Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.618
1.
Neuropsychopharmacology ; 49(6): 1024-1032, 2024 May.
Article En | MEDLINE | ID: mdl-38431758

The 22q11.2 locus contains genes critical for brain development. Reciprocal Copy Number Variations (CNVs) at this locus impact risk for neurodevelopmental and psychiatric disorders. Both 22q11.2 deletions (22qDel) and duplications (22qDup) are associated with autism, but 22qDel uniquely elevates schizophrenia risk. Understanding brain phenotypes associated with these highly penetrant CNVs can provide insights into genetic pathways underlying neuropsychiatric disorders. Human neuroimaging and animal models indicate subcortical brain alterations in 22qDel, yet little is known about developmental differences across specific nuclei between reciprocal 22q11.2 CNV carriers and typically developing (TD) controls. We conducted a longitudinal MRI study in a total of 385 scans from 22qDel (n = 96, scans = 191, 53.1% female), 22qDup (n = 37, scans = 64, 45.9% female), and TD controls (n = 80, scans = 130, 51.2% female), across a wide age range (5.5-49.5 years). Volumes of the thalamus, hippocampus, amygdala, and anatomical subregions were estimated using FreeSurfer, and the linear effects of 22q11.2 gene dosage and non-linear effects of age were characterized with generalized additive mixed models (GAMMs). Positive gene dosage effects (volume increasing with copy number) were observed for total intracranial and whole hippocampus volumes, but not whole thalamus or amygdala volumes. Several amygdala subregions exhibited similar positive effects, with bi-directional effects found across thalamic nuclei. Distinct age-related trajectories were observed across the three groups. Notably, both 22qDel and 22qDup carriers exhibited flattened development of hippocampal CA2/3 subfields relative to TD controls. This study provides novel insights into the impact of 22q11.2 CNVs on subcortical brain structures and their developmental trajectories.


DNA Copy Number Variations , DiGeorge Syndrome , Gene Dosage , Magnetic Resonance Imaging , Humans , Female , Male , DNA Copy Number Variations/genetics , Adult , Adolescent , Child , Young Adult , Middle Aged , Child, Preschool , DiGeorge Syndrome/genetics , DiGeorge Syndrome/pathology , DiGeorge Syndrome/diagnostic imaging , Longitudinal Studies , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hippocampus/growth & development , Brain/diagnostic imaging , Brain/pathology , Brain/growth & development , Amygdala/diagnostic imaging , Amygdala/pathology , Thalamus/diagnostic imaging , Thalamus/growth & development , Thalamus/pathology , Organ Size
2.
Brain Res ; 1827: 148758, 2024 03 15.
Article En | MEDLINE | ID: mdl-38199308

BACKGROUND: Subarachnoid hemorrhage (SAH) is a life-threatening neurological disease that usually has a poor prognosis. Neurogenesis is a potential therapeutic target for brain injury. Ketone metabolism also plays neuroprotective roles in many neurological disorders. OXCT1 (3-Oxoacid CoA-Transferase 1) is the rate-limiting enzyme of ketone body oxidation. In this study, we explored whether increasing ketone oxidation by upregulating OXCT1 in neurons could promote neurogenesis after SAH, and evaluated the potential mechanism involved in this process. METHODS: The ß-hydroxybutyrate content was measured using an enzymatic colorimetric assay. Adeno-associated virus targeting neurons was injected to overexpress OXCT1, and the expression and localization of proteins were evaluated by western blotting and immunofluorescence staining. Adult hippocampal neurogenesis was evaluated by dual staining with doublecortin and 5-Ethynyl-2'-Deoxyuridine. LY294002 was intracerebroventricularly administered to inhibit Akt activity. The Morris water maze and Y-maze tests were employed to assess cognitive function after SAH. RESULTS: The results showed that OXCT1 expression and hippocampal neurogenesis significantly decreased in the early stage of SAH. Overexpression of OXCT1 successfully increased hippocampal neurogenesis via activation of Akt/GSK-3ß/ß-catenin signaling and improved cognitive function, both of which were reversed by administration of LY294002. CONCLUSIONS: OXCT1 regulated hippocampal ketone body metabolism and increased neurogenesis through mechanisms mediated by the Akt/GSK-3ß/ß-catenin pathway, improving cognitive impairment after SAH.


Coenzyme A-Transferases , Cognitive Dysfunction , Hippocampus , Neurogenesis , Subarachnoid Hemorrhage , 3-Hydroxybutyric Acid , beta Catenin , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/growth & development , Proto-Oncogene Proteins c-akt , Animals , Mice
3.
Commun Biol ; 6(1): 440, 2023 04 21.
Article En | MEDLINE | ID: mdl-37085665

RAB35 is a multifunctional small GTPase that regulates endocytic recycling, cytoskeletal rearrangement, and cytokinesis. However, its physiological functions in mammalian development remain unclear. Here, we generated Rab35-knockout mice and found that RAB35 is essential for early embryogenesis. Interestingly, brain-specific Rab35-knockout mice displayed severe defects in hippocampal lamination owing to impaired distribution of pyramidal neurons, although defects in cerebral cortex formation were not evident. In addition, Rab35-knockout mice exhibited defects in spatial memory and anxiety-related behaviors. Quantitative proteomics indicated that the loss of RAB35 significantly affected the levels of other RAB proteins associated with endocytic trafficking, as well as some neural cell adhesion molecules, such as contactin-2. Collectively, our findings revealed that RAB35 is required for precise neuronal distribution in the developing hippocampus by regulating the expression of cell adhesion molecules, thereby influencing spatial memory.


Hippocampus , Neurons , rab GTP-Binding Proteins , Animals , Mice , Biological Transport , Hippocampus/growth & development , Hippocampus/metabolism , Mammals , Mice, Knockout , Neurons/metabolism , rab GTP-Binding Proteins/metabolism
4.
J Comp Neurol ; 530(15): 2711-2748, 2022 10.
Article En | MEDLINE | ID: mdl-35603771

Little is known about the development of the human entorhinal cortex (EC), a major hub in a widespread network for learning and memory, spatial navigation, high-order processing of object information, multimodal integration, attention and awareness, emotion, motivation, and perception of time. We analyzed a series of 20 fetal and two adult human brains using Nissl stain, acetylcholinesterase (AChE) histochemistry, and immunocytochemistry for myelin basic protein (MBP), neuronal nuclei antigen (NeuN), a pan-axonal neurofilament marker, and synaptophysin, as well as postmortem 3T MRI. In comparison with other parts of the cerebral cortex, the cytoarchitectural differentiation of the EC begins remarkably early, in the 10th week of gestation (w.g.). The differentiation occurs in a superficial magnocellular layer in the deep part of the marginal zone, accompanied by cortical plate (CP) condensation and multilayering of the deep part of CP. These processes last until the 13-14th w.g. At 14 w.g., the superficial lamina dissecans (LD) is visible, which divides the CP into the lamina principalis externa (LPE) and interna (LPI). Simultaneously, the rostral LPE separates into vertical cell-dense islands, whereas in the LPI, the deep LD emerges as a clear acellular layer. In the 16th w.g., the LPE remodels into vertical cell-dense and cell-sparse zones with a caudorostral gradient. At 20 w.g., NeuN immunoreactivity is most pronounced in the islands of layer II cells, whereas migration and differentiation inside-out gradients are seen simultaneously in both the upper (LPE) and the lower (LPI) pyramidal layers. At this stage, the EC adopts for the first time an adult-like cytoarchitectural organization, the superficial LD becomes discernible by 3T MRI, MBP-expressing oligodendrocytes first appear in the fimbria and the perforant path (PP) penetrates the subiculum to reach its molecular layer and travels along through the Cornu Ammonis fields to reach the suprapyramidal blade of the dentate gyrus, whereas the entorhinal-dentate branch perforates the hippocampal sulcus about 2-3 weeks later. The first AChE reactivity appears as longitudinal stripes at 23 w.g. in layers I and II of the rostrolateral EC and then also as AChE-positive in-growing fibers in islands of superficial layer III and layer II neurons. At 40 w.g., myelination of the PP starts as patchy MBP-immunoreactive oligodendrocytes and their processes. Our results refute the possibility of an inside-out pattern of the EC development and support the key role of layer II prospective stellate cells in the EC lamination. As the early cytoarchitectural differentiation of the EC is paralleled by the neurochemical development, these developmental milestones in EC structure and connectivity have implications for understanding its normal function, including its puzzling modular organization and potential contribution to consciousness content (awareness), as well as for its insufficiently explored deficits in developmental, psychiatric, and degenerative brain disorders.


Acetylcholinesterase , Entorhinal Cortex , Fetal Development , Acetylcholinesterase/metabolism , Adult , Entorhinal Cortex/growth & development , Female , Fetus , Hippocampus/growth & development , Humans , Neurons/metabolism , Pregnancy , Prospective Studies
5.
Proc Natl Acad Sci U S A ; 119(22): e2118240119, 2022 05 31.
Article En | MEDLINE | ID: mdl-35613055

Adult hippocampal neurogenesis is critical for learning and memory, and aberrant adult neurogenesis has been implicated in cognitive decline associated with aging and neurological diseases [J. T. Gonçalves, S. T. Schafer, F. H. Gage, Cell 167, 897­914 (2016)]. In previous studies, we observed that the delayed-rectifier voltage-gated potassium channel Kv1.1 controls the membrane potential of neural stem and progenitor cells and acts as a brake on neurogenesis during neonatal hippocampal development [S. M. Chou et al., eLife 10, e58779 (2021)]. To assess the role of Kv1.1 in adult hippocampal neurogenesis, we developed an inducible conditional knockout mouse to specifically remove Kv1.1 from adult neural stem cells via tamoxifen administration. We determined that Kv1.1 deletion in adult neural stem cells causes overproliferation and depletion of radial glia-like neural stem cells, prevents proper adult-born granule cell maturation and integration into the dentate gyrus, and moderately impairs hippocampus-dependent contextual fear learning and memory. Taken together, these findings support a critical role for this voltage-gated ion channel in adult neurogenesis.


Conditioning, Classical , Hippocampus , Kv1.1 Potassium Channel , Neural Stem Cells , Neurogenesis , Neurons , Animals , Fear , Hippocampus/cytology , Hippocampus/growth & development , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/physiology , Mice , Mice, Knockout , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology
6.
J Neurosci ; 42(24): 4812-4827, 2022 06 15.
Article En | MEDLINE | ID: mdl-35589394

Neonatal brain injury renders the developing brain vulnerable to oxidative stress, leading to cognitive deficit. However, oxidative stress-induced damage to hippocampal circuits and the mechanisms underlying long-term changes in memory and learning are poorly understood. We used high oxygen tension or hyperoxia (HO) in neonatal mice of both sexes to investigate the role of oxidative stress in hippocampal damage. Perinatal HO induces reactive oxygen species and cell death, together with reduced interneuron maturation, inhibitory postsynaptic currents, and dentate progenitor proliferation. Postinjury interneuron stimulation surprisingly improved inhibitory activity and memory tasks, indicating reversibility. With decreased hippocampal levels of Wnt signaling components and somatostatin, HO aberrantly activated glycogen synthase kinase 3 ß activity. Pharmacological inhibition or ablation of interneuron glycogen synthase kinase 3 ß during HO challenge restored progenitor cell proliferation, interneuron development, inhibitory/excitatory balance, as well as hippocampal-dependent behavior. Biochemical targeting of interneuron function may benefit learning deficits caused by oxidative damage.SIGNIFICANCE STATEMENT Premature infants are especially vulnerable to oxidative stress, as their antioxidant defenses are underdeveloped. Indeed, high oxygen tension is associated with poor neurologic outcomes. Because of its sustained postnatal development and role in learning and memory, the hippocampus is especially vulnerable to oxidative damage in premature infants. However, the role of oxidative stress in the developing hippocampus has yet to be explored. With ever-rising rates of neonatal brain injury and no universally viable approach to maximize functional recovery, a better understanding of the mechanisms underlying neonatal brain injury is needed. Addressing this need, this study uses perinatal hyperoxia to study cognitive deficits, pathophysiology, and molecular mechanisms of oxidative damage in the developing hippocampus.


Brain Injuries , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Hyperoxia , Oxidative Stress , Animals , Female , Hippocampus/growth & development , Humans , Hyperoxia/metabolism , Male , Mice , Oxygen/metabolism , Pregnancy
7.
Science ; 376(6590): eabn8861, 2022 04 15.
Article En | MEDLINE | ID: mdl-35420933

Terreros-Roncal et al. investigated the impacts of human neurodegeneration on immunostainings assumed to be associated with neurogenesis. However, the study provides no evidence that putative proliferating cells are linked to neurogenesis, that multipolar nestin+ astrocytes are progenitors, or that mature-looking doublecortin+ neurons are adult-born. Their histology-marker expression differs from what is observed in species where adult hippocampal neurogenesis is well documented.


Hippocampus , Neurodegenerative Diseases , Neurogenesis , Adult , Astrocytes , Hippocampus/cytology , Hippocampus/growth & development , Humans , Neurodegenerative Diseases/metabolism , Neurogenesis/physiology , Neurons/physiology
8.
Proc Natl Acad Sci U S A ; 119(13): e2023784119, 2022 03 29.
Article En | MEDLINE | ID: mdl-35333654

Neural stem cells, the source of newborn neurons in the adult hippocampus, are intimately involved in learning and memory, mood, and stress response. Despite considerable progress in understanding the biology of neural stem cells and neurogenesis, regulating the neural stem cell population precisely has remained elusive because we have lacked the specific targets to stimulate their proliferation and neurogenesis. The orphan nuclear receptor TLX/NR2E1 governs neural stem and progenitor cell self-renewal and proliferation, but the precise mechanism by which it accomplishes this is not well understood because its endogenous ligand is not known. Here, we identify oleic acid (18:1ω9 monounsaturated fatty acid) as such a ligand. We first show that oleic acid is critical for neural stem cell survival. Next, we demonstrate that it binds to TLX to convert it from a transcriptional repressor to a transcriptional activator of cell-cycle and neurogenesis genes, which in turn increases neural stem cell mitotic activity and drives hippocampal neurogenesis in mice. Interestingly, oleic acid-activated TLX strongly up-regulates cell cycle genes while only modestly up-regulating neurogenic genes. We propose a model in which sufficient quantities of this endogenous ligand must bind to TLX to trigger the switch to proliferation and drive the progeny toward neuronal lineage. Oleic acid thus serves as a metabolic regulator of TLX activity that can be used to selectively target neural stem cells, paving the way for future therapeutic manipulations to counteract pathogenic impairments of neurogenesis.


Hippocampus , Neurogenesis , Oleic Acid , Receptors, Cytoplasmic and Nuclear , Animals , Cell Proliferation , Hippocampus/growth & development , Hippocampus/metabolism , Ligands , Mice , Neurogenesis/physiology , Oleic Acid/metabolism , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/metabolism
9.
Toxicol Ind Health ; 38(1): 41-52, 2022 Jan.
Article En | MEDLINE | ID: mdl-35075925

Silicon dioxide nanoparticles (SiO2-NPs) are among the most widely used nanoparticles because of their chemical-physical properties. Since most brain maturation occurs in the neonatal period in humans and many mammals, it is important to understand how NPs may affect this process. This study tested the hypothesis that SiO2-NPs from treated dams could affect the hippocampus of neonatal rats during lactation. Twenty-four pregnant rats, after delivery, were divided into three groups of control, SiO2-NPs (25 mg/kg) and SiO2-NPs (100 mg/kg). The rats were treated from 2nd to 21st days post-delivery by gavage and the effects of these NPs were evaluated in the offspring's hippocampi to reveal the effects of maternal exposure to SiO2-NPs during lactation on the offspring's hippocampi. The offspring in the SiO2-NPs groups had higher malondialdehyde concentration and lower antioxidant activity in the hippocampi than the non-treated control group. The mean number of doublecortin positive (DCX+) cells and synaptophysin expression in the hippocampi of the SiO2-NPs groups were significantly lower than the control group, whereas the mean number of dark neurons was significantly higher. Also, animals in the SiO2-NPs groups had a weak cognitive performance in adulthood. In conclusion, maternal exposure to SiO2-NPs via breastfeeding could affect offspring's hippocampal neurogenesis and synaptogenesis, leading to impaired cognitive performance.


Hippocampus/drug effects , Hippocampus/growth & development , Maternal Exposure/adverse effects , Nanoparticles/toxicity , Neurogenesis/drug effects , Silicon Dioxide/toxicity , Animals , Female , Lactation , Pregnancy , Rats
10.
J Comp Neurol ; 530(1): 6-503, 2022 01.
Article En | MEDLINE | ID: mdl-34525221

Increasing interest in studies of prenatal human brain development, particularly using new single-cell genomics and anatomical technologies to create cell atlases, creates a strong need for accurate and detailed anatomical reference atlases. In this study, we present two cellular-resolution digital anatomical atlases for prenatal human brain at postconceptional weeks (PCW) 15 and 21. Both atlases were annotated on sequential Nissl-stained sections covering brain-wide structures on the basis of combined analysis of cytoarchitecture, acetylcholinesterase staining, and an extensive marker gene expression dataset. This high information content dataset allowed reliable and accurate demarcation of developing cortical and subcortical structures and their subdivisions. Furthermore, using the anatomical atlases as a guide, spatial expression of 37 and 5 genes from the brains, respectively, at PCW 15 and 21 was annotated, illustrating reliable marker genes for many developing brain structures. Finally, the present study uncovered several novel developmental features, such as the lack of an outer subventricular zone in the hippocampal formation and entorhinal cortex, and the apparent extension of both cortical (excitatory) and subcortical (inhibitory) progenitors into the prenatal olfactory bulb. These comprehensive atlases provide useful tools for visualization, segmentation, targeting, imaging, and interpretation of brain structures of prenatal human brain, and for guiding and interpreting the next generation of cell census and connectome studies.


Atlases as Topic , Brain/growth & development , Entorhinal Cortex/growth & development , Hippocampus/growth & development , Animals , Female , Humans , Pregnancy
11.
J Neurosci ; 42(4): 601-618, 2022 01 26.
Article En | MEDLINE | ID: mdl-34844990

Precise information flow from the hippocampus (HP) to prefrontal cortex (PFC) emerges during early development and accounts for cognitive processing throughout life. On flip side, this flow is selectively impaired in mental illness. In mouse models of psychiatric risk mediated by gene-environment interaction (GE), the prefrontal-hippocampal coupling is disrupted already shortly after birth. While this impairment relates to local miswiring in PFC and HP, it might be also because of abnormal connectivity between the two brain areas. Here, we test this hypothesis by combining in vivo electrophysiology and optogenetics with in-depth tracing of projections and monitor the morphology and function of hippocampal afferents in the PFC of control and GE mice of either sex throughout development. We show that projections from the hippocampal CA1 area preferentially target layer 5/6 pyramidal neurons and interneurons, and to a lesser extent layer 2/3 neurons of prelimbic cortex (PL), a subdivision of PFC. In neonatal GE mice, sparser axonal projections from CA1 pyramidal neurons with decreased release probability reach the PL. Their ability to entrain layer 5/6 oscillatory activity and firing is decreased. These structural and functional deficits of hippocampal-prelimbic connectivity persist, yet are less prominent in prejuvenile GE mice. Thus, besides local dysfunction of HP and PL, weaker connectivity between the two brain areas is present in GE mice throughout development.SIGNIFICANCE STATEMENT Poor cognitive performance in mental disorders comes along with prefrontal-hippocampal dysfunction. Recent data from mice that model the psychiatric risk mediated by gene-environment (GE) interaction identified the origin of deficits during early development, when the local circuits in both areas are compromised. Here, we show that sparser and less efficient connectivity as well as cellular dysfunction are the substrate of the weaker excitatory drive from hippocampus (HP) to prefrontal cortex (PFC) as well as of poorer oscillatory coupling between the two brain areas in these mice. While the structural and functional connectivity deficits persist during the entire development, their magnitude decreases with age. The results add experimental evidence for the developmental miswiring hypothesis of psychiatric disorders.


Gene-Environment Interaction , Hippocampus/growth & development , Mental Disorders/genetics , Mental Disorders/physiopathology , Nerve Net/growth & development , Prefrontal Cortex/growth & development , Animals , Animals, Newborn , Disease Models, Animal , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/chemistry , Male , Mental Disorders/psychology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/chemistry , Prefrontal Cortex/chemistry , Risk Factors
12.
Neurosci Lett ; 771: 136398, 2022 02 06.
Article En | MEDLINE | ID: mdl-34923042

Aging has been recognized as a major driving force of the Alzheimer's disease's (AD) progression, however, the relationship between brain aging and AD is still unclear. There is also a lack of studies investigating the influence of AD risk factors on brain aging in cognitively normal people. Here, the "Brain Age Gap Estimation" (BrainAGE) framework was applied to investigate the effects of AD risk factors on individual brain aging. Across a total of 165 cognitively normal elderly subjects, although no significant difference was observed in the BrainAGE scores among the three groups, AD risk dose (i.e., the number of AD risk factors) is tend to associated with an increased BrainAGE scores (high-risk > middle risk > low risk). Female exhibited more advanced brain aging (P = 0.004), and higher education years were associated with preserved brain aging (P < 0.001). APOE-ɛ4 (P = 0.846) and family history (FH) of dementia (P = 0.209) did not increase BrainAGE scores. When comparing 52 aMCI patients with 38 cognitively normal controls from ADNI dataset, aMCI patients showed significantly increased BrainAGE scores. BrainAGE scores were negatively correlated with CSF Aß42 levels in the aMCI group (r = -0.275, P = 0.048). With an accuracy of 68.9%, BrainAGE outperformed APOE-ɛ4 and hippocampus gray matter volume (GMV) in predicting aMCI. In conclusion, AD is independently associated with structural changes in the brain that reflect advanced aging. Potentially, BrainAGE combined with APOE-ɛ4 and hippocampus GMV could be used as a pre-screening tool in early-stage AD.


Aging/physiology , Alzheimer Disease/epidemiology , Cognition , Hippocampus/physiology , Aged , Alzheimer Disease/genetics , Apolipoproteins E/genetics , China , Educational Status , Female , Hippocampus/diagnostic imaging , Hippocampus/growth & development , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Sex Factors
13.
Biomolecules ; 13(1)2022 12 22.
Article En | MEDLINE | ID: mdl-36671403

Non-coding RNAs (ncRNAs), including miRNAs, lncRNAs, circRNAs, and piRNAs, do not encode proteins. Nonetheless, they have critical roles in a variety of cellular activities-such as development, neurogenesis, degeneration, and the response to injury to the nervous system-via protein translation, RNA splicing, gene activation, silencing, modifications, and editing; thus, they may serve as potential targets for disease treatment. The activity of adult neural stem cells (NSCs) in the subgranular zone of the hippocampal dentate gyrus critically influences hippocampal function, including learning, memory, and emotion. ncRNAs have been shown to be involved in the regulation of hippocampal neurogenesis, including proliferation, differentiation, and migration of NSCs and synapse formation. The interaction among ncRNAs is complex and diverse and has become a major topic within the life science. This review outlines advances in research on the roles of ncRNAs in modulating NSC bioactivity in the hippocampus and discusses their potential applications in the treatment of illnesses affecting the hippocampus.


Hippocampus , Neural Stem Cells , Neurogenesis , RNA, Long Noncoding , Adult , Humans , Cell Differentiation , Cell Proliferation , Hippocampus/growth & development , Neural Stem Cells/metabolism , Neurogenesis/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
15.
Sci Rep ; 11(1): 23897, 2021 12 13.
Article En | MEDLINE | ID: mdl-34903845

Early-life exposure to environmental toxins like tobacco can permanently re-program body structure and function. Here, we investigated the long-term effects on mouse adult sleep phenotype exerted by early-life exposure to nicotine or to its principal metabolite, cotinine. Moreover, we investigated whether these effects occurred together with a reprogramming of the activity of the hippocampus, a key structure to coordinate the hormonal stress response. Adult male mice born from dams subjected to nicotine (NIC), cotinine (COT) or vehicle (CTRL) treatment in drinking water were implanted with electrodes for sleep recordings. NIC and COT mice spent significantly more time awake than CTRL mice at the transition between the rest (light) and the activity (dark) period. NIC and COT mice showed hippocampal glucocorticoid receptor (GR) downregulation compared to CTRL mice, and NIC mice also showed hippocampal mineralocorticoid receptor downregulation. Hippocampal GR expression significantly and inversely correlated with the amount of wakefulness at the light-to-dark transition, while no changes in DNA methylation were found. We demonstrated that early-life exposure to nicotine (and cotinine) concomitantly entails long-lasting reprogramming of hippocampal activity and sleep phenotype suggesting that the adult sleep phenotype may be modulated by events that occurred during that critical period of life.


Cotinine/toxicity , Hippocampus/drug effects , Nicotine/toxicity , Receptors, Glucocorticoid/metabolism , Sleep Wake Disorders/metabolism , Animals , Down-Regulation , Hippocampus/growth & development , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurogenesis , Receptors, Glucocorticoid/genetics , Sleep Wake Disorders/etiology , Tobacco Smoke Pollution/adverse effects
16.
Nutrients ; 13(11)2021 Oct 28.
Article En | MEDLINE | ID: mdl-34836113

Iron deficiency (ID) anemia is the foremost micronutrient deficiency worldwide, affecting around 40% of pregnant women and young children. ID during the prenatal and early postnatal periods has a pronounced effect on neurodevelopment, resulting in long-term effects such as cognitive impairment and increased risk for neuropsychiatric disorders. Treatment of ID has been complicated as it does not always resolve the long-lasting neurodevelopmental deficits. In animal models, developmental ID results in abnormal hippocampal structure and function associated with dysregulation of genes involved in neurotransmission and synaptic plasticity. Dysregulation of these genes is a likely proximate cause of the life-long deficits that follow developmental ID. However, a direct functional link between iron and gene dysregulation has yet to be elucidated. Iron-dependent epigenetic modifications are one mechanism by which ID could alter gene expression across the lifespan. The jumonji and AT-rich interaction domain-containing (JARID) protein and the Ten-Eleven Translocation (TET) proteins are two families of iron-dependent epigenetic modifiers that play critical roles during neural development by establishing proper gene regulation during critical periods of brain development. Therefore, JARIDs and TETs can contribute to the iron-mediated epigenetic mechanisms by which early-life ID directly causes stable changes in gene regulation across the life span.


Anemia, Iron-Deficiency/genetics , Epigenesis, Genetic/physiology , Hippocampus/metabolism , Infant Nutritional Physiological Phenomena/genetics , Maternal Nutritional Physiological Phenomena/genetics , Anemia, Iron-Deficiency/complications , Animals , Animals, Newborn , Child Development/physiology , Epigenomics , Female , Hippocampus/growth & development , Humans , Infant , Infant, Newborn , Neurodevelopmental Disorders/genetics , Neurogenesis/physiology , Neuronal Plasticity/physiology , Pregnancy , Prenatal Exposure Delayed Effects/genetics , Synaptic Transmission/physiology
17.
Science ; 374(6568): eabk2055, 2021 Nov 05.
Article En | MEDLINE | ID: mdl-34735259

During development, neural circuit formation requires the stabilization of active γ-aminobutyric acid­mediated (GABAergic) synapses and the elimination of inactive ones. Here, we demonstrate that, although the activation of postsynaptic GABA type A receptors (GABAARs) and adenosine A2A receptors (A2ARs) stabilizes GABAergic synapses, only A2AR activation is sufficient. Both GABAAR- and A2AR-dependent signaling pathways act synergistically to produce adenosine 3',5'-monophosphate through the recruitment of the calcium­calmodulin­adenylyl cyclase pathway. Protein kinase A, thus activated, phosphorylates gephyrin on serine residue 303, which is required for GABAAR stabilization. Finally, the stabilization of pre- and postsynaptic GABAergic elements involves the interaction between gephyrin and the synaptogenic membrane protein Slitrk3. We propose that A2ARs act as detectors of active GABAergic synapses releasing GABA, adenosine triphosphate, and adenosine to regulate their fate toward stabilization or elimination.


Adenosine/metabolism , Hippocampus/growth & development , Neurons/physiology , Receptor, Adenosine A2A/metabolism , Signal Transduction , Synapses/physiology , gamma-Aminobutyric Acid/metabolism , Adenosine A2 Receptor Antagonists , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cognition , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Hippocampus/metabolism , Male , Membrane Proteins/metabolism , Mice , Nerve Tissue Proteins , Phosphorylation , Receptor, Adenosine A2A/genetics , Receptors, GABA-A/metabolism
18.
Nat Commun ; 12(1): 6137, 2021 10 21.
Article En | MEDLINE | ID: mdl-34675233

The mammalian brain stores and distinguishes among episodic memories, i.e. memories formed during the personal experience, through a mechanism of pattern separation computed in the hippocampal dentate gyrus. Decision-making for food-related behaviors, such as the choice and intake of food, might be affected in obese subjects by alterations in the retrieval of episodic memories. Adult neurogenesis in the dentate gyrus regulates the pattern separation. Several molecular factors affect adult neurogenesis and exert a critical role in the development and plasticity of newborn neurons. Orexin-A/hypocretin-1 and downstream endocannabinoid 2-arachidonoylglycerol signaling are altered in obese mice. Here, we show that excessive orexin-A/2-arachidonoylglycerol/cannabinoid receptor type-1 signaling leads to the dysfunction of adult hippocampal neurogenesis and the subsequent inhibition of plasticity and impairment of pattern separation. By inhibiting orexin-A action at orexin-1 receptors we rescued both plasticity and pattern separation impairment in obese mice, thus providing a molecular and functional mechanism to explain alterations in episodic memory in obesity.


Endocannabinoids/metabolism , Hippocampus/growth & development , Neurogenesis , Neuronal Plasticity , Obesity/metabolism , Obesity/psychology , Orexins/metabolism , Animals , Female , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Male , Memory, Episodic , Mice , Mice, Obese , Neurons/cytology , Neurons/metabolism , Obesity/genetics , Obesity/physiopathology , Orexin Receptors/genetics , Orexin Receptors/metabolism , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction
19.
Genes (Basel) ; 12(10)2021 09 28.
Article En | MEDLINE | ID: mdl-34680922

Down syndrome (DS), trisomy of the long arm of human chromosome 21 (Hsa21), is the most common genetic cause of intellectual disability (ID). Currently, there are no effective pharmacotherapies. The success of clinical trials to improve cognition depends in part on the design of preclinical evaluations in mouse models. To broaden understanding of the common limitations of experiments in learning and memory, we report performance in context fear conditioning (CFC) in three mouse models of DS, the Dp(16)1Yey, Dp(17)1Yey and Dp(10)1Yey (abbreviated Dp16, Dp17 and Dp10), separately trisomic for the human Hsa21 orthologs mapping to mouse chromosomes 16, 17 and 10, respectively. We examined female and male mice of the three lines on the standard C57BL/6J background at 3 months of age and Dp17 and Dp10 at 18 months of age. We also examined female and male mice of Dp17 and Dp10 at 3 months of age as F1 hybrids obtained from a cross with the DBA/2J background. Results indicate that genotype, sex, age and genetic background affect CFC performance. These data support the need to use both female and male mice, trisomy of sets of all Hsa21 orthologs, and additional ages and genetic backgrounds to improve the reliability of preclinical evaluations of drugs for ID in DS.


Conditioning, Classical , Down Syndrome/physiopathology , Genetic Background , Animals , Down Syndrome/genetics , Fear , Female , Hippocampus/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Sex Characteristics
20.
Int Rev Neurobiol ; 160: 251-280, 2021.
Article En | MEDLINE | ID: mdl-34696875

Adolescence is a period of continued brain development. Regions of the brain, such as the hippocampus, continue to undergo refinement and maturation throughout adolescence and into early adulthood. Adolescence is also a time of heightened sensitivity to novelty and reward, which contribute to an increase in risk-taking behaviors including the use of drugs and alcohol. Importantly, binge drinking is highly prevalent among adolescents and emerging adults. The hippocampus which is important for the integration of emotion, reward, homeostasis, and memory is particularly vulnerable to the neurotoxic effects of alcohol. In this chapter, we cover the fundamentals of hippocampal neuroanatomy and the current state of knowledge of the acute and chronic effects of ethanol in adolescent humans and adolescent rodent models. We focus on the hippocampal-dependent behavioral, structural, and neurochemical changes and identify knowledge gaps in our understanding of age-dependent neurobiological effects of alcohol use.


Ethanol , Hippocampus , Underage Drinking , Adolescent , Ethanol/toxicity , Hippocampus/drug effects , Hippocampus/growth & development , Humans
...