Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Nat Methods ; 17(2): 232-239, 2020 02.
Article in English | MEDLINE | ID: mdl-31768061

ABSTRACT

Autophagy is a degradative program that maintains cellular homeostasis. Autophagy defects have been described in numerous diseases. However, analysis of autophagy rates can be challenging, particularly in rare cell populations or in vivo, due to limitations in currently available tools for measuring autophagy induction. Here, we describe a method to monitor autophagy by measuring phosphorylation of the protein ATG16L1. We developed and characterized a monoclonal antibody that can detect phospho-ATG16L1 endogenously in mammalian cells. Importantly, phospho-ATG16L1 is only present on newly forming autophagosomes. Therefore, its levels are not affected by prolonged stress or late-stage autophagy blocks, which can confound autophagy analysis. Moreover, we show that ATG16L1 phosphorylation is a conserved signaling pathway activated by numerous autophagy-inducing stressors. The described antibody is suitable for western blot, immunofluorescence and immunohistochemistry, and measured phospho-ATG16L1 levels directly correspond to autophagy rates. Taken together, this phospho-antibody represents an exciting tool to study autophagy induction.


Subject(s)
Antibodies/immunology , Autophagy , Animals , Carrier Proteins/metabolism , Humans , Phosphorylation
2.
J Neurosci ; 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34083257

ABSTRACT

Within the perinatal stroke field, there is a need to establish preclinical models where putative biomarkers for motor function can be examined. In a mouse model of perinatal stroke, we evaluated motor map size and movement latency following optogenetic cortical stimulation against three factors of post-stroke biomarker utility: 1) Correlation to chronic impairment on a behavioral test battery; 2) Amenability to change using a skilled motor training paradigm; 3) Ability to distinguish individuals with potential to respond well to training. Thy1-ChR2-YFP mice received a photothrombotic stroke at postnatal day 7 and were evaluated on a battery of motor tests between days 59-70. Following a cranial window implant, mice underwent longitudinal optogenetic motor mapping both before and after 3 weeks of skilled forelimb training. Map size and movement latency of both hemispheres was positively correlated with impaired spontaneous forelimb use, whereas only ipsilesional hemisphere map size was correlated with performance in skilled reaching. Map size and movement latency did not show groupwise changes with training; however, mice with the smallest pre-training map sizes and worst impairments demonstrated the greatest expansion of map size in response to skilled forelimb training. Overall, motor map size showed utility as a potential biomarker for impairment and training-induced modulation in specific individuals. Future assessment of the predictive capacity of post-stroke motor representations for behavioral outcome in animal models opens the possibility of dissecting how plasticity mechanisms contribute to recovery following perinatal stroke.SIGNIFICANCE STATEMENTWe investigated the utility of two cortical motor representation measures (motor map size and movement onset latency) as potential biomarkers for post-stroke motor recovery in a mouse model of perinatal stroke. Both motor map size and movement latency were associated with functional recovery after perinatal stroke, with map size showing an additional association between training responsiveness and severity of impairment. Overall, both motor map size and movement onset latency show potential as neurophysiological correlates of recovery. As such, future studies of perinatal stroke rehabilitation and neuromodulation should include these measures in order to help explain neurophysiological changes that might be occurring in response to treatment.

3.
Hum Mol Genet ; 26(17): 3327-3341, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28595361

ABSTRACT

Mitochondrial dysfunction is a common feature of many genetic disorders that target the brain and cognition. However, the exact role these organelles play in the etiology of such disorders is not understood. Here, we show that mitochondrial dysfunction impairs brain development, depletes the adult neural stem cell (NSC) pool and impacts embryonic and adult neurogenesis. Using deletion of the mitochondrial oxidoreductase AIF as a genetic model of mitochondrial and neurodegenerative diseases revealed the importance of mitochondria in multiple steps of the neurogenic process. Developmentally, impaired mitochondrial function causes defects in NSC self-renewal, neural progenitor cell proliferation and cell cycle exit, as well as neuronal differentiation. Sustained mitochondrial dysfunction into adulthood leads to NSC depletion, loss of adult neurogenesis and manifests as a decline in brain function and cognitive impairment. These data demonstrate that mitochondrial dysfunction, as observed in genetic mitochondrial and neurodegenerative diseases, underlies the decline of brain function and cognition due to impaired stem cell maintenance and neurogenesis.


Subject(s)
Mitochondria/metabolism , Mitochondria/physiology , Neural Stem Cells/metabolism , Animals , Apoptosis Inducing Factor/metabolism , Brain/metabolism , Cell Differentiation , Cell Proliferation , Cognition , Cognitive Dysfunction/metabolism , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/metabolism , Signal Transduction
4.
Am J Physiol Heart Circ Physiol ; 315(5): H1159-H1173, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30052050

ABSTRACT

Patients with heart failure (HF) have a high prevalence of depression associated with a worse prognosis, particularly in older women. The present study evaluated whether sex and estrogens affect depression-like behavior and associated neuroinflammation induced by myocardial infarction (MI) in rats. MI was induced by occlusion of the left anterior descending artery in young adult male and female Wistar rats or in ovariectomized (OVX) female rats without and with estrogen [17ß-estradiol (E2)] replacement. MI groups showed a comparable degree of cardiac dysfunction. Eight weeks post-MI, male rats with HF exhibited depression-like behaviors, including anhedonia and higher immobility in the sucrose preference and forced swim tests, which were not observed in female rats with HF. In the cued fear conditioning test, male but not female rats with HF froze more than sham rats. After OVX, female sham rats developed mild depression-like behaviors that were pronounced in OVX female rats post-MI and were largely prevented by E2 replacement. Cytokine levels in the plasma and paraventricular nucleus increased in both sexes with HF, but only male rats with HF showed an increase in cytokine levels in the prefrontal cortex. OVX alone did not affect cytokine levels, but OVX-MI caused significant increases in the prefrontal cortex, which were shifted to an anti-inflammatory pattern by E2 replacement. These results suggest that estrogens prevent depression-like behavior induced by HF post-MI in young adult female rats by inhibiting proinflammatory cytokine production and actions in the prefrontal cortex. NEW & NOTEWORTHY In contrast to male rats, female rats with heart failure after myocardial infarction do not develop depression-like behavior or increases in prefrontal cortex cytokines. However, after ovariectomy, female rats exhibit similar changes, which are prevented by 17ß-estradiol replacement. Neuroinflammation in the prefrontal cortex in male subjects may contribute to depression-like behavior, whereas its estrogen-dependent absence in female subjects may protect against depression.


Subject(s)
Behavior, Animal/drug effects , Cytokines/metabolism , Depression/prevention & control , Encephalitis/prevention & control , Estradiol/administration & dosage , Inflammation Mediators/metabolism , Myocardial Infarction/complications , Ovariectomy , Prefrontal Cortex/drug effects , Animals , Conditioning, Psychological/drug effects , Depression/etiology , Depression/metabolism , Depression/psychology , Disease Models, Animal , Encephalitis/etiology , Encephalitis/metabolism , Encephalitis/physiopathology , Estrogen Replacement Therapy , Feeding Behavior/drug effects , Female , Male , Maze Learning/drug effects , Myocardial Infarction/physiopathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Rats, Wistar , Sex Factors , Ventricular Function, Left
5.
J Neurosci ; 36(4): 1203-10, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26818508

ABSTRACT

We demonstrated previously that Pannexin 1 (Panx1), an ion and metabolite channel, promotes the growth and proliferation of ventricular zone (VZ) neural precursor cells (NPCs) in vitro. To investigate its role in vivo, we used floxed Panx1 mice in combination with viruses to delete Panx1 in VZ NPCs and to track numbers of Panx1-null and Panx1-expressing VZ NPCs over time. Two days after virus injection, Panx1-null cells were less abundant than Panx1-expressing cells, suggesting that Panx1 is required for the maintenance of VZ NPCs. We also investigated the effect of Panx1 deletion in VZ NPCs after focal cortical stroke via photothrombosis. Panx1 is essential for maintaining elevated VZ NPC numbers after stroke. In contrast, Panx1-null NPCs were more abundant than Panx1-expressing NPCs in the peri-infarct cortex. Together, these findings suggest that Panx1 plays an important role in NPC maintenance in the VZ niche in the naive and stroke brain and could be a key target for improving NPC survival in the peri-infarct cortex. SIGNIFICANCE STATEMENT: Here, we demonstrate that Pannexin 1 (Panx1) maintains a consistent population size of neural precursor cells in the ventricular zone, both in the healthy brain and in the context of stroke. In contrast, Panx1 appears to be detrimental to the survival of neural precursor cells that surround damaged cortical tissue in the stroke brain. This suggests that targeting Panx1 in the peri-infarct cortex, in combination with other therapies, could improve cell survival around the injury site.


Subject(s)
Cerebral Infarction/pathology , Cerebral Ventricles/cytology , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Neurogenesis/physiology , Analysis of Variance , Animals , Caspase 3/metabolism , Cell Count , Cell Survival/physiology , Connexins/genetics , Disease Models, Animal , Doublecortin Domain Proteins , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Ki-67 Antigen/metabolism , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Neuropeptides/metabolism , Stroke/complications
6.
Hum Mol Genet ; 24(16): 4573-83, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26002103

ABSTRACT

Defects in mitochondrial fission and cyclin dependent kinase 5 (CDK5) activation are early events that precede neuronal loss following NMDA-induced neuronal death. Here, we report that the cytoplasmic CDK5 tightly regulates mitochondrial morphology defects associated with NMDA-induced neuronal injury via regulation of the mitochondrial fission protein, dynamin-related protein 1 (DRP1). We show that DRP1 is a direct target of CDK5. CDK5-mediated phosphorylation of DRP1 at a conserved Serine residue, S585, is elevated at the mitochondria and is associated with increased mitochondrial fission. Ectopic expression of a cytoplasmic CDK5 or mutant DRP1-S585D results in increased mitochondrial fragmentation in primary neurons. Conversely, expression of a dominant negative form of cytoplasmic CDK5 or mutant DRP1-S585A results in elongated mitochondria. In addition, pharmacological inhibition of CDK5 by Roscovitine inhibits DRP1 phosphorylation and mitochondrial fission associated with NMDA-induced neuronal loss. Importantly, conditional deletion of CDK5 significantly attenuates DRP1 phosphorylation at S585 and rescues mitochondrial fission defects in neurons exposed to NMDA. Our studies delineate an important mechanism by which CDK5 regulates mitochondrial morphology defects associated with neuronal injury.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Dynamins/metabolism , Mitochondria/metabolism , N-Methylaspartate/toxicity , Neurons/metabolism , Amino Acid Substitution , Animals , Cell Death/drug effects , Cell Death/genetics , Cyclin-Dependent Kinase 5/genetics , Dynamins/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Mutation, Missense , Neurons/pathology , Phosphorylation
7.
J Neural Transm (Vienna) ; 124(6): 721-738, 2017 06.
Article in English | MEDLINE | ID: mdl-28477284

ABSTRACT

Braak and Del Tredici have proposed that typical Parkinson disease (PD) has its origins in the olfactory bulb and gastrointestinal tract. However, the role of the olfactory system has insufficiently been explored in the pathogeneses of PD and Alzheimer disease (AD) in laboratory models. Here, we demonstrate applications of a new method to process mouse heads for microscopy by sectioning, mounting, and staining whole skulls ('holocranohistochemistry'). This technique permits the visualization of the olfactory system from the nasal cavity to mitral cells and dopamine-producing interneurons of glomeruli in the olfactory bulb. We applied this method to two specific goals: first, to visualize PD- and AD-linked gene expression in the olfactory system, where we detected abundant, endogenous α-synuclein and tau expression in the olfactory epithelium. Furthermore, we observed amyloid-ß plaques and proteinase-K-resistant α-synuclein species, respectively, in cranial nerve-I of APP- and human SNCA-over-expressing mice. The second application of the technique was to the modeling of gene-environment interactions in the nasal cavity of mice. We tracked the infection of a neurotropic respiratory-enteric-orphan virus from the nose pad into cranial nerves-I (and -V) and monitored the ensuing brain infection. Given its abundance in the olfactory epithelia, we questioned whether α-synuclein played a role in innate host defenses to modify the outcome of infections. Indeed, Snca-null mice were more likely to succumb to viral encephalitis versus their wild-type littermates. Moreover, using a bacterial sepsis model, Snca-null mice were less able to control infection after intravenous inoculation with Salmonella typhimurium. Together, holocranohistochemistry enabled new discoveries related to α-synuclein expression and its function in mice. Future studies will address: the role of Mapt and mutant SNCA alleles in infection paradigms; the contribution of xenobiotics in the initiation of idiopathic PD; and the safety to the host when systemically targeting α-synuclein by immunotherapy.


Subject(s)
Brain/metabolism , Brain/virology , Encephalitis, Viral/virology , Olfactory Mucosa/anatomy & histology , Olfactory Mucosa/metabolism , Reoviridae Infections/virology , alpha-Synuclein/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/diagnostic imaging , Brain/pathology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/mortality , Encephalitis, Viral/pathology , Female , Head , Humans , Immunohistochemistry , Male , Mammalian orthoreovirus 3 , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/anatomy & histology , Neural Pathways/diagnostic imaging , Neural Pathways/metabolism , Neural Pathways/pathology , Olfactory Mucosa/pathology , Olfactory Receptor Neurons/metabolism , Olfactory Receptor Neurons/virology , Reoviridae Infections/immunology , Salmonella Infections/immunology , Salmonella Infections/pathology , Salmonella typhimurium , Tissue Preservation/methods , alpha-Synuclein/genetics
8.
Hippocampus ; 26(2): 211-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26266948

ABSTRACT

In the adult brain only a small proportion of the neural stem and progenitor cells (NPCs) and their progeny survive to become mature neurons in the hippocampus. Recent studies have elucidated the roles for members of the B-cell lymphoma-2 (Bcl-2) family of proteins in regulating the survival of NPCs and their progeny at different stages of maturation, yet the requirement of Bcl-2 during this process remains unknown. Here we report that inducible removal of Bcl-2 from nestin-expressing neural stem/progenitor cells and their progeny resulted in a reduction in the survival of doublecortin-expressing cells in the absence of changing the number of radial-glial stem cells or dividing NPCs. The requirement of Bcl-2 for the survival of maturing NPCs was confirmed by removal of Bcl-2 through infecting NPCs using a retroviral strategy that resulted in the complete loss of Bcl-2 null cells by 30-day post-viral injection. Furthermore, we observed that the function of Bcl-2 in the adult-generated neurons was dependent on the Bcl-2-associated X (BAX) protein, since Bcl-2 null NPCs were rescued in BAX knockout mice. These results indicate that Bcl-2 is an essential regulator in the survival of doublecortin-expressing immature neurons through a mechanism that is upstream of BAX.


Subject(s)
Microtubule-Associated Proteins/biosynthesis , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neuropeptides/biosynthesis , Proto-Oncogene Proteins c-bcl-2/deficiency , Animals , Doublecortin Domain Proteins , Female , Gene Expression Regulation , Male , Mice , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Neuropeptides/genetics , Proto-Oncogene Proteins c-bcl-2/genetics
9.
Hippocampus ; 26(11): 1379-1392, 2016 11.
Article in English | MEDLINE | ID: mdl-27325572

ABSTRACT

In mammals, hippocampal dentate gyrus granule cells (DGCs) constitute a particular neuronal population produced both during embryogenesis and adult life, and play key roles in neural plasticity and memory. However, the molecular mechanisms regulating neurogenesis in the dentate lineage throughout development and adulthood are still not well understood. The Retinoblastoma protein (RB), a transcriptional repressor primarily involved in cell cycle control and cell death, plays crucial roles during cortical development but its function in the formation and maintenance of DGCs remains unknown. Here, we show that loss of RB during embryogenesis induces massive ectopic proliferation and delayed cell cycle exit of young DGCs specifically at late developmental stages but without affecting stem cells. This phenotype was partially counterbalanced by increased cell death. Similarly, during adulthood, loss of RB causes ectopic proliferation of newborn DGCs and dramatically impairs their survival. These results demonstrate a crucial role for RB in the generation and the survival of DGCs in the embryonic and the adult brain. © 2016 Wiley Periodicals, Inc.


Subject(s)
Dentate Gyrus/cytology , Dentate Gyrus/embryology , Neurogenesis/genetics , Neurons/physiology , Retinoblastoma Protein/metabolism , Stem Cells/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/metabolism , Embryo, Mammalian , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin/genetics , Nestin/metabolism , Retinoblastoma Protein/genetics , SOXB1 Transcription Factors/metabolism
10.
Proc Natl Acad Sci U S A ; 109(39): 15918-23, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23019375

ABSTRACT

DJ-1 mutations cause autosomal recessive early-onset Parkinson disease (PD). We report a model of PD pathology: the DJ1-C57 mouse. A subset of DJ-1-nullizygous mice, when fully backcrossed to a C57BL/6 [corrected] background, display dramatic early-onset unilateral loss of dopaminergic (DA) neurons in their substantia nigra pars compacta, progressing to bilateral degeneration of the nigrostriatal axis with aging. In addition, these mice exhibit age-dependent bilateral degeneration at the locus ceruleus nucleus and display mild motor behavior deficits at aged time points. These findings effectively recapitulate the early stages of PD. Therefore, the DJ1-C57 mouse provides a tool to study the preclinical aspects of neurodegeneration. Importantly, by exome sequencing, we identify candidate modifying genes that segregate with the phenotype, providing potentially critical clues into how certain genes may influence the penetrance of DJ-1-related degeneration in mice.


Subject(s)
Dopaminergic Neurons/pathology , Intracellular Signaling Peptides and Proteins , Locus Coeruleus/pathology , Nerve Tissue Proteins , Oncogene Proteins , Parkinson Disease/genetics , Parkinson Disease/pathology , Substantia Nigra/pathology , Animals , Disease Models, Animal , Dopaminergic Neurons/metabolism , Humans , Locus Coeruleus/metabolism , Mice , Mice, Knockout , Parkinson Disease/metabolism , Substantia Nigra/metabolism
11.
J Neurosci ; 33(13): 5773-84, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23536090

ABSTRACT

Ubiquitous classical (typical) calpains, calpain-1 and calpain-2, are Ca(+2)-dependent cysteine proteases, which have been associated with numerous physiological and pathological cellular functions. However, a clear understanding of the role of calpains in the CNS has been hampered by the lack of appropriate deletion paradigms in the brain. In this study, we describe a unique model of conditional deletion of both calpain-1 and calpain-2 activities in mouse brain, which more definitively assesses the role of these ubiquitous proteases in brain development/function and pathology. Surprisingly, we show that these calpains are not critical for gross CNS development. However, calpain-1/calpain-2 loss leads to reduced dendritic branching complexity and spine density deficits associated with major deterioration in hippocampal long-term potentiation and spatial memory. Moreover, calpain-1/calpain-2-deficient neurons were significantly resistant to injury induced by excitotoxic stress or mitochondrial toxicity. Examination of downstream target showed that the conversion of the Cdk5 activator, p35, to pathogenic p25 form, occurred only in the presence of calpain and that it played a major role in calpain-mediated neuronal death. These findings unequivocally establish two central roles of calpain-1/calpain-2 in CNS function in plasticity and neuronal death.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/pathology , Brain , Calpain/deficiency , Long-Term Potentiation/physiology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Biophysics , Brain/embryology , Brain/growth & development , Brain/pathology , Brain Injuries/chemically induced , Brain Injuries/physiopathology , Bromodeoxyuridine/metabolism , Cell Death/drug effects , Cell Death/genetics , Dendrites/metabolism , Dendrites/pathology , Dendrites/ultrastructure , Disease Models, Animal , Electric Stimulation , Embryo, Mammalian , Evoked Potentials/drug effects , Evoked Potentials/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Female , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , In Vitro Techniques , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurons/cytology , Neurons/metabolism , Patch-Clamp Techniques , Phosphotransferases , Psychomotor Performance , RNA, Messenger/metabolism , Silver Staining , Transfection
12.
Hippocampus ; 24(9): 1120-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24753271

ABSTRACT

Hippocampal shrinkage is a commonly found neuroanatomical change in stress-related mood disorders such as depression and post-traumatic stress disorders (PTSD). Since the onset and severity of these disorders have been found to be closely related to stressful life events, and as stress alone has been shown to reduce hippocampal volume in animal studies, vulnerability to mood disorders may be related to a susceptibility to stress-induced hippocampal shrinkage. However, a smaller hippocampal volume before stress exposure has also been suggested to confer vulnerability of stressed individuals to PTSD or depression. In this study, we examined the contribution of either innate hippocampal volume differences or hippocampal susceptibility to stress-induced shrinkage to the formation of stress-related psychopathology using longitudinal MRI measurements of hippocampal volume in inbred C57 mice before and after chronic social defeat stress. We found that only half of the stressed C57 mice were susceptible to stress and developed psychopathological behaviors such as social avoidance. The other half was resilient to stress and exhibited no social avoidance. Before exposure to stress, we observed a positive correlation between hippocampal volume and social avoidance. After chronic social defeat stress, we found significant increases in left hippocampal volume in resilient and nonstressed control mice. Intriguingly, this increase in hippocampal volume was not found in susceptible mice, suggesting an arrestment of hippocampal growth in these mice. Our findings suggest that both a susceptibility to stress-induced hippocampal volume changes and a larger hippocampus before stress exposure confer vulnerability to psychopathology after chronic stress.


Subject(s)
Hippocampus/pathology , Resilience, Psychological , Stress, Psychological/pathology , Animals , Avoidance Learning , Body Weight , Chronic Disease , Disease Models, Animal , Dominance-Subordination , Functional Laterality , Longitudinal Studies , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Organ Size , Social Behavior , Stress, Psychological/psychology
13.
Dev Neurosci ; 36(1): 44-63, 2014.
Article in English | MEDLINE | ID: mdl-24576816

ABSTRACT

Growth-associated protein-43 (GAP-43) is a presynaptic protein that plays key roles in axonal growth and guidance and in modulating synapse formation. Previous work has demonstrated that mice lacking one allele of this gene (GAP-43+/- mice) exhibit hippocampal structural abnormalities, impaired spatial learning and stress-induced behavioral withdrawal and anxiety, behaviors that are dependent on proper hippocampal circuitry and function. Given the correlation between hippocampal function, synaptic connectivity and neurogenesis, we tested if behaviorally naïve GAP-43+/- mice had alterations in either neurogenesis or synaptic connectivity in the hippocampus during early postnatal development and young adulthood, and following behavior testing in older adults. To test our hypothesis, we examined hippocampal cell proliferation (Ki67), number of immature neuroblasts (doublecortin, DCX) and mossy fiber volume (synaptoporin) in behaviorally naïve postnatal day 9 (P9) and P26, and behaviorally experienced 5- to 7-month-old GAP-43+/- and +/+ littermate mice. P9 GAP-43+/- mice had fewer Ki67+ and DCX+ cells compared to +/+ mice, particularly in the posterior dentate gyrus, and smaller mossy fiber volume in the same region. In young adulthood, however, male GAP-43+/- mice had more Ki67+ and DCX+ cells and greater mossy fiber volume in the posterior dentate gyrus relative to male +/+ mice. These increases were not seen in females. In 5- to 7-month-old GAP-43+/- mice (whose behaviors were the focus of our prior publication), there was no global change in the number of proliferating or immature neurons relative to +/+ mice. However, more detailed analysis revealed fewer proliferative DCX+ cells in the anterior dentate gyrus of male GAP-43+/- mice compared to male +/+ mice. This reduction was not observed in females. These results suggest that young GAP-43+/- mice have decreased hippocampal neurogenesis and synaptic connectivity, but slightly older mice have greater hippocampal neurogenesis and synaptic connectivity. In conjunction with our previous study, these findings suggest that GAP-43 is dynamically involved in early postnatal and adult hippocampal neurogenesis and synaptic connectivity, possibly contributing to the GAP-43+/- behavioral phenotype.


Subject(s)
GAP-43 Protein/metabolism , Hippocampus/metabolism , Mossy Fibers, Hippocampal/metabolism , Neurogenesis/physiology , Neurons/metabolism , Animals , Doublecortin Domain Proteins , Doublecortin Protein , GAP-43 Protein/genetics , Hippocampus/cytology , Mice , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Neuropeptides/metabolism , Synaptophysin/metabolism
14.
J Neurosci ; 32(12): 4271-83, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22442089

ABSTRACT

The LIM domain only 4 (LMO4) transcription cofactor activates gene expression in neurons and regulates key aspects of network formation, but the mechanisms are poorly understood. Here, we show that LMO4 positively regulates ryanodine receptor type 2 (RyR2) expression, thereby suggesting that LMO4 regulates calcium-induced calcium release (CICR) in central neurons. We found that CICR modulation of the afterhyperpolarization in CA3 neurons from mice carrying a forebrain-specific deletion of LMO4 (LMO4 KO) was severely compromised but could be restored by single-cell overexpression of LMO4. In line with these findings, two-photon calcium imaging experiments showed that the potentiation of RyR-mediated calcium release from internal stores by caffeine was absent in LMO4 KO neurons. The overall facilitatory effect of CICR on glutamate release induced during trains of action potentials was likewise defective in LMO4 KO, confirming that CICR machinery is severely compromised in these neurons. Moreover, the magnitude of CA3-CA1 long-term potentiation was reduced in LMO4 KO mice, a defect that appears to be secondary to an overall reduced glutamate release probability. These cellular phenotypes in LMO4 KO mice were accompanied with deficits in hippocampus-dependent spatial learning as determined by the Morris water maze test. Thus, our results establish LMO4 as a key regulator of CICR in central neurons, providing a mechanism for LMO4 to modulate a wide range of neuronal functions and behavior.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium/metabolism , Hippocampus/cytology , LIM Domain Proteins/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Action Potentials/drug effects , Action Potentials/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Analysis of Variance , Animals , Caffeine/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Dizocilpine Maleate/pharmacology , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hybridomas , LIM Domain Proteins/deficiency , LIM Domain Proteins/genetics , Maze Learning/physiology , Mice , Mice, Transgenic , Neuronal Plasticity/genetics , Neurons/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Ryanodine Receptor Calcium Release Channel/genetics , Transfection
15.
J Neurosci ; 32(35): 12051-65, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22933789

ABSTRACT

Adult hippocampal neurogenesis is thought to be essential for learning and memory, and has been implicated in the pathogenesis of several disorders. Although recent studies have identified key factors regulating neuroprogenitor proliferation in the adult hippocampus, the mechanisms that control the migration and integration of adult-born neurons into circuits are largely unknown. Reelin is an extracellular matrix protein that is vital for neuronal development. Activation of the Reelin cascade leads to phosphorylation of Disabled-1, an adaptor protein required for Reelin signaling. Here we used transgenic mouse and retroviral reporters along with Reelin signaling gain-of-function and loss-of-function studies to show that the Reelin pathway regulates migration and dendritic development of adult-generated hippocampal neurons. Whereas overexpression of Reelin accelerated dendritic maturation, inactivation of the Reelin signaling pathway specifically in adult neuroprogenitor cells resulted in aberrant migration, decreased dendrite development, formation of ectopic dendrites in the hilus, and the establishment of aberrant circuits. Our findings support a cell-autonomous and critical role for the Reelin pathway in regulating dendritic development and the integration of adult-generated granule cells and point to this pathway as a key regulator of adult neurogenesis. Moreover, our data reveal a novel role of the Reelin cascade in adult brain function with potential implications for the pathogenesis of several neurological and psychiatric disorders.


Subject(s)
Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Extracellular Matrix Proteins/antagonists & inhibitors , Hippocampus/cytology , Hippocampus/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Neurogenesis/genetics , Signal Transduction/genetics , Age Factors , Aging/genetics , Animals , Cell Adhesion Molecules, Neuronal/physiology , Cell Line , Cells, Cultured , Extracellular Matrix Proteins/physiology , Gene Silencing/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/physiology , Rats , Rats, Sprague-Dawley , Reelin Protein , Serine Endopeptidases/physiology
16.
Hippocampus ; 23(8): 708-19, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23554226

ABSTRACT

Radial glia-like cells (RGCs) are the hypothesized source of adult hippocampal neurogenesis. However, the current model of hippocampal neurogenesis does not fully incorporate the in vivo heterogeneity of RGCs. In order to better understand the contribution of different RGC subtypes to adult hippocampal neurogenesis, we employed widely used transgenic lines (Nestin-CreER(T2) and GLAST::CreER(T2) mice) to explore how RGCs contribute to neurogenesis under basal conditions and after stimulation and depletion of neural progenitor cells. We first used these inducible fate-tracking transgenic lines to define the similarities and differences in the contribution of nestin- and GLAST-lineage cells to basal long-term hippocampal neurogenesis. We then explored the ability of nestin- and GLAST-lineage RGCs to contribute to neurogenesis after experimental manipulations that either ablate neurogenesis (i.c.v. application of the anti-mitotic AraC, cytosine-ß-D-arabinofuranoside) or stimulate neurogenesis (wheel running). Interestingly, in both ablation and stimulation experiments, labeled RGCs in GLAST::CreER(T2) mice appear to contribute to neurogenesis, whereas RGCs in Nestin-CreER(T2) mice do not. Finally, using NestinGFP reporter mice, we expanded on previous research by showing that not all RGCs in the adult dentate gyrus subgranular zone express nestin, and therefore RGCs are antigenically heterogeneous. These findings are important for the field, as they allow appropriately conservative interpretation of existing and future data that emerge from these inducible transgenic lines. These findings also raise important questions about the differences between transgenic driver lines, the heterogeneity of RGCs, and the potential differences in progenitor cell behavior between transgenic lines. As these findings highlight the possible differences in the contribution of cells to long-term neurogenesis in vivo, they indicate that the current models of hippocampal neurogenesis should be modified to include RGC lineage heterogeneity.


Subject(s)
Cell Lineage/physiology , Excitatory Amino Acid Transporter 1/metabolism , Hippocampus/cytology , Nestin/metabolism , Neurogenesis/physiology , Animals , Doublecortin Domain Proteins , Excitatory Amino Acid Transporter 1/genetics , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Motor Activity/physiology , Nerve Tissue Proteins/metabolism , Nestin/genetics , Neuroglia/physiology , Neurons/physiology , Neuropeptides/metabolism , Organ Culture Techniques , SOXB1 Transcription Factors/metabolism , Stem Cells/physiology
17.
Proc Natl Acad Sci U S A ; 107(9): 4436-41, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20176946

ABSTRACT

The long-term response to chronic stress is variable, with some individuals developing maladaptive functioning, although other "resilient" individuals do not. Stress reduces neurogenesis in the dentate gyrus subgranular zone (SGZ), but it is unknown if stress-induced changes in neurogenesis contribute to individual vulnerability. Using a chronic social defeat stress model, we explored whether the susceptibility to stress-induced social avoidance was related to changes in SGZ proliferation and neurogenesis. Immediately after social defeat, stress-exposed mice (irrespective of whether they displayed social avoidance) had fewer proliferating SGZ cells labeled with the S-phase marker BrdU. The decrease was transient, because BrdU cell numbers were normalized 24 h later. The survival of BrdU cells labeled before defeat stress was also not altered. However, 4 weeks later, mice that displayed social avoidance had more surviving dentate gyrus neurons. Thus, dentate gyrus neurogenesis is increased after social defeat stress selectively in mice that display persistent social avoidance. Supporting a functional role for adult-generated dentate gyrus neurons, ablation of neurogenesis via cranial ray irradiation robustly inhibited social avoidance. These data show that the time window after cessation of stress is a critical period for the establishment of persistent cellular and behavioral responses to stress and that a compensatory enhancement in neurogenesis is related to the long-term individual differences in maladaptive responses to stress.


Subject(s)
Avoidance Learning , Hippocampus/pathology , Neurogenesis , Stress, Psychological/pathology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Bromodeoxyuridine/metabolism , Cell Death , Hippocampus/metabolism , Male , Mice , S Phase , Signal Transduction
18.
Exp Neurol ; 362: 114326, 2023 04.
Article in English | MEDLINE | ID: mdl-36682400

ABSTRACT

The high incidence of ischemic stroke worldwide and poor efficacy of neuroprotective drugs has increased the need for novel therapies in stroke recovery. Transcription of the neurosecretory protein VGF (non-acronym) is enhanced following ischemic stroke and proposed to be important for stroke recovery. To determine the requirement for VGF in recovery, we created Vgffl/fl:Nestin-Cre conditional knockout (Vgf cKO) mice and induced a photothrombotic focal ischemic stroke. Naïve Vgf cKO mice had significant less body weight in the absence of gross defects in brain size, cortical lamination, or deficits in locomotor activity compared to wildtype controls. Following a focal stroke, the Vgf cKO mice had greater deficits including impaired recovery of forepaw motor deficits at 2- and 4-weeks post stroke. The increase in deficits occurred in the absence of any difference in lesion size and was accompanied by a striking loss of stroke-induced migration of SVZ-derived immature neurons to the peri-infarct region. Importantly, exogenous adenoviral delivery of VGF (AdVGF) significantly improved recovery in the Vgf cKO mice and was able to rescue the immature neuron migration defect observed. Taken together, our results define a requirement for VGF in post stroke recovery and identify VGF peptides as a potential future therapeutic.


Subject(s)
Ischemic Stroke , Stroke , Mice , Animals , Stroke/drug therapy , Body Weight
19.
Cells ; 11(10)2022 05 13.
Article in English | MEDLINE | ID: mdl-35626670

ABSTRACT

The complexity of adult neurogenesis is becoming increasingly apparent as we learn more about cellular heterogeneity and diversity of the neurogenic lineages and stem cell niches within the adult brain. This complexity has been unraveled in part due to single-cell and single-nucleus RNA sequencing (sc-RNAseq and sn-RNAseq) studies that have focused on adult neurogenesis. This review summarizes 33 published studies in the field of adult neurogenesis that have used sc- or sn-RNAseq methods to answer questions about the three main regions that host adult neural stem cells (NSCs): the subventricular zone (SVZ), the dentate gyrus (DG) of the hippocampus, and the hypothalamus. The review explores the similarities and differences in methodology between these studies and provides an overview of how these studies have advanced the field and expanded possibilities for the future.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Lateral Ventricles , Neurogenesis/genetics , Solitary Nucleus
20.
Methods Mol Biol ; 2515: 75-87, 2022.
Article in English | MEDLINE | ID: mdl-35776346

ABSTRACT

The endothelin-1 (ET-1) model of stroke involves the stereotactic injection of the vasoconstrictor ET-1 to produce a focal ischemic injury. In rats, this model produces consistent deficits, in contrast to more variable results in mice. In this chapter, we describe a new method to induce a murine focal ischemic cortical stroke by injecting L-NAME, another potent vasoconstrictor , in combination with ET-1 into the sensorimotor cortex. This ET-1 /L-NAME stroke induction protocol produces consistent focal cortical infarcts and sensorimotor functional impairments in C57BL/6 mice.


Subject(s)
Endothelin-1 , Stroke , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Stroke/chemically induced , Vasoconstrictor Agents
SELECTION OF CITATIONS
SEARCH DETAIL