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










Publication year range
1.
Mol Psychiatry ; 27(3): 1542-1551, 2022 03.
Article in English | MEDLINE | ID: mdl-35087195

ABSTRACT

Mounting evidence supports the role of the Ras/mitogen-activated protein kinase (Ras/MAPK) pathway in neurodevelopmental disorders. Here, the authors used a genetics-first approach to examine how Ras/MAPK pathogenic variants affect the functional organization of the brain and cognitive phenotypes including weaknesses in attention and inhibition. Functional MRI was used to examine resting state functional connectivity (RSFC) in association with Ras/MAPK pathogenic variants in children with Noonan syndrome (NS). Participants (age 4-12 years) included 39 children with NS (mean age 8.44, SD = 2.20, 25 females) and 49 typically developing (TD) children (mean age 9.02, SD = 9.02, 33 females). Twenty-eight children in the NS group and 46 in the TD group had usable MRI data and were included in final analyses. The results indicated significant hyperconnectivity for the NS group within canonical visual, ventral attention, left frontoparietal and limbic networks (p < 0.05 FWE). Higher connectivity within canonical left frontoparietal and limbic networks positively correlated with cognitive function within the NS but not the TD group. Further, the NS group demonstrated significant group differences in seed-based striatal-frontal connectivity (Z > 2.6, p < 0.05 FWE). Hyperconnectivity within canonical brain networks may represent an intermediary phenotype between Ras/MAPK pathogenic variants and cognitive phenotypes, including weaknesses in attention and inhibition. Altered striatal-frontal connectivity corresponds with smaller striatal volume and altered white matter connectivity previously documented in children with NS. These results may indicate delayed maturation and compensatory mechanisms and they are important for understanding the pathophysiology underlying cognitive phenotypes in NS and in the broader population of children with neurodevelopmental disorders.


Subject(s)
MAP Kinase Signaling System , Mitogen-Activated Protein Kinases , White Matter , ras Proteins , Attention/physiology , Brain/enzymology , Brain/pathology , Female , Humans , Magnetic Resonance Imaging , White Matter/enzymology , White Matter/pathology , ras Proteins/metabolism
2.
Biochemistry (Mosc) ; 86(5): 540-550, 2021 05.
Article in English | MEDLINE | ID: mdl-33993861

ABSTRACT

Ischemia/reperfusion (I/R) is among the most frequent neurological problems and early intervention to limit the damage is crucial in decreasing mortality and morbidity. Based on reports regarding beneficial effects of melatonin, we investigated its impact on Na+-K+/Mg2+ ATPase and Ca2+/Mg2+ ATPase activities and ultrastructure of gray and white matter in the rat forebrain I/R model. Adult Wistar-albino rats (n = 78), were randomized into control, ischemia (I), ischemia/reperfusion (I/R), low (I/R + melatonin 400 µg/kg), moderate (I/R + melatonin 1200 µg/kg), and high (I/R + melatonin 2400 µg/kg) dose melatonin. Two-vessel occlusion combined with hypotension (15 min) induced ischemia and reperfusion (75 min) achieved by blood reinfusion were performed. Activities of the membrane-bound enzyme, brain malondialdehyde levels, and brain matter ultrastructure were examined in frontoparietal cortices. Melatonin lowered production of malondialdehyde in a dose-dependently. The enzyme activities attenuated under I and I/R, improved with melatonin treatment. I and I/R severely disturbed gray and white matter morphology. Melatonin, in all applied doses, decreased ultrastructural damages in both gray and white matter. Favorable effects of melatonin can be attributed to its antioxidant properties suggesting that it could be a promising neuroprotective agent against I/R injury being effective both for gray and white matter due to favorable biological properties.


Subject(s)
Adenosine Triphosphatases/metabolism , Gray Matter/enzymology , Melatonin/pharmacology , Reperfusion Injury/drug therapy , White Matter/enzymology , Animals , Brain Ischemia , Disease Models, Animal , Gray Matter/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/metabolism , White Matter/metabolism
3.
J Neuropathol Exp Neurol ; 78(6): 492-500, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31058279

ABSTRACT

Leukocyte infiltration is an important pathological hallmark of multiple sclerosis (MS) and is therefore targeted by current MS therapies. The enzyme tissue transglutaminase (TG2) contributes to monocyte/macrophage migration and is present in MS lesions and could be a potential therapeutic target. We examined the cellular identity of TG2-expressing cells by immunohistochemistry in white matter lesions of 13 MS patients; 9 active and chronic active lesions from 4 patients were analyzed in detail. In these active MS lesions, TG2 is predominantly expressed in leukocytes (CD45+) but not in cells of the lymphocyte lineage, that is, T cells (CD3+) and B cells (CD20+). In general, cells of the monocyte/macrophage lineage (CD11b+ or CD68+) are TG2+ but no further distinction could be made regarding pro- or anti-inflammatory macrophage subtypes. In conclusion, TG2 is abundantly present in cells of the monocyte/macrophage lineage in active white matter MS lesions. We consider that TG2 can play a role in MS as it is associated with macrophage infiltration into the CNS. As such, TG2 potentially presents a novel target for therapeutic intervention that can support available MS therapies targeting lymphocyte infiltration.


Subject(s)
GTP-Binding Proteins/metabolism , Lymphocytes/enzymology , Macrophages/enzymology , Monocytes/enzymology , Multiple Sclerosis/enzymology , Transglutaminases/metabolism , White Matter/enzymology , Adult , Aged , Cell Lineage , Female , Humans , Immunohistochemistry , Lymphocytes/pathology , Macrophages/pathology , Male , Middle Aged , Monocytes/pathology , Multiple Sclerosis/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Tissue Banks , White Matter/pathology
4.
Psychiatry Res Neuroimaging ; 286: 4-10, 2019 04 30.
Article in English | MEDLINE | ID: mdl-30822678

ABSTRACT

Considerable evidence suggests that the tryptophan hydroxylase-2 (TPH2) gene is associated with the pathophysiology of major depressive disorder (MDD). In the present study, we investigated alterations of white matter (WM) integrity and the impact of TPH2 polymorphism on WM in a sample of 118 first-episode, medication-naïve, MDD patients and 118 well-matched healthy controls. Whole brain analyses of fractional anisotropy (FA) were performed using tract-based spatial statistics (TBSS). The results showed that the MDD group had significantly reduced FA values for the genu and body of the corpus callosum (CC) and the bilateral anterior corona radiate (ACR). In the MDD patient group, the GG homozygote subgroup exhibited a widespread reduction of FA (uncorrected) and significantly reduced FA in the left retrolenticular portion of the internal capsule and left superior longitudinal fasciculus (SLF) compared with those of the T carriers (GT/TT) (FWE corrected). No significant correlation was found between the FA values in any brain region and the patients' clinical variables. Our findings demonstrate the presence of abnormal white matter integrity in untreated patients with first-episode depression. TPH2-rs4570625 polymorphisms may be involved in the pathological mechanism of WM microarchitecture in patients.


Subject(s)
Corpus Callosum/diagnostic imaging , Depressive Disorder, Major/diagnostic imaging , Depressive Disorder, Major/genetics , Polymorphism, Single Nucleotide/genetics , Tryptophan Hydroxylase/genetics , White Matter/diagnostic imaging , Adult , Corpus Callosum/enzymology , Depressive Disorder, Major/enzymology , Diffusion Tensor Imaging/methods , Female , Humans , Internal Capsule/diagnostic imaging , Internal Capsule/enzymology , Male , Middle Aged , Tryptophan Hydroxylase/metabolism , White Matter/enzymology , Young Adult
5.
Neuroscience ; 398: 274-294, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30031123

ABSTRACT

The α3 Na+,K+-ATPase (α3NKA) is one of four known α isoforms of the mammalian transporter. A deficiency in α3NKA is linked to severe movement control disorders. Understanding the pathogenesis of these disorders is limited by an incomplete knowledge of α3NKA expression in the brain as well as the challenges associated with identifying living cells that express the isoform for subsequent electrophysiological studies. To address this problem, transgenic mice were generated on the C57BL/6 genetic background, which utilize the mouse α3 subunit gene (Atp1a3) promoter to drive the expression of ZsGreen1 fluorescent protein. Consistent with published results on α3NKA distribution, a ZsGreen1 signal was detected in the brain, but not in the liver, with Atp1a3-ZsGreen1 transgenic mice. The intensity of ZsGreen1 fluorescence in neuronal cell bodies varied considerably in the brain, being highest in the brainstem, deep cerebellar and select thalamic nuclei, and relatively weak in cortical regions. Fluorescence was not detected in astrocytes or white matter areas. ZsGreen1-positive neurons were readily observed in fresh (unfixed) brain sections, which were amenable to patch-clamp recordings. Thus, the α3NKA-ZsGreen1 mouse model provides a powerful tool for studying the distribution and functional properties of α3NKA-expressing neurons in the brain.


Subject(s)
Brain/cytology , Brain/enzymology , Mice, Transgenic , Models, Theoretical , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Astrocytes/cytology , Astrocytes/enzymology , Female , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Inbred C57BL , Neurons/cytology , Neurons/enzymology , Patch-Clamp Techniques , Promoter Regions, Genetic , Sodium-Potassium-Exchanging ATPase/genetics , Tissue Culture Techniques , White Matter/cytology , White Matter/enzymology
6.
Mol Neurobiol ; 56(1): 648-657, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29779173

ABSTRACT

Glutaric acidemia type I (GA-I) is a neurometabolic disease caused by deficient activity of glutaryl-CoA dehydrogenase (GCDH) that results in accumulation of metabolites derived from lysine (Lys), hydroxylysine, and tryptophan catabolism. GA-I patients typically develop encephalopatic crises with striatal degeneration and progressive white matter defects. However, late onset patients as well as Gcdh-/- mice only suffer diffuse myelinopathy, suggesting that neuronal death and white matter defects are different pathophysiological events. To test this hypothesis, striatal myelin was studied in Gcdh-/- mice fed from 30 days of age during up to 60 days with a diet containing normal or moderately increased amounts of Lys (2.8%), which ensure sustained elevated levels of GA-I metabolites. Gcdh-/- mice fed with 2.8% Lys diet showed a significant decrease in striatal-myelinated areas and progressive vacuolation of white matter tracts, as compared with animals fed with normal diet. Myelin pathology increased with the time of exposure to high Lys diet and was also detected in 90-day old Gcdh-/- mice fed with normal diet, suggesting that dietary Lys accelerated the undergoing white matter damage. Gcdh-/- mice fed with 2.8% Lys diet also showed increased GRP78/BiP immunoreactivity in oligodendrocytes and neurons, denoting ER stress. However, the striatal and cortical neuronal density was unchanged with respect to normal diet. Thus, myelin damage seen in Gcdh-/- mice fed with 2.8% Lys seems to be mediated by a long-term increased levels of GA-I metabolites having deleterious effects in myelinating oligodendrocytes over neurons.


Subject(s)
Diet , Glutaryl-CoA Dehydrogenase/deficiency , Lysine/adverse effects , White Matter/enzymology , White Matter/injuries , Animals , Cell Count , Cell Death/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Endoplasmic Reticulum Chaperone BiP , Glutaryl-CoA Dehydrogenase/metabolism , Mice , Myelin Sheath/metabolism , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , White Matter/pathology
7.
J Neuropathol Exp Neurol ; 78(2): 191-194, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30590671

ABSTRACT

Cerebral small vessel disease in deep penetrating arteries is a major cause of lacunar infarcts, white matter lesions and vascular cognitive impairment. Local cerebral blood flow in these small vessels is controlled by endothelial-derived nitric oxide, which exerts a primary vasodilator stimulus on vascular myocytes, via cytoplasmic cyclic GMP. Here, we investigated whether the cGMP-degrading enzyme phosphodiesterase-5 (PDE5) is present in small penetrating arteries in the deep subcortical white matter of older people. Frontal cortical tissue blocks were examined from donated brains of older people (n = 42, 24 male: 18 female, median age 81, range: 59-100 years). PDE5, detected by immunohistochemical labeling, was graded as absent, sparse, or abundant in vascular cells within small arteries in subcortical white matter (vessel outer diameter: 20-100 µm). PDE5 labeling within arterial myocytes was detected in all cases. Degree of PDE5 expression (absent, sparse, or abundant) was not associated with age or with neuropathological diagnosis of small vessel disease. In conclusion, PDE5 is present in vascular myocytes within small penetrating arteries in older people. This is a potential molecular target for pharmacological interventions.


Subject(s)
Brain/enzymology , Cerebral Arteries/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Muscle, Smooth, Vascular/enzymology , White Matter/enzymology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Muscle Cells/enzymology
8.
Neurosci Lett ; 687: 37-42, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30125643

ABSTRACT

Strokes occur predominantly in the elderly and white matter (WM) is injured in most strokes, contributing to the disability associated with clinical deficits. Casein kinase 2 (CK2) is expressed in neuronal cells and was reported to be neuroprotective during cerebral ischemia. Recently, we reported that CK2 is abundantly expressed by glial cells and myelin. However, in contrast to its role in cerebral (gray matter) ischemia, CK2 activation during ischemia mediated WM injury via the CDK5 and AKT/GSK3ß signaling pathways (Bastian et al., 2018). Subsequently, CK2 inhibition using the small molecule inhibitor CX-4945 correlated with preservation of oligodendrocytes as well as conservation of axon structure and axonal mitochondria, leading to improved functional recovery. Notably, CK2 inhibition promoted WM function when applied before or after ischemic injury by differentially regulating the CDK5 and AKT/GSK3ß pathways. Specifically, blockade of the active conformation of AKT conferred post-ischemic protection to young, aging, and old WM, suggesting a common therapeutic target across age groups. CK2 inhibitors are currently being used in clinical trials for cancer patients; therefore, it is important to consider the potential benefits of CK2 inhibitors during an ischemic attack.


Subject(s)
Brain Ischemia/enzymology , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Naphthyridines/therapeutic use , Neuroprotective Agents/therapeutic use , White Matter/enzymology , Animals , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Humans , Naphthyridines/pharmacology , Neuroprotective Agents/pharmacology , Phenazines , White Matter/drug effects , White Matter/pathology
9.
Glia ; 66(9): 1960-1971, 2018 09.
Article in English | MEDLINE | ID: mdl-29726608

ABSTRACT

Myelinating glial cells (MGCs), oligodendrocytes (OLs) in the central nervous system (CNS) and Schwann cells (SCs) in the peripheral nervous system (PNS), generate myelin sheaths that insulate axons. After myelination is completed in adulthood, MGC functions independent from myelin are required to support axon survival, but the underlying mechanisms are still unclear. Dicer is a key enzyme that is responsible for generating functional micro-RNAs (miRNAs). Despite the importance of Dicer in initiating myelination, the role of Dicer in mature MGCs is still unclear. Here, Dicer was specifically deleted in mature MGCs in 2-month old mice (PLP-CreERT; Dicer fl/fl) by tamoxifen administration. Progressive motor dysfunction was observed in the Dicer conditional knockout mice, which displayed hind limb ataxia at 3 months post recombination that deteriorated into paralysis within 5 months. Massive axonal degeneration/atrophy in peripheral nerves was responsible for this phenomenon, but overt demyelination was not observed in either the CNS or PNS. In contrast to the PNS, signs of axonal degeneration were not observed in the CNS of these animals. We induced a Dicer deletion in oligodendroglia at postnatal day 5 in NG2-CreERT; Dicer fl/fl mice to evaluate whether Dicer expression in OLs is essential for axonal survival. Dicer deletion in oligodendroglia did not cause motor dysfunction at the age of 7 months. Neither axonal atrophy nor demyelination was observed in the CNS. Based on our results, Dicer expression in SCs is required to maintain axon integrity in adult PNS, and Dicer is dispensable for maintaining myelin sheaths in MGCs.


Subject(s)
Axons/enzymology , DEAD-box RNA Helicases/deficiency , Myelin Sheath/enzymology , Nerve Degeneration/enzymology , Ribonuclease III/deficiency , Animals , Ataxia/enzymology , Ataxia/pathology , Atrophy , Axons/pathology , DEAD-box RNA Helicases/genetics , Disease Progression , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Myelin Sheath/pathology , Nerve Degeneration/pathology , Optic Nerve/enzymology , Optic Nerve/pathology , Paralysis/enzymology , Paralysis/pathology , Ribonuclease III/genetics , Sciatic Nerve/enzymology , Sciatic Nerve/pathology , Spinal Cord/enzymology , Spinal Cord/pathology , White Matter/enzymology , White Matter/pathology
10.
J Stroke Cerebrovasc Dis ; 27(7): 1743-1751, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29602614

ABSTRACT

BACKGROUND AND PURPOSE: Estradiol is a sex steroid hormone known to protect the brain against damage related to transient and global cerebral ischemia. In the present study, we leverage an experimental murine model of bilateral carotid artery stenosis (BCAS) to examine the putative effects of estradiol therapy on chronic cerebral hypoperfusion. We hypothesize that long-term estradiol therapy protects against white matter injury and declarative memory deficits associated with chronic cerebral hypoperfusion. METHODS: Adult male C57BL/6J mice underwent either surgical BCAS or sham procedures. Two days after surgery, the mice were given oral estradiol (Sham+E, BCAS+E) or placebo (Sham+P, BCAS+P) treatments daily for 31-34 days. All mice underwent Novel Object Recognition (NOR) testing 31-34 days after the start of oral treatments. Following sacrifice, blood was collected and brains fixed, sliced, and prepared for histological examination of white matter injury and extracellular signal-regulated kinase (ERK) expression. RESULTS: Animals receiving long-term oral estradiol therapy (BCAS-E2 and Sham-E2) had higher plasma estradiol levels than those receiving placebo treatment (BCAS-P and Sham-P). BCAS-E2 mice demonstrated less white matter injury (Klüver-Barrera staining) and performed better on the NOR task compared to BCAS-P mice. ERK expression in the brain was increased in the BCAS compared to sham cohorts. Among the BCAS mice, the BCAS-E2 cohort had a greater number of ERK + cells. CONCLUSION: This study demonstrates a potentially protective role for oral estradiol therapy in the setting of white matter injury and declarative memory deficits secondary to murine chronic cerebral hypoperfusion.


Subject(s)
Carotid Stenosis/drug therapy , Estradiol/pharmacology , Memory Disorders/prevention & control , Neuroprotective Agents/pharmacology , White Matter/drug effects , Administration, Oral , Animals , Carotid Stenosis/complications , Carotid Stenosis/enzymology , Carotid Stenosis/pathology , Cerebrovascular Circulation , Disease Models, Animal , Estradiol/blood , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Memory Disorders/enzymology , Memory Disorders/etiology , Memory Disorders/pathology , Mice, Inbred C57BL , Neuroprotective Agents/blood , Random Allocation , Recognition, Psychology/drug effects , White Matter/diagnostic imaging , White Matter/enzymology , White Matter/pathology
11.
Glia ; 66(5): 920-933, 2018 05.
Article in English | MEDLINE | ID: mdl-29350438

ABSTRACT

Human mesial temporal lobe epilepsy (MTLE) features subregion-specific hippocampal neurodegeneration and reactive astrogliosis, including up-regulation of the glial fibrillary acidic protein (GFAP) and down-regulation of glutamine synthetase (GS). However, the regional astrocytic expression pattern of GFAP and GS upon MTLE-associated neurodegeneration still remains elusive. We assessed GFAP and GS expression in strict correlation with the local neuronal number in cortical and hippocampal surgical specimens from 16 MTLE patients using immunohistochemistry, stereology and high-resolution image analysis for digital pathology and whole-slide imaging. In the cortex, GS-positive (GS+) astrocytes are dominant in all neuronal layers, with a neuron to GS+ cell ratio of 2:1. GFAP-positive (GFAP+) cells are widely spaced, with a GS+ to GFAP+ cell ratio of 3:1-5:1. White matter astrocytes, on the contrary, express mainly GFAP and, to a lesser extent, GS. In the hippocampus, the neuron to GS+ cell ratio is approximately 1:1. Hippocampal degeneration is associated with a reduction of GS+ astrocytes, which is proportional to the degree of neuronal loss and primarily present in the hilus. Up-regulation of GFAP as a classical hallmark of reactive astrogliosis does not follow the GS-pattern and is prominent in the CA1. Reactive alterations were proportional to the neuronal loss in the neuronal somatic layers (stratum pyramidale and hilus), while observed to a lesser extent in the axonal/dendritic layers (stratum radiatum, molecular layer). We conclude that astrocytic GS is expressed in the neuronal somatic layers and, upon neurodegeneration, is down-regulated proportionally to the degree of neuronal loss.


Subject(s)
Astrocytes/enzymology , Cerebral Cortex/enzymology , Epilepsy, Temporal Lobe/enzymology , Glutamate-Ammonia Ligase/metabolism , Neurons/enzymology , Adult , Astrocytes/pathology , Cell Death/physiology , Cerebral Cortex/pathology , Drug Resistant Epilepsy/enzymology , Drug Resistant Epilepsy/pathology , Drug Resistant Epilepsy/surgery , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/surgery , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/enzymology , Gliosis/pathology , Humans , Immunohistochemistry , Male , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurons/pathology , White Matter/enzymology , White Matter/pathology
12.
Neurobiol Aging ; 53: 27-35, 2017 05.
Article in English | MEDLINE | ID: mdl-28208063

ABSTRACT

In Alzheimer's disease (AD), white matter lesions (WMLs) are associated with an increased risk of progression from mild cognitive impairment (MCI) to dementia, while memory deficits have, at least in part, been linked to a cholinergic deficit. We investigated the relationship between WML load assessed with the Scheltens scale, cerebral acetylcholinesterase (AChE) activity measured with [11C]N-methyl-4-piperidyl acetate PET, and neuropsychological performance in 17 patients with MCI due to AD and 18 cognitively normal older participants. Only periventricular, not nonperiventricular, WML load negatively correlated with AChE activity in both groups. Memory performance depended on periventricular and total WML load across groups. Crucially, AChE activity predicted memory function better than WML load, gray matter atrophy, or age. The effects of WML load on memory were fully mediated by AChE activity. Data suggest that the contribution of WML to the dysfunction of the cholinergic system in MCI due to AD depends on WML distribution. Pharmacologic studies are warranted to explore whether this influences the response to cholinergic treatment.


Subject(s)
Acetylcholinesterase/metabolism , Aging/metabolism , Aging/psychology , Cognitive Dysfunction/etiology , White Matter/enzymology , Acetates , Aged , Aged, 80 and over , Alzheimer Disease/complications , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/psychology , Female , Humans , Male , Memory/physiology , Middle Aged , Neuropsychological Tests , Piperidines , Positron-Emission Tomography , White Matter/diagnostic imaging
13.
Int J Neuropsychopharmacol ; 20(5): 363-373, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28034960

ABSTRACT

Background: Pathology of white matter in brains of patients with major depressive disorder (MDD) is well-documented, but the cellular and molecular basis of this pathology are poorly understood. Methods: Levels of DNA oxidation and gene expression of DNA damage repair enzymes were measured in Brodmann area 10 (BA10) and/or amygdala (uncinate fasciculus) white matter tissue from brains of MDD (n=10) and psychiatrically normal control donors (n=13). DNA oxidation was also measured in BA10 white matter of schizophrenia donors (n=10) and in prefrontal cortical white matter from control rats (n=8) and rats with repeated stress-induced anhedonia (n=8). Results: DNA oxidation in BA10 white matter was robustly elevated in MDD as compared to control donors, with a smaller elevation occurring in schizophrenia donors. DNA oxidation levels in psychiatrically affected donors that died by suicide did not significantly differ from DNA oxidation levels in psychiatrically affected donors dying by other causes (non-suicide). Gene expression levels of two base excision repair enzymes, PARP1 and OGG1, were robustly elevated in oligodendrocytes laser captured from BA10 and amygdala white matter of MDD donors, with smaller but significant elevations of these gene expressions in astrocytes. In rats, repeated stress-induced anhedonia, as measured by a reduction in sucrose preference, was associated with increased DNA oxidation in white, but not gray, matter. Conclusions: Cellular residents of brain white matter demonstrate markers of oxidative damage in MDD. Medications that interfere with oxidative damage or pathways activated by oxidative damage have potential to improve treatment for MDD.


Subject(s)
DNA Glycosylases/metabolism , Deoxyguanosine/analogs & derivatives , Depressive Disorder, Major/pathology , Gene Expression Regulation, Enzymologic/physiology , Poly (ADP-Ribose) Polymerase-1/metabolism , White Matter/enzymology , 8-Hydroxy-2'-Deoxyguanosine , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Astrocytes/metabolism , Deoxyguanosine/metabolism , Depressive Disorder, Major/psychology , Disease Models, Animal , Female , Humans , Interpersonal Relations , Male , Middle Aged , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats , Rats, Sprague-Dawley , Schizophrenia/pathology , Young Adult
14.
PLoS One ; 11(10): e0164301, 2016.
Article in English | MEDLINE | ID: mdl-27723767

ABSTRACT

BACKGROUND: The SLC6A15 gene has been identified as a novel candidate gene for major depressive disorder (MDD). It is presumed to be involved in the pathophysiology of MDD through regulation of glutamate transmission in the brain. However, the involvement of this gene in microstructural changes in white matter (WM) tracts remains unclear. We aimed to investigate the influence of a polymorphism of this gene (rs1545853) on the structural integrity of WM tracts in the cortico-limbic network. METHODS: Eighty-six patients with MDD and 64 healthy controls underwent T1-weighted structural magnetic resonance imaging, including diffusion tensor imaging (DTI), and genotype analysis. We selected the genu of the corpus callosum, the uncinate fasciculus, cingulum, and fornix as regions of interest, and extracted fractional anisotropy (FA) values using the FMRIB Diffusion Toolbox software. RESULTS: FA values for the left parahippocampal cingulum (PHC) was significantly reduced in the patients with MDD compared to healthy control participants (p = 0.004). We also found that MDD patients with the A allele showed reduced FA values for the left PHC than did healthy controls with the A allele (p = 0.012). There was no significant difference in the FA value of left PHC for the comparison between the G homozygotes of MDD and healthy control group. CONCLUSIONS: We observed an association between the risk allele of the SLC6A15 gene rs1545843 and the WM integrity of the PHC in MDD patients, which is known to play an important role in the neural circuit involved in emotion processing.


Subject(s)
Alleles , Amino Acid Transport Systems, Neutral/genetics , Depressive Disorder, Major/diagnostic imaging , Diffusion Tensor Imaging , Nerve Tissue Proteins/genetics , Polymorphism, Genetic , White Matter/diagnostic imaging , Adult , Depressive Disorder, Major/enzymology , Female , Humans , Male , Middle Aged , White Matter/enzymology
15.
Cell Mol Neurobiol ; 36(2): 195-202, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26993507

ABSTRACT

Vascular cognitive impairment (VCI) is a heterogeneous group of diseases linked together by cerebrovascular disease. Treatment of VCI has been hindered by the lack of a coherent pathophysiological process that could provide molecular targets. Of the several forms of VCI, the small vessel disease form is both the most prevalent and generally has a progressive course. Binswanger's disease (BD) is the small vessel form of VCI that involves extensive injury to the deep white matter. Growing evidence suggests that there is disruption of the blood-brain barrier (BBB) secondary to an inflammatory state. Matrix metalloproteinases (MMPs) are increased in the brain and CSF of patients with BD, and have been shown to disrupt the BBB in animal studies, suggesting that they may be biomarkers and therapeutic targets. Multimodal biomarkers derived from clinical, neuropsychological, imaging, and biochemical data can be used to narrow the VCI population to the progressive inflammatory form that will be optimal for treatment trials. This review describes the role of the MMPs in pathophysiology and their use as biomarkers.


Subject(s)
Brain/blood supply , Brain/pathology , Cerebrovascular Disorders/enzymology , Cerebrovascular Disorders/pathology , Cognitive Dysfunction/enzymology , Inflammation/enzymology , Matrix Metalloproteinases/metabolism , Animals , Brain/enzymology , Brain/physiopathology , Cerebrovascular Disorders/physiopathology , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Humans , Inflammation/pathology , Inflammation/physiopathology , White Matter/enzymology , White Matter/pathology , White Matter/physiopathology
16.
Neurochem Int ; 94: 90-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26921668

ABSTRACT

White matter lesions (WML) are thought to contribute to vascular cognitive impairment in elderly patients. Growing evidence show that failure of myelin formation arising from the disruption of oligodendrocyte progenitor cell (OPC) differentiation is a cause of chronic vascular white matter damage. The sphingosine kinase (SphK)/sphingosine-1-phosphate (S1P) signaling pathway regulates oligodendroglia differentiation and function, and is known to be altered in hypoxia. In this study, we measured SphK, S1P as well as markers of WML, hypoxia and OPC (NG2) in a mouse bilateral carotid artery stenosis (BCAS) model of chronic cerebral hypoperfusion. Our results indicated that BCAS induced hypoxia inducible factor (HIF)-1α, Sphk2, S1P, and NG2 up-regulation together with accumulation of WML. In contrast, BCAS mice treated with the SphK inhibitor, SKI-II, showed partial reversal of SphK2, S1P and NG2 elevation and amelioration of WML. In an in vitro model of hypoxia, SKI-II reversed the suppression of OPC differentiation. Our study suggests a mechanism for hypoperfusion-associated WML involving HIF-1α-SphK2-S1P-mediated disruption of OPC differentiation, and proposes the SphK signaling pathway as a potential therapeutic target for white matter disease.


Subject(s)
Brain/blood supply , Brain/enzymology , Carotid Artery Diseases/enzymology , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , White Matter/enzymology , Animals , Brain/drug effects , Carotid Artery Diseases/drug therapy , Carotid Artery Diseases/pathology , Cells, Cultured , Chronic Disease , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Lysophospholipids/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sphingosine/antagonists & inhibitors , Sphingosine/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use , White Matter/drug effects , White Matter/pathology
17.
J Cereb Blood Flow Metab ; 34(10): 1613-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25005877

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT) has been implicated in neuroprotection against ischemic brain injury, but the mechanism underlying its protective effect remains largely unknown. To further examine the protective effect of NAMPT against ischemic stroke and its potential mechanism of action, we generated a novel neuron-specific NAMPT transgenic mouse line. Transgenic mice and wild-type littermates were subjected to transient occlusion of the middle cerebral artery (MCAO) for 60 minutes. Neuron-specific NAMPT overexpression significantly reduced infarct volume by 65% (P=0.018) and improved long-term neurologic outcomes (P≤0.05) compared with littermates. Interestingly, neuronal overexpression of NAMPT increased the area of myelinated fibers in the striatum and corpus callosum, indicating that NAMPT protects against white matter injury. The mechanism of protection appeared to be through extracellular release of NAMPT. First, NAMPT was secreted into the extracellular medium by primary cortical neurons exposed to ischemia-like oxygen-glucose deprivation (OGD) in vitro. Second, conditioned medium from NAMPT-overexpressing neurons exposed to OGD protected cultured oligodendrocytes from OGD. Third, the protective effects of conditioned medium were abolished by antibody-mediated NAMPT depletion, strongly suggesting that the protective effect is mediated by the extracellular NAMPT released into in the medium. These data suggest a novel neuroprotective role for secreted NAMPT in the protection of white matter after ischemic injury.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/pathology , Nicotinamide Phosphoribosyltransferase/metabolism , White Matter/enzymology , White Matter/pathology , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , Nicotinamide Phosphoribosyltransferase/genetics , Up-Regulation , White Matter/metabolism
18.
PLoS One ; 9(6): e100574, 2014.
Article in English | MEDLINE | ID: mdl-24959868

ABSTRACT

Infiltration of leukocytes is a major pathological event in white matter lesion formation in the brain of multiple sclerosis (MS) patients. In grey matter lesions, less infiltration of these cells occur, but microglial activation is present. Thus far, the interaction of ß-integrins with extracellular matrix proteins, e.g. fibronectin, is considered to be of importance for the influx of immune cells. Recent in vitro studies indicate a possible role for the enzyme tissue Transglutaminase (TG2) in mediating cell adhesion and migration. In the present study we questioned whether TG2 is present in white and grey matter lesions observed in the marmoset model for MS. To this end, immunohistochemical studies were performed. We observed that TG2, expressed by infiltrating monocytes in white matter lesions co-expressed ß1-integrin and is located in close apposition to deposited fibronectin. These data suggest an important role for TG2 in the adhesion and migration of infiltrating monocytes during white matter lesion formation. Moreover, in grey matter lesions, TG2 is mainly present in microglial cells together with some ß1-integrin, whereas fibronectin is absent in these lesions. These data imply an alternative role for microglial-derived TG2 in grey matter lesions, e.g. cell proliferation. Further research should clarify the functional role of TG2 in monocytes or microglial cells in MS lesion formation.


Subject(s)
GTP-Binding Proteins/metabolism , Multiple Sclerosis/enzymology , Multiple Sclerosis/pathology , Transglutaminases/metabolism , Animals , Astrocytes/metabolism , Callithrix , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/pathology , Fibronectins/immunology , Fibronectins/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Gene Expression , Gray Matter/enzymology , Gray Matter/immunology , Gray Matter/pathology , Immunohistochemistry , Integrin beta1/immunology , Integrin beta1/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Microglia/metabolism , Monocytes/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Oligodendroglia , Organ Specificity , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/genetics , Transglutaminases/immunology , White Matter/enzymology , White Matter/immunology , White Matter/pathology
19.
J Neurochem ; 131(2): 229-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24934403

ABSTRACT

Serotonin/norepinephrine reuptake inhibitors antidepressants exert their effects by increasing serotonin and norepinephrine in the synaptic cleft. Studies show it takes 2-3 weeks for the mood-enhancing effects, which indicate other mechanisms may underlie their treatment effects. Here, we investigated the role of white matter in treatment and pathogenesis of depression using an unpredictable chronic mild stress (UCMS) mouse model. Desvenlafaxine (DVS) was orally administrated to UCMS mice at the dose of 10 mg/kg/day 1 week before they went through a 7-week stress procedure and lasted for over 8 weeks before the mice were killed. No significant changes were found for protein markers of neurons and astrocytes in UCMS mice. However, myelin and oligodendrocyte-related proteins were significantly reduced in UCMS mice. DVS prevented the stress-induced injury to white matter and the decrease of phosphorylated 5'-AMP-activated protein kinase and 3-hydroxy-3-methyl-glutaryl-CoA reductase protein expression. DVS increased open arm entries in an elevated plus-maze test, sucrose consumption in the sucrose preference test and decreased immobility in tail suspension and forced swimming tests. These findings suggest that stress induces depression-like behaviors and white matter deficits in UCMS mice. DVS may ameliorate the oligodendrocyte dysfunction by affecting cholesterol synthesis, alleviating the depression-like phenotypes in these mice. We examined the possible role of oligodendrocyte and myelin in the pathological changes of depression with an unpredictable chronic mild stress (UCMS) mouse model. Oligodendrocyte-related proteins in the mouse brain were specifically changed during the stress period. The depressive-like behaviors and oligodendrocyte deficits could be prevented by the administration of desvenlafaxine. Oligodendrocyte and myelin may be an essential target of desvenlafaxine for the treatment of depression.


Subject(s)
Cholesterol/biosynthesis , Cyclohexanols/therapeutic use , Depression/drug therapy , Depression/enzymology , Disease Models, Animal , White Matter/enzymology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cyclohexanols/pharmacology , Depression/pathology , Desvenlafaxine Succinate , Female , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Phosphorylation/physiology , Random Allocation , White Matter/drug effects , White Matter/pathology
20.
Neurochem Int ; 77: 17-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24657831

ABSTRACT

Histone deacetylases (HDACs) constitute a super-family of enzymes grouped into four major classes (Class I-IV) that deacetylate histone tails leading to chromatin condensation and gene repression. Whether stroke-induced oligodendrogenesis is related to the expression of individual HDACs in the oligodendrocyte lineage has not been investigated. We found that 2 days after stroke, oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes (OLGs) were substantially reduced in the peri-infarct corpus callosum, whereas at 7 days after stroke, a robust increase in OPCs and OLGs was observed. Ischemic brains isolated from rats sacrificed 7 days after stroke were used to test levels of individual members of Class I (1 and 2) and Class II (4 and 5) HDACs in white matter oligodendrocytes during stroke-induced oligodendrogenesis. Double immunohistochemistry analysis revealed that stroke substantially increased the number of NG2+OPCs with nuclear HDAC1 and HDAC2 immunoreactivity and cytoplasmic HDAC4 which were associated with augmentation of proliferating OPCs, as determined by BrdU and Ki67 double reactive cells after stroke. A decrease in HDAC1 and an increase in HDAC2 immunoreactivity were detected in mature adenomatous polyposis coli (APC) positive OLGs, which paralleled an increase in newly generated BrdU positive OLGs in the peri-infarct corpus callosum. Concurrently, stroke substantially decreased the acetylation levels of histones H3 and H4 in both OPCs and OLGs. Taken together, these findings demonstrate that stroke induces distinct profiles of Class I and Class II HDACs in white matter OPCs and OLGs, suggesting that the individual members of Class I and II HDACs play divergent roles in the regulation of OPC proliferation and differentiation during brain repair after stroke.


Subject(s)
Histone Deacetylases/biosynthesis , Histone Deacetylases/genetics , Oligodendroglia/enzymology , Stroke/enzymology , White Matter/enzymology , Acetylation , Animals , Cell Differentiation , Cell Proliferation , Cerebral Infarction/enzymology , Cerebral Infarction/pathology , Gene Expression Regulation, Enzymologic/genetics , Histones/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Rats , Rats, Wistar , Stem Cells/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...