Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Neurobiol ; 59(4): 2389-2406, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35084654

ABSTRACT

Focal cortical dysplasia (FCD) is associated with blood-brain barrier (BBB) dysfunction in patients with difficult-to-treat epilepsy. However, the underlying cellular and molecular factors in cortical dysplasia (CD) associated with progressive neurovascular challenges during the pro-epileptic phase, post-seizure, and during epileptogenesis remain unclear. We studied the BBB function in a rat model of congenital (in utero radiation-induced, first hit) CD and longitudinally examined the cortical brain tissues at baseline and the progressive neurovascular alterations, glucose transporter-1 (GLUT1) expression, and glucose metabolic activity at 2, 15, and 30 days following a second hit using pentylenetetrazole-induced seizure. Our study revealed through immunoblotting, immunohistochemistry, and biochemical analysis that (1) altered vascular density and prolongation of BBB albumin leakages in CD rats continued through 30 days post-seizure; (2) CD brain tissues showed elevated matrix metalloproteinase-9 levels at 2 days post-seizure and microglial overactivation through 30 days post-seizure; (3) BBB tight junction protein and GLUT1 levels were decreased and neuronal monocarboxylate transporter-2 (MCT2) and mammalian target of rapamycin (mTOR) levels were increased in the CD rat brain: (4) ATPase activity is elevated and a low glucose/high lactate imbalance exists in CD rats; and (5) the mTOR pathway is activated and MCT2 levels are elevated in the presence of high lactate during glucose starvation in vitro. Together, this study suggests that BBB dysfunction, including decreased GLUT1 expression and metabolic disturbance, may contribute to epileptogenesis in this CD rat model through multiple mechanisms that could be translated to FCD therapy in medically refractory epilepsy.


Subject(s)
Epilepsy , Malformations of Cortical Development , Animals , Blood-Brain Barrier/metabolism , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Lactic Acid , Mammals/metabolism , Rats , Seizures , TOR Serine-Threonine Kinases/metabolism
2.
Mol Neurobiol ; 57(11): 4511-4529, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32748370

ABSTRACT

Pharmacoresistance in epilepsy is a major challenge to successful clinical therapy. Glucocorticoid receptor (GR) dysregulation can affect the underlying disease pathogenesis. We recently reported that local drug biotransformation at the blood-brain barrier is upregulated by GR, which controls drug-metabolizing enzymes (e.g., cytochrome P450s, CYPs) and efflux drug transporters (MDR1) in human epileptic brain endothelial cells (EPI-ECs). Here, we establish that this mechanism is influenced upstream by GR and its association with heat shock proteins/co-chaperones (Hsps) during maturation, which differentially affect human epileptic (EPI) tissue and brain endothelial cells. Overexpressed GR, Hsp90, Hsp70, and Hsp40 were found in EPI vs. NON-EPI brain regions. Elevated neurovascular GR expression and co-localization with Hsps was evident in the EPI regions with cortical dysplasia, predominantly in the brain micro-capillaries and neurons. A corresponding increase in ATPase activity (*p < 0.05) was found in the EPI regions. The GR-Hsp90/Hsp70 binding patterns indicated a faster chaperone-promoted maturation of GR, leading to its overactivation in both the tissue and EPI-ECs derived from EPI/focal regions and GR silencing in EPI-ECs slowed such GR-Hsp interactions. Significantly accelerated GR nuclear translocation was determined in EPI-ECs following treatment with GR modulators/ligands dexamethasone, rifampicin, or phenytoin. Our findings reveal that overexpressed GR co-localizes with Hsps in the neurovasculature of EPI brain, increased GR maturation by Hsps accelerates EPI GR machinery, and furthermore this change in EPI and NON-EPI GR-Hsp interaction alters with the age of seizure onset in epileptic patients, together affecting the pathophysiology and drug regulation in the epileptic brain endothelium.


Subject(s)
Brain/pathology , Drug Resistant Epilepsy/metabolism , Drug Resistant Epilepsy/pathology , Endothelial Cells/metabolism , Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adenosine Triphosphatases/metabolism , Adolescent , Adult , Aged , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Child , Cytochrome P-450 Enzyme System/metabolism , Female , Gene Silencing/drug effects , Humans , Male , Middle Aged , Oxcarbazepine/pharmacology , Permeability , Phenytoin/pharmacology , Protein Transport/drug effects , Young Adult
3.
Drug Discov Today ; 25(1): 89-106, 2020 01.
Article in English | MEDLINE | ID: mdl-31541713

ABSTRACT

Glucocorticoid receptors (GRs) are ubiquitous transcription factors widely studied for their role in controlling events related to inflammation, stress and homeostasis. Recently, GRs have reemerged as crucial targets of investigation in neurological disorders, with a focus on pharmacological strategies to direct complex mechanistic GR regulation and improve therapy. In the brain, GRs control functions necessary for neurovascular integrity, including responses to stress, neurological changes mediated by the hypothalamic-pituitary-adrenal axis and brain-specific responses to corticosteroids. Therefore, this review will examine GR regulation at the neurovascular interface in normal and pathological conditions, pharmacological GR modulation and glucocorticoid insensitivity in neurological disorders.


Subject(s)
Nervous System Diseases , Receptors, Glucocorticoid , Animals , Brain/blood supply , Brain/metabolism , Brain/physiology , Drug Resistance , Glucocorticoids/therapeutic use , Humans , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/physiology
4.
Mol Neurobiol ; 56(12): 8392-8407, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31243719

ABSTRACT

Pharmacoresistance is a major clinical challenge for approximately 30% of patients with epilepsy. Previous studies indicate nuclear receptors (NRs), drug efflux transporters, and cytochrome P450 enzymes (CYPs) control drug passage across the blood-brain barrier (BBB) in drug-resistant epilepsy. Here, we (1) evaluate BBB changes, neurovascular nuclear receptors, and drug transporters in lesional/epileptic (EPI) and non-lesional/non-epileptic (NON-EPI) regions of the same brain, (2) examine regional CYP expression and activity, and (3) investigate the association among CYP brain expression, seizure frequency, duration of epilepsy, and antiepileptic drug (AED) combination. We used surgically resected brain specimens from patients with medically intractable epilepsy (n = 22) where the epileptogenic loci were well-characterized by invasive and non-invasive methods; histology confirmed distinction of small NON-EPI regions from EPI tissues. NRs, transporters, CYPs, and tight-junction proteins were assessed by western blots/immunohistochemistry, and CYP metabolic activity was determined and compared. The relationship of CYP expression with seizure frequency, duration of epilepsy, and prescribed AEDs was evaluated. Decreased BBB tight-junction proteins accompanied IgG leakage in EPI regions and correlated with upregulated NR and efflux transporter levels. CYP expression and activity significantly increased in EPI compared to NON-EPI tissues. Change in EPI and NON-EPI CYP3A4 expression increased in patients taking AEDs that were CYP substrates, was downregulated when CYP- and non-CYP-substrate AEDs were given together, and correlated with seizure frequency. Our studies suggest focal neurovascular CYP-NR-transporter alterations, as demonstrated by the relationship of seizure frequency and AED combination to brain CYP3A4, might together impact biotransformation machinery of human pharmacoresistant epilepsy.


Subject(s)
Anticonvulsants/therapeutic use , Brain/enzymology , Epilepsy/drug therapy , Epilepsy/enzymology , Seizures/drug therapy , Seizures/enzymology , Adolescent , Adult , Aged , Anticonvulsants/pharmacology , Biotransformation , Blood-Brain Barrier/pathology , Brain/pathology , Child , Child, Preschool , Cytochrome P-450 CYP3A/metabolism , Female , Humans , Male , Membrane Transport Proteins/metabolism , Middle Aged , Receptors, Cytoplasmic and Nuclear/metabolism , Tight Junction Proteins/metabolism , Young Adult
5.
Mol Neurobiol ; 56(7): 4904-4915, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30414085

ABSTRACT

Death-associated protein kinase (DAPK) is a key player in various cell death signaling pathways. Prolonged seizures induce neuronal stress; thus, we studied DAPK expression in resected brain tissues from patients with refractory epilepsy and the pathophysiological relevance of neurovascular DAPK. We used brain resections from temporal lobe epilepsy (TLE), tumor (BT), arteriovenous malformation (AVM), and autopsy, and isolated human endothelial cells (EPI-ECs) and glial cells (EPI-Astro) from epileptic brains compared to control brain endothelial cells (HBMECs) and astrocytes. DAPK and phosphorylated DAPK (p-DAPK) expression was evaluated by immunohistochemistry and western blot. Subcellular localization of DAPK in epileptic brain was explored; DAPK mRNA/protein levels in EPI-ECs/EPI-Astro were evaluated. We assessed DAPK localization with hypoxic inducible factor (HIF-1α) and vascular endothelial growth factor (VEGF) in epilepsy, BT, and AVM. We found DAPK overexpression across neurons, microcapillaries, and astrocytes in TLE vs controls; DAPK and p-DAPK levels significantly increased only in microsomal fractions of epileptic brain. DAPK mRNA remained unchanged, although increased DAPK and p-DAPK protein expression was observed in EPI-ECs. DAPK inhibition reduced p-DAPK, HIF-1α, and VEGF expression, but increased cytotoxicity and decreased cell viability in EPI-ECs and EPI-astro vs. controls. DAPK staining in TLE resembled BT and AVM, with predominant DAPK/p-DAPK expression in neurons and vasculature. Taken together, these findings suggest DAPK could be a potential molecular target in neuronal death and vascular changes in epilepsy. Increased brain endothelial and astrocytic DAPK in epilepsy, identified for the first time, may have relevance to angiogenesis, hypoxia, and cell survival in pathological conditions.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Death-Associated Protein Kinases/biosynthesis , Drug Resistant Epilepsy/metabolism , Endothelial Cells/metabolism , Astrocytes/pathology , Brain/pathology , Cells, Cultured , Death-Associated Protein Kinases/genetics , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/pathology , Endothelial Cells/pathology , Epilepsy, Temporal Lobe/genetics , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/pathology , Gene Expression , Humans , Neurons/metabolism , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...