Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.764
Filter
1.
J Psychopharmacol ; 38(5): 489-499, 2024 May.
Article in English | MEDLINE | ID: mdl-38680011

ABSTRACT

BACKGROUND: Psilocybin offers new hope for treating mood disorders due to its rapid and sustained antidepressant effects, as standard medications require weeks or months to exert their effects. However, the mechanisms underlying this action of psilocybin have not been identified. AIMS: To investigate whether psilocybin has rapid and sustained antidepressant-like effects in mice and investigate whether its potential mechanisms of action are related to promoted neuroplasticity. METHODS: We first examined the antidepressant-like effects of psilocybin in normal mice by the forced swimming test and in chronic corticosterone (CORT)-exposed mice by the sucrose preference test and novelty-suppressed feeding test. Furthermore, to explore the role of neuroplasticity in mediating the antidepressant-like effects of psilocybin, we measured structural neuroplasticity and neuroplasticity-associated protein levels in the prefrontal cortex (PFC) and hippocampus. RESULTS: We observed that a single dose of psilocybin had rapid and sustained antidepressant-like effects in both healthy mice and chronic CORT-exposed mice. Moreover, psilocybin ameliorated chronic CORT exposure-induced inhibition of neuroplasticity in the PFC and hippocampus, including by increasing neuroplasticity (total number of dendritic branches and dendritic spine density), synaptic protein (p-GluA1, PSD95 and synapsin-1) levels, BDNF-mTOR signalling pathway activation (BDNF, TrkB and mTOR levels), and promoting neurogenesis (number of DCX-positive cells). CONCLUSIONS: Our results demonstrate that psilocybin elicits robust, rapid and sustained antidepressant-like effects which is accompanied by the promotion of neuroplasticity in the PFC and hippocampus.


Subject(s)
Antidepressive Agents , Brain-Derived Neurotrophic Factor , Corticosterone , Hippocampus , Neuronal Plasticity , Prefrontal Cortex , Psilocybin , Animals , Neuronal Plasticity/drug effects , Antidepressive Agents/pharmacology , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Male , Psilocybin/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism , Doublecortin Protein , Behavior, Animal/drug effects , Disease Models, Animal
2.
Neurosci Lett ; 832: 137787, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38641312

ABSTRACT

BACKGROUND: Salidroside (Sal) has been found to protect against multiple impairments caused by diabetes, and we designed this study to investigate the effect of Sal on gestational hypertension (GHP)-induced impairment of offspring learning and memory. METHODS: We established a GHP rat model by intraperitoneal injection of NG-nitro-L-arginine methyl ester (L-NAME), and treated with Sal by daily gavage. We used Morris Water Maze test to evaluate the learning and memory ability of offspring rats. HE staining was used to measured the pathological changes in hippocampus of offspring. Immunohistochemistry, cellular immunofluorescence and western blot were used to detect the protein expression. RESULTS: The learning and memory abilities of GHP offspring rats were significantly lower than those of normal rat offspring, while Sal treatment could significantly improve the learning and memory abilities of GHP offspring rats. HE staining did not reveal pathological differences in the hippocampus of normal rats, GHP rats and Sal-treated GHP offspring rats. However, Sal treatment can significantly increase the expression of Wnt1 and Skp2 protein, and decrease the expression of P27kiwf and P21waf1 protein in the hippocampus of GHP offspring rats. In vitro, Sal significantly promoted the proliferation and differentiation on neural stem cell, while Wnt1 knockdown could reverse these promotions by Sal. In the hippocampus of GHP offspring rats, Sal treatment significantly increased the expression of Tuj1, SOX2, Ki67 and DCX protein. CONCLUSION: Salidroside significantly improves the learning and memory impairment of offspring caused by GHP, and its mechanism may be related to the fact that Salidroside promotes the proliferation and differentiation of neural stem cells by activating the Wnt1/Skp2 signaling pathway.


Subject(s)
Glucosides , Hippocampus , Hypertension, Pregnancy-Induced , Phenols , Rats, Sprague-Dawley , Wnt Signaling Pathway , Animals , Glucosides/pharmacology , Glucosides/therapeutic use , Phenols/pharmacology , Pregnancy , Female , Rats , Wnt Signaling Pathway/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Hypertension, Pregnancy-Induced/metabolism , Hypertension, Pregnancy-Induced/prevention & control , Memory/drug effects , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/prevention & control , Doublecortin Protein , Memory Disorders/prevention & control , Memory Disorders/metabolism , Memory Disorders/drug therapy , Male
3.
Cells ; 13(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38667284

ABSTRACT

This study investigates the combined effects of the neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31-Pro34]NPY at a dose of 132 µg and Ketamine at 10 mg/Kg on cognitive functions and neuronal proliferation, against a backdrop where neurodegenerative diseases present an escalating challenge to global health systems. Utilizing male Sprague-Dawley rats in a physiological model, this research employed a single-dose administration of these compounds and assessed their impact 24 h after treatment on object-in-place memory tasks, alongside cellular proliferation within the dorsal hippocampus dentate gyrus. Methods such as the in situ proximity ligation assay and immunohistochemistry for proliferating a cell nuclear antigen (PCNA) and doublecortin (DCX) were utilized. The results demonstrated that co-administration significantly enhanced memory consolidation and increased neuronal proliferation, specifically neuroblasts, without affecting quiescent neural progenitors and astrocytes. These effects were mediated by the potential formation of NPY1R-TrkB heteroreceptor complexes, as suggested by receptor co-localization studies, although further investigation is required to conclusively prove this interaction. The findings also highlighted the pivotal role of brain-derived neurotrophic factor (BDNF) in mediating these effects. In conclusion, this study presents a promising avenue for enhancing cognitive functions and neuronal proliferation through the synergistic action of the NPY1R agonist and Ketamine, potentially via NPY1R-TrkB heteroreceptor complex formation, offering new insights into therapeutic strategies for neurodegenerative diseases.


Subject(s)
Cell Proliferation , Cognition , Doublecortin Protein , Ketamine , Neurons , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide Y , Receptors, Neuropeptide , Animals , Male , Ketamine/pharmacology , Ketamine/administration & dosage , Cognition/drug effects , Rats , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Neurons/drug effects , Neurons/metabolism , Cell Proliferation/drug effects , Receptor, trkB/agonists , Receptor, trkB/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Neurogenesis/drug effects
4.
Medicina (Kaunas) ; 60(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38674304

ABSTRACT

Background and Objectives. Neurogenesis is an integral process in post-stroke recovery, involving the recruitment of proliferating neuroblasts from neurogenic niches of the mammal brain. However, the role of neurogenesis in the long-term restoration following ischemic stroke is fragmented. Post-stroke motor dysfunction includes challenges in the proper, coordinated use of hands and is present in roughly two-thirds of human patients. In this study, we investigated chronic behavioral and biochemical alterations after transient cerebral ischemia in adult male mice. Materials and Methods: Twelve-week-old C57BL/6N male mice were used, and fMCAo lasting 60 min was induced. At multiple timepoints after fMCAo induction, a single pellet reaching task was performed. Six months after the procedure, we immunohistochemically determined the number of proliferating neuroblasts (BrdU and DCX-positive) and the number of differentiated astrocytes (GFAP-positive) in both brain hemispheres. Results: The reaching ability of fMCAo mice was impaired from one month to six months after the induction of ischemia. Neuroblast proliferation was increased in the ipsilateral SVZ, whereas GFAP+ cell count was elevated in the hippocampal DG of both hemispheres of the fMCAo group mice. Conclusions: Our current report demonstrates the long-term effects of transient cerebral ischemia on mice functional parameters and neurogenesis progression. Our data demonstrate that transient cerebral ischemia promotes a long-lasting regenerative response in the ipsilateral brain hemisphere, specifically in the neurogenic SVZ and DG regions.


Subject(s)
Astrocytes , Doublecortin Protein , Mice, Inbred C57BL , Motor Skills , Neurogenesis , Animals , Neurogenesis/physiology , Mice , Male , Astrocytes/physiology , Motor Skills/physiology , Disease Models, Animal , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/complications
5.
Aging Cell ; 22(12): e13974, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37649323

ABSTRACT

Beyond the canonical neurogenic niches, there are dormant neuronal precursors in several regions of the adult mammalian brain. Dormant precursors maintain persisting post-mitotic immaturity from birth to adulthood, followed by staggered awakening, in a process that is still largely unresolved. Strikingly, due to the slow rate of awakening, some precursors remain immature until old age, which led us to question whether their awakening and maturation are affected by aging. To this end, we studied the maturation of dormant precursors in transgenic mice (DCX-CreERT2 /flox-EGFP) in which immature precursors were labelled permanently in vivo at different ages. We found that dormant precursors are capable of awakening at young age, becoming adult-matured neurons (AM), as well as of awakening at old age, becoming late AM. Thus, protracted immaturity does not prevent late awakening and maturation. However, late AM diverged morphologically and functionally from AM. Moreover, AM were functionally most similar to neonatal-matured neurons (NM). Conversely, late AM were endowed with high intrinsic excitability and high input resistance, and received a smaller amount of spontaneous synaptic input, implying their relative immaturity. Thus, late AM awakening still occurs at advanced age, but the maturation process is slow.


Subject(s)
Doublecortin Protein , Neurons , Mice , Animals , Neurons/metabolism , Brain/metabolism , Mice, Transgenic , Neurogenesis/physiology , Microtubule-Associated Proteins/metabolism , Mammals/metabolism
6.
Hippocampus ; 33(10): 1075-1093, 2023 10.
Article in English | MEDLINE | ID: mdl-37421207

ABSTRACT

We investigated the mechanisms underlying the effects of the antidepressant fluoxetine on behavior and adult hippocampal neurogenesis (AHN). After confirming our earlier report that the signaling molecule ß-arrestin-2 (ß-Arr2) is required for the antidepressant-like effects of fluoxetine, we found that the effects of fluoxetine on proliferation of neural progenitors and survival of adult-born granule cells are absent in the ß-Arr2 knockout (KO) mice. To our surprise, fluoxetine induced a dramatic upregulation of the number of doublecortin (DCX)-expressing cells in the ß-Arr2 KO mice, indicating that this marker can be increased even though AHN is not. We discovered two other conditions where a complex relationship occurs between the number of DCX-expressing cells compared to levels of AHN: a chronic antidepressant model where DCX is upregulated and an inflammation model where DCX is downregulated. We concluded that assessing the number of DCX-expressing cells alone to quantify levels of AHN can be complex and that caution should be applied when label retention techniques are unavailable.


Subject(s)
Doublecortin Protein , Fluoxetine , Animals , Mice , Antidepressive Agents/pharmacology , Fluoxetine/pharmacology , Hippocampus/physiology , Neurogenesis/physiology , Neurons
7.
Neurotox Res ; 41(4): 349-361, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37074591

ABSTRACT

We aimed to test the effects of connexin43 (Cx43) on ischemic neurogenesis and examined whether it was dependent on aquaporin-4 (AQP4). We detected the expression of Cx43 and AQP4 in the ipsilateral subventricular zone (SVZ) and peri-infarct cortex after middle cerebral artery occlusion (MCAO). Also, we examined neurogenesis in the above regions via co-labeling of 5-bromo-2-deoxyuridine (BrdU)/neuronal nuclear antigen (NeuN) and BrdU/doublecortin (DCX). The effects of Cx43 and AQP4 were investigated by using two transgenic animals: heterozygous Cx43 (Cx43±) mice and AQP4 knockout (AQP4-/-) mice, and connexin mimetic peptide (CMP), a selective Cx43 blocker. We demonstrated AQP4 and Cx43 were co-expressed in the astrocytes after MCAO and the expression was highly increased in ipsilateral SVZ and peri-infarct cortex. Cx43± mice had larger infarction volumes and worse neurological function. Both BrdU/NeuN and BrdU/DCX co-labeled cells in the two regions were reduced in Cx43± and AQP4-/- mice compared to wild-type (WT) mice, suggesting Cx43 and AQP4 participated in neurogenesis of neural stem cells. Moreover, CMP decreased AQP4 expression and inhibited neurogenesis in WT mice, while the latter failed to be observed in AQP4-/- mice. Besides, higher levels of IL-1ß and TNF-α were detected in the SVZ and peri-infarct cortex of AQP4-/- and Cx43± mice than those in WT mice. In conclusion, our data suggest that Cx43 elicits neuroprotective effects after cerebral ischemia through promoting neurogenesis in the SVZ to regenerate the injured neurons, which is AQP4 dependent and associated with down-regulation of inflammatory cytokines IL-1ß and TNF-α.


Subject(s)
Aquaporin 4 , Brain Ischemia , Connexin 43 , Animals , Mice , Brain Ischemia/complications , Bromodeoxyuridine/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Doublecortin Protein , Infarction, Middle Cerebral Artery/complications , Neurogenesis , Tumor Necrosis Factor-alpha/metabolism , Aquaporin 4/genetics , Aquaporin 4/metabolism , Neuroprotection
8.
Bull Exp Biol Med ; 176(2): 246-252, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38194066

ABSTRACT

We studied the effect of human lactoferrin on cells of the hippocampal dentate gyrus of 2-2.5-month-old male C57BL/6 mice after acute gamma irradiation of the head in a dose of 8 Gy from a 60Co source. Immediately after irradiation some animals received an intraperitoneal injection of human lactoferrin (4 mg/mouse). The appearance of TUNEL+ cells in the subgranular zone 6 h after irradiation was accompanied by a corresponding decrease in the number of Ki-67- and DCX-immunoreactive cells. Administration of lactoferrin had a protective effect on mouse brain cells, which manifested in a decrease in the number of TUNEL+ cells (by 77% relative to the irradiation alone) and an increase in the number of proliferating cells (from 16 to 61% relative to control animals) and immature neurons (from 14 to 22% relative to control animals) in the dentate gyrus of the hippocampus.


Subject(s)
Dentate Gyrus , Lactoferrin , Humans , Mice , Male , Animals , Infant , Lactoferrin/pharmacology , Doublecortin Protein , Mice, Inbred C57BL , Hippocampus , Brain , Neurogenesis/physiology , Cell Proliferation
10.
Pharmacol Res ; 179: 106226, 2022 05.
Article in English | MEDLINE | ID: mdl-35460881

ABSTRACT

BKM120 is an inhibitor of class I phosphoinositide 3-kinases and its anti-cancer effects have been demonstrated in various solid cancer models. BKM120 is highly brain permeable and has been reported to induce mood disturbances in clinical trials. Therefore, we examined whether BKM120 produces anxiety- and depression-like behaviors in mice, as with patients receiving BKM120 in clinical trials. In this study, repeated BKM120 treatment (2.0 or 5.0 mg/kg, i.p., five times at 12-h interval) significantly induced anxiety- and depression-like behaviors in mice. Although abnormal changes in hippocampal neurogenesis have been suggested to, at least in part, associated with the pathogenesis of depression and anxiety, BKM120 did not affect the incorporation of 5-bromo-2'-deoxyuridine or the expression of doublecortin (DCX); however, it significantly enhanced the radial migration of DCX-positive cells in the dentate gyrus. BKM120-induced changes in migration were not accompanied by obvious neuronal damage in the hippocampus. Importantly, BKM120-induced anxiety- and depression-like behaviors were positively correlated with the extent of DCX-positive cell migration. Concomitantly, p-Akt expression was significantly decreased in the dentate gyrus. Moreover, the expression of p-c-Jun N-terminal kinase (JNK), p-DCX, and Ras homolog family member A (RhoA)-GTP decreased significantly, particularly in aberrantly migrated DCX-positive cells. Together, the results suggest that repeated BKM120 treatment enhances the radial migration of DCX-positive cells and induces anxiety- and depression-like behaviors by regulating the activity of Akt, JNK, DCX, and RhoA in the dentate gyrus. It also suggests that the altered migration of adult-born neurons in the dentate gyrus plays a role in mood disturbances.


Subject(s)
Dentate Gyrus , Neuropeptides , Aminopyridines , Animals , Dentate Gyrus/metabolism , Doublecortin Domain Proteins , Doublecortin Protein , Hippocampus/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Morpholines , Neuropeptides/metabolism , Proto-Oncogene Proteins c-akt/metabolism
11.
Elife ; 112022 04 29.
Article in English | MEDLINE | ID: mdl-35485925

ABSTRACT

Doublecortin (DCX) is a microtubule (MT)-associated protein that regulates MT structure and function during neuronal development and mutations in DCX lead to a spectrum of neurological disorders. The structural properties of MT-bound DCX that explain these disorders are incompletely determined. Here, we describe the molecular architecture of the DCX-MT complex through an integrative modeling approach that combines data from X-ray crystallography, cryo-electron microscopy, and a high-fidelity chemical crosslinking method. We demonstrate that DCX interacts with MTs through its N-terminal domain and induces a lattice-dependent self-association involving the C-terminal structured domain and its disordered tail, in a conformation that favors an open, domain-swapped state. The networked state can accommodate multiple different attachment points on the MT lattice, all of which orient the C-terminal tails away from the lattice. As numerous disease mutations cluster in the C-terminus, and regulatory phosphorylations cluster in its tail, our study shows that lattice-driven self-assembly is an important property of DCX.


Subject(s)
Neuropeptides , Cryoelectron Microscopy , Doublecortin Domain Proteins , Doublecortin Protein , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Neuropeptides/metabolism
12.
Neurobiol Dis ; 168: 105702, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35339680

ABSTRACT

Human doublecortin (DCX) mutations are associated with severe brain malformations leading to aberrant neuron positioning (heterotopia), intellectual disability and epilepsy. DCX is a microtubule-associated protein which plays a key role during neurodevelopment in neuronal migration and differentiation. Dcx knockout (KO) mice show disorganized hippocampal pyramidal neurons. The CA2/CA3 pyramidal cell layer is present as two abnormal layers and disorganized CA3 KO pyramidal neurons are also more excitable than wild-type (WT) cells. To further identify abnormalities, we characterized Dcx KO hippocampal neurons at subcellular, molecular and ultrastructural levels. Severe defects were observed in mitochondria, affecting number and distribution. Also, the Golgi apparatus was visibly abnormal, increased in volume and abnormally organized. Transcriptome analyses from laser microdissected hippocampal tissue at postnatal day 60 (P60) highlighted organelle abnormalities. Ultrastructural studies of CA3 cells performed in P60 (young adult) and > 9 months (mature) tissue showed that organelle defects are persistent throughout life. Locomotor activity and fear memory of young and mature adults were also abnormal: Dcx KO mice consistently performed less well than WT littermates, with defects becoming more severe with age. Thus, we show that disruption of a neurodevelopmentally-regulated gene can lead to permanent organelle anomalies contributing to abnormal adult behavior.


Subject(s)
Doublecortin Protein/genetics , Neuropeptides , Animals , Doublecortin Domain Proteins , Golgi Apparatus , Hippocampus/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mutation , Neuropeptides/genetics , Neuropeptides/metabolism , Pyramidal Cells/metabolism
13.
Cells ; 11(5)2022 02 24.
Article in English | MEDLINE | ID: mdl-35269413

ABSTRACT

Background: Adult-born neurons of the hippocampal dentate gyrus play a role in specific forms of learning, and disturbed neurogenesis seems to contribute to the development of neuropsychiatric disorders, such as major depression. Neuroinflammation inhibits adult neurogenesis, but the effect of peripheral inflammation on this form of neuroplasticity is ambiguous. Objective: Our aim was to investigate the influence of acute and chronic experimental arthritis on adult hippocampal neurogenesis and to elucidate putative regulatory mechanisms. Methods: Arthritis was triggered by subcutaneous injection of complete Freund's adjuvant (CFA) into the hind paws of adult male mice. The animals were killed either seven days (acute inflammation) or 21 days (chronic inflammation) after the CFA injection. Behavioral tests were used to demonstrate arthritis-related hypersensitivity to painful stimuli. We used in vivo bioluminescence imaging to verify local inflammation. The systemic inflammatory response was assessed by complete blood cell counts and by measurement of the cytokine/chemokine concentrations of TNF-α, IL-1α, IL-4, IL-6, IL-10, KC and MIP-2 in the inflamed hind limbs, peripheral blood and hippocampus to characterize the inflammatory responses in the periphery and in the brain. In the hippocampal dentate gyrus, the total number of newborn neurons was determined with quantitative immunohistochemistry visualizing BrdU- and doublecortin-positive cells. Microglial activation in the dentate gyrus was determined by quantifying the density of Iba1- and CD68-positive cells. Results: Both acute and chronic arthritis resulted in paw edema, mechanical and thermal hyperalgesia. We found phagocytic infiltration and increased levels of TNF-α, IL-4, IL-6, KC and MIP-2 in the inflamed hind paws. Circulating neutrophil granulocytes and IL-6 levels increased in the blood solely during the acute phase. In the dentate gyrus, chronic arthritis reduced the number of doublecortin-positive cells, and we found increased density of CD68-positive macrophages/microglia in both the acute and chronic phases. Cytokine levels, however, were not altered in the hippocampus. Conclusions: Our data suggest that acute peripheral inflammation initiates a cascade of molecular and cellular changes that eventually leads to reduced adult hippocampal neurogenesis, which was detectable only in the chronic inflammatory phase.


Subject(s)
Arthritis, Experimental , Tumor Necrosis Factor-alpha , Animals , Cytokines/metabolism , Doublecortin Protein , Freund's Adjuvant , Hippocampus/metabolism , Inflammation , Interleukin-4 , Interleukin-6 , Male , Mice , Neurogenesis/physiology
14.
Epilepsia ; 63(5): 1253-1265, 2022 05.
Article in English | MEDLINE | ID: mdl-35213059

ABSTRACT

OBJECTIVE: Pathogenic variants in DCX on the X chromosome lead to lissencephaly and subcortical band heterotopia (SBH), brain malformations caused by neuronal migration defects. Its product doublecortin (DCX) binds to microtubules to modulate microtubule polymerization. How pathogenic DCX variants affect these activities remains not fully investigated. METHODS: DCX variants were identified using whole exome and Sanger sequencing from six families with lissencephaly/SBH. We examined how these variants affect DCX functions using microtubule binding, regrowth, and colocalization assays. RESULTS: We found novel DCX variants p.Val177AlafsTer31 and p.Gly188Trp, as well as reported variants p.Arg196His, p.Lys202Met, and p.Thr203Ala. Incidentally, all of the missense variants were clustered on the C-terminal DCX domain. The microtubule binding ability was significantly decreased in p.Val177AlafsTer31, p.Gly188Trp, p.Lys202Met, and previously reported p.Asp262Gly variants. Furthermore, expression of p.Val177AlafsTer31, p.Gly188Trp, p.Arg196His, p.Lys202Met, and p.Asp262Gly variants hindered microtubule growth in cells. There were also decreases in the colocalization of p.Val177AlafsTer31, p.Thr203Ala, and p.Asp262Gly variants to microtubules. SIGNIFICANCE: Our results indicate that these variants in the C-terminal DCX domain altered microtubule binding and dynamics, which may underlie neuronal migration defects during brain development.


Subject(s)
Classical Lissencephalies and Subcortical Band Heterotopias , Lissencephaly , Neuropeptides , Doublecortin Domain Proteins , Doublecortin Protein , Humans , Lissencephaly/genetics , Microtubule-Associated Proteins/genetics , Microtubules , Neuropeptides/genetics
15.
Cells ; 11(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-35011720

ABSTRACT

Williams syndrome (WS) is a multisystem neurodevelopmental disorder caused by a de novo hemizygous deletion of ~26 genes from chromosome 7q11.23, among them the general transcription factor II-I (GTF2I). By studying a novel murine model for the hypersociability phenotype associated with WS, we previously revealed surprising aberrations in myelination and cell differentiation properties in the cortices of mutant mice compared to controls. These mutant mice had selective deletion of Gtf2i in the excitatory neurons of the forebrain. Here, we applied diffusion magnetic resonance imaging and fiber tracking, which showed a reduction in the number of streamlines in limbic outputs such as the fimbria/fornix fibers and the stria terminalis, as well as the corpus callosum of these mutant mice compared to controls. Furthermore, we utilized next-generation sequencing (NGS) analysis of cortical small RNAs' expression (RNA-Seq) levels to identify altered expression of microRNAs (miRNAs), including two from the miR-34 cluster, known to be involved in prominent processes in the developing nervous system. Luciferase reporter assay confirmed the direct binding of miR-34c-5p to the 3'UTR of PTPRU-a gene involved in neural development that was elevated in the cortices of mutant mice relative to controls. Moreover, we found an age-dependent variation in the expression levels of doublecortin (Dcx)-a verified miR-34 target. Thus, we demonstrate the substantial effect a single gene deletion can exert on miRNA regulation and brain structure, and advance our understanding and, hopefully, treatment of WS.


Subject(s)
Brain/growth & development , Doublecortin Protein/metabolism , MicroRNAs/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , White Matter/physiopathology , Williams Syndrome/genetics , Animals , Disease Models, Animal , Female , Humans , Mice , Williams Syndrome/pathology
16.
Neuron ; 110(3): 452-469.e14, 2022 02 02.
Article in English | MEDLINE | ID: mdl-34798047

ABSTRACT

The hippocampal-entorhinal system supports cognitive functions, has lifelong neurogenic capabilities in many species, and is selectively vulnerable to Alzheimer's disease. To investigate neurogenic potential and cellular diversity, we profiled single-nucleus transcriptomes in five hippocampal-entorhinal subregions in humans, macaques, and pigs. Integrated cross-species analysis revealed robust transcriptomic and histologic signatures of neurogenesis in the adult mouse, pig, and macaque but not humans. Doublecortin (DCX), a widely accepted marker of newly generated granule cells, was detected in diverse human neurons, but it did not define immature neuron populations. To explore species differences in cellular diversity and implications for disease, we characterized subregion-specific, transcriptomically defined cell types and transitional changes from the three-layered archicortex to the six-layered neocortex. Notably, METTL7B defined subregion-specific excitatory neurons and astrocytes in primates, associated with endoplasmic reticulum and lipid droplet proteins, including Alzheimer's disease-related proteins. This resource reveals cell-type- and species-specific properties shaping hippocampal-entorhinal neurogenesis and function.


Subject(s)
Macaca , Transcriptome , Animals , Doublecortin Protein , Hippocampus/pathology , Humans , Mice , Neurogenesis/genetics , Swine
17.
Int J Mol Sci ; 22(24)2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34948016

ABSTRACT

Glioblastoma (GBM) remains the leading cause of cancer-related deaths with the lowest five-year survival rates among all of the human cancers. Multiple factors contribute to its poor outcome, including intratumor heterogeneity, along with migratory and invasive capacities of tumour cells. Over the last several years Doublecortin (DCX) has been one of the debatable factors influencing GBM cells' migration. To resolve DCX's ambiguous role in GBM cells' migration, we set to analyse the expression patterns of DCX along with Nestin (NES) and Oligodendrocyte lineage transcription factor 2 (OLIG2) in 17 cases of GBM, using immunohistochemistry, followed by an analysis of single-cell RNA-seq data. Our results showed that only a small subset of DCX positive (DCX+) cells was present in the tumour. Moreover, no particular pattern emerged when analysing DCX+ cells relative position to the tumour margin. By looking into single-cell RNA-seq data, the majority of DCX+ cells were classified as non-cancerous, with a small subset of cells that could be regarded as glioma stem cells. In conclusion, our findings support the notion that glioma cells express DCX; however, there is no clear evidence to prove that DCX participates in GBM cell migration.


Subject(s)
Brain Neoplasms/metabolism , Doublecortin Protein/metabolism , Gene Expression Profiling/methods , Glioblastoma/metabolism , Nestin/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Brain Neoplasms/genetics , Cell Movement , Doublecortin Protein/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Heuristics , Humans , Image Processing, Computer-Assisted , Microscopy, Confocal , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Nestin/genetics , Oligodendrocyte Transcription Factor 2/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Survival Analysis
18.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34830044

ABSTRACT

BMPs regulate synovial quiescence and adult neurogenesis in the hippocampus in non-stress conditions. However, changes in BMP expression that are induced by inflammation during rheumatoid arthritis (RA) have not yet been reported. Here, we show that signalling with synovial BMPs (BMP-4 and -7) mediates the effect of systemic inflammation on adult neurogenesis in the hippocampus during pristane-induced arthritis (PIA) in Dark Agouti (DA) rats, an animal model of RA. Moreover, we show gender differences in BMP expressions and their antagonists (Noggin and Gremlin) during PIA and their correlations with the clinical course and IL-17A and TNF-α levels in serum. Our results indicate gender differences in the clinical course, where male rats showed earlier onset and earlier recovery but a worse clinical course in the first two phases of the disease (onset and peak), which correlates with the initial increase of serum IL-17A level. The clinical course of the female rats worsened in remission. Their prolonged symptoms could be a reflection of an increased TNF-α level in serum during remission. Synovial inflammation was greater in females in PIA-remission with greater synovial BMP and antagonist expressions. More significant correlations between serum cytokines (IL-17A and TNF-α), and synovial BMPs and their antagonists were found in females than in males. On the other hand, males showed an increase in hippocampal BMP-4 expression during the acute phase, but both genders showed a decrease in antagonist expressions during PIA in general. Both genders showed a decrease in the number of Ki-67+ and SOX-2+ and DCX+ cells and in the ratio of DCX+ to Ki67+ cells in the dentate gyrus during PIA. However, in PIA remission, females showed a faster increase in the number of Ki67+, SOX-2+, and DCX+ cells and a faster increase in the DCX/Ki67 ratio than males. Both genders showed an increase of hippocampal BMP-7 expression during remission, although males constantly showed greater BMP-7 expression at all time points. Our data show that gender differences exist in the BMP expressions in the periphery-hippocampus axis and in the IL-17A and TNF-α levels in serum, which could imply differences in the mechanisms for the onset and progression of the disease, the clinical course severity, and adult neurogenesis with subsequent neurological complications between genders.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Bone Morphogenetic Proteins/metabolism , Hippocampus/metabolism , Joints/metabolism , Neurogenesis , Aging , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 7/metabolism , Carrier Proteins/metabolism , Cytokines/metabolism , Doublecortin Protein/metabolism , Female , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-17/blood , Ki-67 Antigen/metabolism , Male , Rats , SOXB1 Transcription Factors/metabolism , Sex Factors , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Terpenes/toxicity , Tumor Necrosis Factor-alpha/blood
19.
Sci Rep ; 11(1): 21499, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728755

ABSTRACT

Adolescence represents a crucial period for maturation of brain structures involved in cognition. Early in life unhealthy dietary patterns are associated with inferior cognitive outcomes at later ages; conversely, healthy diet is associated with better cognitive results. In this study we analyzed the effects of a short period of hypercaloric diet on newborn hippocampal doublecortin+ (DCX) immature neurons in adolescent mice. Male mice received high fat diet (HFD) or control low fat diet (LFD) from the 5th week of age for 1 or 2 weeks, or 1 week HFD followed by 1 week LFD. After diet supply, mice were either perfused for immunohistochemical (IHC) analysis or their hippocampi were dissected for biochemical assays. Detailed morphometric analysis was performed in DCX+ cells that displayed features of immature neurons. We report that 1 week-HFD was sufficient to dramatically reduce dendritic tree complexity of DCX+ cells. This effect occurred specifically in dorsal and not ventral hippocampus and correlated with reduced BDNF expression levels in dorsal hippocampus. Both structural and biochemical changes were reversed by a return to LFD. Altogether these studies increase our current knowledge on potential consequences of hypercaloric diet on brain and in particular on dorsal hippocampal neuroplasticity.


Subject(s)
Diet, High-Fat/adverse effects , Doublecortin Protein/metabolism , Hippocampus/pathology , Neural Stem Cells/pathology , Neurogenesis , Neuronal Plasticity , Neurons/pathology , Animals , Body Weight , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurons/metabolism
20.
Int J Mol Sci ; 22(20)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34681636

ABSTRACT

Depression is a neuropsychiatric disorder with a high impact on the worldwide population. To overcome depression, antidepressant drugs are the first line of treatment. However, pre-clinical studies have pointed out that antidepressants are not entirely efficacious and that the quality of the living environment after stress cessation may play a relevant role in increasing their efficacy. As it is unknown whether a short daily exposure to environmental enrichment during chronic stress and antidepressant treatment will be more effective than just the pharmacological treatment, this study analyzed the effects of fluoxetine, environmental enrichment, and their combination on depressive-associated behavior. Additionally, we investigated hippocampal neurogenesis in mice exposed to chronic mild stress. Our results indicate that fluoxetine reversed anhedonia. Besides, fluoxetine reversed the decrement of some events of the hippocampal neurogenic process caused by chronic mild stress. Conversely, short daily exposure to environmental enrichment changed the deterioration of the coat and anhedonia. Although, this environmental intervention did not produce significant changes in the neurogenic process affected by chronic mild stress, fluoxetine plus environmental enrichment showed similar effects to those caused by environmental enrichment to reverse depressive-like behaviors. Like fluoxetine, the combination reversed the declining number of Ki67, doublecortin, calretinin cells and mature newborn neurons. Finally, this study suggests that short daily exposure to environmental enrichment improves the effects of fluoxetine to reverse the deterioration of the coat and anhedonia in chronically stressed mice. In addition, the combination of fluoxetine with environmental enrichment produces more significant effects than those caused by fluoxetine alone on some events of the neurogenic process. Thus, environmental enrichment improves the benefits of pharmacological treatment by mechanisms that need to be clarified.


Subject(s)
Anhedonia/drug effects , Fluoxetine/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/physiopathology , Anhedonia/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Calbindin 2/metabolism , Cell Proliferation , Doublecortin Protein/metabolism , Environment , Female , Hippocampus/metabolism , Hippocampus/pathology , Ki-67 Antigen/metabolism , Mice , Mice, Inbred BALB C , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...