Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 980
Filter
1.
Brain Commun ; 6(4): fcae204, 2024.
Article in English | MEDLINE | ID: mdl-38978722

ABSTRACT

Cholinergic innervation in the brain is involved in modulating neurovascular function including cerebral blood flow haemodynamics in response to neuronal activity. Cholinergic deficit is associated with pathophysiology in Alzheimer's disease, albeit the aetiology remains to be clarified. In the current study, neocortex cerebral blood flow response to acetylcholine was evaluated by Laser-Doppler Flowmetry (LDF) in 3xTgAD Alzheimer's disease model) and wild-type mice of two age groups. The peak of cerebral blood flow to acetylcholine (i.v.) from baseline levels (% ΔrCBF) was higher in young 3xTgAD versus in wild-type mice (48.35; 95% CI:27.03-69.67 versus 22.70; CI:15.5-29.91, P < 0.05); this was reversed in old 3xTgAD mice (21.44; CI:2.52-40.35 versus 23.25; CI:23.25-39). Choline acetyltransferase protein was reduced in neocortex, while cerebrovascular reactivity to acetylcholine was preserved in young 3×TgAD mice. This suggests endogenous acetylcholine deficit and possible cholinergic denervation from selected cholinergic nuclei within the basal forebrain. The early deposition of tauopathy moieties (mutant hTau and pTau181) and its coincidence in cholinergic cell clusters (occasionaly), were observed at the basal forebrain of 3xTgAD mice including substantia innominate, nucleus Basalis of Meynert and nucleus of horizontal limb diagonal band of Broca. A prominent feature was microglia interacting tauopathy and demonstrated a variety of morphology changes particularly when located in proximity to tauopathy. The microglia ramified phenotype was reduced as evaluated by the ramification index and Fractal analysis. Increased microglia senescence, identified as SASP (senescence-associated secretory phenotype), was colocalization with p16Ink4ɑ, a marker of irreversible cell-cycle arrest in old 3xTgAD versus wild-type mice (P = 0.001). The p16Ink4ɑ was also observed in neuronal cells bearing tauopathy within the basal forebrain of 3xTgAD mice. TNF-ɑ, the pro-inflammatory cytokine elevated persistently in microglia (Pearson's correlation coefficient = 0.62) and the loss of cholinergic cells in vulnerable basal forebrain environment, was indicated by image analysis in 3xTgAD mice, which linked to the cholinergic deficits in neocortex rCBF haemodynamics. Our study revealed the early change of CBF haemodynamics to acetylcholine in 3xTgAD model. As a major effector of brain innate immune activation, microglia SASP with age-related disease progression is indicative of immune cell senescence, which contributes to chronic inflammation and cholinergic deficits at the basal forebrain. Targeting neuroinflammation and senescence may mitigate cholinergic pathophysiology in Alzheimer's disease.

2.
Int J Mol Sci ; 25(11)2024 May 26.
Article in English | MEDLINE | ID: mdl-38891978

ABSTRACT

Binge alcohol consumption during adolescence can produce lasting deficits in learning and memory while also increasing the susceptibility to substance use disorders. The adolescent intermittent ethanol (AIE) rodent model mimics human adolescent binge drinking and has identified the nucleus basalis magnocellularis (NbM) as a key site of pathology. The NbM is a critical regulator of prefrontal cortical (PFC) cholinergic function and attention. The cholinergic phenotype is controlled pro/mature neurotrophin receptor activation. We sought to determine if p75NTR activity contributes to the loss of cholinergic phenotype in AIE by using a p75NTR modulator (LM11A-31) to inhibit prodegenerative signaling during ethanol exposure. Male and female rats underwent 5 g/kg ethanol (AIE) or water (CON) exposure following 2-day-on 2-day-off cycles from postnatal day 25-57. A subset of these groups also received a protective dose of LM11A-31 (50 mg/kg) during adolescence. Rats were trained on a sustained attention task (SAT) and behaviorally relevant acetylcholine (ACh) activity was recorded in the PFC with a fluorescent indicator (AChGRAB 3.0). AIE produced learning deficits on the SAT, which were spared with LM11A-31. In addition, PFC ACh activity was blunted by AIE, which LM11A-31 corrected. Investigation of NbM ChAT+ and TrkA+ neuronal expression found that AIE led to a reduction of ChAT+TrkA+ neurons, which again LM11A-31 protected. Taken together, these findings demonstrate the p75NTR activity during AIE treatment is a key regulator of cholinergic degeneration.


Subject(s)
Acetylcholine , Cholinergic Neurons , Ethanol , Prefrontal Cortex , Animals , Female , Male , Rats , Acetylcholine/metabolism , Atrophy , Behavior, Animal/drug effects , Cholinergic Neurons/metabolism , Cholinergic Neurons/drug effects , Disease Models, Animal , Ethanol/toxicity , Nerve Tissue Proteins , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Rats, Sprague-Dawley , Receptors, Growth Factor , Receptors, Nerve Growth Factor/metabolism
3.
Cells ; 13(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891027

ABSTRACT

Sleep disruption is a frequent problem of advancing age, often accompanied by low-grade chronic central and peripheral inflammation. We examined whether chronic neuroinflammation in the preoptic and basal forebrain area (POA-BF), a critical sleep-wake regulatory structure, contributes to this disruption. We developed a targeted viral vector designed to overexpress tumor necrosis factor-alpha (TNFα), specifically in astrocytes (AAV5-GFAP-TNFα-mCherry), and injected it into the POA of young mice to induce heightened neuroinflammation within the POA-BF. Compared to the control (treated with AAV5-GFAP-mCherry), mice with astrocytic TNFα overproduction within the POA-BF exhibited signs of increased microglia activation, indicating a heightened local inflammatory milieu. These mice also exhibited aging-like changes in sleep-wake organization and physical performance, including (a) impaired sleep-wake functions characterized by disruptions in sleep and waking during light and dark phases, respectively, and a reduced ability to compensate for sleep loss; (b) dysfunctional VLPO sleep-active neurons, indicated by fewer neurons expressing c-fos after suvorexant-induced sleep; and (c) compromised physical performance as demonstrated by a decline in grip strength. These findings suggest that inflammation-induced dysfunction of sleep- and wake-regulatory mechanisms within the POA-BF may be a critical component of sleep-wake disturbances in aging.


Subject(s)
Aging , Astrocytes , Basal Forebrain , Preoptic Area , Sleep , Tumor Necrosis Factor-alpha , Animals , Astrocytes/metabolism , Astrocytes/pathology , Aging/metabolism , Preoptic Area/metabolism , Mice , Tumor Necrosis Factor-alpha/metabolism , Sleep/physiology , Basal Forebrain/metabolism , Basal Forebrain/pathology , Wakefulness , Male , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Sleep Wake Disorders/metabolism , Sleep Wake Disorders/pathology
4.
Neuropeptides ; 107: 102449, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38908356

ABSTRACT

Previous research has demonstrated that basal forebrain (BF) regulates arousal during propofol anesthesia. However, as the BF comprises cholinergic neurons alongside two other types of neurons, the specific role of cholinergic neurons has not been definitively elucidated. In our study, calcium signal imaging was utilized to monitor the real-time activities of cholinergic neurons in the BF during propofol anesthesia. Additionally, we selectively stimulated these neurons to investigate EEG and behavioral responses during propofol anesthesia. Furthermore, we specifically lesioned cholinergic neurons in the BF to investigate the sensitivity to propofol and the induction time. The results revealed that propofol suppressed calcium signals of cholinergic neurons within the BF following intraperitoneal injection. Notably, upon recovery of the righting reflex, the calcium signals partially recovered. Spectral analysis of the EEG elucidated that optical stimulation of cholinergic neurons led to a decrease in δ power underlie propofol anesthesia. Conversely, depletion of cholinergic neurons in the BF enhanced sensitivity to propofol and shortened the induction time. These findings clarify the role of cholinergic neurons in the anesthesia-arousal process, as well as the depth and the sensitivity of propofol anesthesia.

5.
bioRxiv ; 2024 May 17.
Article in English | MEDLINE | ID: mdl-38798371

ABSTRACT

Inhibitory control is a critical executive function that allows animals to suppress their impulsive behavior in order to achieve certain goals or avoid punishment. We investigated norepinephrine (NE) and acetylcholine (ACh) dynamics and population neuronal activity in the prefrontal cortex during inhibitory control. Using fluorescent sensors to measure extracellular levels of NE and ACh, we simultaneously recorded the dynamics of prefrontal NE and ACh in mice performing an inhibitory control task. The prefrontal NE and ACh signals exhibited strong coherence at 0.4-0.8 Hz. Chemogenetic inhibition of locus coeruleus (LC) neurons that project to the basal forebrain region reduced inhibitory control performance to chance levels. However, this manipulation did not diminish the difference in NE/ACh signals between successful and failed trials; instead, it abolished the difference in NE-ACh phase synchrony between the successful and failed trials, indicating that NE-ACh phase synchrony is a task-relevant neuromodulatory feature. Chemogenetic inhibition of cholinergic neurons that project to the LC region did not impair the inhibitory control performance, nor did it abolish the difference in NE-ACh phase synchrony between successful or failed trials, further confirming the relevance of NE-ACh phase synchrony to inhibitory control. To understand the possible effect of NE-ACh synchrony on prefrontal population activity, we employed Neuropixels to record from the prefrontal cortex with and without inhibiting LC neurons that project to the basal forebrain during inhibitory control. The LC inhibition reduced the number of prefrontal neurons encoding inhibitory control. Demixed principal component analysis (dPCA) further revealed that population firing patterns representing inhibitory control were impaired by the LC inhibition. Disparities in NE-ACh phase synchrony relevant to inhibitory control occurred only in the prefrontal cortex, but not in the parietal cortex, somatosensory cortex, and the somatosensory thalamus. Taken together, these findings suggest that the LC modulates inhibitory control through its collective effect with cholinergic systems on population activity in the prefrontal cortex. Our results further revealed that NE-ACh phase synchrony is a critical neuromodulatory feature with important implications for cognitive control.

6.
Sci Rep ; 14(1): 10422, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38710727

ABSTRACT

Anticipating positive outcomes is a core cognitive function in the process of reward prediction. However, no neurophysiological method objectively assesses reward prediction in basic medical research. In the present study, we established a physiological paradigm using cortical direct current (DC) potential responses in rats to assess reward prediction. This paradigm consisted of five daily 1-h sessions with two tones, wherein the rewarded tone was followed by electrical stimulation of the medial forebrain bundle (MFB) scheduled at 1000 ms later, whereas the unrewarded tone was not. On day 1, both tones induced a negative DC shift immediately after auditory responses, persisting up to MFB stimulation. This negative shift progressively increased and peaked on day 4. Starting from day 3, the negative shift from 600 to 1000 ms was significantly larger following the rewarded tone than that following the unrewarded tone. This negative DC shift was particularly prominent in the frontal cortex, suggesting its crucial role in discriminative reward prediction. During the extinction sessions, the shift diminished significantly on extinction day 1. These findings suggest that cortical DC potential is related to reward prediction and could be a valuable tool for evaluating animal models of depression, providing a testing system for anhedonia.


Subject(s)
Extinction, Psychological , Reward , Animals , Rats , Male , Extinction, Psychological/physiology , Electric Stimulation , Acoustic Stimulation , Medial Forebrain Bundle/physiology , Rats, Sprague-Dawley
7.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38755010

ABSTRACT

Cholinergic neurons of the basal forebrain represent the main source of cholinergic innervation of large parts of the neocortex and are involved in adults in the modulation of attention, memory, and arousal. During the first postnatal days, they play a crucial role in the development of cortical neurons and cortical cytoarchitecture. However, their characteristics, during this period have not been studied. To understand how they can fulfill this role, we investigated the morphological and electrophysiological maturation of cholinergic neurons of the substantia innominata-nucleus basalis of Meynert (SI/NBM) complex in the perinatal period in mice. We show that cholinergic neurons, whether or not they express gamma-aminobutyric acid (GABA) as a cotransmitter, are already functional at Embryonic Day 18. Until the end of the first postnatal week, they constitute a single population of neurons with a well developed dendritic tree, a spontaneous activity including bursting periods, and a short-latency response to depolarizations (early-firing). They are excited by both their GABAergic and glutamatergic afferents. During the second postnatal week, a second, less excitable, neuronal population emerges, with a longer delay response to depolarizations (late-firing), together with the hyperpolarizing action of GABAA receptor-mediated currents. This classification into early-firing (40%) and late-firing (60%) neurons is again independent of the coexpression of GABAergic markers. These results strongly suggest that during the first postnatal week, the specific properties of developing SI/NBM cholinergic neurons allow them to spontaneously release acetylcholine (ACh), or ACh and GABA, into the developing cortex.


Subject(s)
Basal Forebrain , Cholinergic Neurons , gamma-Aminobutyric Acid , Animals , Cholinergic Neurons/physiology , Cholinergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Basal Forebrain/physiology , Basal Forebrain/metabolism , Animals, Newborn , Mice, Inbred C57BL , Female , Basal Nucleus of Meynert/physiology , Basal Nucleus of Meynert/metabolism , Substantia Innominata/physiology , Substantia Innominata/metabolism , Mice , Receptors, GABA-A/metabolism , Action Potentials/physiology , Patch-Clamp Techniques , Glutamic Acid/metabolism
8.
Cogn Neurodyn ; 18(2): 757-767, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38699625

ABSTRACT

The ability to learn by observing the behavior of others is energy efficient and brings high survival value, making it an important learning tool that has been documented in a myriad of species in the animal kingdom. In the laboratory, rodents have proven useful models for studying different forms of observational learning, however, the most robust learning paradigms typically rely on aversive stimuli, like foot shocks, to drive the social acquisition of fear. Non-fear-based tasks have also been used but they rarely succeed in having observer animals perform a new behavior de novo. Consequently, little known regarding the cellular mechanisms supporting non-aversive types of learning, such as visuomotor skill acquisition. To address this we developed a reward-based observational learning paradigm in adult rats, in which observer animals learn to tap lit spheres in a specific sequence by watching skilled demonstrators, with successful trials leading to rewarding intracranial stimulation in both observers and performers. Following three days of observation and a 24-hour delay, observer animals outperformed control animals on several metrics of task performance and efficiency, with a subset of observers demonstrating correct performance immediately when tested. This paradigm thus introduces a novel tool to investigate the neural circuits supporting observational learning and memory for visuomotor behavior, a phenomenon about which little is understood, particularly in rodents.

9.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38819455

ABSTRACT

The vertebrate Dlx gene family encode homeobox transcription factors that are related to the Drosophila Distal-less (Dll) gene and are crucial for development. Over the last ∼35 years detailed information has accrued about the redundant and unique expression and function of the six mammalian Dlx family genes. DLX proteins interact with general transcriptional regulators, and co-bind with other transcription factors to enhancer elements with highly specific activity in the developing forebrain. Integration of the genetic and biochemical data has yielded a foundation for a gene regulatory network governing the differentiation of forebrain GABAergic neurons. In this Primer, we describe the discovery of vertebrate Dlx genes and their crucial roles in embryonic development. We largely focus on the role of Dlx family genes in mammalian forebrain development revealed through studies in mice. Finally, we highlight questions that remain unanswered regarding vertebrate Dlx genes despite over 30 years of research.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins , Prosencephalon , Transcription Factors , Animals , Prosencephalon/metabolism , Prosencephalon/embryology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Humans , Mammals/genetics , Mice
10.
Neurobiol Aging ; 139: 54-63, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608458

ABSTRACT

Nucleus Basalis of Meynert (NbM), a crucial source of cholinergic projection to the entorhinal cortex (EC) and hippocampus (HC), has shown sensitivity to neurofibrillary degeneration in the early stages of Alzheimer's Disease. Using deformation-based morphometry (DBM) on up-sampled MRI scans from 1447 Alzheimer's Disease Neuroimaging Initiative participants, we aimed to quantify NbM degeneration along the disease trajectory. Results from cross-sectional analysis revealed significant differences of NbM volume between cognitively normal and early mild cognitive impairment cohorts, confirming recent studies suggesting that NbM degeneration happens before degeneration in the EC or HC. Longitudinal linear mixed-effect models were then used to compare trajectories of volume change after realigning all participants into a common timeline based on their cognitive decline. Results indicated the earliest deviations in NbM volumes from the cognitively healthy trajectory, challenging the prevailing idea that Alzheimer's originates in the EC. Converging evidence from cross-sectional and longitudinal models suggest that the NbM may be a focal target of early AD progression, which is often obscured by normal age-related decline.


Subject(s)
Alzheimer Disease , Basal Nucleus of Meynert , Disease Progression , Magnetic Resonance Imaging , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Humans , Female , Male , Aged , Cross-Sectional Studies , Basal Nucleus of Meynert/pathology , Basal Nucleus of Meynert/diagnostic imaging , Aged, 80 and over , Cognitive Dysfunction/pathology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/etiology , Entorhinal Cortex/pathology , Entorhinal Cortex/diagnostic imaging , Longitudinal Studies , Organ Size , Hippocampus/pathology , Hippocampus/diagnostic imaging
11.
Biochem Pharmacol ; 224: 116201, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608783

ABSTRACT

Intestinal barrier dysfunction, leaky gut, is implicated in various diseases, including irritable bowel syndrome (IBS) and neurodegenerative conditions like Alzheimer's disease. Our recent investigation revealed that basal forebrain cholinergic neurons (BFCNs), critical for cognitive function, receive signals from butyrate and orexin, playing a role in regulating intestinal barrier function through adenosine A2B signaling and the vagus. This study explores the involvement and function of brain histamine, linked to BFCNs, in the regulation of intestinal barrier function. Colonic permeability, assessed by quantifying absorbed Evans blue in rat colonic tissue, showed that histamine did not affect increased colonic permeability induced by LPS when administered subcutaneously. However, intracisternal histamine administration improved colonic hyperpermeability. Elevating endogenous histamine levels in the brain with SKF91488, a histamine N-methyltransferase inhibitor, also improved colonic hyperpermeability. This effect was abolished by intracisternal chlorpheniramine, an histamine H1 receptor antagonist, not ranitidine, an H2 receptor antagonist. The SKF91488-induced improvement in colonic hyperpermeability was blocked by vagotomy, intracisternal pirenzepine (suppressing BFCNs activity), or alloxazine (an adenosine A2B receptor antagonist). Additionally, intracisternal chlorpheniramine injection eliminated butyrate-induced improvement in colonic hyperpermeability. These findings suggest that brain histamine, acting via the histamine H1 receptor, regulates intestinal barrier function involving BFCNs, adenosine A2B signaling, and the vagus. Brain histamine appears to centrally regulate intestinal barrier function influenced by butyrate, differentiating its actions from peripheral histamine in conditions like IBS, where mast cell-derived histamine induces leaky gut. Brain histamine emerges as a potential pharmacological target for diseases associated with leaky gut, such as dementia and IBS.


Subject(s)
Cholinergic Neurons , Colon , Histamine , Permeability , Rats, Sprague-Dawley , Receptor, Adenosine A2B , Vagus Nerve , Animals , Histamine/metabolism , Histamine/pharmacology , Rats , Male , Receptor, Adenosine A2B/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cholinergic Neurons/physiology , Vagus Nerve/drug effects , Vagus Nerve/physiology , Vagus Nerve/metabolism , Colon/metabolism , Colon/drug effects , Permeability/drug effects , Prosencephalon/drug effects , Prosencephalon/metabolism
12.
J Alzheimers Dis ; 99(1): 145-159, 2024.
Article in English | MEDLINE | ID: mdl-38640150

ABSTRACT

Background: Degeneration of cholinergic basal forebrain (BF) neurons characterizes Alzheimer's disease (AD). However, what role the BF plays in the dynamics of AD pathophysiology has not been investigated precisely. Objective: To investigate the baseline and longitudinal roles of BF along with core neuropathologies in AD. Methods: In this retrospective cohort study, we enrolled 113 subjects (38 amyloid [Aß]-negative cognitively unimpaired, 6 Aß-positive cognitively unimpaired, 39 with prodromal AD, and 30 with AD dementia) who performed brain MRI for BF volume and cortical thickness, 18F-florbetaben PET for Aß, 18F-flortaucipir PET for tau, and detailed cognitive testing longitudinally. We investigated the baseline and longitudinal association of BF volume with Aß and tau standardized uptake value ratio and cognition. Results: Cross-sectionally, lower BF volume was not independently associated with higher cortical Aß, but it was associated with tau burden. Tau burden in the orbitofrontal, insular, lateral temporal, inferior temporo-occipital, and anterior cingulate cortices were associated with progressive BF atrophy. Lower BF volume was associated with faster Aß accumulation, mainly in the prefrontal, anterior temporal, cingulate, and medial occipital cortices. BF volume was associated with progressive decline in language and memory functions regardless of baseline Aß and tau burden. Conclusions: Tau deposition affected progressive BF atrophy, which in turn accelerated amyloid deposition, leading to a vicious cycle. Also, lower baseline BF volume independently predicted deterioration in cognitive function.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Basal Forebrain , Cognition , Magnetic Resonance Imaging , Positron-Emission Tomography , tau Proteins , Humans , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/diagnostic imaging , Male , Female , Aged , tau Proteins/metabolism , Basal Forebrain/pathology , Basal Forebrain/metabolism , Basal Forebrain/diagnostic imaging , Amyloid beta-Peptides/metabolism , Retrospective Studies , Cognition/physiology , Cross-Sectional Studies , Aged, 80 and over , Longitudinal Studies , Middle Aged , Neuropsychological Tests , Cohort Studies
13.
Mol Neurobiol ; 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38676810

ABSTRACT

It is established that neurogenesis of dentate gyrus is increased after ischemic insult, although the regulatory mechanisms have not yet been elucidated. In this study, we focused on Ezh2 which suppresses gene expression through catalyzing trimethylation of lysine 27 of histone 3. Male gerbils were injected with adeno-associated virus (AAV) carrying shRNA targeting to Ezh2 into right dentate gyrus 2 weeks prior to forebrain ischemia. One week after ischemia, animals were injected with thymidine analogue to label proliferating cells. Three weeks after ischemia, animals were killed for histological analysis. AAV-mediated knockdown of Ezh2 significantly decreased the ischemia-induced increment of proliferating cells, and the proliferated cells after ischemia showed significantly longer migration from subgranular zone (SGZ), compared to the control group. Furthermore, the number of neural stem cells in SGZ significantly decreased after ischemia with Ezh2 knockdown group. Of note, Ezh2 knockdown did not affect the number of proliferating cells or the migration from SGZ in the non-ischemic condition. Our data showed that, specifically after ischemia, Ezh2 knockdown shifted the balance between self-renewal and differentiation toward differentiation in adult dentate gyrus.

14.
Biomedicines ; 12(4)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38672235

ABSTRACT

The malfunctioning of the brain synucleins is associated with pathogenesis of Parkinson's disease. Synucleins' ability to modulate various pre-synaptic processes suggests their modifying effects on the electroencephalogram (EEG) recorded from different brain structures. Disturbances in interrelations between them are critical for the onset and evolution of neurodegenerative diseases. Recently, we have shown that, in mice lacking several synucleins, differences between the frequency spectra of EEG from different brain structures are correlated with specificity of synucleins' combinations. Given that EEG spectra are indirect characteristics of inter-structural relations, in this study, we analyzed a coherence of instantaneous values for EEGs recorded from different structures as a direct measure of "functional connectivity" between them. METHODS: EEG data from seven groups of knock-out (KO) mice with combined deletions of alpha, beta, and gamma synucleins versus a group of wild-type (WT) mice were compared. EEG coherence was estimated between the cortex (MC), putamen (Pt), ventral tegmental area (VTA), and substantia nigra (SN) in all combinations. RESULTS: EEG coherence suppression, predominantly in the beta frequency band, was observed in KO mice versus WT littermates. The suppression was minimal in MC-Pt and VTA-SN interrelations in all KO groups and in all inter-structural relations in mice lacking either all synucleins or only beta synuclein. In other combinations of deleted synucleins, significant EEG coherence suppression in KO mice was dominant in relations with VTA and SN. CONCLUSION: Deletions of the synucleins produced significant attenuation of intra-cerebral EEG coherence depending on the imbalance of different types of synucleins.

15.
Cell ; 187(9): 2129-2142.e17, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38670071

ABSTRACT

Interspecies blastocyst complementation (IBC) provides a unique platform to study development and holds the potential to overcome worldwide organ shortages. Despite recent successes, brain tissue has not been achieved through IBC. Here, we developed an optimized IBC strategy based on C-CRISPR, which facilitated rapid screening of candidate genes and identified that Hesx1 deficiency supported the generation of rat forebrain tissue in mice via IBC. Xenogeneic rat forebrain tissues in adult mice were structurally and functionally intact. Cross-species comparative analyses revealed that rat forebrain tissues developed at the same pace as the mouse host but maintained rat-like transcriptome profiles. The chimeric rate of rat cells gradually decreased as development progressed, suggesting xenogeneic barriers during mid-to-late pre-natal development. Interspecies forebrain complementation opens the door for studying evolutionarily conserved and divergent mechanisms underlying brain development and cognitive function. The C-CRISPR-based IBC strategy holds great potential to broaden the study and application of interspecies organogenesis.


Subject(s)
Prosencephalon , Animals , Prosencephalon/metabolism , Prosencephalon/embryology , Mice , Rats , Blastocyst/metabolism , Female , CRISPR-Cas Systems/genetics , Transcriptome , Organogenesis , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Male , Mice, Inbred C57BL
16.
J Neurophysiol ; 131(5): 872-875, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38533940

ABSTRACT

A recent study by Tran et al. (Tran LN, Loew SK, Franco SJ. J Neurosci 43: 6854-6871, 2023) investigated the cellular processes underlying the timing and regulation of oligodendrocyte production, focusing on the role of Notch signaling in the dorsal forebrain of mouse embryos. They found that although Notch signaling is required to specify oligodendrocyte precursor cell fate during embryonic development, overexpression prevents oligodendrogenesis through several mechanisms. This critical review highlights their findings and offers suggestions for future research investigating the precise spatiotemporal control of Notch signaling throughout the development of the central nervous system.


Subject(s)
Receptors, Notch , Signal Transduction , Animals , Signal Transduction/physiology , Receptors, Notch/metabolism , Neurons/physiology , Neurons/metabolism , Neuroglia/physiology , Neuroglia/metabolism , Oligodendroglia/physiology , Oligodendroglia/metabolism
17.
J Neurosurg ; : 1-15, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38518292

ABSTRACT

OBJECTIVE: The ventral amygdalofugal pathway (VAFP) provides afferent and efferent connections to the amygdala and spans along some of the frequently traversed intra-axial surgical corridors as a dominant fiber bundle. This study aimed to reveal the frequently overlooked VAFP fibers by examining their courses and connections to the basal forebrain, septal region, hypothalamus, thalamus, tegmentum, and brainstem. METHODS: Ten postmortem human brains were used to display the characteristics of the VAFP, and fiber dissection results were compared with those of tractography. RESULTS: From anterior to posterior, the VAFP was separated into 5 different portions: 1) amygdala-substantia innominata; 2) amygdaloseptal (diagonal band of Broca); 3) amygdalo-thalamic; 4) amygdalo-hypothalamic, intermingling with the medial forebrain bundle and extending to the bed nucleus of stria terminalis; and 5) amygdalotegmental. The results of fiber dissections were confirmed with findings obtained from diffusion tensor tractography. CONCLUSIONS: This study supports the concept that interconnected forebrain, diencephalic, mesencephalic, and brainstem connections of the VAFP form an integrated surgically important network. The fiber dissection findings also provide the neuroanatomical basis for VAFP segmentation, which may help neurosurgeons better appreciate the complex microsurgical anatomy of the amygdalar connections. Amygdala-substantia innominata and amygdalotegmental connections are demonstrated for the first time and clarified within the structure of the VAFP.

19.
Methods Mol Biol ; 2761: 491-498, 2024.
Article in English | MEDLINE | ID: mdl-38427257

ABSTRACT

Robust preclinical models of Parkinson's disease (PD) are valuable tools for understanding the biology and treatment of this complex disease. 6-Hydroxydopamine (6-OHDA) is a selective catecholaminergic drug injected into the substantia nigra pars compacta (SNc), medial forebrain bundle (MFB), or striatum, which is then metabolized to induce parkinsonism. Unilateral injection of 6-OHDA produces loss of dopaminergic (DAergic) neurons on the injected side with a marked motor asymmetry known as hemiparkinsonism, typically characterized by a rotational behavior to the impaired side. The present work describes a stable unilateral 6-OHDA-lesioned rat model of PD. 6-OHDA was administered into the MFB, leading to the consistent loss of striatal dopamine (DA) and behavioral imbalance in unilateral 6-OHDA-lesioned rats to establish the model of PD. This model of PD is a valuable tool for understanding the mechanisms underlying the generation of parkinsonian symptoms.


Subject(s)
Parkinson Disease , Rats , Male , Animals , Parkinson Disease/metabolism , Oxidopamine/pharmacology , Rats, Wistar , Dopamine/metabolism , Medial Forebrain Bundle/metabolism , Corpus Striatum/metabolism , Substantia Nigra/metabolism , Disease Models, Animal
20.
Dev Neurobiol ; 84(2): 47-58, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38466218

ABSTRACT

In sexually dimorphic zebra finches (Taeniopygia guttata), only males learn to sing their father's song, whereas females learn to recognize the songs of their father or mate but cannot sing themselves. Memory of learned songs is behaviorally expressed in females by preferring familiar songs over unfamiliar ones. Auditory association regions such as the caudomedial mesopallium (CMM; or caudal mesopallium) have been shown to be key nodes in a network that supports preferences for learned songs in adult females. However, much less is known about how song preferences develop during the sensitive period of learning in juvenile female zebra finches. In this study, we used blood-oxygen level-dependent (BOLD) functional magnetic resonance imaging (fMRI) to trace the development of a memory-based preference for the father's song in female zebra finches. Using BOLD fMRI, we found that only in adult female zebra finches with a preference for learned song over novel conspecific song, neural selectivity for the father's song was localized in the thalamus (dorsolateral nucleus of the medial thalamus; part of the anterior forebrain pathway, AFP) and in CMM. These brain regions also showed a selective response in juvenile female zebra finches, although activation was less prominent. These data reveal that neural responses in CMM, and perhaps also in the AFP, are shaped during development to support behavioral preferences for learned songs.


Subject(s)
Finches , Vocalization, Animal , Male , Animals , Female , Vocalization, Animal/physiology , alpha-Fetoproteins/metabolism , Finches/metabolism , Acoustic Stimulation/methods , Auditory Perception/physiology , Prosencephalon/metabolism , Magnetic Resonance Imaging/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...