , Cation Transport Proteins/metabolism , Neurons/metabolism , Animals , Blotting, Western , Cerebral Cortex , Immunohistochemistry , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction
20.
Drug Discov Today ; 15(3-4): 137-41, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20036755

ABSTRACT

Schizophrenia is characterized by a diverse symptomatology that often includes positive, cognitive and negative symptoms. Current anti-schizophrenic drugs act at multiple receptors, but little is known about how each of these receptors contributes to their mechanisms of action. Screening of novel anti-schizophrenic drug candidates targeting single receptors will be based on biomarker assays that measure signalling pathways, transcriptional factors, epigenetic mechanisms and synaptic function and translate these effects to behavioural effects in animals and humans. This review discusses current states of the validity of biomarkers in the identification of novel anti-schizophrenic drug candidates.


Subject(s)
Antipsychotic Agents/therapeutic use , Biomarkers, Pharmacological/metabolism , Drug Discovery/methods , Schizophrenia , Animals , Antipsychotic Agents/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Epigenesis, Genetic/drug effects , Genetic Predisposition to Disease , Humans , Models, Biological , Nerve Degeneration/metabolism , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...