Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.050
Filtrar
1.
Sci Adv ; 10(19): eadj9911, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38728406

RESUMO

During cerebral cortex development, excitatory pyramidal neurons (PNs) establish specific projection patterns while receiving inputs from GABAergic inhibitory interneurons (INs). Whether these inhibitory inputs can shape PNs' projection patterns is, however, unknown. While layer 4 (L4) PNs of the primary somatosensory (S1) cortex are all born as long-range callosal projection neurons (CPNs), most of them acquire local connectivity upon activity-dependent elimination of their interhemispheric axons during postnatal development. Here, we demonstrate that precise developmental regulation of inhibition is key for the retraction of S1L4 PNs' callosal projections. Ablation of somatostatin INs leads to premature inhibition from parvalbumin INs onto S1L4 PNs and prevents them from acquiring their barrel-restricted local connectivity pattern. As a result, adult S1L4 PNs retain interhemispheric projections responding to tactile stimuli, and the mice lose whisker-based texture discrimination. Overall, we show that temporally ordered IN activity during development is key to shaping local ipsilateral S1L4 PNs' projection pattern, which is required for fine somatosensory processing.


Assuntos
Neurônios GABAérgicos , Interneurônios , Córtex Somatossensorial , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Interneurônios/citologia , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Neurônios GABAérgicos/citologia , Córtex Somatossensorial/fisiologia , Córtex Somatossensorial/metabolismo , Córtex Somatossensorial/citologia , Camundongos , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Parvalbuminas/metabolismo
2.
Elife ; 132024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38748470

RESUMO

Acetylcholine is widely believed to modulate the release of dopamine in the striatum of mammals. Experiments in brain slices clearly show that synchronous activation of striatal cholinergic interneurons is sufficient to drive dopamine release via axo-axonal stimulation of nicotinic acetylcholine receptors. However, evidence for this mechanism in vivo has been less forthcoming. Mohebi, Collins and Berke recently reported that, in awake behaving rats, optogenetic activation of striatal cholinergic interneurons with blue light readily evokes dopamine release measured with the red fluorescent sensor RdLight1 (Mohebi et al., 2023). Here, we show that blue light alone alters the fluorescent properties of RdLight1 in a manner that may be misconstrued as phasic dopamine release, and that this artefactual photoactivation can account for the effects attributed to cholinergic interneurons. Our findings indicate that measurements of dopamine using the red-shifted fluorescent sensor RdLight1 should be interpreted with caution when combined with optogenetics. In light of this and other publications that did not observe large acetylcholine-evoked dopamine transients in vivo, the conditions under which such release occurs in behaving animals remain unknown.


Assuntos
Neurônios Colinérgicos , Dopamina , Interneurônios , Optogenética , Dopamina/metabolismo , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/fisiologia , Ratos , Optogenética/métodos , Motivação , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiologia , Acetilcolina/metabolismo
3.
Proc Natl Acad Sci U S A ; 121(21): e2406565121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38753507

RESUMO

While depolarization of the neuronal membrane is known to evoke the neurotransmitter release from synaptic vesicles, hyperpolarization is regarded as a resting state of chemical neurotransmission. Here, we report that hyperpolarizing neurons can actively signal neural information by employing undocked hemichannels. We show that UNC-7, a member of the innexin family in Caenorhabditis elegans, functions as a hemichannel in thermosensory neurons and transmits temperature information from the thermosensory neurons to their postsynaptic interneurons. By monitoring neural activities in freely behaving animals, we find that hyperpolarizing thermosensory neurons inhibit the activity of the interneurons and that UNC-7 hemichannels regulate this process. UNC-7 is required to control thermotaxis behavior and functions independently of synaptic vesicle exocytosis. Our findings suggest that innexin hemichannels mediate neurotransmission from hyperpolarizing neurons in a manner that is distinct from the synaptic transmission, expanding the way of neural circuitry operations.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Interneurônios , Neurônios , Transmissão Sináptica , Animais , Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Transmissão Sináptica/fisiologia , Interneurônios/metabolismo , Interneurônios/fisiologia , Neurônios/fisiologia , Neurônios/metabolismo , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/fisiologia , Resposta Táctica/fisiologia , Conexinas/metabolismo , Conexinas/genética , Proteínas de Membrana
4.
Cereb Cortex ; 34(5)2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38760318

RESUMO

Cortical parvalbumin interneurons (PV+) are major regulators of excitatory/inhibitory information processing, and their maturation is associated with the opening of developmental critical periods (CP). Recent studies reveal that cortical PV+ axons are myelinated, and that myelination along with perineuronal net (PNN) maturation around PV+ cells is associated with the closures of CP. Although PV+ interneurons are susceptible to early-life stress, their relationship between their myelination and PNN coverage remains unexplored. This study compared the fine features of PV+ interneurons in well-characterized human post-mortem ventromedial prefrontal cortex samples (n = 31) from depressed suicides with or without a history of child abuse (CA) and matched controls. In healthy controls, 81% of all sampled PV+ interneurons displayed a myelinated axon, while a subset (66%) of these cells also displayed a PNN, proposing a relationship between both attributes. Intriguingly, a 3-fold increase in the proportion of unmyelinated PV+ interneurons with a PNN was observed in CA victims, along with greater PV-immunofluorescence intensity in myelinated PV+ cells with a PNN. This study, which is the first to provide normative data on myelination and PNNs around PV+ interneurons in human neocortex, sheds further light on the cellular and molecular consequences of early-life adversity on cortical PV+ interneurons.


Assuntos
Interneurônios , Parvalbuminas , Córtex Pré-Frontal , Humanos , Córtex Pré-Frontal/patologia , Córtex Pré-Frontal/metabolismo , Parvalbuminas/metabolismo , Interneurônios/patologia , Interneurônios/metabolismo , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , Bainha de Mielina/patologia , Bainha de Mielina/metabolismo , Suicídio , Idoso , Autopsia , Maus-Tratos Infantis/psicologia , Adulto Jovem
5.
Acta Neuropathol ; 147(1): 80, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714540

RESUMO

GABAergic interneurons play a critical role in maintaining neural circuit balance, excitation-inhibition regulation, and cognitive function modulation. In tuberous sclerosis complex (TSC), GABAergic neuron dysfunction contributes to disrupted network activity and associated neurological symptoms, assumingly in a cell type-specific manner. This GABAergic centric study focuses on identifying specific interneuron subpopulations within TSC, emphasizing the unique characteristics of medial ganglionic eminence (MGE)- and caudal ganglionic eminence (CGE)-derived interneurons. Using single-nuclei RNA sequencing in TSC patient material, we identify somatostatin-expressing (SST+) interneurons as a unique and immature subpopulation in TSC. The disrupted maturation of SST+ interneurons may undergo an incomplete switch from excitatory to inhibitory GABAergic signaling during development, resulting in reduced inhibitory properties. Notably, this study reveals markers of immaturity specifically in SST+ interneurons, including an abnormal NKCC1/KCC2 ratio, indicating an imbalance in chloride homeostasis crucial for the postsynaptic consequences of GABAergic signaling as well as the downregulation of GABAA receptor subunits, GABRA1, and upregulation of GABRA2. Further exploration of SST+ interneurons revealed altered localization patterns of SST+ interneurons in TSC brain tissue, concentrated in deeper cortical layers, possibly linked to cortical dyslamination. In the epilepsy context, our research underscores the diverse cell type-specific roles of GABAergic interneurons in shaping seizures, advocating for precise therapeutic considerations. Moreover, this study illuminates the potential contribution of SST+ interneurons to TSC pathophysiology, offering insights for targeted therapeutic interventions.


Assuntos
Neurônios GABAérgicos , Interneurônios , Esclerose Tuberosa , Interneurônios/patologia , Interneurônios/metabolismo , Esclerose Tuberosa/patologia , Esclerose Tuberosa/metabolismo , Humanos , Neurônios GABAérgicos/patologia , Neurônios GABAérgicos/metabolismo , Masculino , Feminino , Eminência Mediana/patologia , Eminência Mediana/metabolismo , Somatostatina/metabolismo , Criança , Pré-Escolar , Receptores de GABA-A/metabolismo , Adolescente , Eminência Ganglionar
6.
Nat Commun ; 15(1): 2823, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38561349

RESUMO

Dysfunction in fast-spiking parvalbumin interneurons (PV-INs) may represent an early pathophysiological perturbation in Alzheimer's Disease (AD). Defining early proteomic alterations in PV-INs can provide key biological and translationally-relevant insights. We used cell-type-specific in-vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state PV-IN proteomes. PV-IN proteomic signatures include high metabolic and translational activity, with over-representation of AD-risk and cognitive resilience-related proteins. In bulk proteomes, PV-IN proteins were associated with cognitive decline in humans, and with progressive neuropathology in humans and the 5xFAD mouse model of Aß pathology. PV-IN CIBOP in early stages of Aß pathology revealed signatures of increased mitochondria and metabolism, synaptic and cytoskeletal disruption and decreased mTOR signaling, not apparent in whole-brain proteomes. Furthermore, we demonstrated pre-synaptic defects in PV-to-excitatory neurotransmission, validating our proteomic findings. Overall, in this study we present native-state proteomes of PV-INs, revealing molecular insights into their unique roles in cognitive resiliency and AD pathogenesis.


Assuntos
Doença de Alzheimer , Camundongos , Humanos , Animais , Doença de Alzheimer/metabolismo , Parvalbuminas/metabolismo , Proteômica , Proteoma/metabolismo , Interneurônios/metabolismo , Camundongos Transgênicos
7.
Cells ; 13(8)2024 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-38667267

RESUMO

The differential expression of transcription factors during embryonic development has been selected as the main feature to define the specific subclasses of spinal interneurons. However, recent studies based on single-cell RNA sequencing and transcriptomic experiments suggest that this approach might not be appropriate in the adult spinal cord, where interneurons show overlapping expression profiles, especially in the ventral region. This constitutes a major challenge for the identification and direct targeting of specific populations that could be involved in locomotor recovery after a traumatic spinal cord injury in adults. Current experimental therapies, including electrical stimulation, training, pharmacological treatments, or cell implantation, that have resulted in improvements in locomotor behavior rely on the modulation of the activity and connectivity of interneurons located in the surroundings of the lesion core for the formation of detour circuits. However, very few publications clarify the specific identity of these cells. In this work, we review the studies where premotor interneurons were able to create new intraspinal circuits after different kinds of traumatic spinal cord injury, highlighting the difficulties encountered by researchers, to classify these populations.


Assuntos
Interneurônios , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal , Adulto , Animais , Humanos , Interneurônios/metabolismo , Medula Espinal/citologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/terapia , Traumatismos da Medula Espinal/fisiopatologia
8.
Cell Rep ; 43(4): 114115, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38607918

RESUMO

In the CA1 hippocampus, vasoactive intestinal polypeptide-expressing interneurons (VIP-INs) play a prominent role in disinhibitory circuit motifs. However, the specific behavioral conditions that lead to circuit disinhibition remain uncertain. To investigate the behavioral relevance of VIP-IN activity, we employed wireless technologies allowing us to monitor and manipulate their function in freely behaving mice. Our findings reveal that, during spatial exploration in new environments, VIP-INs in the CA1 hippocampal region become highly active, facilitating the rapid encoding of novel spatial information. Remarkably, both VIP-INs and pyramidal neurons (PNs) exhibit increased activity when encountering novel changes in the environment, including context- and object-related alterations. Concurrently, somatostatin- and parvalbumin-expressing inhibitory populations show an inverse relationship with VIP-IN and PN activity, revealing circuit disinhibition that occurs on a timescale of seconds. Thus, VIP-IN-mediated disinhibition may constitute a crucial element in the rapid encoding of novelty and the acquisition of recognition memory.


Assuntos
Região CA1 Hipocampal , Interneurônios , Reconhecimento Psicológico , Peptídeo Intestinal Vasoativo , Animais , Interneurônios/metabolismo , Interneurônios/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Região CA1 Hipocampal/fisiologia , Região CA1 Hipocampal/metabolismo , Região CA1 Hipocampal/citologia , Camundongos , Masculino , Reconhecimento Psicológico/fisiologia , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Camundongos Endogâmicos C57BL , Memória/fisiologia , Parvalbuminas/metabolismo , Comportamento Exploratório/fisiologia , Somatostatina/metabolismo
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167178, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38636614

RESUMO

Pitt-Hopkins syndrome (PTHS) is a neurodevelopmental disorder caused by haploinsufficiency of transcription factor 4 (TCF4). In this work, we focused on the cerebral cortex and investigated in detail the progenitor cell dynamics and the outcome of neurogenesis in a PTHS mouse model. Labeling and quantification of progenitors and newly generated neurons at various time points during embryonic development revealed alterations affecting the dynamic of cortical progenitors since the earliest stages of cortex formation in PTHS mice. Consequently, establishment of neuronal populations and layering of the cortex were found to be altered in heterozygotes subjects at birth. Interestingly, defective layering process of pyramidal neurons was partially rescued by reintroducing TCF4 expression using focal in utero electroporation in the cerebral cortex. Coincidentally with a defective dorsal neurogenesis, we found that ventral generation of interneurons was also defective in this model, which may lead to an excitation/inhibition imbalance in PTHS. Overall, sex-dependent differences were detected with more marked effects evidenced in males compared with females. All of this contributes to expand our understanding of PTHS, paralleling the advances of research in autism spectrum disorder and further validating the PTHS mouse model as an important tool to advance preclinical studies.


Assuntos
Córtex Cerebral , Modelos Animais de Doenças , Hiperventilação , Deficiência Intelectual , Neurogênese , Fator de Transcrição 4 , Animais , Fator de Transcrição 4/metabolismo , Fator de Transcrição 4/genética , Feminino , Masculino , Camundongos , Hiperventilação/metabolismo , Hiperventilação/genética , Hiperventilação/patologia , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Deficiência Intelectual/metabolismo , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Fácies , Caracteres Sexuais , Interneurônios/metabolismo , Interneurônios/patologia , Células Piramidais/metabolismo , Células Piramidais/patologia , Haploinsuficiência
10.
Dis Model Mech ; 17(5)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38616770

RESUMO

Dystonia is thought to arise from abnormalities in the motor loop of the basal ganglia; however, there is an ongoing debate regarding cerebellar involvement. We adopted an established cerebellar dystonia mouse model by injecting ouabain to examine the contribution of the cerebellum. Initially, we examined whether the entopeduncular nucleus (EPN), substantia nigra pars reticulata (SNr), globus pallidus externus (GPe) and striatal neurons were activated in the model. Next, we examined whether administration of a dopamine D1 receptor agonist and dopamine D2 receptor antagonist or selective ablation of striatal parvalbumin (PV, encoded by Pvalb)-expressing interneurons could modulate the involuntary movements of the mice. The cerebellar dystonia mice had a higher number of cells positive for c-fos (encoded by Fos) in the EPN, SNr and GPe, as well as a higher positive ratio of c-fos in striatal PV interneurons, than those in control mice. Furthermore, systemic administration of combined D1 receptor agonist and D2 receptor antagonist and selective ablation of striatal PV interneurons relieved the involuntary movements of the mice. Abnormalities in the motor loop of the basal ganglia could be crucially involved in cerebellar dystonia, and modulating PV interneurons might provide a novel treatment strategy.


Assuntos
Corpo Estriado , Modelos Animais de Doenças , Distonia , Interneurônios , Parvalbuminas , Proteínas Proto-Oncogênicas c-fos , Receptores de Dopamina D2 , Animais , Interneurônios/metabolismo , Interneurônios/efeitos dos fármacos , Parvalbuminas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Distonia/patologia , Distonia/metabolismo , Distonia/fisiopatologia , Corpo Estriado/patologia , Corpo Estriado/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D1/metabolismo , Cerebelo/patologia , Cerebelo/metabolismo , Ouabaína/farmacologia , Camundongos Endogâmicos C57BL , Camundongos , Masculino
11.
Nature ; 629(8011): 402-409, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38632412

RESUMO

Throughout life, neuronal networks in the mammalian neocortex maintain a balance of excitation and inhibition, which is essential for neuronal computation1,2. Deviations from a balanced state have been linked to neurodevelopmental disorders, and severe disruptions result in epilepsy3-5. To maintain balance, neuronal microcircuits composed of excitatory and inhibitory neurons sense alterations in neural activity and adjust neuronal connectivity and function. Here we identify a signalling pathway in the adult mouse neocortex that is activated in response to increased neuronal network activity. Overactivation of excitatory neurons is signalled to the network through an increase in the levels of BMP2, a growth factor that is well known for its role as a morphogen in embryonic development. BMP2 acts on parvalbumin-expressing (PV) interneurons through the transcription factor SMAD1, which controls an array of glutamatergic synapse proteins and components of perineuronal nets. PV-interneuron-specific disruption of BMP2-SMAD1 signalling is accompanied by a loss of glutamatergic innervation in PV cells, underdeveloped perineuronal nets and decreased excitability. Ultimately, this impairment of the functional recruitment of PV interneurons disrupts the cortical excitation-inhibition balance, with mice exhibiting spontaneous epileptic seizures. Our findings suggest that developmental morphogen signalling is repurposed to stabilize cortical networks in the adult mammalian brain.


Assuntos
Proteína Morfogenética Óssea 2 , Interneurônios , Neocórtex , Parvalbuminas , Transdução de Sinais , Proteína Smad1 , Animais , Proteína Smad1/metabolismo , Camundongos , Interneurônios/metabolismo , Neocórtex/metabolismo , Neocórtex/citologia , Parvalbuminas/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Masculino , Feminino , Neurônios/metabolismo , Inibição Neural , Epilepsia/metabolismo , Epilepsia/fisiopatologia , Sinapses/metabolismo , Rede Nervosa/metabolismo
12.
eNeuro ; 11(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38658137

RESUMO

The primary motor cortex (M1) integrates sensory and cognitive inputs to generate voluntary movement. Its functional impairments have been implicated in the pathophysiology of motor symptoms in Parkinson's disease (PD). Specifically, dopaminergic degeneration and basal ganglia dysfunction entrain M1 neurons into the abnormally synchronized bursting pattern of activity throughout the cortico-basal ganglia-thalamocortical network. However, how degeneration of the midbrain dopaminergic neurons affects the anatomy, microcircuit connectivity, and function of the M1 network remains poorly understood. The present study examined whether and how the loss of dopamine (DA) affects the morphology, cellular excitability, and synaptic physiology of Layer 5 parvalbumin-expressing (PV+) cells in the M1 of mice of both sexes. Here, we reported that loss of midbrain dopaminergic neurons does not alter the number, morphology, and physiology of Layer 5 PV+ cells in M1. Moreover, we demonstrated that the number of perisomatic PV+ puncta of M1 pyramidal neurons as well as their functional innervation of cortical pyramidal neurons were not altered following the loss of DA. Together, the present study documents an intact GABAergic inhibitory network formed by PV+ cells following the loss of midbrain dopaminergic neurons.


Assuntos
Neurônios Dopaminérgicos , Interneurônios , Mesencéfalo , Camundongos Transgênicos , Córtex Motor , Parvalbuminas , Animais , Parvalbuminas/metabolismo , Córtex Motor/metabolismo , Neurônios Dopaminérgicos/metabolismo , Interneurônios/metabolismo , Masculino , Feminino , Mesencéfalo/metabolismo , Neurônios GABAérgicos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos , Inibição Neural/fisiologia
13.
Elife ; 122024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38655918

RESUMO

Obstructive sleep apnea (OSA) is a prevalent sleep-related breathing disorder that results in multiple bouts of intermittent hypoxia. OSA has many neurological and systemic comorbidities, including dysphagia, or disordered swallow, and discoordination with breathing. However, the mechanism in which chronic intermittent hypoxia (CIH) causes dysphagia is unknown. Recently, we showed the postinspiratory complex (PiCo) acts as an interface between the swallow pattern generator (SPG) and the inspiratory rhythm generator, the preBötzinger complex, to regulate proper swallow-breathing coordination (Huff et al., 2023). PiCo is characterized by interneurons co-expressing transporters for glutamate (Vglut2) and acetylcholine (ChAT). Here we show that optogenetic stimulation of ChATcre:Ai32, Vglut2cre:Ai32, and ChATcre:Vglut2FlpO:ChR2 mice exposed to CIH does not alter swallow-breathing coordination, but unexpectedly disrupts swallow behavior via triggering variable swallow motor patterns. This suggests that glutamatergic-cholinergic neurons in PiCo are not only critical for the regulation of swallow-breathing coordination, but also play an important role in the modulation of swallow motor patterning. Our study also suggests that swallow disruption, as seen in OSA, involves central nervous mechanisms interfering with swallow motor patterning and laryngeal activation. These findings are crucial for understanding the mechanisms underlying dysphagia, both in OSA and other breathing and neurological disorders.


Assuntos
Deglutição , Hipóxia , Animais , Camundongos , Deglutição/fisiologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Masculino , Optogenética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Apneia Obstrutiva do Sono/fisiopatologia , Apneia Obstrutiva do Sono/metabolismo , Neurônios Colinérgicos/fisiologia , Neurônios Colinérgicos/metabolismo , Interneurônios/fisiologia , Interneurônios/metabolismo , Respiração , Feminino
14.
Proc Natl Acad Sci U S A ; 121(18): e2322550121, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38657053

RESUMO

Pronounced differences in neurotransmitter release from a given presynaptic neuron, depending on the synaptic target, are among the most intriguing features of cortical networks. Hippocampal pyramidal cells (PCs) release glutamate with low probability to somatostatin expressing oriens-lacunosum-moleculare (O-LM) interneurons (INs), and the postsynaptic responses show robust short-term facilitation, whereas the release from the same presynaptic axons onto fast-spiking INs (FSINs) is ~10-fold higher and the excitatory postsynaptic currents (EPSCs) display depression. The mechanisms underlying these vastly different synaptic behaviors have not been conclusively identified. Here, we applied a combined functional, pharmacological, and modeling approach to address whether the main difference lies in the action potential-evoked fusion or else in upstream priming processes of synaptic vesicles (SVs). A sequential two-step SV priming model was fitted to the peak amplitudes of unitary EPSCs recorded in response to complex trains of presynaptic stimuli in acute hippocampal slices of adult mice. At PC-FSIN connections, the fusion probability (Pfusion) of well-primed SVs is 0.6, and 44% of docked SVs are in a fusion-competent state. At PC-O-LM synapses, Pfusion is only 40% lower (0.36), whereas the fraction of well-primed SVs is 6.5-fold smaller. Pharmacological enhancement of fusion by 4-AP and priming by PDBU was recaptured by the model with a selective increase of Pfusion and the fraction of well-primed SVs, respectively. Our results demonstrate that the low fidelity of transmission at PC-O-LM synapses can be explained by a low occupancy of the release sites by well-primed SVs.


Assuntos
Neurotransmissores , Vesículas Sinápticas , Animais , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/fisiologia , Camundongos , Neurotransmissores/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Transmissão Sináptica/fisiologia , Interneurônios/metabolismo , Interneurônios/fisiologia , Células Piramidais/metabolismo , Células Piramidais/fisiologia , Sinapses/metabolismo , Sinapses/fisiologia , Modelos Neurológicos
15.
ACS Chem Neurosci ; 15(9): 1738-1754, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38613458

RESUMO

Iboga alkaloids, also known as coronaridine congeners, have shown promise in the treatment of alcohol and opioid use disorders. The objective of this study was to evaluate the effects of catharanthine and 18-methoxycoronaridine (18-MC) on dopamine (DA) transmission and cholinergic interneurons in the mesolimbic DA system, nicotine-induced locomotor activity, and nicotine-taking behavior. Utilizing ex vivo fast-scan cyclic voltammetry (FSCV) in the nucleus accumbens core of male mice, we found that catharanthine or 18-MC differentially inhibited evoked DA release. Catharanthine inhibition of evoked DA release was significantly reduced by both α4 and α6 nicotinic acetylcholine receptors (nAChRs) antagonists. Additionally, catharanthine substantially increased DA release more than vehicle during high-frequency stimulation, although less potently than an α4 nAChR antagonist, which confirms previous work with nAChR antagonists. Interestingly, while catharanthine slowed DA reuptake measured via FSCV ex vivo, it also increased extracellular DA in striatal dialysate from anesthetized mice in vivo in a dose-dependent manner. Superfusion of catharanthine or 18-MC inhibited the firing rate of striatal cholinergic interneurons in a concentration dependent manner, which are known to potently modulate presynaptic DA release. Catharanthine or 18-MC suppressed acetylcholine currents in oocytes expressing recombinant rat α6/α3ß2ß3 or α6/α3ß4 nAChRs. In behavioral experiments using male Sprague-Dawley rats, systemic administration of catharanthine or 18-MC blocked nicotine enhancement of locomotor activity. Importantly, catharanthine attenuated nicotine self-administration in a dose-dependent manner while having no effect on food reinforcement. Lastly, administration of catharanthine and nicotine together greatly increased head twitch responses, indicating a potential synergistic hallucinogenic effect. These findings demonstrate that catharanthine and 18-MC have similar, but not identical effects on striatal DA dynamics, striatal cholinergic interneuron activity and nicotine psychomotor effects.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina , Dopamina , Ibogaína , Ibogaína/análogos & derivados , Nicotina , Receptores Nicotínicos , Animais , Dopamina/metabolismo , Masculino , Receptores Nicotínicos/metabolismo , Receptores Nicotínicos/efeitos dos fármacos , Nicotina/farmacologia , Ibogaína/farmacologia , Camundongos , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Camundongos Endogâmicos C57BL , Antagonistas Nicotínicos/farmacologia , Oócitos/efeitos dos fármacos , Agonistas Nicotínicos/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Autoadministração , Xenopus laevis , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Relação Dose-Resposta a Droga , Atividade Motora/efeitos dos fármacos
16.
eNeuro ; 11(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38637152

RESUMO

Canonically, action potentials of most mammalian neurons initiate at the axon initial segment (AIS) and propagate bidirectionally: orthodromically along the distal axon and retrogradely into the soma and dendrites. Under some circumstances, action potentials may initiate ectopically, at sites distal to the AIS, and propagate antidromically along the axon. These "ectopic action potentials" (EAPs) have been observed in experimental models of seizures and chronic pain, and more rarely in nonpathological forebrain neurons. Here we report that a large majority of parvalbumin-expressing (PV+) interneurons in the upper layers of mouse neocortex, from both orbitofrontal and primary somatosensory areas, fire EAPs after sufficient activation of their somata. Somatostatin-expressing interneurons also fire EAPs, though less robustly. Ectopic firing in PV+ cells occurs in varying temporal patterns and can persist for several seconds. PV+ cells evoke strong synaptic inhibition in pyramidal neurons and interneurons and play critical roles in cortical function. Our results suggest that ectopic spiking of PV+ interneurons is common and may contribute to both normal and pathological network functions of the neocortex.


Assuntos
Potenciais de Ação , Interneurônios , Camundongos Transgênicos , Neocórtex , Parvalbuminas , Animais , Parvalbuminas/metabolismo , Interneurônios/fisiologia , Interneurônios/metabolismo , Neocórtex/fisiologia , Potenciais de Ação/fisiologia , Masculino , Camundongos , Feminino , Camundongos Endogâmicos C57BL , Células Piramidais/fisiologia , Somatostatina/metabolismo
17.
PLoS One ; 19(4): e0301592, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38635806

RESUMO

Hippocampal interneurons are a very diverse population of cells. Using single-cell quantitative PCR to analyze rat CA1 hippocampal interneurons, we quantified neuronal nicotinic acetylcholine receptor (nAChR) mRNA subunit expression and detailed possible nAChR subtype combinations for the α2, α3, α4, α5, α7, ß2, ß3, and ß4 subunits. We also compared the expression detected in the stratum oriens and the stratum radiatum hippocampal layers. We show that the majority of interneurons in the CA1 of the rat hippocampus contain detectable levels of nAChR subunit mRNA. Our results highlight the complexity of the CA1 nAChR population. Interestingly, the α3 nAChR subunit is one of the highest expressed subunit mRNAs in this population, while the α4 is one of the least likely subunits to be detected in CA1 interneurons. The ß2 nAChR subunit is the highest expressed beta subunit mRNA in these cells. In addition, Pearson's correlation coefficient values are calculated to identify significant differences between the nAChR subunit combinations expressed in the CA1 stratum oriens and the stratum radiatum. Statistical analysis also indicates that there are likely over 100 different nAChR subunit mRNA combinations expressed in rat CA1 interneurons. These results provide a valid avenue for identifying nAChR subtype targets that may be effective hippocampus-specific pharmacological targets.


Assuntos
Receptores Nicotínicos , Ratos , Animais , RNA Mensageiro/metabolismo , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Interneurônios/metabolismo , Neurônios/metabolismo , Hipocampo/metabolismo
18.
J Nutr Sci Vitaminol (Tokyo) ; 70(2): 164-173, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38684387

RESUMO

Bitterness and astringency are the aversive tastes in mammals. In humans, aversion to bitterness and astringency may be reduced depending on the eating experience. However, the cellular and molecular mechanisms underlying plasticity in preference to bitter and astringent tastants remain unknown. This study aimed to investigate the preference plasticity to bitter and astringent tea polyphenols, including catechins and tannic acids, in the model animal Caenorhabditis elegans. C. elegans showed avoidance behavior against epigallocatechin gallate (EGCG), tannic acid, and theaflavin. However, they displayed diminishing avoidance against EGCG depending on their EGCG-feeding regime at larval stages. Additionally, the behavioral plasticity in avoiding EGCG required the transcription factor DAF-16/FOXO. Isoform-specific deletion mutant analysis and cell-specific rescue analysis revealed that the function of daf-16 isoform b in AIY interneurons is necessary for experience-dependent behavioral plasticity to EGCG.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Catequina , Fatores de Transcrição Forkhead , Interneurônios , Animais , Catequina/análogos & derivados , Catequina/farmacologia , Caenorhabditis elegans/efeitos dos fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Fatores de Transcrição Forkhead/metabolismo , Interneurônios/efeitos dos fármacos , Interneurônios/metabolismo , Aprendizagem da Esquiva/efeitos dos fármacos , Biflavonoides/farmacologia , Paladar/efeitos dos fármacos , Chá/química , Comportamento Animal/efeitos dos fármacos , Larva/efeitos dos fármacos
19.
PLoS One ; 19(3): e0298105, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38551921

RESUMO

The nematode Caenorhabditis elegans is a widely used model organism for neuroscience. Although its nervous system has been fully reconstructed, the physiological bases of single-neuron functioning are still poorly explored. Recently, many efforts have been dedicated to measuring signals from C. elegans neurons, revealing a rich repertoire of dynamics, including bistable responses, graded responses, and action potentials. Still, biophysical models able to reproduce such a broad range of electrical responses lack. Realistic electrophysiological descriptions started to be developed only recently, merging gene expression data with electrophysiological recordings, but with a large variety of cells yet to be modeled. In this work, we contribute to filling this gap by providing biophysically accurate models of six classes of C. elegans neurons, the AIY, RIM, and AVA interneurons, and the VA, VB, and VD motor neurons. We test our models by comparing computational and experimental time series and simulate knockout neurons, to identify the biophysical mechanisms at the basis of inter and motor neuron functioning. Our models represent a step forward toward the modeling of C. elegans neuronal networks and virtual experiments on the nematode nervous system.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Humanos , Animais , Caenorhabditis elegans/metabolismo , Interneurônios/metabolismo , Neurônios Motores/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sistema Nervoso/metabolismo
20.
Curr Biol ; 34(7): 1561-1568.e4, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38479389

RESUMO

The basolateral amygdala (BLA) mediates both fear and reward learning.1,2 Previous work has shown that parvalbumin (PV) interneurons in the BLA contribute to BLA oscillatory states integral to fear expression.3,4,5,6,7 However, despite it being critical to our understanding of reward behaviors, it is unknown whether BLA oscillatory states and PV interneurons similarly contribute to reward processing. Local field potentials in the BLA were collected as male and female mice consumed sucrose reward, where prominent changes in the beta band (15-30 Hz) emerged with reward experience. During consumption of one water bottle during a two-water-bottle choice test, rhythmic optogenetic stimulation of BLA PVs produced a robust bottle preference, showing that PVs can sufficiently drive reward seeking. Finally, to demonstrate that PV activity is necessary for reward value use, PVs were chemogenetically inhibited following outcome devaluation, rendering mice incapable of using updated reward representations to guide their behavior. Taken together, these experiments provide novel information about the physiological signatures of reward while highlighting BLA PV interneuron contributions to behaviors that are BLA dependent. This work builds upon established knowledge of PV involvement in fear expression and provides evidence that PV orchestration of unique BLA network states is involved in both learning types.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Camundongos , Masculino , Feminino , Animais , Complexo Nuclear Basolateral da Amígdala/fisiologia , Parvalbuminas/metabolismo , Aprendizagem/fisiologia , Interneurônios/metabolismo , Recompensa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA