Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.105
Filter
1.
J Neurochem ; 160(3): 356-375, 2022 02.
Article in English | MEDLINE | ID: mdl-34837396

ABSTRACT

Neurodegeneration with brain iron accumulation (NBIA) is a clinically and genetically heterogeneous group of neurodegenerative diseases characterized by the abnormal accumulation of brain iron and the progressive degeneration of the nervous system. One of the recently identified subtypes of NBIA is ß-propeller protein-associated neurodegeneration (BPAN). BPAN is caused by de novo mutations in the WDR45/WIPI4 (WD repeat domain 45) gene. WDR45 is one of the four mammalian homologs of yeast Atg18, a regulator of autophagy. WDR45 deficiency in BPAN patients and animal models may result in defects in autophagic flux. However, how WDR45 deficiency leads to brain iron overload remains unclear. To elucidate the role of WDR45, we generated a WDR45-knockout (KO) SH-SY5Y neuroblastoma cell line using CRISPR-Cas9-mediated genome editing. Using these cells, we demonstrated that the non-TF (transferrin)-bound iron pathway dominantly mediated the accumulation of iron. Moreover, the loss of WDR45 led to defects in ferritinophagy, a form of autophagy that degrades the iron storage protein ferritin. We showed that impaired ferritinophagy contributes to iron accumulation in WDR45-KO cells. Iron accumulation was also detected in the mitochondria, which was accompanied by impaired mitochondrial respiration, elevated reactive oxygen species, and increased cell death. Thus, our study links WDR45 to specific iron acquisition pathways and ferritinophagy. Cover Image for this issue: https://doi.org/10.1111/jnc.15388.


Subject(s)
Autophagy/genetics , Carrier Proteins/genetics , Iron Overload/genetics , Neurodegenerative Diseases/genetics , Brain Chemistry/genetics , Cell Death , Cell Line , Gene Knockout Techniques , Humans , Iron/metabolism , Iron Overload/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species , Transferrin/metabolism
2.
Front Endocrinol (Lausanne) ; 12: 750797, 2021.
Article in English | MEDLINE | ID: mdl-34759891

ABSTRACT

The melanocortin receptors are defined as a series of vital pharmaceutical targets to regulate neuronal appetite and maintain controllable body weight for mammals and teleosts. Melanocortin receptor accessory protein 2 (MRAP2) functions as an essential accessory player that modulates the surface translocation and binding to a variety of endogenous or synthetic hormones of central melanocortin-4 receptor (MC4R) signaling. MRAP2 is a single-transmembrane protein and could form a functional symmetric antiparallel homodimer topology. Here, we inverted the N-terminal, transmembrane, and C-terminal domains and generated six distinct conformational variants of the mouse MRAP2 to explore the functional orientations and the internal symmetry of MRAP2 dimers. These remolded MRAP2 mutants showed proper assembly of the antiparallel homodimer and binding to the MC4R, but slightly altered the regulatory profile on the surface expression and the ligand-stimulated cAMP cascades of MC4R. This study elucidated the importance of the orientation of each domain of the single-transmembrane protein and revealed the pharmacological properties of the internal symmetry of the antiparallel homodimer for MRAP2.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Animals , Body Weight , Brain Chemistry/genetics , Cyclic AMP/metabolism , HEK293 Cells , Humans , Mice , Mutation , Protein Conformation , Receptor, Melanocortin, Type 4 , Signal Transduction
3.
CNS Neurosci Ther ; 27(11): 1289-1299, 2021 11.
Article in English | MEDLINE | ID: mdl-34347369

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disease, and its pathogenesis is unclear. Previous studies mainly focus on the lesions of substantia nigra (SN) and striatum (Str) in PD. However, lesions are not limited. The olfactory bulb (OB), subventricular zone (SVZ), and hippocampus (Hippo) are also affected in PD. AIM: To reveal gene expression changes in the five brain regions (OB, SVZ, Str, SN, and Hippo), and to look for potential candidate genes and pathways that may be correlated with the pathogenesis of PD. MATERIALS AND METHODS: We established control group and 6-hydroxydopamine (6-OHDA) PD model group, and detected gene expressions in the five brain regions using RNA-seq and real-time quantitative polymerase chain reaction (RT-qPCR). We further analyzed the RNA-seq data by bioinformatics. RESULTS: We identified differentially expressed genes (DEGs) in all five brain regions. The DEGs were significantly enriched in the "dopaminergic synapse" and "retrograde endocannabinoid signaling," and Gi/o-GIRK is the shared cascade in the two pathways. We further identified Ephx2, Fam111a, and Gng2 as the potential candidate genes in the pathogenesis of PD for further studies. CONCLUSION: Our study suggested that gene expressions change in the five brain regions following exposure to 6-OHDA. The "dopaminergic synapse," "retrograde endocannabinoid signaling," and Gi/o-GIRK may be the key pathways and cascade of the synaptic damage in 6-OHDA PD rats. Ephx2, Fam111a, and Gng2 may play critical roles in the pathogenesis of PD.


Subject(s)
Brain Chemistry/genetics , Gene Expression Profiling , Oxidopamine , Parkinson Disease, Secondary/genetics , Transcriptome , Animals , Computational Biology , Dopaminergic Neurons , Endocannabinoids/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Gene Expression Regulation , Parkinson Disease, Secondary/chemically induced , Polymerase Chain Reaction , RNA-Seq , Rats , Rats, Sprague-Dawley
4.
Brain Res Bull ; 175: 158-167, 2021 10.
Article in English | MEDLINE | ID: mdl-34339779

ABSTRACT

Fabry disease (FD) is an X-linked inherited disorder characterized by glycosphingolipid accumulation due to deficiency of α-galactosidase A (α-Gal A) enzyme. Chronic pain and mood disorders frequently coexist in FD clinical setting, however underlying pathophysiologic mechanisms are still unclear. Here we investigated the mechanical and thermal sensitivity in α-Gal A (-/0) hemizygous male and the α-Gal A (-/-) homozygous female mice. We also characterized the gene expression of dynorphinergic, nociceptinergic and CRFergic systems, known to be involved in pain control and mood disorders, in the prefrontal cortex, amygdala and thalamus of α-Gal A (-/0) hemizygous male and the α-Gal A (-/-) homozygous female mice. Moreover, KOP receptor protein levels were evaluated in the same areas. Fabry knock-out male, but not female, mice displayed a decreased pain threshold in both mechanical and thermal tests compared to their wild type littermates. In the amygdala and prefrontal cortex, we observed a decrease of pDYN mRNA levels in males, whereas an increase was assessed in females, thus suggesting sex-related dysregulation of stress coping and pain mechanisms. Elevated mRNA levels for pDYN/KOP and CRF/CRFR1 systems were observed in male and female thalamus, a critical crossroad for both painful signals and cognitive/emotional processes. KOP receptor protein level changes assessed in the investigated areas, appeared mostly in agreement with KOP gene expression alterations. Our data suggest that α-Gal A enzyme deficiency in male and female mice is associated with distinct neuropeptide gene and protein expression dysregulations of investigated systems, possibly related to the neuroplasticity underlying the neurological features of FD.


Subject(s)
Behavior, Animal , Fabry Disease/psychology , Neuropeptides/metabolism , Nociception , Animals , Brain Chemistry/genetics , Corticotropin-Releasing Hormone , Dynorphins/genetics , Female , Gene Expression , Male , Mice , Mice, Knockout , Nociceptors , Pain Threshold , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Opioid, kappa/genetics , Sex Characteristics
5.
Brain Res Bull ; 175: 186-195, 2021 10.
Article in English | MEDLINE | ID: mdl-34333052

ABSTRACT

Puberty includes a highly stress-sensitive period with significant sex differences in the neurophysiological and behavioural outcomes of a peripheral immune challenge. Sex differences in the pubertal neuroimmune network's responses to systemic LPS may explain some of these enduring sex-specific outcomes of a pubertal immune challenge. However, the functional implications of these sex-specific neuroimmune responses on the local microenvironment are unclear. Western blots were used to examine treatment- and sex-related changes in expression of regulatory proteins in inflammation (NFκB), cell death (AIF), oxidative stress (SOD-1), and synaptic plasticity (PSD-95) following symptomatic recovery (i.e., one week post-treatment) from pubertal immune challenge. Across the four examined brain regions (i.e., hippocampus, PFC, hypothalamus, and cerebellum), only PSD-95 levels were altered one week post-treatment by the pubertal LPS treatment. Unlike their female counterparts, seven-week-old males showed increased PSD-95 expression in the hippocampus (p < .05). AIF, SOD-1, and NFκB levels in both sexes were unaffected by treatment (all p > .05), which suggests appropriate resolution of NFκB-mediated immune responses to pubertal LPS without stimulating AIF-mediated apoptosis and oxidative stress. We also report a significant male-biased sex difference in PSD-95 levels in the PFC and in cerebellar expression of SOD-1 during puberty (all p < .05). These findings highlight the sex-specific vulnerability of the pubertal hippocampus to systemic LPS and suggest that a pubertal immune challenge may expedite neurodevelopment in the hippocampus in a sex-specific manner.


Subject(s)
Disks Large Homolog 4 Protein/biosynthesis , Lipopolysaccharides/pharmacology , Sexual Maturation , Animals , Apoptosis/drug effects , Apoptosis/genetics , Body Weight/drug effects , Brain Chemistry/drug effects , Brain Chemistry/genetics , Disks Large Homolog 4 Protein/genetics , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , NF-kappa B/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Sex Characteristics , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics
6.
J Neurochem ; 159(2): 273-291, 2021 10.
Article in English | MEDLINE | ID: mdl-34218440

ABSTRACT

Cerebral amyloid angiopathy (CAA), a prevalent cerebral small vessel disease in the elderly and a common comorbidity of Alzheimer's disease, is characterized by cerebral vascular amyloid accumulation, cerebral infarction, microbleeds, and intracerebral hemorrhages and is a prominent contributor to vascular cognitive impairment and dementia. Here, we investigate proteome changes associated with specific pathological features in several brain regions of rTg-DI rats, a preclinical model of CAA. Whereas varying degrees of microvascular amyloid and associated neuroinflammation are found in several brain regions, the presence of microbleeds and occluded small vessels is largely restricted to the thalamic region of rTg-DI rats, indicating different levels of CAA and associated pathologies occur in distinct brain regions in this model. Here, using SWATHLC-MS/MS, we report specific proteomic analysis of isolated brain regions and employ pathway analysis to correlate regionally specific proteomic changes with uniquely implicated molecular pathways. Pathway analysis suggested common activation of tumor necrosis factor α (TNFα), abnormal nervous system morphology, and neutrophil degranulation in all three regions. Activation of transforming growth factor-ß1 (TGF-ß1) was common to the hippocampus and thalamus, which share high CAA loads, while the thalamus, which uniquely exhibits thrombotic events, additionally displayed activation of thrombin and aggregation of blood cells. Thus, we present significant and new insight into the cerebral proteome changes found in distinct brain regions with differential CAA-related pathologies of rTg-DI rats and provide new information on potential pathogenic mechanisms associated with these regional disease processes.


Subject(s)
Brain Chemistry/genetics , Cerebral Amyloid Angiopathy/genetics , Proteome/genetics , Animals , Capillaries/pathology , Cell Degranulation , Computational Biology , Disease Models, Animal , Female , Humans , Male , Mass Spectrometry , Neutrophils/pathology , Pathology, Molecular , Proteomics , Rats , Rats, Transgenic , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Brain Res Bull ; 175: 99-106, 2021 10.
Article in English | MEDLINE | ID: mdl-34303767

ABSTRACT

BACKGROUND: Cognitive dysfunction is increasingly recognized as an important complication of diabetes mellitus (DM). Accumulating evidence indicates that the abnormality of cerebrovascular structure and function plays an essential role in diabetic cognitive impairment (DCI), however, changes in cerebrovascular factors have been blurred during the development of diabetes. OBJECTIVE: To evaluate the changes in the structure and function of cerebrovascular in DCI mice and to investigate the changes of cerebral angiogenesis and stability factors during the development of DM. METHODS: Diabetes was induced by feeding with high-fat diet combined with intraperitoneal injection of streptozotocin (STZ,120 mg/kg). Cognitive function was evaluated at different stages of DM, cerebral neovascularization, blood-brain barrier (BBB) permeability and hippocampal neurons were measured of DCI mice, and the expression of vascular endothelial growth factor (VEGF) and platelet-derived growth factor receptor ß (PDGFRß) in hippocampus was detected during the development of DM. RESULTS: With the progress of diabetes, the learning and memory ability of mice gradually decreased, and DCI mice showed neuronal degeneration, increased BBB permeability and pathological cerebral neovascularization. Moreover, the expression of VEGF in the hippocampus increased first and then decreased at DM+8week, PDGFRß decreased continuously with the development of diabetes. CONCLUSIONS: Our results demonstrate that DCI may be attributed to the dynamic expression of VEGF/PDGFRß in diabetic hippocampus, and pathological cerebral neovascularization, increased BBB permeability and neuronal degeneration are the key links.


Subject(s)
Brain Chemistry/genetics , Cognition Disorders/genetics , Cognition Disorders/psychology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/psychology , Receptor, Platelet-Derived Growth Factor beta/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Blood-Brain Barrier , Diet, High-Fat , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Psychomotor Performance
8.
J Neurochem ; 158(2): 391-412, 2021 07.
Article in English | MEDLINE | ID: mdl-33704768

ABSTRACT

Behavioral flexibility is an important cornerstone for the ecological success of animals. Social Cataglyphis nodus ants with their age-related polyethism characterized by age-related behavioral phenotypes represent a prime example for behavioral flexibility. We propose neuropeptides as powerful candidates for the flexible modulation of age-related behavioral transitions in individual ants. As the neuropeptidome of C. nodus was unknown, we collected a comprehensive peptidomic data set obtained by transcriptome analysis of the ants' central nervous system combined with brain extract analysis by Q-Exactive Orbitrap mass spectrometry (MS) and direct tissue profiling of different regions of the brain by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS. In total, we identified 71 peptides with likely bioactive function, encoded on 49 neuropeptide-, neuropeptide-like, and protein hormone prepropeptide genes, including a novel neuropeptide-like gene (fliktin). We next characterized the spatial distribution of a subset of peptides encoded on 16 precursor proteins with high resolution by MALDI MS imaging (MALDI MSI) on 14 µm brain sections. The accuracy of our MSI data were confirmed by matching the immunostaining patterns for tachykinins with MSI ion images from consecutive brain sections. Our data provide a solid framework for future research into spatially resolved qualitative and quantitative peptidomic changes associated with stage-specific behavioral transitions and the functional role of neuropeptides in Cataglyphis ants.


Subject(s)
Ants/physiology , Brain Chemistry/genetics , Brain/diagnostic imaging , Gene Expression Profiling , Neuropeptides/genetics , Proteomics , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Immunohistochemistry , Mass Spectrometry , Neuropeptides/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcriptome
9.
J Neuroendocrinol ; 33(3): e12963, 2021 03.
Article in English | MEDLINE | ID: mdl-33733541

ABSTRACT

Oxytocin (OT) is a neuropeptide hormone. Single and repetitive administration of OT increases social interaction and maternal behaviour in humans and mammals. Recently, it was found that the receptor for advanced glycation end-products (RAGE) is an OT-binding protein and plays a critical role in the uptake of OT to the brain after peripheral OT administration. Here, we address some unanswered questions on RAGE-dependent OT transport. First, we found that, after intranasal OT administration, the OT concentration increased in the extracellular space of the medial prefrontal cortex (mPFC) of wild-type male mice, as measured by push-pull microperfusion. No increase of OT in the mPFC was observed in RAGE knockout male mice. Second, in a reconstituted in vitro blood-brain barrier system, inclusion of the soluble form of RAGE (endogenous secretory RAGE [esRAGE]), an alternative splicing variant, in the luminal (blood) side had no effect on the transport of OT to the abluminal (brain) chamber. Third, OT concentrations in the cerebrospinal fluid after i.p. OT injection were slightly higher in male mice overexpressing esRAGE (esRAGE transgenic) compared to those in wild-type male mice, although this did not reach statistical significance. Although more extensive confirmation is necessary because of the small number of experiments in the present study, the reported data support the hypothesis that RAGE may be involved in the transport of OT to the mPFC from the circulation. These results suggest that the soluble form of RAGE in the plasma does not function as a decoy in vitro.


Subject(s)
Brain Chemistry/genetics , Oxytocin/metabolism , Receptor for Advanced Glycation End Products/genetics , Alternative Splicing , Animals , Antigens, Neoplasm/genetics , Biological Transport/genetics , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Extracellular Space/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinases/genetics , Oxytocin/cerebrospinal fluid
10.
J Neurochem ; 158(2): 197-216, 2021 07.
Article in English | MEDLINE | ID: mdl-33576044

ABSTRACT

The forebrain includes the cerebral cortex, the thalamus, and the striatum and globus pallidus (GP) in the subpallium. The formation of these structures and their interconnections by specific axonal tracts take place in a precise and orchestrated time and spatial-dependent manner during development. However, the knowledge of the molecular and cellular mechanisms that are involved is rather limited. Moreover, while many extracellular cues and specific receptors have been shown to play a role in different aspects of nervous system development, including neuron migration and axon guidance, examples of intracellular signaling effectors involved in these processes are sparse. In the present work, we have shown that the atypical RhoGTPase, Rnd3, is expressed very early during brain development and keeps a dynamic expression in several brain regions including the cortex, the thalamus, and the subpallium. By using a gene-trap allele (Rnd3gt ) and immunological techniques, we have shown that Rnd3gt/gt embryos display severe defects in striatal and thalamocortical axonal projections (SAs and TCAs, respectively) and defects in GP formation already at early stages. Surprisingly, the corridor, an important intermediate target for TCAs is still present in these mutants. Mechanistically, a conditional genetic deletion approach revealed that Rnd3 is primarily required for the normal development of Medial Ganglionic Eminence-derived structures, such as the GP, and therefore acts non-cell autonomously in SAs and TCAs. In conclusion, we have demonstrated the important role of Rnd3 as an early regulator of subpallium development in vivo and revealed new insights about SAs and TCAs development.


Subject(s)
Globus Pallidus/abnormalities , Internal Capsule/abnormalities , rho GTP-Binding Proteins/genetics , Animals , Axons/pathology , Brain/growth & development , Brain Chemistry/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Median Eminence/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neostriatum/abnormalities , Neural Pathways/abnormalities
11.
Ann Neurol ; 89(4): 813-822, 2021 04.
Article in English | MEDLINE | ID: mdl-33527515

ABSTRACT

OBJECTIVE: Perinatal intracranial hemorrhage (pICH) is a rare event that occurs during the fetal/neonatal period with potentially devastating neurological outcome. However, the etiology of pICH is frequently hard to depict. We investigated the role of rare genetic variations in unexplained cases of pICH. METHODS: We performed whole-exome sequencing (WES) in fetuses and term neonates with otherwise unexplained pICH and their parents. Variant causality was determined according to the American College of Medical Genetics and Genomics (ACMG) criteria, consistency between suggested genes and phenotypes, and mode of inheritance. RESULTS: Twenty-six probands (25 families) were included in the study (9 with a prenatal diagnosis and 17 with a postnatal diagnosis). Intraventricular hemorrhage (IVH) was the most common type of hemorrhage (n = 16, 62%), followed by subpial (n = 4, 15%), subdural (n = 4, 15%), and parenchymal (n = 2, 8%) hemorrhage. Causative/likely causative variants were found in 4 subjects from 3 of the 25 families (12%) involving genes related to the brain microenvironment (COL4A1, COL4A2, and TREX-1). Additionally, potentially causative variants were detected in genes related to coagulation (GP1BA, F11, Von Willebrand factor [VWF], FGA, and F7; n = 4, 16%). A potential candidate gene for phenotypic expansion related to microtubular function (DNAH5) was identified in 1 case (4%). Fifty-five percent of the variants were inherited from an asymptomatic parent. Overall, these findings showed a monogenic cause for pICH in 12% to 32% of the families. INTERPRETATION: Our findings reveal a clinically significant diagnostic yield of WES in apparently idiopathic pICH and support the use of WES in the evaluation of these cases. ANN NEUROL 2021;89:813-822.


Subject(s)
Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/genetics , Adult , Brain Chemistry/genetics , Cerebral Ventricles , DNA/genetics , Exome , Female , Fetus , Genetic Variation , Genotype , Humans , Infant, Newborn , Intracranial Hemorrhages/diagnostic imaging , Magnetic Resonance Imaging , Male , Phenotype , Pregnancy , Prenatal Diagnosis , Exome Sequencing
12.
Nat Neurosci ; 24(2): 197-203, 2021 02.
Article in English | MEDLINE | ID: mdl-33432194

ABSTRACT

Although germline de novo copy number variants (CNVs) are known causes of autism spectrum disorder (ASD), the contribution of mosaic (early-developmental) copy number variants (mCNVs) has not been explored. In this study, we assessed the contribution of mCNVs to ASD by ascertaining mCNVs in genotype array intensity data from 12,077 probands with ASD and 5,500 unaffected siblings. We detected 46 mCNVs in probands and 19 mCNVs in siblings, affecting 2.8-73.8% of cells. Probands carried a significant burden of large (>4-Mb) mCNVs, which were detected in 25 probands but only one sibling (odds ratio = 11.4, 95% confidence interval = 1.5-84.2, P = 7.4 × 10-4). Event size positively correlated with severity of ASD symptoms (P = 0.016). Surprisingly, we did not observe mosaic analogues of the short de novo CNVs recurrently observed in ASD (eg, 16p11.2). We further experimentally validated two mCNVs in postmortem brain tissue from 59 additional probands. These results indicate that mCNVs contribute a previously unexplained component of ASD risk.


Subject(s)
Autism Spectrum Disorder/genetics , DNA Copy Number Variations , Mosaicism , Adult , Autism Spectrum Disorder/epidemiology , Autopsy , Brain Chemistry/genetics , Child , Child Development Disorders, Pervasive/genetics , Cohort Studies , Genetic Predisposition to Disease , Genotype , Germ-Line Mutation , Humans , Risk Assessment , Tissue Banks
13.
Psychopharmacology (Berl) ; 238(4): 1213-1222, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33496816

ABSTRACT

RATIONALE: B vitamins play essential roles in brain development and functionality; however, the effects of their deficiency during early life on mental health are not thoroughly understood. OBJECTIVES: The objective of this study is to investigate the effects of a maternal deficiency of vitamin B6, B9 (folate), and B12 on behavioral changes in adult offspring. METHODS: Female C57BL/6 J mice were put on a diet lacking vitamin B6, B9, B12, or the above three vitamins from pregnancy to weaning. The growth and developmental characteristics of both the pregnant mothers and offspring were collected. In the adult offspring, the serum levels of neuroactive substances were measured using an enzyme-linked immunosorbent assay. The level of BDNF and dimethylated lysine 9 on histone H3 (H3K9me2) was detected by immunohistochemical staining. In addition, their depressive-like behaviors, anxiety-like behaviors, and sociability were recorded using sucrose preference, a forced swim, social interaction, tail suspension, and open field tests. RESULTS: The maternal deficiency of the three B vitamins delayed offspring development. Compared to the controls, all of the groups showed decreased serum levels of 5-HT and neuropeptide Y. In the groups with deficiency of B9 or the three B vitamins, there were significant changes in sociability and social novelty preference. In groups with deficiencies in B9, B12, or all three B vitamins, the expression levels of BDNF and H3K9me2 in the hippocampus were significantly decreased. CONCLUSIONS: Maternal deficiencies of the major B vitamins caused changes in social behaviors in adult mice accompanied with epigenetic alterations in the brain and changes in the serum levels of neuroactive substances.


Subject(s)
Behavior, Animal , Epigenesis, Genetic/genetics , Vitamin B Deficiency/genetics , Vitamin B Deficiency/psychology , Animals , Brain Chemistry/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Female , Hindlimb Suspension , Histones/metabolism , Mice , Mice, Inbred C57BL , Motor Activity , Neuropeptide Y/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Social Interaction
14.
J Neurochem ; 158(4): 943-959, 2021 08.
Article in English | MEDLINE | ID: mdl-32813270

ABSTRACT

Signaling pathways mediated by corticotropin-releasing factor and its receptor 1 (CRF1) play a central role in stress responses. Dysfunction of the CRF system has been associated with neuropsychiatric disorders. However, dynamic changes in the CRF system during brain development and aging are not well investigated. In this study, we characterized CRF1, CRF, and corticotropin-releasing factor binding protein (CRFBP) expression in different brain regions in both male and female C57BL/6J mice from 1 to 18 months of age under basal conditions as well as after an acute 2-hr-restraint stress. We found that CRF and CRF1 levels tended to increase in the hippocampus and hypothalamus, and to decrease in the prefrontal cortex with aging, especially at 18 months of age, whereas CRFBP expression followed an opposite direction in these brain areas. We also observed area-specific sex differences in the expression of these three proteins. For example, CRF expression was lower in females than in males in all the brain regions examined except the prefrontal cortex. After acute stress, CRF and CRF1 were up-regulated at 1, 6, and 12 months of age, and down-regulated at 18 months of age. Females showed more robust changes compared to males of the same age. CRFBP expression either decreased or remained unchanged in most of the brain areas following acute stress. Our findings suggest that brain CRF1, CRF, and CRFBP expression changes dynamically across the lifespan and under stress condition in a sex- and regional-specific manner. Sex differences in the CRF system in response to stress may contribute to the etiology of stress-related neuropsychiatric disorders.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Corticotropin-Releasing Hormone/biosynthesis , Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Stress, Psychological/genetics , Stress, Psychological/metabolism , Animals , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Restraint, Physical , Sex Characteristics , Stress, Psychological/psychology
15.
Nat Neurosci ; 24(1): 24-33, 2021 01.
Article in English | MEDLINE | ID: mdl-33349712

ABSTRACT

Despite extensive study of the neurobiological correlates of post-traumatic stress disorder (PTSD), little is known about its molecular determinants. Here, differential gene expression and network analyses of four prefrontal cortex subregions from postmortem tissue of people with PTSD demonstrate extensive remodeling of the transcriptomic landscape. A highly connected downregulated set of interneuron transcripts is present in the most significant gene network associated with PTSD. Integration of this dataset with genotype data from the largest PTSD genome-wide association study identified the interneuron synaptic gene ELFN1 as conferring significant genetic liability for PTSD. We also identified marked transcriptomic sexual dimorphism that could contribute to higher rates of PTSD in women. Comparison with a matched major depressive disorder cohort revealed significant divergence between the molecular profiles of individuals with PTSD and major depressive disorder despite their high comorbidity. Our analysis provides convergent systems-level evidence of genomic networks within the prefrontal cortex that contribute to the pathophysiology of PTSD in humans.


Subject(s)
Brain Chemistry/genetics , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/physiopathology , Transcriptome , Adult , Autopsy , Cohort Studies , Depressive Disorder, Major/genetics , Female , Gene Expression Regulation/genetics , Gene Regulatory Networks , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Interneurons/metabolism , Male , Middle Aged , Nerve Tissue Proteins/genetics , Sex Characteristics , Young Adult
16.
Brain ; 143(12): 3827-3841, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33155043

ABSTRACT

The aetiology of late-onset neurodegenerative diseases is largely unknown. Here we investigated whether de novo somatic variants for semantic dementia can be detected, thereby arguing for a more general role of somatic variants in neurodegenerative disease. Semantic dementia is characterized by a non-familial occurrence, early onset (<65 years), focal temporal atrophy and TDP-43 pathology. To test whether somatic variants in neural progenitor cells during brain development might lead to semantic dementia, we compared deep exome sequencing data of DNA derived from brain and blood of 16 semantic dementia cases. Somatic variants observed in brain tissue and absent in blood were validated using amplicon sequencing and digital PCR. We identified two variants in exon one of the TARDBP gene (L41F and R42H) at low level (1-3%) in cortical regions and in dentate gyrus in two semantic dementia brains, respectively. The pathogenicity of both variants is supported by demonstrating impaired splicing regulation of TDP-43 and by altered subcellular localization of the mutant TDP-43 protein. These findings indicate that somatic variants may cause semantic dementia as a non-hereditary neurodegenerative disease, which might be exemplary for other late-onset neurodegenerative disorders.


Subject(s)
DNA-Binding Proteins/genetics , Frontotemporal Dementia/etiology , Frontotemporal Dementia/genetics , TDP-43 Proteinopathies/complications , TDP-43 Proteinopathies/genetics , Alternative Splicing , Brain Chemistry/genetics , DNA/genetics , Exome , Exons/genetics , Female , Frontotemporal Dementia/psychology , Genetic Variation/genetics , Germ-Line Mutation , Humans , Male , Middle Aged , Mutation/genetics , Semantics , TDP-43 Proteinopathies/psychology , Exome Sequencing
17.
Neurochem Int ; 141: 104888, 2020 12.
Article in English | MEDLINE | ID: mdl-33199267

ABSTRACT

System xc- is a heterodimeric amino acid antiporter that, in the central nervous system, is best known for linking the import of L-cystine (CySS) with the export of L-glutamate for the production and maintenance of cellular glutathione (GSH) and extracellular glutamate levels, respectively. Yet, mice that are null for system xc- are healthy, fertile, and, morphologically, their brains are grossly normal. This suggests other glutamate and/or cyst(e)ine transport mechanisms may be upregulated in compensation. To test this, we measured the plasma membrane expression of Excitatory Amino Acid Transporters (EAATs) 1-3, the Alanine-Serine-Cysteine-Transporter (ASCT) 1, the sodium-coupled neutral amino acid transporter (SNAT) 3 and the L Amino Acid Transporter (LAT) 2 in striatum, hippocampus and cortex of male and female mice using Western Blot analysis. Present results demonstrate brain region and transporter-specific changes occurs in female system xc- null mice with increased expression of EAAT1 and ASCT1 occurring in the striatum and cortex, respectively, and decreased SNAT 3 expression in cortex. In male system xc- null brain, only SNAT3 was altered significantly - increasing in the cortex, but decreasing in the striatum. Total levels of GSH and CyS were similar to that found in age and sex-matched littermate control mice, however, reductions in the ratio of reduced to oxidized GSH (GSH/GSSG) - a hallmark of oxidative stress - were found in all three brain regions in female system xc- null mice, whereas this occurred exclusively in the striatum of males. Protein levels of Superoxide dismutase (SOD) 1 were reduced, whereas SOD2 was enhanced in the hippocampus of male xc- null mice only. Finally, striatal vulnerability to 3-nitropropionic acid (3-NP)-mediated oxidative stress in either sex showed no genotype difference, although 3-NP was more toxic to female mice of either genotype, as evidenced by an increase in moribundity as compared to males.


Subject(s)
Amino Acid Transport System y+/genetics , Amino Acid Transport Systems/genetics , Brain Chemistry/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Animals , Cerebral Cortex/metabolism , Cystine/metabolism , Female , Glutamic Acid/metabolism , Glutathione/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Knockout , Neostriatum/metabolism , Oxidative Stress/genetics , Sex Characteristics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism
18.
J Neuroendocrinol ; 32(11): e12901, 2020 11.
Article in English | MEDLINE | ID: mdl-33000513

ABSTRACT

Pregnancy represents a period of remarkable adaptive physiology throughout the body, with many of these important adaptations mediated by changes in gene transcription in the brain. A marked activation of the transcription factor signal transducer and activator of transcription 5 (STAT5) has been described in the brain during pregnancy and likely drives some of these changes. We aimed to investigate the physiological mechanism causing this increase in phosphorylated STAT5 (pSTAT5) during pregnancy. In various tissues, STAT5 is known to be activated by a number of different cytokines, including erythropoietin, growth hormone and prolactin. Because the lactogenic hormones that act through the prolactin receptor (PRLR), prolactin and its closely-related placental analogue placental lactogen, are significantly increased during pregnancy, we hypothesised that this receptor was primarily responsible for the pregnancy-induced increase in pSTAT5 in the brain. By examining temporal changes in plasma prolactin levels and the pattern of pSTAT5 immunoreactivity in the hypothalamus during early pregnancy, we found that the level of pSTAT5 was sensitive to circulating levels of endogenous prolactin. Using a transgenic model to conditionally delete PRLRs from forebrain neurones (Prlrlox/lox /CamK-Cre), we assessed the relative contribution of the PRLR to the up-regulation of pSTAT5 in the brain of pregnant mice. In the absence of PRLRs on most forebrain neurones, a significant reduction in pSTAT5 was observed throughout the hypothalamus and amygdala in late pregnancy, confirming that PRLR is key in mediating this response. The exception to this was the hypothalamic paraventricular nucleus, where only 17% of pSTAT5 immunoreactivity during pregnancy was in PRLR-expressing cells. Taken together, these data indicate that, although there are region-specific mechanisms involved, lactogenic activity through the PRLR is the primary signal activating STAT5 in the brain during pregnancy.


Subject(s)
Brain Chemistry/physiology , Receptors, Prolactin/physiology , STAT5 Transcription Factor/metabolism , Amygdala/metabolism , Animals , Brain Chemistry/genetics , Cytokines/metabolism , Female , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Phosphorylation , Placenta/metabolism , Placental Lactogen/metabolism , Pregnancy , Prolactin/metabolism , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects
19.
J Neurosci ; 40(45): 8669-8682, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33046550

ABSTRACT

Serotonergic neurons in the dorsal raphe (DR) nucleus are associated with several psychiatric disorders including depression and anxiety disorders, which often have a neurodevelopmental component. During embryonic development, GATA transcription factors GATA2 and GATA3 operate as serotonergic neuron fate selectors and regulate the differentiation of serotonergic neuron subtypes of DR. Here, we analyzed the requirement of GATA cofactor ZFPM1 in the development of serotonergic neurons using Zfpm1 conditional mouse mutants. Our results demonstrated that, unlike the GATA factors, ZFPM1 is not essential for the early differentiation of serotonergic precursors in the embryonic rhombomere 1. In contrast, in perinatal and adult male and female Zfpm1 mutants, a lateral subpopulation of DR neurons (ventrolateral part of the DR) was lost, whereas the number of serotonergic neurons in a medial subpopulation (dorsal region of the medial DR) had increased. Additionally, adult male and female Zfpm1 mutants had reduced serotonin concentration in rostral brain areas and displayed increased anxiety-like behavior. Interestingly, female Zfpm1 mutant mice showed elevated contextual fear memory that was abolished with chronic fluoxetine treatment. Altogether, these results demonstrate the importance of ZFPM1 for the development of DR serotonergic neuron subtypes involved in mood regulation. It also suggests that the neuronal fate selector function of GATAs is modulated by their cofactors to refine the differentiation of neuronal subtypes.SIGNIFICANCE STATEMENT Predisposition to anxiety disorders has both a neurodevelopmental and a genetic basis. One of the brainstem nuclei involved in the regulation of anxiety is the dorsal raphe, which contains different subtypes of serotonergic neurons. We show that inactivation of a transcriptional cofactor ZFPM1 in mice results in a developmental failure of laterally located dorsal raphe serotonergic neurons and changes in serotonergic innervation of rostral brain regions. This leads to elevated anxiety-like behavior and contextual fear memory, alleviated by chronic fluoxetine treatment. Our work contributes to understanding the neurodevelopmental mechanisms that may be disturbed in the anxiety disorder.


Subject(s)
Anxiety/genetics , Anxiety/psychology , Dorsal Raphe Nucleus/growth & development , GATA Transcription Factors/genetics , Serotonergic Neurons , Transcription Factors/genetics , Animals , Behavior, Animal , Brain Chemistry/genetics , Dorsal Raphe Nucleus/cytology , Fear/psychology , Female , Fluoxetine/pharmacology , Male , Memory , Mice , Mice, Knockout , Mutation/genetics , Pregnancy , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
20.
Elife ; 92020 09 21.
Article in English | MEDLINE | ID: mdl-32955435

ABSTRACT

The underlying cell types mediating predisposition to obesity remain largely obscure. Here, we integrated recently published single-cell RNA-sequencing (scRNA-seq) data from 727 peripheral and nervous system cell types spanning 17 mouse organs with body mass index (BMI) genome-wide association study (GWAS) data from >457,000 individuals. Developing a novel strategy for integrating scRNA-seq data with GWAS data, we identified 26, exclusively neuronal, cell types from the hypothalamus, subthalamus, midbrain, hippocampus, thalamus, cortex, pons, medulla, pallidum that were significantly enriched for BMI heritability (p<1.6×10-4). Using genes harboring coding mutations associated with obesity, we replicated midbrain cell types from the anterior pretectal nucleus and periaqueductal gray (p<1.2×10-4). Together, our results suggest that brain nuclei regulating integration of sensory stimuli, learning and memory are likely to play a key role in obesity and provide testable hypotheses for mechanistic follow-up studies.


Subject(s)
Brain Chemistry/genetics , Brain , Computational Biology/methods , Obesity , Animals , Body Mass Index , Brain/cytology , Brain/metabolism , Genetic Techniques , Genome-Wide Association Study , Mice , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Organ Specificity/genetics , RNA/chemistry , RNA/metabolism , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...