Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Nature ; 607(7918): 321-329, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35676479

RESUMO

Although bradykinesia, tremor and rigidity are the hallmark motor defects in patients with Parkinson's disease (PD), patients also experience motor learning impairments and non-motor symptoms such as depression1. The neural circuit basis for these different symptoms of PD are not well understood. Although current treatments are effective for locomotion deficits in PD2,3, therapeutic strategies targeting motor learning deficits and non-motor symptoms are lacking4-6. Here we found that distinct parafascicular (PF) thalamic subpopulations project to caudate putamen (CPu), subthalamic nucleus (STN) and nucleus accumbens (NAc). Whereas PF→CPu and PF→STN circuits are critical for locomotion and motor learning, respectively, inhibition of the PF→NAc circuit induced a depression-like state. Whereas chemogenetically manipulating CPu-projecting PF neurons led to a long-term restoration of locomotion, optogenetic long-term potentiation (LTP) at PF→STN synapses restored motor learning behaviour in an acute mouse model of PD. Furthermore, activation of NAc-projecting PF neurons rescued depression-like phenotypes. Further, we identified nicotinic acetylcholine receptors capable of modulating PF circuits to rescue different PD phenotypes. Thus, targeting PF thalamic circuits may be an effective strategy for treating motor and non-motor deficits in PD.


Assuntos
Afeto , Destreza Motora , Vias Neurais , Doença de Parkinson , Tálamo , Animais , Modelos Animais de Doenças , Aprendizagem , Locomoção , Potenciação de Longa Duração , Camundongos , Neurônios/fisiologia , Núcleo Accumbens , Optogenética , Doença de Parkinson/fisiopatologia , Doença de Parkinson/psicologia , Doença de Parkinson/terapia , Putamen , Receptores Nicotínicos , Núcleo Subtalâmico , Sinapses , Tálamo/citologia , Tálamo/patologia
2.
Nature ; 593(7857): 114-118, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33790466

RESUMO

Innate social behaviours, such as mating and fighting, are fundamental to animal reproduction and survival1. However, social engagements can also put an individual at risk2. Little is known about the neural mechanisms that enable appropriate risk assessment and the suppression of hazardous social interactions. Here we identify the posteromedial nucleus of the cortical amygdala (COApm) as a locus required for the suppression of male mating when a female mouse is unhealthy. Using anatomical tracing, functional imaging and circuit-level epistatic analyses, we show that suppression of mating with an unhealthy female is mediated by the COApm projections onto the glutamatergic population of the medial amygdalar nucleus (MEA). We further show that the role of the COApm-to-MEA connection in regulating male mating behaviour relies on the neuromodulator thyrotropin-releasing hormone (TRH). TRH is expressed in the COApm, whereas the TRH receptor (TRHR) is found in the postsynaptic MEA glutamatergic neurons. Manipulating neural activity of TRH-expressing neurons in the COApm modulated male mating behaviour. In the MEA, activation of the TRHR pathway by ligand infusion inhibited mating even towards healthy female mice, whereas genetic ablation of TRHR facilitated mating with unhealthy individuals. In summary, we reveal a neural pathway that relies on the neuromodulator TRH to modulate social interactions according to the health status of the reciprocating individual. Individuals must balance the cost of social interactions relative to the benefit, as deficits in the ability to select healthy mates may lead to the spread of disease.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Preferência de Acasalamento Animal/fisiologia , Vias Neurais/fisiologia , Comportamento Social , Animais , Copulação/fisiologia , Complexo Nuclear Corticomedial/citologia , Complexo Nuclear Corticomedial/metabolismo , Feminino , Ácido Glutâmico/metabolismo , Saúde , Ligantes , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Neurônios/metabolismo , Receptores do Hormônio Liberador da Tireotropina/metabolismo , Hormônio Liberador de Tireotropina/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(7): e2023481120, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-37053554

RESUMO

Monosynaptic tracing using rabies virus is an important technique in neuroscience, allowing brain-wide labeling of neurons directly presynaptic to a targeted neuronal population. A 2017 article reported the development of a noncytotoxic version-a major advance-based on attenuating the rabies virus by the addition of a destabilization domain to the C terminus of a viral protein. However, this modification did not appear to hinder the ability of the virus to spread between neurons. We analyzed two viruses provided by the authors and show here that both were mutants that had lost the intended modification, explaining the paper's paradoxical results. We then made a virus that actually did have the intended modification in at least the majority of virions and found that it did not spread efficiently under the conditions described in the original paper, namely, without an exogenous protease being expressed in order to remove the destabilization domain. We found that it did spread when the protease was supplied, although this also appeared to result in the deaths of most source cells by 3 wk postinjection. We conclude that the new approach is not robust but that it could become a viable technique given further optimization and validation.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/metabolismo , Neurônios/metabolismo , Proteínas Virais/metabolismo , Encéfalo/metabolismo , Peptídeo Hidrolases/metabolismo
4.
J Transl Med ; 21(1): 258, 2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37061718

RESUMO

BACKGROUND: Alzheimer's disease (AD) is characterized by a progressive loss of memory that cannot be efficiently managed by currently available AD therapeutics. So far, most treatments for AD that have the potential to improve memory target neural circuits to protect their integrity. However, the vulnerable neural circuits and their dynamic remodeling during AD progression remain largely undefined. METHODS: Circuit-based approaches, including anterograde and retrograde tracing, slice electrophysiology, and fiber photometry, were used to investigate the dynamic structural and functional remodeling of a GABAergic circuit projected from the medial septum (MS) to the dentate gyrus (DG) in 3xTg-AD mice during AD progression. RESULTS: We identified a long-distance GABAergic circuit that couples highly connected MS and DG GABAergic neurons during spatial memory encoding. Furthermore, we found hyperactivity of DG interneurons during early AD, which persisted into late AD stages. Interestingly, MS GABAergic projections developed a series of adaptive strategies to combat DG interneuron hyperactivity. During early-stage AD, MS-DG GABAergic projections exhibit increased inhibitory synaptic strength onto DG interneurons to inhibit their activities. During late-stage AD, MS-DG GABAergic projections form higher anatomical connectivity with DG interneurons and exhibit aberrant outgrowth to increase the inhibition onto DG interneurons. CONCLUSION: We report the structural and functional remodeling of the MS-DG GABAergic circuit during disease progression in 3xTg-AD mice. Dynamic MS-DG GABAergic circuit remodeling represents a compensatory mechanism to combat DG interneuron hyperactivity induced by reduced GABA transmission.


Assuntos
Doença de Alzheimer , Camundongos , Animais , Camundongos Transgênicos , Hipocampo
5.
Nature ; 548(7669): 582-587, 2017 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-28847002

RESUMO

Multiple populations of wake-promoting neurons have been characterized in mammals, but few sleep-promoting neurons have been identified. Wake-promoting cell types include hypocretin and GABA (γ-aminobutyric-acid)-releasing neurons of the lateral hypothalamus, which promote the transition to wakefulness from non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. Here we show that a subset of GABAergic neurons in the mouse ventral zona incerta, which express the LIM homeodomain factor Lhx6 and are activated by sleep pressure, both directly inhibit wake-active hypocretin and GABAergic cells in the lateral hypothalamus and receive inputs from multiple sleep-wake-regulating neurons. Conditional deletion of Lhx6 from the developing diencephalon leads to decreases in both NREM and REM sleep. Furthermore, selective activation and inhibition of Lhx6-positive neurons in the ventral zona incerta bidirectionally regulate sleep time in adult mice, in part through hypocretin-dependent mechanisms. These studies identify a GABAergic subpopulation of neurons in the ventral zona incerta that promote sleep.


Assuntos
Neurônios GABAérgicos/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Sono/fisiologia , Fatores de Transcrição/metabolismo , Zona Incerta/citologia , Ácido gama-Aminobutírico/metabolismo , Animais , Linhagem da Célula , Neurônios GABAérgicos/efeitos dos fármacos , Deleção de Genes , Hipocampo/citologia , Hipocampo/fisiologia , Proteínas com Homeodomínio LIM/deficiência , Proteínas com Homeodomínio LIM/efeitos dos fármacos , Proteínas com Homeodomínio LIM/genética , Masculino , Camundongos , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Orexinas/metabolismo , Terminações Pré-Sinápticas/metabolismo , Sono/efeitos dos fármacos , Sono/genética , Sono REM/efeitos dos fármacos , Sono REM/genética , Sono REM/fisiologia , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética , Vigília/efeitos dos fármacos , Vigília/genética , Vigília/fisiologia , Zona Incerta/efeitos dos fármacos , Zona Incerta/fisiologia
7.
Nature ; 549(7673): 482-487, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28902835

RESUMO

Viral infection during pregnancy is correlated with increased frequency of neurodevelopmental disorders, and this is studied in mice prenatally subjected to maternal immune activation (MIA). We previously showed that maternal T helper 17 cells promote the development of cortical and behavioural abnormalities in MIA-affected offspring. Here we show that cortical abnormalities are preferentially localized to a region encompassing the dysgranular zone of the primary somatosensory cortex (S1DZ). Moreover, activation of pyramidal neurons in this cortical region was sufficient to induce MIA-associated behavioural phenotypes in wild-type animals, whereas reduction in neural activity rescued the behavioural abnormalities in MIA-affected offspring. Sociability and repetitive behavioural phenotypes could be selectively modulated according to the efferent targets of S1DZ. Our work identifies a cortical region primarily, if not exclusively, centred on the S1DZ as the major node of a neural network that mediates behavioural abnormalities observed in offspring exposed to maternal inflammation.


Assuntos
Comportamento Animal , Inflamação/fisiopatologia , Transtornos Mentais/etiologia , Complicações Infecciosas na Gravidez/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Células Th17 , Animais , Feminino , Masculino , Transtornos Mentais/psicologia , Camundongos , Mães , Fenótipo , Gravidez , Células Piramidais/patologia , Células Piramidais/fisiologia , Comportamento Social , Córtex Somatossensorial/anormalidades , Córtex Somatossensorial/patologia , Córtex Somatossensorial/fisiopatologia , Células Th17/fisiologia
8.
Nature ; 520(7549): 675-8, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25925480

RESUMO

The ability to differentiate stimuli predicting positive or negative outcomes is critical for survival, and perturbations of emotional processing underlie many psychiatric disease states. Synaptic plasticity in the basolateral amygdala complex (BLA) mediates the acquisition of associative memories, both positive and negative. Different populations of BLA neurons may encode fearful or rewarding associations, but the identifying features of these populations and the synaptic mechanisms of differentiating positive and negative emotional valence have remained unknown. Here we show that BLA neurons projecting to the nucleus accumbens (NAc projectors) or the centromedial amygdala (CeM projectors) undergo opposing synaptic changes following fear or reward conditioning. We find that photostimulation of NAc projectors supports positive reinforcement while photostimulation of CeM projectors mediates negative reinforcement. Photoinhibition of CeM projectors impairs fear conditioning and enhances reward conditioning. We characterize these functionally distinct neuronal populations by comparing their electrophysiological, morphological and genetic features. Overall, we provide a mechanistic explanation for the representation of positive and negative associations within the amygdala.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Vias Neurais , Neurônios/fisiologia , Recompensa , Animais , Condicionamento Clássico , Medo/psicologia , Perfilação da Expressão Gênica , Potenciação de Longa Duração , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Motivação , Núcleo Accumbens/citologia , Núcleo Accumbens/fisiologia , Núcleo Accumbens/efeitos da radiação , Reforço Psicológico , Transcrição Gênica
9.
J Physiol ; 596(16): 3695-3707, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29808928

RESUMO

KEY POINTS: There are two electrophysiological dichotomous populations of parvalbumin (PV) interneurons located in the dorsal striatum. Striatal PV interneurons in medial and lateral regions differ significantly in their intrinsic excitability. Parvalbumin interneurons in the dorsomedial striatum, but not in the dorsolateral striatum, receive afferent glutamatergic input from cingulate cortex. ABSTRACT: Dorsomedial striatum circuitry is involved in goal-directed actions or movements that become habits upon repetition, as encoded by the dorsolateral striatum. An inability to shift from habits can compromise action-control and prevent behavioural adaptation. Although these regions appear to be clearly behaviourally distinct, little is known about their distinct physiology. Parvalbumin (PV) interneurons are a major source of striatal inhibition and are usually considered as a homogeneous population in the entire dorsal striatum. In the present study, we recorded PV interneurons in dorsal striatum slices from wild-type male mice and suggest the existence of two electrophysiological dichotomous populations. We found that PV interneurons located at the dorsomedial striatum region have increased intrinsic excitability compared to PV interneurons in dorsolateral region. We also found that PV interneurons in the dorsomedial region, but not in the dorsolateral striatum region, receive short-latency excitatory inputs from cingulate cortex. Therefore, the results of the present study demonstrate the importance of considering region specific parvalbumin interneuron populations when studying dorsal striatal function.


Assuntos
Corpo Estriado/fisiologia , Lateralidade Funcional , Ácido Glutâmico/metabolismo , Interneurônios/fisiologia , Parvalbuminas/fisiologia , Vias Aferentes , Animais , Corpo Estriado/citologia , Interneurônios/citologia , Masculino , Camundongos , Camundongos Knockout
10.
J Neurosci ; 33(37): 14889-98, 2013 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-24027288

RESUMO

The enormous potential of modern molecular neuroanatomical tools lies in their ability to determine the precise connectivity of the neuronal cell types comprising the innate circuitry of the brain. We used transgenically targeted viral tracing to identify the monosynaptic inputs to the projection neurons of layer II of medial entorhinal cortex (MEC-LII) in mice. These neurons are not only major inputs to the hippocampus, the structure most clearly implicated in learning and memory, they also are "grid cells." Here we address the question of what kinds of inputs are specifically targeting these MEC-LII cells. Cell-specific infection of MEC-LII with recombinant rabies virus results in unambiguous labeling of monosynaptic inputs. Furthermore, ratios of labeled neurons in different regions are largely consistent between animals, suggesting that label reflects density of innervation. While the results mostly confirm prior anatomical work, they also reveal a novel major direct input to MEC-LII from hippocampal pyramidal neurons. Interestingly, the vast majority of these direct hippocampal inputs arise not from the major hippocampal subfields of CA1 and CA3, but from area CA2, a region that has historically been thought to merely be a transitional zone between CA3 and CA1. We confirmed this unexpected result using conventional tracing techniques in both rats and mice.


Assuntos
Região CA2 Hipocampal/citologia , Córtex Entorrinal/fisiologia , Vias Neurais/fisiologia , Animais , Mapeamento Encefálico , Região CA2 Hipocampal/fisiologia , Contagem de Células , Córtex Entorrinal/citologia , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Vírus da Raiva/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
11.
Nat Neurosci ; 27(2): 373-383, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38212587

RESUMO

Rabies-virus-based monosynaptic tracing is a widely used technique for mapping neural circuitry, but its cytotoxicity has confined it primarily to anatomical applications. Here we present a second-generation system for labeling direct inputs to targeted neuronal populations with minimal toxicity, using double-deletion-mutant rabies viruses. Viral spread requires expression of both deleted viral genes in trans in postsynaptic source cells. Suppressing this expression with doxycycline following an initial period of viral replication reduces toxicity to postsynaptic cells. Longitudinal two-photon imaging in vivo indicated that over 90% of both presynaptic and source cells survived for the full 12-week course of imaging. Ex vivo whole-cell recordings at 5 weeks postinfection showed that the second-generation system perturbs input and source cells much less than the first-generation system. Finally, two-photon calcium imaging of labeled networks of visual cortex neurons showed that their visual response properties appeared normal for 10 weeks, the longest we followed them.


Assuntos
Vírus da Raiva , Vírus da Raiva/genética , Neurônios/fisiologia , Replicação Viral
12.
J Neurosci ; 32(14): 4992-5001, 2012 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-22492054

RESUMO

The generation of purposive movement by mammals involves coordinated activity in the corticospinal and corticostriatal systems, which are involved in different aspects of motor control. In the motor cortex, corticospinal and corticostriatal neurons are closely intermingled, raising the question of whether and how information flows intracortically within and across these two channels. To explore this, we developed an optogenetic technique based on retrograde transfection of neurons with deletion-mutant rabies virus encoding channelrhodopsin-2, and used this in conjunction with retrograde anatomical labeling to stimulate and record from identified projection neurons in mouse motor cortex. We also used paired recordings to measure unitary connections. Both corticospinal and callosally projecting corticostriatal neurons in layer 5B formed within-class (recurrent) connections, with higher connection probability among corticostriatal than among corticospinal neurons. In contrast, across-class connectivity was extraordinarily asymmetric, essentially unidirectional from corticostriatal to corticospinal. Corticostriatal neurons in layer 5A and corticocortical neurons (callosal projection neurons similar to corticostriatal neurons) similarly received a paucity of corticospinal input. Connections involving presynaptic corticostriatal neurons had greater synaptic depression, and those involving postsynaptic corticospinal neurons had faster decaying EPSPs. Consequently, the three connections displayed a diversity of dynamic properties reflecting the different combinations of presynaptic and postsynaptic projection neurons. Collectively, these findings delineate a four-way specialized excitatory microcircuit formed by corticospinal and corticostriatal neurons. The "rectifying" corticostriatal-to-corticospinal connectivity implies a hierarchical organization and functional compartmentalization of corticospinal activity via unidirectional signaling from higher-order (corticostriatal) to lower-order (corticospinal) output neurons.


Assuntos
Corpo Estriado/fisiologia , Córtex Motor/fisiologia , Rede Nervosa/fisiologia , Tratos Piramidais/fisiologia , Animais , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/fisiologia
13.
J Neurosci ; 32(20): 7021-33, 2012 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22593070

RESUMO

In motor cortex, long-range output to subcortical motor circuits depends on excitatory and inhibitory inputs converging on projection neurons in layers 5A/B. How interneurons interconnect with these projection neurons, and whether these microcircuits are interneuron and/or projection specific, is unclear. We found that fast-spiking interneurons received strong intralaminar (horizontal) excitation from pyramidal neurons in layers 5A/B including corticostriatal and corticospinal neurons, implicating them in mediating disynaptic recurrent, feedforward, and feedback inhibition within and across the two projection classes. Low-threshold-spiking (LTS) interneurons were instead strongly excited by descending interlaminar (vertical) input from layer 2/3 pyramidal neurons, implicating them in mediating disynaptic feedforward inhibition to both projection classes. Furthermore, in a novel pattern, lower layer 2/3 preferentially excited interneurons in one layer (5A/LTS) and excitatory neurons in another (5B/corticospinal). Thus, these inhibitory microcircuits in mouse motor cortex follow an orderly arrangement that is laminarly orthogonalized by interneuron-specific, projection-nonspecific connectivity.


Assuntos
Corpo Estriado/fisiologia , Interneurônios/fisiologia , Córtex Motor/fisiologia , Tratos Piramidais/fisiologia , Potenciais de Ação/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Inibição Neural/fisiologia , Vias Neurais/fisiologia , Técnicas de Rastreamento Neuroanatômico/métodos , Células Piramidais/fisiologia
14.
Proc Natl Acad Sci U S A ; 107(50): 21848-53, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21115815

RESUMO

We describe a powerful system for revealing the direct monosynaptic inputs to specific cell types in Cre-expressing transgenic mice through the use of Cre-dependent helper virus and a modified rabies virus. We generated helper viruses that target gene expression to Cre-expressing cells, allowing us to control initial rabies virus infection and subsequent monosynaptic retrograde spread. Investigators can use this system to elucidate the connections onto a desired cell type in a high-throughput manner, limited only by the availability of Cre mouse lines. This method allows for identification of circuits that would be extremely tedious or impossible to study with other methods and can be used to build subcircuit maps of inputs onto many different types of cells within the same brain region. Furthermore, by expressing various transgenes from the rabies genome, this system also has the potential to allow manipulation of targeted neuronal circuits without perturbing neighboring cells.


Assuntos
Integrases/metabolismo , Vírus da Raiva/metabolismo , Coloração e Rotulagem/métodos , Sinapses/metabolismo , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Vírus Auxiliares/genética , Vírus Auxiliares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/citologia , Neurônios/fisiologia , Vírus da Raiva/genética , Sinapses/ultraestrutura , Transgenes
15.
bioRxiv ; 2023 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-36993334

RESUMO

Mapping the connectivity of diverse neuronal types provides the foundation for understanding the structure and function of neural circuits. High-throughput and low-cost neuroanatomical techniques based on RNA barcode sequencing have the potential to map circuits at cellular resolution and a brain-wide scale, but existing Sindbis virus-based techniques can only map long-range projections using anterograde tracing approaches. Rabies virus can complement anterograde tracing approaches by enabling either retrograde labeling of projection neurons or monosynaptic tracing of direct inputs to genetically targeted postsynaptic neurons. However, barcoded rabies virus has so far been only used to map non-neuronal cellular interactions in vivo and synaptic connectivity of cultured neurons. Here we combine barcoded rabies virus with single-cell and in situ sequencing to perform retrograde labeling and transsynaptic labeling in the mouse brain. We sequenced 96 retrogradely labeled cells and 295 transsynaptically labeled cells using single-cell RNA-seq, and 4,130 retrogradely labeled cells and 2,914 transsynaptically labeled cells in situ. We found that the transcriptomic identities of rabies virus-infected cells can be robustly identified using both single-cell RNA-seq and in situ sequencing. By associating gene expression with connectivity inferred from barcode sequencing, we distinguished long-range projecting cortical cell types from multiple cortical areas and identified cell types with converging or diverging synaptic connectivity. Combining in situ sequencing with barcoded rabies virus complements existing sequencing-based neuroanatomical techniques and provides a potential path for mapping synaptic connectivity of neuronal types at scale.

16.
Neuron ; 111(9): 1486-1503.e7, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-36893756

RESUMO

Extracting the valence of environmental cues is critical for animals' survival. How valence in sensory signals is encoded and transformed to produce distinct behavioral responses remains not well understood. Here, we report that the mouse pontine central gray (PCG) contributes to encoding both negative and positive valences. PCG glutamatergic neurons were activated selectively by aversive, but not reward, stimuli, whereas its GABAergic neurons were preferentially activated by reward signals. The optogenetic activation of these two populations resulted in avoidance and preference behavior, respectively, and was sufficient to induce conditioned place aversion/preference. Suppression of them reduced sensory-induced aversive and appetitive behaviors, respectively. These two functionally opponent populations, receiving a broad range of inputs from overlapping yet distinct sources, broadcast valence-specific information to a distributed brain network with distinguishable downstream effectors. Thus, PCG serves as a critical hub to process positive and negative valences of incoming sensory signals and drive valence-specific behaviors with distinct circuits.


Assuntos
Encéfalo , Neurônios GABAérgicos , Camundongos , Animais , Substância Cinzenta Periaquedutal , Afeto , Sinais (Psicologia)
17.
Cell Rep Methods ; 3(11): 100644, 2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37989085

RESUMO

Rabies viral vectors have become important components of the systems neuroscience toolkit, allowing both direct retrograde targeting of projection neurons and monosynaptic tracing of inputs to defined postsynaptic populations, but the rapid cytotoxicity of first-generation (ΔG) vectors limits their use to short-term experiments. We recently introduced second-generation, double-deletion-mutant (ΔGL) rabies viral vectors, showing that they efficiently retrogradely infect projection neurons and express recombinases effectively but with little to no detectable toxicity; more recently, we have shown that ΔGL viruses can be used for monosynaptic tracing with far lower cytotoxicity than the first-generation system. Here, we introduce third-generation (ΔL) rabies viral vectors, which appear to be as nontoxic as second-generation ones but have the major advantage of growing to much higher titers, resulting in significantly increased numbers of retrogradely labeled neurons in vivo.


Assuntos
Vírus da Raiva , Raiva , Humanos , Vírus da Raiva/genética , Interneurônios , Vetores Genéticos/genética , Neurônios
18.
J Comp Neurol ; 530(11): 1992-2013, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35383929

RESUMO

The rodent homolog of the primate pulvinar, the lateral posterior (LP) thalamus, is extensively interconnected with multiple cortical areas. While these cortical interactions can span the entire LP, subdivisions of the LP are characterized by differential connections with specific cortical regions. In particular, the medial LP has reciprocal connections with frontoparietal cortical areas, including the anterior cingulate cortex (ACC). The ACC plays an integral role in top-down sensory processing and attentional regulation, likely exerting some of these functions via the LP. However, little is known about how ACC and LP interact, and about the information potentially integrated in this reciprocal network. Here, we address this gap by employing a projection-specific monosynaptic rabies tracing strategy to delineate brain-wide inputs to bottom-up LP→ACC and top-down ACC→LP neurons. We find that LP→ACC neurons receive inputs from widespread cortical regions, including primary and higher order sensory and motor cortical areas. LP→ACC neurons also receive extensive subcortical inputs, particularly from the intermediate and deep layers of the superior colliculus (SC). Sensory inputs to ACC→LP neurons largely arise from visual cortical areas. In addition, ACC→LP neurons integrate cross-hemispheric prefrontal cortex inputs as well as inputs from higher order medial cortex. Our brain-wide anatomical mapping of inputs to the reciprocal LP-ACC pathways provides a roadmap for understanding how LP and ACC communicate different sources of information to mediate attentional control and visuomotor functions.


Assuntos
Pulvinar , Animais , Giro do Cíngulo , Camundongos , Pulvinar/fisiologia , Colículos Superiores/fisiologia , Tálamo/fisiologia , Vias Visuais/fisiologia
19.
Nat Commun ; 13(1): 4545, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35927275

RESUMO

Anterior cingulate cortex mediates the flexible updating of an animal's choice responses upon rule changes in the environment. However, how anterior cingulate cortex entrains motor cortex to reorganize rule representations and generate required motor outputs remains unclear. Here, we demonstrate that chemogenetic silencing of the terminal projections of cingulate cortical neurons in secondary motor cortex in the rat disrupts choice performance in trials immediately following rule switches, suggesting that these inputs are necessary to update rule representations for choice decisions stored in the motor cortex. Indeed, the silencing of cingulate cortex decreases rule selectivity of secondary motor cortical neurons. Furthermore, optogenetic silencing of cingulate cortical neurons that is temporally targeted to error trials immediately after rule switches exacerbates errors in the following trials. These results suggest that cingulate cortex monitors behavioral errors and updates rule representations in motor cortex, revealing a critical role for cingulate-motor circuits in adaptive choice behaviors.


Assuntos
Giro do Cíngulo , Córtex Motor , Animais , Giro do Cíngulo/fisiologia , Córtex Motor/fisiologia , Neurônios/fisiologia , Ratos
20.
Neuron ; 110(19): 3091-3105.e9, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-35987206

RESUMO

A major pathological hallmark of neurodegenerative diseases, including Alzheimer's, is a significant reduction in the white matter connecting the two cerebral hemispheres, as well as in the correlated activity between anatomically corresponding bilateral brain areas. However, the underlying circuit mechanisms and the cognitive relevance of cross-hemispheric (CH) communication remain poorly understood. Here, we show that novelty discrimination behavior activates CH neurons and enhances homotopic synchronized neural oscillations in the visual cortex. CH neurons provide excitatory drive required for synchronous neural oscillations between hemispheres, and unilateral inhibition of the CH circuit is sufficient to impair synchronous oscillations and novelty discrimination behavior. In the 5XFAD and Tau P301S mouse models, CH communication is altered, and novelty discrimination is impaired. These data reveal a hitherto uncharacterized CH circuit in the visual cortex, establishing a causal link between this circuit and novelty discrimination behavior and highlighting its impairment in mouse models of neurodegeneration.


Assuntos
Hipocampo , Córtex Visual , Animais , Modelos Animais de Doenças , Hipocampo/fisiologia , Interneurônios/fisiologia , Camundongos , Neurônios/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa