Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.394
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(17): e2306382121, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38640347

RESUMO

Hippocampal somatostatin-expressing (Sst) GABAergic interneurons (INs) exhibit considerable anatomical and functional heterogeneity. Recent single-cell transcriptome analyses have provided a comprehensive Sst-IN subpopulations census, a plausible molecular ground truth of neuronal identity whose links to specific functionality remain incomplete. Here, we designed an approach to identify and access subpopulations of Sst-INs based on transcriptomic features. Four mouse models based on single or combinatorial Cre- and Flp- expression differentiated functionally distinct subpopulations of CA1 hippocampal Sst-INs that largely tiled the morpho-functional parameter space of the Sst-INs superfamily. Notably, the Sst;;Tac1 intersection revealed a population of bistratified INs that preferentially synapsed onto fast-spiking interneurons (FS-INs) and were sufficient to interrupt their firing. In contrast, the Ndnf;;Nkx2-1 intersection identified a population of oriens lacunosum-moleculare INs that predominantly targeted CA1 pyramidal neurons, avoiding FS-INs. Overall, our results provide a framework to translate neuronal transcriptomic identity into discrete functional subtypes that capture the diverse specializations of hippocampal Sst-INs.


Assuntos
Hipocampo , Interneurônios , Camundongos , Animais , Interneurônios/fisiologia , Hipocampo/metabolismo , Neurônios/metabolismo , Células Piramidais/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
2.
Ageing Res Rev ; 96: 102270, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38484981

RESUMO

Among the central features of Alzheimer's disease (AD) progression are altered levels of the neuropeptide somatostatin (SST), and the colocalisation of SST-positive interneurons (SST-INs) with amyloid-ß plaques, leading to cell death. In this theoretical review, I propose a molecular model for the pathogenesis of AD based on SST-IN hypofunction and hyperactivity. Namely, hypofunctional and hyperactive SST-INs struggle to control hyperactivity in medial regions in early stages, leading to axonal Aß production through excessive presynaptic GABAB inhibition, GABAB1a/APP complex downregulation and internalisation. Concomitantly, excessive SST-14 release accumulates near SST-INs in the form of amyloids, which bind to Aß to form toxic mixed oligomers. This leads to differential SST-IN death through excitotoxicity, further disinhibition, SST deficits, and increased Aß release, fibrillation and plaque formation. Aß plaques, hyperactive networks and SST-IN distributions thereby tightly overlap in the brain. Conversely, chronic stimulation of postsynaptic SST2/4 on gulutamatergic neurons by hyperactive SST-INs promotes intense Mitogen-Activated Protein Kinase (MAPK) p38 activity, leading to somatodendritic p-tau staining and apoptosis/neurodegeneration - in agreement with a near complete overlap between p38 and neurofibrillary tangles. This model is suitable to explain some of the principal risk factors and markers of AD progression, including mitochondrial dysfunction, APOE4 genotype, sex-dependent vulnerability, overactive glial cells, dystrophic neurites, synaptic/spine losses, inter alia. Finally, the model can also shed light on qualitative aspects of AD neuropsychology, especially within the domains of spatial and declarative (episodic, semantic) memory, under an overlying pattern of contextual indiscrimination, ensemble instability, interference and generalisation.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Somatostatina/metabolismo , Neurônios/metabolismo , Emaranhados Neurofibrilares/patologia
3.
J Neurosci ; 44(17)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38438258

RESUMO

Acetylcholine (ACh) is released from basal forebrain cholinergic neurons in response to salient stimuli and engages brain states supporting attention and memory. These high ACh states are associated with theta oscillations, which synchronize neuronal ensembles. Theta oscillations in the basolateral amygdala (BLA) in both humans and rodents have been shown to underlie emotional memory, yet their mechanism remains unclear. Here, using brain slice electrophysiology in male and female mice, we show large ACh stimuli evoke prolonged theta oscillations in BLA local field potentials that depend upon M3 muscarinic receptor activation of cholecystokinin (CCK) interneurons (INs) without the need for external glutamate signaling. Somatostatin (SOM) INs inhibit CCK INs and are themselves inhibited by ACh, providing a functional SOM→CCK IN circuit connection gating BLA theta. Parvalbumin (PV) INs, which can drive BLA oscillations in baseline states, are not involved in the generation of ACh-induced theta, highlighting that ACh induces a cellular switch in the control of BLA oscillatory activity and establishes an internally BLA-driven theta oscillation through CCK INs. Theta activity is more readily evoked in BLA over the cortex or hippocampus, suggesting preferential activation of the BLA during high ACh states. These data reveal a SOM→CCK IN circuit in the BLA that gates internal theta oscillations and suggest a mechanism by which salient stimuli acting through ACh switch the BLA into a network state enabling emotional memory.


Assuntos
Acetilcolina , Colecistocinina , Camundongos Endogâmicos C57BL , Ritmo Teta , Ritmo Teta/efeitos dos fármacos , Ritmo Teta/fisiologia , Animais , Masculino , Camundongos , Feminino , Acetilcolina/farmacologia , Acetilcolina/metabolismo , Colecistocinina/farmacologia , Colecistocinina/metabolismo , Interneurônios/fisiologia , Interneurônios/efeitos dos fármacos , Somatostatina/metabolismo , Somatostatina/farmacologia , Tonsila do Cerebelo/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/fisiologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Rede Nervosa/fisiologia , Rede Nervosa/efeitos dos fármacos , Receptor Muscarínico M3/fisiologia , Receptor Muscarínico M3/metabolismo , Parvalbuminas/metabolismo
4.
J Neurosci ; 44(13)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38378274

RESUMO

Inhibitory modulation of glutamatergic information processing is a prerequisite for proper network function. Among the many groups of interneurons (INs), somatostatin-expressing interneurons (SOM-INs) play an important role in the maintenance of physiological brain activity. We have previously shown that somatostatin (SOM) causes a reduction in pyramidal cell (PC) excitability. However, the mechanisms of action of the peptide on cortical synaptic circuits are still unclear. To understand the effects of the neuropeptide SOM on cortical synaptic circuits, we performed a detailed side-by-side comparison of its postsynaptic effects on PCs, SOM-INs, and layer 1 interneurons (L1-INs) in the anterior cingulate cortex of male and female mice and found that SOM produced pronounced postsynaptic effects in PCs while having little to no effect on either IN type. This comparison allowed us to link the observed postsynaptic effects to SOM-induced modulations of glutamatergic and GABAergic synaptic transmission and to trace the impact of the neuropeptide on the neuronal circuitry between these three cell types. We show here that SOM depresses glutamatergic synaptic transmission via a presynaptic mechanism while exerting a differential impact on GABAA receptor- and GABAB receptor-mediated transmission at the pre- and postsynaptic level resulting in a shift of inhibition in L2/3 PCs from L1-INs to SOM-INs. In summary, this study unravels a novel aspect by which SOM modulates synaptic signaling between PCs, L1-INs, and SOM-INs.


Assuntos
Giro do Cíngulo , Transmissão Sináptica , Camundongos , Masculino , Animais , Feminino , Giro do Cíngulo/metabolismo , Transmissão Sináptica/fisiologia , Células Piramidais/metabolismo , Interneurônios/fisiologia , Somatostatina/metabolismo
5.
J Neurosci ; 44(10)2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38326037

RESUMO

The inferior colliculus (IC) represents a crucial relay station in the auditory pathway, located in the midbrain's tectum and primarily projecting to the thalamus. Despite the identification of distinct cell classes based on various biomarkers in the IC, their specific contributions to the organization of auditory tectothalamic pathways have remained poorly understood. In this study, we demonstrate that IC neurons expressing parvalbumin (ICPV+) or somatostatin (ICSOM+) represent two minimally overlapping cell classes throughout the three IC subdivisions in mice of both sexes. Strikingly, regardless of their location within the IC, these neurons predominantly project to the primary and secondary auditory thalamic nuclei, respectively. Cell class-specific input tracing suggested that ICPV+ neurons primarily receive auditory inputs, whereas ICSOM+ neurons receive significantly more inputs from the periaqueductal gray and the superior colliculus (SC), which are sensorimotor regions critically involved in innate behaviors. Furthermore, ICPV+ neurons exhibit significant heterogeneity in both intrinsic electrophysiological properties and presynaptic terminal size compared with ICSOM+ neurons. Notably, approximately one-quarter of ICPV+ neurons are inhibitory neurons, whereas all ICSOM+ neurons are excitatory neurons. Collectively, our findings suggest that parvalbumin and somatostatin expression in the IC can serve as biomarkers for two functionally distinct, parallel tectothalamic pathways. This discovery suggests an alternative way to define tectothalamic pathways and highlights the potential usefulness of Cre mice in understanding the multifaceted roles of the IC at the circuit level.


Assuntos
Colículos Inferiores , Parvalbuminas , Feminino , Masculino , Camundongos , Animais , Parvalbuminas/metabolismo , Colículos Inferiores/fisiologia , Neurônios/fisiologia , Vias Auditivas/fisiologia , Somatostatina/metabolismo
6.
Int J Mol Sci ; 25(3)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38339198

RESUMO

The overexpression of one or more somatostatin receptors (SST1-5R) in human tumors has provided an opportunity for diagnosis and therapy with somatostatin-like radionuclide carriers. The application of "pansomatostatin" analogs is expected to broaden the clinical indications and upgrade the diagnostic/therapeutic efficacy of currently applied SST2R-prefering radioligands. In pursuit of this goal, we now introduce two bicyclic somatostatin-14 (SS14) analogs, AT5S (DOTA-Ala1-Gly2-c[Cys3-Lys4-Asn5-c[Cys6-Phe7-DTrp8-Lys9-Thr10-Cys11]-Thr12-Ser13-Cys14]) and AT6S (DOTA-Ala1-Gly2-c[Cys3-Lys4-c[Cys5-Phe6-Phe7-DTrp8-Lys9-Thr10-Phe11-Cys12]-Ser13-Cys14]), suitable for labeling with trivalent radiometals and designed to sustain in vivo degradation. Both AT5S and AT6S and the respective [111In]In-AT5S and [111In]In-AT6S were evaluated in a series of in vitro assays, while radioligand stability and biodistribution were studied in mice. The 8/12-mer bicyclic AT6S showed expanded affinity for all SST1-5R and agonistic properties at the SST2R, whereas AT5S lost all affinity to SST1-5R. Both [111In]In-AT5S and [111In]In-AT6S remained stable in the peripheral blood of mice, while [111In]In-AT6S displayed low, but specific uptake in AR4-2J tumors and higher uptake in HEK293-SST3R tumors in mice. In summary, high radioligand stability was acquired by the two disulfide bridges introduced into the SS14 motif, but only the 8/12-mer ring AT6S retained a pansomatostatin profile. In consequence, [111In]In-AT6S targeted SST2R-/SST3R-positive xenografts in mice. These results call for further research on pansomatostatin-like radioligands for cancer theranostics.


Assuntos
Neoplasias , Somatostatina , Animais , Humanos , Camundongos , Células HEK293 , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Distribuição Tecidual
7.
Sci Rep ; 14(1): 4047, 2024 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-38374188

RESUMO

Somatostatin analogues have recently been used as therapeutic targets for metastatic or surgically unresectable gastroenteropancreatic (GEP) neuroendocrine tumors (NETs), and associated somatostatin receptor (SSTR) expression has been well demonstrated in most GEP NETs, with the exception of rectal NETs. SSTR2 immunohistochemical expressions were evaluated in 350 surgically or endoscopically resected rectal NETs and compared to clinicopathologic factors. SSTR2 expression was observed in 234 (66.9%) rectal NET cases and associated tumors with smaller size (p = 0.001), low pT classification (p = 0.030), low AJCC tumor stage (p = 0.012), and absence of chromogranin expression (p = 0.009). Patients with rectal NET and SSTR2 expression had significantly better overall survival than those without SSTR2 expression both by univariable (p = 0.006) and multivariable (p = 0.014) analyses. In summary, approximately two-thirds of rectal NETs expressed SSTR2. SSTR2 expression was significantly associated with favorable behavior and good overall survival in patients with rectal NETs. Furthermore, SSTR2 expression can be used as prognostic factors. When metastatic disease occurs, SSTR2 expression can be used a possible target for somatostatin analogues.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Neoplasias Pancreáticas , Neoplasias Retais , Neoplasias Gástricas , Humanos , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Prognóstico , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Neoplasias Retais/genética , Neoplasias Retais/cirurgia , Neoplasias Retais/tratamento farmacológico , Somatostatina/metabolismo
8.
Neuron ; 112(7): 1182-1195.e5, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38266646

RESUMO

Emotional responses arise from limbic circuits including the hippocampus and amygdala. In the human brain, beta-frequency communication between these structures correlates with self-reported mood and anxiety. However, both the mechanism and significance of this biomarker as a readout vs. driver of emotional state remain unknown. Here, we show that beta-frequency communication between ventral hippocampus and basolateral amygdala also predicts anxiety-related behavior in mice, both on long timescales (∼30 min) and immediately preceding behavioral choices. Genetically encoded voltage indicators reveal that this biomarker reflects synchronization between somatostatin interneurons across both structures. Indeed, synchrony between these neurons dynamically predicts approach-avoidance decisions, and optogenetically shifting the phase of synchronization by just 25 ms is sufficient to bidirectionally modulate anxiety-related behaviors. Thus, back-translation establishes a human biomarker as a causal determinant (not just predictor) of emotional state, revealing a novel mechanism whereby interregional synchronization that is frequency, phase, and cell type specific controls emotional processing.


Assuntos
Tonsila do Cerebelo , Interneurônios , Camundongos , Humanos , Animais , Tonsila do Cerebelo/fisiologia , Interneurônios/fisiologia , Ansiedade , Hipocampo/fisiologia , Somatostatina/metabolismo
9.
Neuropsychopharmacology ; 49(6): 961-973, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38182776

RESUMO

Distinguishing between cues predicting safety and danger is crucial for survival. Impaired learning of safety cues is a central characteristic of anxiety-related disorders. Despite recent advances in dissecting the neural circuitry underlying the formation and extinction of conditioned fear, the neuronal basis mediating safety learning remains elusive. Here, we showed that safety learning reduces the responses of paraventricular thalamus (PVT) neurons to safety cues, while activation of these neurons controls both the formation and expression of safety memory. Additionally, the PVT preferentially activates prefrontal cortex somatostatin interneurons (SOM-INs), which subsequently inhibit parvalbumin interneurons (PV-INs) to modulate safety memory. Importantly, we demonstrate that acute stress impairs the expression of safety learning, and this impairment can be mitigated when the PVT is inhibited, indicating PVT mediates the stress effect. Altogether, our findings provide insights into the mechanism by which acute stress modulates safety learning.


Assuntos
Núcleos da Linha Média do Tálamo , Córtex Pré-Frontal , Estresse Psicológico , Animais , Estresse Psicológico/fisiopatologia , Masculino , Núcleos da Linha Média do Tálamo/fisiologia , Núcleos da Linha Média do Tálamo/efeitos dos fármacos , Camundongos , Interneurônios/fisiologia , Medo/fisiologia , Camundongos Endogâmicos C57BL , Sinais (Psicologia) , Parvalbuminas/metabolismo , Somatostatina/metabolismo , Aprendizagem/fisiologia
10.
Commun Biol ; 7(1): 104, 2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-38228886

RESUMO

Glucose homeostasis is maintained by hormones secreted from different cell types of the pancreatic islets and controlled by manifold input including signals mediated through G protein-coupled receptors (GPCRs). RNA-seq analyses revealed expression of numerous GPCRs in mouse and human pancreatic islets, among them Gpr116/Adgrf5. GPR116 is an adhesion GPCR mainly found in lung and required for surfactant secretion. Here, we demonstrate that GPR116 is involved in the somatostatin release from pancreatic delta cells using a whole-body as well as a cell-specific knock-out mouse model. Interestingly, the whole-body GPR116 deficiency causes further changes such as decreased beta-cell mass, lower number of small islets, and reduced pancreatic insulin content. Glucose homeostasis in global GPR116-deficient mice is maintained by counter-acting mechanisms modulating insulin degradation. Our data highlight an important function of GPR116 in controlling glucose homeostasis.


Assuntos
Ilhotas Pancreáticas , Humanos , Animais , Camundongos , Ilhotas Pancreáticas/metabolismo , Somatostatina/metabolismo , Insulina/metabolismo , Pulmão/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Camundongos Knockout , Glucose/metabolismo
11.
Neuron ; 112(2): 201-208.e4, 2024 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-37944516

RESUMO

Despite recent advancements in identifying engram cells, our understanding of their regulatory and functional mechanisms remains in its infancy. To provide mechanistic insight into engram cell functioning, we introduced a novel local microcircuit labeling technique that enables the labeling of intraregional synaptic connections. Utilizing this approach, we discovered a unique population of somatostatin (SOM) interneurons in the mouse basolateral amygdala (BLA). These neurons are activated during fear memory formation and exhibit a preference for forming synapses with excitatory engram neurons. Post-activation, these SOM neurons displayed varying excitability based on fear memory retrieval. Furthermore, when we modulated these SOM neurons chemogenetically, we observed changes in the expression of fear-related behaviors, both in a fear-associated context and in a novel setting. Our findings suggest that these activated SOM interneurons play a pivotal role in modulating engram cell activity. They influence the expression of fear-related behaviors through a mechanism that is dependent on memory cues.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Interneurônios , Camundongos , Animais , Interneurônios/fisiologia , Memória/fisiologia , Neurônios/fisiologia , Complexo Nuclear Basolateral da Amígdala/fisiologia , Somatostatina/metabolismo
12.
Mol Neurobiol ; 61(2): 567-580, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37642935

RESUMO

Aging is often associated with a decline in cognitive function. A reduction in the number of somatostatin-positive (SOM+) interneurons in the dentate gyrus (DG) has been described in cognitively impaired but not in unimpaired aged rodents. However, it remains unclear whether the reduction in SOM + interneurons in the DG hilus is causal for age-related cognitive dysfunction. We hypothesized that hilar SOM+ interneurons play an essential role in maintaining cognitive function and that a reduction in the number of hilar SOM + interneurons might be sufficient to induce cognitive dysfunction. Hilar SOM+ interneurons were ablated by expressing a diphtheria toxin transgene specifically in these interneurons, which resulted in a reduction in the number of SOM+ /GAD-67+ neurons and dendritic spine density in the DG. C-fos and Iba-1 immunostainings were increased in DG and CA3, but not CA1, and BDNF protein expression in the hippocampus was decreased. Behavioral testing showed a reduced recognition index in the novel object recognition test, decreased alternations in the Y maze test, and longer latencies and path lengths in the learning and reversal learning phases of the Morris water maze. Our results show that partial genetic ablation of SOM+ hilar interneurons is sufficient to increase activity in DG and CA3, as has been described to occur with aging and to induce an impairment of learning and memory functions. Thus, partial ablation of hilar SOM + interneurons may be a significant contributing factor to age-related cognitive dysfunction. These mice may also be useful as a cellularly defined model of hippocampal aging.


Assuntos
Disfunção Cognitiva , Interneurônios , Camundongos , Animais , Interneurônios/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Somatostatina/metabolismo
13.
Dev Neurobiol ; 84(1): 3-17, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38072668

RESUMO

Transient expression of somatostatin (SST) has been observed in the olfactory epithelium (OE) and nerves of chick embryos. Intense expression of SST in these regions on embryonic days (E) 5-8 coincides with the migration of neurons producing gonadotropin-releasing hormone (GnRH) from the OE to the forebrain (FB), suggesting that SST plays a role in the development of GnRH neurons. Using in ovo electroporation of small interfering RNA, we found that the suppression of SST mRNA in the olfactory placode (OP) of E3.5 chick embryos significantly reduced the number of GnRH and Islet-1-immunoreactive neurons in the nasal region without affecting the entry of GnRH neurons into the FB at E5.5-6. SST knockdown did not lead to changes in the number of apoptotic, proliferating, or HuC/D-positive neuronal cells in the OE; therefore, it is possible that SST is involved in the neurogenesis/differentiation of GnRH neurons and OP-derived GnRH-negative migratory neurons. In whole OP explant cultures, we also found that SST or its analog octreotide treatment significantly increased the number of migratory GnRH neurons and the migratory distance from the explants. The co-application of an SST antagonist blocked the octreotide-induced increase in the number of GnRH neurons. Furthermore, the fasciculation of polysialylated neural cell adhesion molecule-immunoreactive fibers emerging from the explants was dependent on octreotide. Taken together, our results provide evidence that SST exerts facilitatory effects on the development of neurons expressing GnRH or Islet-1 and on GnRH neuronal migration, in addition to olfactory-related fiber fasciculation.


Assuntos
Hormônio Liberador de Gonadotropina , Octreotida , Animais , Embrião de Galinha , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Octreotida/metabolismo , Octreotida/farmacologia , Fasciculação/metabolismo , Neurônios/fisiologia , Somatostatina/farmacologia , Somatostatina/metabolismo , Movimento Celular/fisiologia
14.
Proc Natl Acad Sci U S A ; 120(51): e2312752120, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38091292

RESUMO

Somatostatin-expressing interneurons (SOMIs) in the mouse dentate gyrus (DG) receive feedforward excitation from granule cell (GC) mossy fiber (MF) synapses and provide feedback lateral inhibition onto GC dendrites to support environment representation in the DG network. Although this microcircuitry has been implicated in memory formation, little is known about activity-dependent plastic changes at MF-SOMI synapses and their influence on behavior. Here, we report that the metabotropic glutamate receptor 1α (mGluR1α) is required for the induction of associative long-term potentiation (LTP) at MF-SOMI synapses. Pharmacological block of mGluR1α, but not mGluR5, prevented synaptic weight changes. LTP at MF-SOMI synapses was postsynaptically induced, required increased intracellular Ca2+, involved G-protein-mediated and Ca2+-dependent (extracellular signal-regulated kinase) ERK1/2 pathways, and the activation of NMDA receptors. Specific knockdown of mGluR1α in DG-SOMIs by small hairpin RNA expression prevented MF-SOMI LTP, reduced SOMI recruitment, and impaired object location memory. Thus, postsynaptic mGluR1α-mediated MF-plasticity at SOMI input synapses critically supports DG-dependent mnemonic functions.


Assuntos
Fibras Musgosas Hipocampais , Plasticidade Neuronal , Camundongos , Animais , Fibras Musgosas Hipocampais/fisiologia , Plasticidade Neuronal/fisiologia , Interneurônios/fisiologia , Potenciação de Longa Duração/fisiologia , Sinapses/metabolismo , Somatostatina/metabolismo , Giro Denteado/metabolismo , Transmissão Sináptica
15.
Sci Rep ; 13(1): 19473, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945756

RESUMO

Abnormal dopamine neurotransmission is a common trait of some psychiatric diseases, like schizophrenia or bipolar disorder. Excessive dopaminergic tone in subcortical brain regions is associated with psychotic episodes, while reduced prefrontal dopaminergic activity is associated with impaired cognitive performance and reduced motivation, among other symptoms. Inhibitory interneurons expressing the calcium binding protein parvalbumin are particularly affected in both schizophrenia and bipolar disorder, as they set a fine-tuned physiological inhibitory/excitatory balance. Parvalbumin and somatostatin interneuron subtypes, are born from the medial ganglionic eminence and require the sequential expression of specific transcription factors for their specification, such as Nkx6.2. Here, we aimed at characterizing in detail interneuron subtypes derived from Nkx6.2 expressing progenitors by the generation of an Nkx6.2 Cre transgenic mouse line. We show that Nkx6.2 specifies over a third part of the total population of cortical somatostatin interneurons, preferentially at early developmental time points, whereas at late developmental stages, Nkx6.2 expressing progenitors shift to parvalbumin interneuron specification. Dopamine D2 receptor deletion from Nkx6.2 expressing progenitors causes abnormal phenotypes restricted to cognitive, motivation and anxiety domains. Our results show that Nkx6.2 have the potential to specify both somatostatin and parvalbumin interneurons in an opposite timed program and that DRD2 expression is required in Nkx6.2 expressing progenitors to avoid impaired phenotypes commonly associated to the pathophysiology of psychiatric diseases.


Assuntos
Motivação , Parvalbuminas , Animais , Camundongos , Ansiedade/genética , Cognição , Interneurônios/metabolismo , Camundongos Transgênicos , Parvalbuminas/metabolismo , Fenótipo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
16.
Nat Commun ; 14(1): 7165, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37935668

RESUMO

Somatostatin-positive (SOM) interneurons are integral for shaping cortical processing and their dynamic recruitment is likely necessary for adaptation to sensory experience and contextual information. We found that excitatory synapses on SOMs in layer 2/3 (L2/3) of primary visual cortex (V1) of mice can be categorized into fast (F)- and slow (S)-Types based on the kinetics of the AMPA receptor-mediated current. Each SOM contains both types of synapses in varying proportions. The majority of local pyramidal neurons (PCs) make unitary connections with SOMs using both types, followed by those utilizing only S-Type, and a minority with only F-Type. Sensory experience differentially regulates synapses on SOMs, such that local F-Type synapses change with visual deprivation and S-Type synapses undergo plasticity with crossmodal auditory deprivation. Our results demonstrate that the two types of excitatory synapses add richness to the SOM circuit recruitment and undergo selective plasticity enabling dynamic adaptation of the adult V1.


Assuntos
Sinapses , Córtex Visual , Camundongos , Animais , Sinapses/metabolismo , Interneurônios/metabolismo , Células Piramidais/metabolismo , Somatostatina/metabolismo , Córtex Visual/fisiologia
17.
Genes (Basel) ; 14(11)2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-38002975

RESUMO

In this study, to explore the effect of growth hormone changes on the related genes and regulatory roles of the turtle, PCR amplification, real-time fluorescence quantitative analysis, and enzyme cutting technology were used to clone and sequence the somatostatin (SS) gene, growth hormone receptor (GHR), and insulin-like growth factor-1 (IGF-I) sequence of Chinemys reevesii. The effects of human growth hormone on the mRNA expression of growth-axis-related genes SS, GHR, and IGF-1 in different sexes were observed. The study of the SS gene in turtles using real-time fluorescence quantitative PCR showed that the SS gene was mainly expressed in the nervous system and the digestive system, with the highest expression found in the brain, while the GHR gene and the IGF-I gene were expressed in all tissues of Chinemys reevesii. The SS gene was expressed in the brain, pituitary, liver, stomach, and intestine, with the highest expression in the brain and the lowest expression in the liver. Within 4 weeks of the injection of exogenous growth hormone, the expression level of the SS gene in the brain of both sexes first increased and then decreased, showing a parabolic trend, and the expression level of the experimental group was lower than that of the control group. After the injection of growth hormone (GH), the expression of the GHR gene in the liver of both sexes showed a significant increase in the first week, decreasing to the control group level in the second week, and then gradually increasing. Finally, a significant level of difference in the expression of the GHR gene was reached at 3 and 4 weeks. In terms of the IGF-I gene, the changing trend of the expression level in the liver was the same as that of the GHR gene. After the injection of exogenous growth hormone, although the expression of the SS gene increased the inhibition of the secretion of the GHR gene by the Reeves' turtle, exogenous growth hormone could replace the synthesis of GH and GHR, accelerating the growth of the turtle. The experiments showed that the injection of recombinant human growth hormone affects the expression of SS, GHR, and IGF-1 genes, and promotes the growth of the Reeves' turtle.


Assuntos
Hormônio do Crescimento Humano , Tartarugas , Masculino , Animais , Feminino , Humanos , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Receptores da Somatotropina/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/farmacologia , Regulação da Expressão Gênica , Somatostatina/genética , Somatostatina/metabolismo
18.
Life Sci ; 334: 122173, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37907154

RESUMO

AIMS: The aim of our study was to determine the effect of histone deacetylase (HDAC) inhibitors (HDACis) on somatostatin type-2 receptor (SSTR2) expression and [111In]In-/[177Lu]Lu-DOTA-TATE uptake in vitro and in vivo. MATERIALS AND METHODS: The human cell lines NCI-H69 (small-cell lung carcinoma) and BON-1 (pancreatic neuroendocrine tumor) were treated with HDACis (i.e. entinostat, mocetinostat (MOC), LMK-235, CI-994 or panobinostat (PAN)), and SSTR2 mRNA expression levels and [111In]In-DOTA-TATE uptake were measured. Furthermore, vehicle- and HDACi-treated NCI-H69 and BON-1 tumor-bearing mice were injected with radiolabeled DOTA-TATE followed by biodistribution studies. Additionally, SSTR2 and HDAC mRNA expression of xenografts, and of NCI-H69, BON-1, NCI-H727 (human pulmonary carcinoid) and GOT1 (human midgut neuroendocrine tumor) cells were determined. KEY FINDINGS: HDACi treatment resulted in the desired effects in vitro. However, no significant increase in tumoral DOTA-TATE uptake was observed after HDACi treatment in NCI-H69 tumor-bearing animals, whereas tumoral SSTR2 mRNA and/or protein expression levels were significantly upregulated after treatment with MOC, CI-994 and PAN, i.e. a maximum of 2.1- and 1.3-fold, respectively. Analysis of PAN-treated BON-1 xenografts solely demonstrated increased SSTR2 mRNA expression levels. Comparison of HDACs and SSTR2 expression in BON-1 and NCI-H69 xenografts showed a significantly higher expression of 6/11 HDACs in BON-1 xenografts. Of these HDACs, a significant inverse correlation was found between HDAC3 and SSTR2 expression (Pearson r = -0.92) in the studied cell lines. SIGNIFICANCE: To conclude, tumoral uptake levels of radiolabeled DOTA-TATE were not enhanced after HDACi treatment in vivo, but, depending on the applied inhibitor, increased SSTR2 expression levels were observed.


Assuntos
Receptores de Somatostatina , Somatostatina , Humanos , Camundongos , Animais , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Distribuição Tecidual , Somatostatina/metabolismo , Linhagem Celular Tumoral , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
19.
J Neurosci ; 43(43): 7101-7118, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37709538

RESUMO

Somatostatin (SST) interneurons produce delayed inhibition because of the short-term facilitation of their excitatory inputs created by the expression of metabotropic glutamate receptor 7 (mGluR7) and presynaptic GluK2-containing kainate receptors (GluK2-KARs). Using mice of both sexes, we find that as synaptic facilitation at layer (L)2/3 SST cell inputs increases during the first few postnatal weeks, so does GluK2-KAR expression. Removal of sensory input by whisker trimming does not affect mGluR7 but prevents the emergence of presynaptic GluK2-KARs, which can be restored by allowing whisker regrowth or by acute calmodulin activation. Conversely, late trimming or acute inhibition of Ca2+/calmodulin-dependent protein kinase II is sufficient to reduce GluK2-KAR activity. This developmental and activity-dependent regulation also produces a specific reduction of L4 GluK2-KARs that advances in parallel with the maturation of sensory processing in L2/3. Finally, we find that removal of both GluK2-KARs and mGluR7 from the synapse eliminates short-term facilitation and reduces sensory adaptation to repetitive stimuli, first in L4 of somatosensory cortex, then later in development in L2/3. The dynamic regulation of presynaptic GluK2-KARs potentially allows for flexible scaling of late inhibition and sensory adaptation.SIGNIFICANCE STATEMENT Excitatory synapses onto somatostatin (SST) interneurons express presynaptic, calcium-permeable kainate receptors containing the GluK2 subunit (GluK2-KARs), activated by high-frequency activity. In this study we find that their presence on L2/3 SST synapses in the barrel cortex is not based on a hardwired genetic program but instead is regulated by sensory activity, in contrast to that of mGluR7. Thus, in addition to standard synaptic potentiation and depression mechanisms, excitatory synapses onto SST neurons undergo an activity-dependent presynaptic modulation that uses GluK2-KARs. Further, we present evidence that loss of the frequency-dependent synaptic components (both GluK2-KARs and mGluR7 via Elfn1 deletion) contributes to a decrease in the sensory adaptation commonly seen on repetitive stimulus presentation.


Assuntos
Ácido Caínico , Receptores de Ácido Caínico , Masculino , Feminino , Camundongos , Animais , Receptores de Ácido Caínico/metabolismo , Receptores Pré-Sinápticos/metabolismo , Sinapses/fisiologia , Interneurônios/fisiologia , Somatostatina/metabolismo
20.
Nat Neurosci ; 26(10): 1805-1819, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37735497

RESUMO

The prefrontal cortex (PFC) enables mammals to respond to situations, including internal states, with appropriate actions. One such internal state could be 'tiredness'. Here, using activity tagging in the mouse PFC, we identified particularly excitable, fast-spiking, somatostatin-expressing, γ-aminobutyric acid (GABA) (PFCSst-GABA) cells that responded to sleep deprivation. These cells projected to the lateral preoptic (LPO) hypothalamus and the lateral hypothalamus (LH). Stimulating PFCSst-GABA terminals in the LPO hypothalamus caused sleep-preparatory behavior (nesting, elevated theta power and elevated temperature), and stimulating PFCSst-GABA terminals in the LH mimicked recovery sleep (non-rapid eye-movement sleep with higher delta power and lower body temperature). PFCSst-GABA terminals had enhanced activity during nesting and sleep, inducing inhibitory postsynaptic currents on diverse cells in the LPO hypothalamus and the LH. The PFC also might feature in deciding sleep location in the absence of excessive fatigue. These findings suggest that the PFC instructs the hypothalamus to ensure that optimal sleep takes place in a suitable place.


Assuntos
Região Hipotalâmica Lateral , Neurônios , Camundongos , Animais , Região Hipotalâmica Lateral/metabolismo , Neurônios/fisiologia , Somatostatina/metabolismo , Sono/fisiologia , Hipotálamo/fisiologia , Ácido gama-Aminobutírico , Córtex Pré-Frontal/fisiologia , Mamíferos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...