Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.465
Filter
1.
Brain Res Bull ; 213: 110981, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38777132

ABSTRACT

INTRODUCTION: The medial prefrontal cortex (mPFC) forms output pathways through projection neurons, inversely receiving adjacent and long-range inputs from other brain regions. However, how afferent neurons of mPFC are affected by chronic stress needs to be clarified. In this study, the effects of chronic restraint stress (CRS) on the distribution density of mPFC dendrites/dendritic spines and the projections from the cortex and subcortical brain regions to the mPFC were investigated. METHODS: In the present study, C57BL/6 J transgenic (Thy1-YFP-H) mice were subjected to CRS to establish an animal model of depression. The infralimbic (IL) of mPFC was selected as the injection site of retrograde AAV using stereotactic technique. The effects of CRS on dendrites/dendritic spines and afferent neurons of the mPFC IL were investigaed by quantitatively assessing the distribution density of green fluorescent (YFP) positive dendrites/dendritic spines and red fluorescent (retrograde AAV recombinant protein) positive neurons, respectively. RESULTS: The results revealed that retrograde tracing virus labeled neurons were widely distributed in ipsilateral and contralateral cingulate cortex (Cg1), second cingulate cortex (Cg2), prelimbic cortex (PrL), infralimbic cortex, medial orbital cortex (MO), and dorsal peduncular cortex (DP). The effects of CRS on the distribution density of mPFC red fluorescence positive neurons exhibited regional differences, ranging from rostral to caudal or from top to bottom. Simultaneously, CRS resulted a decrease in the distribution density of basal, proximal and distal dendrites, as well as an increase in the loss of dendritic spines of the distal dendrites in the IL of mPFC. Furthermore, varying degrees of red retrograde tracing virus fluorescence signals were observed in other cortices, amygdala, hippocampus, septum/basal forebrain, hypothalamus, thalamus, mesencephalon, and brainstem in both ipsilateral and contralateral brain. CRS significantly reduced the distribution density of red fluorescence positive neurons in other cortices, hippocampus, septum/basal forebrain, hypothalamus, and thalamus. Conversely, CRS significantly increased the distribution density of red fluorescence positive neurons in amygdala. CONCLUSION: Our results suggest a possible mechanism that CRS leads to disturbances in synaptic plasticity by affecting multiple inputs to the mPFC, which is characterized by a decrease in the distribution density of dendrites/dendritic spines in the IL of mPFC and a reduction in input neurons of multiple cortices to the IL of mPFC as well as an increase in input neurons of amygdala to the IL of mPFC, ultimately causing depression-like behaviors.


Subject(s)
Depression , Mice, Inbred C57BL , Mice, Transgenic , Prefrontal Cortex , Restraint, Physical , Stress, Psychological , Animals , Prefrontal Cortex/pathology , Prefrontal Cortex/metabolism , Stress, Psychological/pathology , Stress, Psychological/metabolism , Mice , Depression/pathology , Male , Dendritic Spines/pathology , Disease Models, Animal , Afferent Pathways , Dendrites/pathology , Dendrites/metabolism , Neurons, Afferent/pathology , Neurons, Afferent/metabolism , Brain/pathology , Brain/metabolism
2.
J Biol Chem ; 300(5): 107237, 2024 May.
Article in English | MEDLINE | ID: mdl-38552740

ABSTRACT

Tauopathies are neurodegenerative disorders characterized by the deposition of aggregates of the microtubule-associated protein tau, a main component of neurofibrillary tangles. Alzheimer's disease (AD) is the most common type of tauopathy and dementia, with amyloid-beta pathology as an additional hallmark feature of the disease. Besides its role in stabilizing microtubules, tau is localized at postsynaptic sites and can regulate synaptic plasticity. The activity-regulated cytoskeleton-associated protein (Arc) is an immediate early gene that plays a key role in synaptic plasticity, learning, and memory. Arc has been implicated in AD pathogenesis and regulates the release of amyloid-beta. We found that decreased Arc levels correlate with AD status and disease severity. Importantly, Arc protein was upregulated in the hippocampus of Tau KO mice and dendrites of Tau KO primary hippocampal neurons. Overexpression of tau decreased Arc stability in an activity-dependent manner, exclusively in neuronal dendrites, which was coupled to an increase in the expression of dendritic and somatic surface GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. The tau-dependent decrease in Arc was found to be proteasome-sensitive, yet independent of Arc ubiquitination and required the endophilin-binding domain of Arc. Importantly, these effects on Arc stability and GluA1 localization were not observed in the commonly studied tau mutant, P301L. These observations provide a potential molecular basis for synaptic dysfunction mediated through the accumulation of tau in dendrites. Our findings confirm that Arc is misregulated in AD and further show a physiological role for tau in regulating Arc stability and AMPA receptor targeting.


Subject(s)
Cytoskeletal Proteins , Dendrites , Nerve Tissue Proteins , Proteasome Endopeptidase Complex , tau Proteins , Animals , Humans , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Dendrites/metabolism , Dendrites/pathology , Hippocampus/metabolism , Hippocampus/pathology , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , tau Proteins/metabolism , tau Proteins/genetics , Ubiquitin/metabolism , Ubiquitination
3.
Cell Rep ; 42(12): 113573, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38096054

ABSTRACT

Huntington's disease (HD) usually causes cognitive disorders, including learning difficulties, that emerge before motor symptoms. Mutations related to lysosomal trafficking are linked to the pathogenesis of neurological diseases, whereas the cellular mechanisms remain elusive. Here, we discover a reduction in the dendritic density of lysosomes in the hippocampus that correlates with deficits in synaptic plasticity and spatial learning in early CAG-140 HD model mice. We directly manipulate intraneuronal lysosomal positioning with light-induced CRY2:CIB1 dimerization and demonstrate that lysosomal abundance in dendrites positively modulates long-term potentiation of glutamatergic synapses onto the neuron. This modulation depends on lysosomal Ca2+ release, which further promotes endoplasmic reticulum (ER) entry into spines. Importantly, optogenetically restoring lysosomal density in dendrites rescues the synaptic plasticity deficit in hippocampal slices of CAG-140 mice. Our data reveal dendritic lysosomal density as a modulator of synaptic plasticity and suggest a role of lysosomal mispositioning in cognitive decline in HD.


Subject(s)
Huntington Disease , Mice , Animals , Huntington Disease/genetics , Neuronal Plasticity/physiology , Neurons/pathology , Hippocampus/pathology , Synapses/pathology , Lysosomes/pathology , Dendrites/pathology , Dendritic Spines/pathology
4.
J Comp Neurol ; 531(6): 663-680, 2023 04.
Article in English | MEDLINE | ID: mdl-36629001

ABSTRACT

Facial nerve injury in rats have been widely used to study functional and structural changes that occur in the injured motoneurons and other central nervous system structures related with sensorimotor processing. A decrease in long-term potentiation of hippocampal CA3-to-CA1 commissural synapse has recently been reported related to this peripheral injury. Additionally, it has been found increased corticosterone plasmatic levels, impairment in spatial memory consolidation, and hippocampal microglial activation in animals with facial nerve axotomy. In this work, we analyzed the neuronal morphology of hippocampal CA1 and CA3 pyramidal neurons in animals with either reversible or irreversible facial nerve injury. For this purpose, brain tissues of injured animals sacrificed at different postlesion times, were stained with the Golgi-Cox method and compared with control brains. It was found that both reversible and irreversible facial nerve injury-induced significant decreases in dendritic tree complexity, dendritic length, branch points, and spine density of hippocampal neurons. However, such changes' timing varied according to hippocampal area (CA1 vs. CA3), dendritic area (apical vs. basal), and lesion type (reversible vs. irreversible). In general, the observed changes were transient when animals had the possibility of motor recovery (reversible injury), but perdurable if the recovery from the lesion was impeded (irreversible injury). CA1 apical and CA3 basal dendritic tree morphology were more sensible to irreversible injury. It is concluded that facial nerve injury induced significant changes in hippocampal CA1 and CA3 pyramidal neurons morphology, which could be related to LTP impairments and microglial activation in the hippocampal formation, previously described.


Subject(s)
Facial Nerve Injuries , Rats , Animals , Facial Nerve Injuries/pathology , Facial Nerve , Axotomy , Pyramidal Cells/physiology , Hippocampus/physiology , Motor Neurons , Dendrites/pathology
5.
J Cell Biol ; 222(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36547519

ABSTRACT

Disruptions in membrane trafficking are associated with neurodevelopmental disorders, but underlying pathological mechanisms remain largely unknown. In this issue, O'Brien et al. (2023. J. Cell Biol.https://doi.org/10.1083/jcb.202112108) show how GARP regulates sterol transfer critical for remodeling of dendrites in flies.


Subject(s)
Dendrites , Membrane Proteins , Neurodevelopmental Disorders , Sterols , Dendrites/pathology , Membranes , Neurodevelopmental Disorders/physiopathology , Sterols/metabolism , Membrane Proteins/metabolism
6.
Hum Vaccin Immunother ; 18(6): 2121568, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36113067

ABSTRACT

Bacillus Calmette - Guerin (BCG) is an immune regulator that can enhance hippocampal synaptic plasticity in rats; however, it is unclear whether it can improve synaptic function in a mouse model with Alzheimer's disease (AD). We hypothesized that BCG plays a protective role in AD mice and investigated its effect on dendritic morphology. The results obtained show that BCG immunization significantly increases dendritic complexity, as indicated by the increased number of dendritic intersections and branch points, as well as the increase in the fractal dimension. Furthermore, the number of primary neurites and dendritic length also increased following BCG immunization, which increased the number of spines and promoted maturation. IFN-γ and IL-4 levels increased, while TNF-α levels decreased following BCG immunization; expression levels of p-JAK2, P-STAT3, SYN, and PSD-95 also increased. Therefore, this study demonstrates that BCG immunization in APP/PS1 mice mitigated hippocampal dendritic spine pathology, especially after the third round of immunization. This effect could possibly be attributed to; changes in dendritic arborization and spine morphology or increases in SYN and PSD-95 expression levels. It could also be related to mechanisms of BCG-induced increases in IFN-γ or IL-4/JAK2/STAT3 levels.


BCG immunization in a mouse model for Alzheimer's disease significantly increased dendritic complexity, as indicated by an increase in the number of dendritic intersections and branch points, as well as an increase in the fractal dimension of hippocampal CA1 neurons.


Subject(s)
Alzheimer Disease , BCG Vaccine , Dendrites , Animals , Mice , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Dendritic Spines/immunology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Hippocampus/metabolism , Interleukin-4/metabolism , Mice, Transgenic , BCG Vaccine/therapeutic use , Dendrites/immunology , Dendrites/metabolism , Dendrites/pathology , Tumor Necrosis Factor-alpha/metabolism
7.
Mol Neurobiol ; 59(5): 2962-2976, 2022 May.
Article in English | MEDLINE | ID: mdl-35249200

ABSTRACT

Amyotrophic lateral sclerosis (ALS) attacks the corticomotor system, with motor cortex function affected early in disease. Younger females have a lower relative risk of succumbing to ALS than males and older females, implicating a role for female sex hormones in disease progression. However, the mechanisms driving this dimorphic incidence are still largely unknown. We endeavoured to determine if estrogen mitigates disease progression and pathogenesis, focussing upon the dendritic spine as a site of action. Using two-photon live imaging we identify, in the prpTDP-43A315T mouse model of ALS, that dendritic spines in the male motor cortex have a reduced capacity for remodelling than their wild-type controls. In contrast, females show higher capacity for remodelling, with peak plasticity corresponding to highest estrogen levels during the estrous cycle. Estrogen manipulation through ovariectomies and estrogen replacement with 17ß estradiol in vivo was found to significantly alter spine density and mitigate disease severity. Collectively, these findings reveal that synpatic plasticity is reduced in ALS, which can be amelioriated with estrogen, in conjuction with improved disease outcomes.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/pathology , Animals , Dendrites/pathology , Disease Models, Animal , Disease Progression , Estrogens/pharmacology , Female , Male , Mice , Mice, Transgenic , Neuronal Plasticity
8.
J Biol Chem ; 298(3): 101614, 2022 03.
Article in English | MEDLINE | ID: mdl-35101447

ABSTRACT

Microbial infections have been linked to the onset and severity of neurodegenerative diseases such as amyotrophic lateral sclerosis, multiple sclerosis, Alzheimer's disease, but the underlying mechanisms remain largely unknown. Here, we used a genetic screen for genes involved in protection from infection-associated neurodegeneration and identified the gene mtm-10. We then validated the role of the encoded myotubularin-related protein, MTM-10, in protecting the dendrites of Caenorhabditis elegans from degeneration mediated by oxidative stress or Pseudomonas aeruginosa infection. Further experiments indicated that mtm-10 is expressed in the AWC neurons of C. elegans, where it functions in a cell-autonomous manner to protect the dendrite degeneration caused by pathogen infection. We also confirm that the changes observed in the dendrites of the animals were not because of premature death or overall sickness. Finally, our studies indicated that mtm-10 functions in AWC neurons to preserve chemosensation after pathogen infection. These results reveal an essential role for myotubularin-related protein 10 in the protection of dendrite morphology and function against the deleterious effects of oxidative stress or infection.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Neurodegenerative Diseases , Neurons , Protein Tyrosine Phosphatases, Non-Receptor , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dendrites/metabolism , Dendrites/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism
9.
Sci Rep ; 11(1): 22568, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799629

ABSTRACT

WDR45 plays an essential role in the early stage of autophagy. De novo heterozygous mutations in WDR45 have been known to cause ß-propeller protein-associated neurodegeneration (BPAN), a subtype of neurodegeneration with brain iron accumulation (NBIA). Although BPAN patients display global developmental delay with intellectual disability, the neurodevelopmental pathophysiology of BPAN remains largely unknown. In the present study, we analyzed the physiological role of Wdr45 and pathophysiological significance of the gene abnormality during mouse brain development. Morphological and biochemical analyses revealed that Wdr45 is expressed in a developmental stage-dependent manner in mouse brain. Wdr45 was also found to be located in excitatory synapses by biochemical fractionation. Since WDR45 mutations are thought to cause protein degradation, we conducted acute knockdown experiments by in utero electroporation in mice to recapitulate the pathophysiological conditions of BPAN. Knockdown of Wdr45 caused abnormal dendritic development and synaptogenesis during corticogenesis, both of which were significantly rescued by co-expression with RNAi-resistant version of Wdr45. In addition, terminal arbors of callosal axons were less developed in Wdr45-deficient cortical neurons of adult mouse when compared to control cells. These results strongly suggest a pathophysiological significance of WDR45 gene abnormalities in neurodevelopmental aspects of BPAN.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Iron Metabolism Disorders/metabolism , Nerve Degeneration , Neuroaxonal Dystrophies/metabolism , Neurogenesis , Animals , Axons/metabolism , Axons/pathology , Brain/embryology , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , Dendrites/metabolism , Dendrites/pathology , Electrical Synapses/metabolism , Electrical Synapses/pathology , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Gestational Age , Iron Metabolism Disorders/embryology , Iron Metabolism Disorders/genetics , Iron Metabolism Disorders/pathology , Mice, Inbred ICR , Neuroaxonal Dystrophies/embryology , Neuroaxonal Dystrophies/genetics , Neuroaxonal Dystrophies/pathology , Signal Transduction
10.
Nature ; 599(7886): 650-656, 2021 11.
Article in English | MEDLINE | ID: mdl-34732887

ABSTRACT

Loss of functional mitochondrial complex I (MCI) in the dopaminergic neurons of the substantia nigra is a hallmark of Parkinson's disease1. Yet, whether this change contributes to Parkinson's disease pathogenesis is unclear2. Here we used intersectional genetics to disrupt the function of MCI in mouse dopaminergic neurons. Disruption of MCI induced a Warburg-like shift in metabolism that enabled neuronal survival, but triggered a progressive loss of the dopaminergic phenotype that was first evident in nigrostriatal axons. This axonal deficit was accompanied by motor learning and fine motor deficits, but not by clear levodopa-responsive parkinsonism-which emerged only after the later loss of dopamine release in the substantia nigra. Thus, MCI dysfunction alone is sufficient to cause progressive, human-like parkinsonism in which the loss of nigral dopamine release makes a critical contribution to motor dysfunction, contrary to the current Parkinson's disease paradigm3,4.


Subject(s)
Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Cell Death , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Disease Progression , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , Levodopa/pharmacology , Levodopa/therapeutic use , Male , Mice , Motor Skills/drug effects , NADH Dehydrogenase/deficiency , NADH Dehydrogenase/genetics , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Phenotype , Substantia Nigra/cytology , Substantia Nigra/drug effects , Substantia Nigra/metabolism
11.
J Clin Invest ; 131(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34651584

ABSTRACT

CDKL5 deficiency disorder (CDD) is an early onset, neurodevelopmental syndrome associated with pathogenic variants in the X-linked gene encoding cyclin-dependent kinase-like 5 (CDKL5). CDKL5 has been implicated in neuronal synapse maturation, yet its postdevelopmental necessity and the reversibility of CDD-associated impairments remain unknown. We temporally manipulated endogenous Cdkl5 expression in male mice and found that postdevelopmental loss of CDKL5 disrupts numerous behavioral domains, hippocampal circuit communication, and dendritic spine morphology, demonstrating an indispensable role for CDKL5 in the adult brain. Accordingly, restoration of Cdkl5 after the early stages of brain development using a conditional rescue mouse model ameliorated CDD-related behavioral impairments and aberrant NMDA receptor signaling. These findings highlight the requirement of CDKL5 beyond early development, underscore the potential for disease reversal in CDD, and suggest that a broad therapeutic time window exists for potential treatment of CDD-related deficits.


Subject(s)
Epileptic Syndromes/psychology , Protein Serine-Threonine Kinases/physiology , Spasms, Infantile/psychology , Animals , Dendrites/pathology , Evoked Potentials/physiology , Mice , Mice, Inbred C57BL , Receptors, N-Methyl-D-Aspartate/physiology
12.
Nat Rev Neurosci ; 22(11): 685-702, 2021 11.
Article in English | MEDLINE | ID: mdl-34599308

ABSTRACT

The sympathetic nervous system prepares the body for 'fight or flight' responses and maintains homeostasis during daily activities such as exercise, eating a meal or regulation of body temperature. Sympathetic regulation of bodily functions requires the establishment and refinement of anatomically and functionally precise connections between postganglionic sympathetic neurons and peripheral organs distributed widely throughout the body. Mechanistic studies of key events in the formation of postganglionic sympathetic neurons during embryonic and early postnatal life, including axon growth, target innervation, neuron survival, and dendrite growth and synapse formation, have advanced the understanding of how neuronal development is shaped by interactions with peripheral tissues and organs. Recent progress has also been made in identifying how the cellular and molecular diversity of sympathetic neurons is established to meet the functional demands of peripheral organs. In this Review, we summarize current knowledge of signalling pathways underlying the development of the sympathetic nervous system. These findings have implications for unravelling the contribution of sympathetic dysfunction stemming, in part, from developmental perturbations to the pathophysiology of peripheral neuropathies and cardiovascular and metabolic disorders.


Subject(s)
Axons/physiology , Dendrites/physiology , Neurons/physiology , Peripheral Nervous System Diseases/physiopathology , Sympathetic Nervous System/growth & development , Sympathetic Nervous System/physiopathology , Animals , Axons/pathology , Dendrites/pathology , Humans , Neuronal Plasticity/physiology , Neurons/pathology , Peripheral Nervous System Diseases/pathology , Sympathetic Nervous System/cytology
13.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34360985

ABSTRACT

Neurodevelopmental disorders can derive from a complex combination of genetic variation and environmental pressures on key developmental processes. Despite this complex aetiology, and the equally complex array of syndromes and conditions diagnosed under the heading of neurodevelopmental disorder, there are parallels in the neuropathology of these conditions that suggest overlapping mechanisms of cellular injury and dysfunction. Neuronal arborisation is a process of dendrite and axon extension that is essential for the connectivity between neurons that underlies normal brain function. Disrupted arborisation and synapse formation are commonly reported in neurodevelopmental disorders. Here, we summarise the evidence for disrupted neuronal arborisation in these conditions, focusing primarily on the cortex and hippocampus. In addition, we explore the developmentally specific mechanisms by which neuronal arborisation is regulated. Finally, we discuss key regulators of neuronal arborisation that could link to neurodevelopmental disease and the potential for pharmacological modification of arborisation and the formation of synaptic connections that may provide therapeutic benefit in the future.


Subject(s)
Brain/growth & development , Neurodevelopmental Disorders/pathology , Neuronal Outgrowth , Animals , Brain/physiopathology , Dendrites/metabolism , Dendrites/pathology , Humans , Neurodevelopmental Disorders/drug therapy , Neurodevelopmental Disorders/physiopathology , Neuroprotective Agents/therapeutic use
14.
J Neurosci ; 41(39): 8111-8125, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34400520

ABSTRACT

The size and structure of the dendritic arbor play important roles in determining how synaptic inputs of neurons are converted to action potential output. The regulatory mechanisms governing the development of dendrites, however, are insufficiently understood. The evolutionary conserved Ste20/Hippo kinase pathway has been proposed to play an important role in regulating the formation and maintenance of dendritic architecture. A key element of this pathway, Ste20-like kinase (SLK), regulates cytoskeletal dynamics in non-neuronal cells and is strongly expressed throughout neuronal development. However, its function in neurons is unknown. We show that, during development of mouse cortical neurons, SLK has a surprisingly specific role for proper elaboration of higher, ≥ third-order dendrites both in male and in female mice. Moreover, we demonstrate that SLK is required to maintain excitation-inhibition balance. Specifically, SLK knockdown caused a selective loss of inhibitory synapses and functional inhibition after postnatal day 15, whereas excitatory neurotransmission was unaffected. Finally, we show that this mechanism may be relevant for human disease, as dysmorphic neurons within human cortical malformations revealed significant loss of SLK expression. Overall, the present data identify SLK as a key regulator of both dendritic complexity during development and inhibitory synapse maintenance.SIGNIFICANCE STATEMENT We show that dysmorphic neurons of human epileptogenic brain lesions have decreased levels of the Ste20-like kinase (SLK). Decreasing SLK expression in mouse neurons revealed that SLK has essential functions in forming the neuronal dendritic tree and in maintaining inhibitory connections with neighboring neurons.


Subject(s)
Cerebral Cortex/metabolism , Dendrites/genetics , Neural Inhibition/genetics , Protein Serine-Threonine Kinases/genetics , Synapses/genetics , Synaptic Transmission/physiology , Adolescent , Adult , Aged , Animals , Cerebral Cortex/pathology , Child , Child, Preschool , Dendrites/metabolism , Dendrites/pathology , Female , HEK293 Cells , Humans , Male , Mice , Middle Aged , Protein Serine-Threonine Kinases/metabolism , Synapses/metabolism , Synapses/pathology , Young Adult
15.
Cell Rep ; 36(1): 109315, 2021 07 06.
Article in English | MEDLINE | ID: mdl-34233200

ABSTRACT

Neurodevelopmental disorders are often caused by chromosomal microdeletions comprising numerous contiguous genes. A subset of neurofibromatosis type 1 (NF1) patients with severe developmental delays and intellectual disability harbors such a microdeletion event on chromosome 17q11.2, involving the NF1 gene and flanking regions (NF1 total gene deletion [NF1-TGD]). Using patient-derived human induced pluripotent stem cell (hiPSC)-forebrain cerebral organoids (hCOs), we identify both neural stem cell (NSC) proliferation and neuronal maturation abnormalities in NF1-TGD hCOs. While increased NSC proliferation results from decreased NF1/RAS regulation, the neuronal differentiation, survival, and maturation defects are caused by reduced cytokine receptor-like factor 3 (CRLF3) expression and impaired RhoA signaling. Furthermore, we demonstrate a higher autistic trait burden in NF1 patients harboring a deleterious germline mutation in the CRLF3 gene (c.1166T>C, p.Leu389Pro). Collectively, these findings identify a causative gene within the NF1-TGD locus responsible for hCO neuronal abnormalities and autism in children with NF1.


Subject(s)
Cerebrum/pathology , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Induced Pluripotent Stem Cells/pathology , Models, Biological , Neurogenesis/genetics , Organoids/pathology , Receptors, Cytokine/metabolism , Autistic Disorder/genetics , Cell Line , Cell Proliferation , Dendrites/metabolism , Dendrites/pathology , Enzyme Activation , Gene Deletion , Genes, Neurofibromatosis 1 , Humans , Mutation/genetics , Signal Transduction , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
16.
Mol Neurodegener ; 16(1): 43, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34187514

ABSTRACT

BACKGROUND: The maintenance of complex dendritic arbors and synaptic transmission are processes that require a substantial amount of energy. Bioenergetic decline is a prominent feature of chronic neurodegenerative diseases, yet the signaling mechanisms that link energy stress with neuronal dysfunction are poorly understood. Recent work has implicated energy deficits in glaucoma, and retinal ganglion cell (RGC) dendritic pathology and synapse disassembly are key features of ocular hypertension damage. RESULTS: We show that adenosine monophosphate-activated protein kinase (AMPK), a conserved energy biosensor, is strongly activated in RGC from mice with ocular hypertension and patients with primary open angle glaucoma. Our data demonstrate that AMPK triggers RGC dendrite retraction and synapse elimination. We show that the harmful effect of AMPK is exerted through inhibition of the mammalian target of rapamycin complex 1 (mTORC1). Attenuation of AMPK activity restores mTORC1 function and rescues dendrites and synaptic contacts. Strikingly, AMPK depletion promotes recovery of light-evoked retinal responses, improves axonal transport, and extends RGC survival. CONCLUSIONS: This study identifies AMPK as a critical nexus between bioenergetic decline and RGC dysfunction during pressure-induced stress, and highlights the importance of targeting energy homeostasis in glaucoma and other neurodegenerative diseases.


Subject(s)
Adenylate Kinase/metabolism , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Animals , Dendrites/pathology , Enzyme Activation/physiology , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Synapses/pathology
17.
Cell Rep ; 35(5): 109080, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33951432

ABSTRACT

Although an imbalance between neuronal excitation and inhibition underlies seizures, clinical approaches that target these mechanisms are insufficient in containing seizures in patients with epilepsy, raising the need for alternative approaches. Brain-resident microglia contribute to the development and stability of neuronal structure and functional networks that are perturbed during seizures. However, the extent of microglial contributions in response to seizures in vivo remain to be elucidated. Using two-photon in vivo imaging to visualize microglial dynamics, we show that severe seizures induce formation of microglial process pouches that target but rarely engulf beaded neuronal dendrites. Microglial process pouches are stable for hours, although they often shrink in size. We further find that microglial process pouches are associated with a better structural resolution of beaded dendrites. These findings provide evidence for the structural resolution of injured dendrites by microglia as a form of neuroprotection.


Subject(s)
Dendrites/pathology , Microglia/ultrastructure , Seizures/physiopathology , Animals , Humans , Mice
18.
J Mol Neurosci ; 71(9): 1849-1862, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34041687

ABSTRACT

A decline of estrogen level leads to spatial learning and memory impairments, which mediated by hippocampus and cortex. Accumulating evidences demonstrated that aerobic exercise improved memory of postmenopausal women and ovariectomized (OVX) mice. However, the molecular mechanisms for this protection of exercise are not completely clear. Accordingly, the present study was designed to examine the effect of aerobic exercise on the dendritic morphology in the hippocampus and cerebral cortex, as well as the BNDF-mTOR signaling pathway of OVX mice. Adult female C57BL/6 mice were divided into four groups (n = 10/group): sham-operated (SHAM/CON), sham-operated with 8-week treadmill exercise (SHAM/EX), ovariectomized operated (OVX/CON), and ovariectomized operated with exercise (OVX/EX). Aerobic exercise improved the impairment of dendritic morphology significantly induced by OVX that was tested by Golgi staining, and it also upregulated the synaptic plasticity-related protein expression of PSD95 and GluR1 as well as activated BDNF-mTOR signaling pathway in the hippocampus and cerebral cortex. In conclusion, aerobic exercise reversed the change of dendritic morphology and increased the synaptic plasticity-related protein expression in the hippocampus and cerebral cortex of OVX mice. The positive effects induced by exercise might be mediated through the BDNF-mTOR signaling pathway.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Dendrites/pathology , Estrogens/deficiency , Hippocampus/metabolism , Running , TOR Serine-Threonine Kinases/metabolism , Animals , Cerebral Cortex/physiology , Dendrites/metabolism , Dendrites/physiology , Estrogens/metabolism , Female , Hippocampus/physiology , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Ovariectomy/adverse effects , Physical Conditioning, Animal , Signal Transduction
19.
J Neurovirol ; 27(3): 403-421, 2021 06.
Article in English | MEDLINE | ID: mdl-34003469

ABSTRACT

HIV-1 infection affects approximately 37 million individuals, and approximately 50% of seropositive individuals will develop symptoms of clinical depression and/or apathy. Dysfunctions of both serotonergic and dopaminergic neurotransmission have been implicated in the pathogenesis of motivational alterations. The present study evaluated the efficacy of a SSRI (escitalopram) in the HIV-1 transgenic (Tg) rat. Behavioral, neurochemical, and neuroanatomical outcomes with respect to HIV-1 and sex were evaluated to determine the efficacy of chronic escitalopram treatment. Escitalopram treatment restored function in each of the behavioral tasks that were sensitive to HIV-1-induced impairments. Further, escitalopram treatment restored HIV-1-mediated synaptodendritic damage in the nucleus accumbens; treatment with escitalopram significantly increased dendritic proliferation in HIV-1 Tg rats. However, restoration did not consistently occur with the neurochemical analysis in the HIV-1 rat. Taken together, these results suggest a role for SSRI therapies in repairing long-term HIV-1 protein-mediated neuronal damage and restoring function.


Subject(s)
Antidepressive Agents/pharmacology , Apathy/drug effects , Depression/drug therapy , Escitalopram/pharmacology , HIV Infections/drug therapy , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Choice Behavior/drug effects , Dendrites/drug effects , Dendrites/pathology , Dendrites/virology , Depression/complications , Depression/physiopathology , Depression/virology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dopaminergic Neurons/virology , Female , HIV Infections/complications , HIV Infections/physiopathology , HIV Infections/virology , HIV-1/growth & development , HIV-1/pathogenicity , Humans , Male , Maze Learning/drug effects , Nucleus Accumbens/drug effects , Nucleus Accumbens/pathology , Nucleus Accumbens/virology , Rats , Rats, Transgenic , Serotonergic Neurons/drug effects , Serotonergic Neurons/pathology , Serotonergic Neurons/virology , Synapses/drug effects , Synapses/pathology , Synapses/virology , Synaptic Transmission/drug effects , Treatment Outcome
20.
Sci Rep ; 11(1): 7615, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33828151

ABSTRACT

Modeling long-term neuronal dynamics may require running long-lasting simulations. Such simulations are computationally expensive, and therefore it is advantageous to use simplified models that sufficiently reproduce the real neuronal properties. Reducing the complexity of the neuronal dendritic tree is one option. Therefore, we have developed a new reduced-morphology model of the rat CA1 pyramidal cell which retains major dendritic branch classes. To validate our model with experimental data, we used HippoUnit, a recently established standardized test suite for CA1 pyramidal cell models. The HippoUnit allowed us to systematically evaluate the somatic and dendritic properties of the model and compare them to models publicly available in the ModelDB database. Our model reproduced (1) somatic spiking properties, (2) somatic depolarization block, (3) EPSP attenuation, (4) action potential backpropagation, and (5) synaptic integration at oblique dendrites of CA1 neurons. The overall performance of the model in these tests achieved higher biological accuracy compared to other tested models. We conclude that, due to its realistic biophysics and low morphological complexity, our model captures key physiological features of CA1 pyramidal neurons and shortens computational time, respectively. Thus, the validated reduced-morphology model can be used for computationally demanding simulations as a substitute for more complex models.


Subject(s)
CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiology , Dendrites/physiology , Action Potentials/physiology , Animals , Computer Simulation , Databases, Factual , Dendrites/pathology , Hippocampus/physiology , Models, Neurological , Neuronal Plasticity/physiology , Neurons/metabolism , Pyramidal Cells/physiology , Rats , Synapses/physiology , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...