Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 495
Filtrar
1.
Neuropharmacology ; 203: 108885, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34798130

RESUMO

Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that mediates a wide spectrum of biological processes including apoptosis, immune response and inflammation. Here, we sought to understand how S1P signaling affects neuronal excitability in the central amygdala (CeA), which is a brain region associated with fear learning, aversive memory, and the affective dimension of pain. Because the G-protein coupled S1P receptor 1 (S1PR1) has been shown to be the primary mediator of S1P signaling, we utilized S1PR1 agonist SEW2871 and S1PR1 antagonist NIBR to determine a potential role of S1PR1 in altering the cellular physiology of neurons in the lateral division of the CeA (CeL) that share the neuronal lineage marker somatostatin (Sst). CeL-Sst neurons play a critical role in expression of conditioned fear and pain modulation. Here we used transgenic breeding strategies to identify fluorescently labeled CeL-Sst neurons for electrophysiological recordings. Using principal component analysis, we identified two primary subtypes of Sst neurons within the CeL in both male and female mice. We denoted the two types regular-firing (type A) and late-firing (type B) CeL-Sst neurons. In response to SEW2871 application, Type A neurons exhibited increased input resistance, while type B neurons displayed a depolarized resting membrane potential and voltage threshold, increased current threshold, and decreased voltage height. NIBR application had no effect on CeL Sst neurons, indicating the absence of tonic S1P-induced S1PR1. Our findings reveal subtypes of Sst neurons within the CeL that are uniquely affected by S1PR1 activation, which may have implications for how S1P alters supraspinal circuits.


Assuntos
Núcleo Central da Amígdala/metabolismo , Potenciais da Membrana/fisiologia , Oxidiazóis/farmacologia , Somatostatina/biossíntese , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacologia , Receptores de Esfingosina-1-Fosfato/metabolismo , Tiofenos/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Núcleo Central da Amígdala/efeitos dos fármacos , Feminino , Expressão Gênica , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Somatostatina/genética , Receptores de Esfingosina-1-Fosfato/agonistas
2.
Epigenomics ; 13(15): 1205-1219, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34318683

RESUMO

Aim: Our study aimed at investigating how LINC01133 functions in gastric cancer (GC) progression. Materials & methods: Gain-of-function and loss-of-function approaches were applied to analyze the effects of LINC01133, microRNA-576-5p (miR-576-5p) and somatostatin (SST) on the biological behaviors of GC cells and in tumor-bearing nude mice. Results: GC tissues and cells showed low expression of LINC01133, and LINC01133 overexpression decreased malignant phenotypes of GC cells. Moreover, LINC01133 upregulated SST through binding to miR-576-5p. Overexpressing miR-576-5p or suppressing SST reversed the functions of LINC01133 in biological potentials of GC cells and tumor growth. Conclusion: LINC01133 overexpression may inhibit GC development by downregulation of miR-576-5p and upregulation of SST, which suggests new therapeutic targets for GC.


Assuntos
MicroRNAs/genética , Interferência de RNA , RNA Longo não Codificante/genética , Somatostatina/biossíntese , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animais , Linhagem Celular Tumoral , Bases de Dados Genéticas , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Modelos Biológicos , Somatostatina/genética , Neoplasias Gástricas/patologia , Transcriptoma
3.
J Chem Neuroanat ; 117: 102007, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34314850

RESUMO

The study was designed to examine the distribution and chemical coding of somatostatin-immunoreactive (SOM-IR) nerve fibers supplying the urinary bladder wall and to establish the distribution and immunohistochemical characteristics of the subpopulation of paracervical ganglion (PCG) SOM-IR neurons projecting to this organ in female pigs. The PCG-urinary bladder projecting neurons (PCG-UBPN) were visualized with retrograde neuronal tracer Fast Blue (FB). Double-labeling immunohistochemistry performed on cryostat sections from the urinary bladder wall revealed that the greatest density of SOM-IR nerve fibers was found in the muscle layer and around blood vessels, a moderate number of these nerve terminals supplied the submucosa and only single SOM-IR axons were encountered beneath the urothelium. In all the investigated sections the vast majority of SOM-IR nerve fibers were immunopositive to vesicular acetylcholine transporter (VAChT) and many SOM-IR axons contained immunoreactivity to neuropeptide Y (NPY). Approximately 65 % of FB-positive (FB+) PCG-UBPN were immunoreactive to SOM. Moreover, PCG FB+/SOM + nerve cells were simultaneously immunoreactive to choline acetyltransferase (ChAT; 64.6 ± 0.6 %), NPY (59.7 ± 1.2 %), neuronal nitric oxide synthase (nNOS; 46.1 ± 0.7 %), vasoactive intestinal polypeptide (VIP; 29.9 ± 2.2 %), Leu5-enkephalin (L-ENK; 19.5 ± 6.3 %), dopamine ß-hydroxylase (DßH; 14.9 ± 1.9 %) or pituitary adenylate cyclase-activating polypeptide (PACAP; 14.8 ± 2.4 %). The present study reveals the extensive expression of SOM in both the nerve fibres supplying the porcine urinary bladder wall and the PCG neurons projecting to this organ, indicating an important regulatory role of SOM in the control of the urinary bladder function.


Assuntos
Colo do Útero/química , Gânglios Autônomos/química , Fibras Nervosas/química , Neurônios/química , Somatostatina/análise , Bexiga Urinária/química , Animais , Colo do Útero/inervação , Colo do Útero/metabolismo , Feminino , Gânglios Autônomos/metabolismo , Fibras Nervosas/metabolismo , Neurônios/metabolismo , Somatostatina/biossíntese , Suínos , Bexiga Urinária/inervação , Bexiga Urinária/metabolismo
4.
Acta Histochem ; 122(8): 151650, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33161374

RESUMO

Huntingtin-associated protein 1 (HAP1) is a neuronal cytoplasmic protein that is predominantly expressed in the brain and spinal cord. In addition to the central nervous system, HAP1 is also expressed in the peripheral organs including endocrine system. Different types of enteroendocrine cells (EEC) are present in the digestive organs. To date, the characterization of HAP1-immunoreactive (ir) cells remains unreported there. In the present study, the expression of HAP1 in pyloric stomach in adult male rats and its relationships with different chemical markers for EEC [gastrin, marker of gastrin (G) cells; somatostatin, marker of delta (D) cells; 5-HT, marker of enterochromaffin (EC) cells; histamine, marker of enterochromaffin-like (ECL) cells] were examined employing single- or double-labelled immunohistochemistry and with light-, fluorescence- or electron-microscopy. HAP1-ir cells were abundantly expressed in the glandular mucosa but were very few or none in the surface epithelium. Double-labelled immunofluorescence staining for HAP1 and markers for EECs showed that almost all the G-cells expressed HAP1. In contrast, HAP1 was completely lacking in D-cells, EC-cells or ECL-cells. Our current study is the first to clarify that HAP1 is selectively expressed in G-cells in rat pyloric stomach, which probably reflects HAP1's involvement in regulation of the secretion of gastrin.


Assuntos
Células Enterocromafins/metabolismo , Celulas Tipo Enterocromafim/metabolismo , Mucosa Gástrica/metabolismo , Proteínas do Tecido Nervoso/genética , Piloro/metabolismo , Células Secretoras de Somatostatina/metabolismo , Animais , Biomarcadores/metabolismo , Células Enterocromafins/citologia , Celulas Tipo Enterocromafim/citologia , Mucosa Gástrica/citologia , Gastrinas/biossíntese , Expressão Gênica , Histamina/biossíntese , Imuno-Histoquímica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Especificidade de Órgãos , Piloro/citologia , Ratos , Ratos Wistar , Somatostatina/biossíntese , Células Secretoras de Somatostatina/citologia
5.
J Neurosci ; 40(47): 9043-9054, 2020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33067362

RESUMO

The central amygdala (CeA) is critically involved in a range of adaptive behaviors, including defensive behaviors. Neurons in the CeA send long-range projections to a number of extra-amygdala targets, but the functions of these projections remain elusive. Here, we report that a previously neglected CeA-to-globus pallidus external segment (GPe) circuit plays an essential role in classical fear conditioning. By anatomic tracing, in situ hybridization and channelrhodopsin (ChR2)-assisted circuit mapping in both male and female mice, we found that a subset of CeA neurons send projections to the GPe, and the majority of these GPe-projecting CeA neurons express the neuropeptide somatostatin. Notably, chronic inhibition of GPe-projecting CeA neurons with the tetanus toxin light chain (TeLC) completely blocks auditory fear conditioning. In vivo fiber photometry revealed that these neurons are selectively excited by the unconditioned stimulus (US) during fear conditioning. Furthermore, transient optogenetic inactivation or activation of these neurons selectively during US presentation impairs or promotes, respectively, fear learning. Our results suggest that a major function of GPe-projecting CeA neurons is to represent and convey US-related information through the CeA-GPe circuit, thereby regulating learning in fear conditioning.SIGNIFICANCE STATEMENT The central amygdala (CeA) has been implicated in the establishment of defensive behaviors toward threats, but the underlying circuit mechanisms remain unclear. Here, we found that a subpopulation of neurons in the CeA, which are mainly those that express the neuropeptide somatostatin, send projections to the globus pallidus external segment (GPe), and this CeA-GPe circuit conveys unconditioned stimulus (US)-related information during classical fear conditioning, thereby having an indispensable role in learning. Our results reveal a previously unknown circuit mechanism for fear learning.


Assuntos
Núcleo Central da Amígdala/fisiologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Medo/psicologia , Globo Pálido/fisiologia , Rede Nervosa/fisiologia , Estimulação Acústica , Animais , Núcleo Central da Amígdala/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Medo/efeitos dos fármacos , Feminino , Globo Pálido/efeitos dos fármacos , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/efeitos dos fármacos , Optogenética , Somatostatina/biossíntese , Somatostatina/genética , Toxina Tetânica/farmacologia
6.
Elife ; 92020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32749220

RESUMO

The cellular architecture of the ventral tegmental area (VTA), the main hub of the brain reward system, remains only partially characterized. To extend the characterization to inhibitory neurons, we have identified three distinct subtypes of somatostatin (Sst)-expressing neurons in the mouse VTA. These neurons differ in their electrophysiological and morphological properties, anatomical localization, as well as mRNA expression profiles. Importantly, similar to cortical Sst-containing interneurons, most VTA Sst neurons express GABAergic inhibitory markers, but some of them also express glutamatergic excitatory markers and a subpopulation even express dopaminergic markers. Furthermore, only some of the proposed marker genes for cortical Sst neurons were expressed in the VTA Sst neurons. Physiologically, one of the VTA Sst neuron subtypes locally inhibited neighboring dopamine neurons. Overall, our results demonstrate the remarkable complexity and heterogeneity of VTA Sst neurons and suggest that these cells are multifunctional players in the midbrain reward circuitry.


Assuntos
Neurônios/metabolismo , Somatostatina/biossíntese , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Animais , Fenômenos Eletrofisiológicos , Feminino , Perfilação da Expressão Gênica , Interneurônios/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/classificação , Neurônios/citologia , Neurotransmissores/metabolismo
7.
Hum Pathol ; 102: 79-87, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32668277

RESUMO

A small fraction of pancreatic neuroendocrine tumors (PanNETs) shows a solid, paraganglioma-like (PG-like) histology. We wanted to know whether these PanNETs have a special hormone expression and are related to paragangliomas (PGs)/pheochromocytomas (PCs). We screened a series of 48 surgically resected PanNETs for their histological growth patterns and their association with expression of islet hormones. The PanNETs were divided into PG-like and non-PG-like tumors and immunohistochemically monitored for the expression of islet hormones, cytokeratins, and S100. The results were correlated to histological pattern, lymph node status, and data in 28 PGs/PCs, including 2 PGs attached to the pancreas. All PanNETs, in contrast to PGs/PCs, were cytokeratin positive. A PG-like growth pattern was identified in 9 of 48 PanNETs and correlated with somatostatin expression. Only half of the non-PG-like PanNETs also contained somatostatin-positive cells. Eight of 28 PGs/PCs expressed somatostatin, mostly in individual cells. PG-like PanNETs and non-PG-like PanNETs infiltrated the adjacent pancreatic tissue, whereas 2 to the pancreas-associated PGs were well demarcated. Lymph node metastases were detected in 58%, 39%, 57%, and 53% of the somatostatin-producing, somatostatin-negative, PG-like, and non-PG-like PanNETs, respectively. PG-like PanNETs, in contrast to PG/PCs, are characterized by the expression of cytokeratin and somatostatin, the development of lymph node metastasis, and the infiltration into pancreatic parenchyma. Non-PG-like PanNETs may also express somatostatin and show lymph node metastases to the same extent. A literature review of cases reported as PG of the pancreas reveals that only a small fraction of these tumors probably represents true pancreatic PGs.


Assuntos
Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Somatostatina/biossíntese , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Paraganglioma/patologia , Adulto Jovem
8.
Toxicology ; 442: 152534, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32622971

RESUMO

We showed previously that chlormequat chloride, a widely used plant growth regulator, could affect embryonic growth and growth hormone (GH)-insulin-like growth factor 1 (IGF-1) axis of rats. However, the potential effects of low dose chlormequat chloride exposure during pregnancy on embryonic and postnatal growth and development remain unclear. To further assess the risk of chlormequat chloride to human embryonic growth and postnatal health, we exposed maternal rats orally to the chemical during pregnancy at 5 mg/kg bw, a dose corresponding to the human acceptable daily intake (ADI) level set by World Health Organization (WHO), and determined the effects of chlormequat on embryo growth and postnatal health. We found that chlormequat chloride increased embryonic growth parameters, GH, and GH-releasing hormone (GHRH) levels, but did not affect somatostatin and IGF-1 on gestational day (GD) 11. In the pups of postnatal day (PD) 7, we observed increased head length, decreased body fat percentage, hypoglycemia, hyperlipidemia and hyperproteinemia. In conclusion, maternal exposure to chlormequat chloride during pregnancy disrupts the embryonic growth probably through its effects on growth regulators and even has adverse effects on postnatal health.


Assuntos
Anormalidades Induzidas por Medicamentos/patologia , Clormequat/toxicidade , Desenvolvimento Embrionário/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Composição Corporal/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento/biossíntese , Hormônio Liberador de Hormônio do Crescimento/biossíntese , Fator de Crescimento Insulin-Like I/biossíntese , Masculino , Exposição Materna , Reguladores de Crescimento de Plantas/toxicidade , Gravidez , Ratos , Ratos Sprague-Dawley , Somatostatina/biossíntese
9.
Front Neural Circuits ; 14: 16, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32395103

RESUMO

Transgenic Cre-recombinase expressing mouse lines are widely used to express fluorescent proteins and opto-/chemogenetic actuators, making them a cornerstone of modern neuroscience. The investigation of interneurons in particular has benefitted from the ability to genetically target specific cell types. However, the specificity of some Cre driver lines has been called into question. Here, we show that nonspecific expression in a subset of hippocampal neurons can have substantial nonspecific functional effects in a somatostatin-Cre (SST-Cre) mouse line. Nonspecific targeting of CA3 pyramidal cells caused large optogenetically evoked excitatory currents in remote brain regions. Similar, but less severe patterns of nonspecific expression were observed in a widely used SST-IRES-Cre line, when crossed with a reporter mouse line. Viral transduction on the other hand yielded more specific expression but still resulted in nonspecific expression in a minority of pyramidal layer cells. These results suggest that a careful analysis of specificity is mandatory before the use of Cre driver lines for opto- or chemogenetic manipulation approaches.


Assuntos
Região CA3 Hipocampal/citologia , Região CA3 Hipocampal/metabolismo , Integrases/biossíntese , Interneurônios/metabolismo , Optogenética/métodos , Somatostatina/biossíntese , Animais , Região CA3 Hipocampal/química , Expressão Gênica , Integrases/análise , Integrases/genética , Interneurônios/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Somatostatina/análise , Somatostatina/genética
10.
J Neurosci ; 40(18): 3564-3575, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32220950

RESUMO

Sensory systems integrate multiple stimulus features to generate coherent percepts. Spectral surround suppression, the phenomenon by which sound-evoked responses of auditory neurons are suppressed by stimuli outside their receptive field, is an example of this integration taking place in the auditory system. While this form of global integration is commonly observed in auditory cortical neurons, and potentially used by the nervous system to separate signals from noise, the mechanisms that underlie this suppression of activity are not well understood. We evaluated the contributions to spectral surround suppression of the two most common inhibitory cell types in the cortex, parvalbumin-expressing (PV+) and somatostatin-expressing (SOM+) interneurons, in mice of both sexes. We found that inactivating SOM+ cells, but not PV+ cells, significantly reduces sustained spectral surround suppression in excitatory cells, indicating a dominant causal role for SOM+ cells in the integration of information across multiple frequencies. The similarity of these results to those from other sensory cortices provides evidence of common mechanisms across the cerebral cortex for generating global percepts from separate features.SIGNIFICANCE STATEMENT To generate coherent percepts, sensory systems integrate simultaneously occurring features of a stimulus, yet the mechanisms by which this integration occurs are not fully understood. Our results show that neurochemically distinct neuronal subtypes in the primary auditory cortex have different contributions to the integration of different frequency components of an acoustic stimulus. Together with findings from other sensory cortices, our results provide evidence of a common mechanism for cortical computations used for global integration of stimulus features.


Assuntos
Estimulação Acústica/métodos , Córtex Auditivo/metabolismo , Interneurônios/metabolismo , Somatostatina/biossíntese , Potenciais de Ação/fisiologia , Animais , Córtex Auditivo/citologia , Eletrodos Implantados , Feminino , Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Somatostatina/genética
11.
Front Neural Circuits ; 13: 56, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507383

RESUMO

Conscious memories are critically dependent upon bilateral hippocampal formation, and interhemispheric commissural projections made by mossy cells and CA3 pyramidal cells. GABAergic interneurons also make long-range axonal projections, but little is known regarding their commissural, inter-hippocampal connections. We used retrograde and adeno-associated viral tracing, immunofluorescence and electron microscopy, and in vitro optogenetics to assess contralateral projections of neurochemically defined interneuron classes. We found that contralateral-projecting interneurons were 24-fold less common compared to hilar mossy cells, and mostly consisted of somatostatin- and parvalbumin-expressing types. Somatostatin-expressing cells made denser contralateral axonal projections than parvalbumin-expressing cells, although this was typically 10-fold less than the ipsilateral projection density. Somatostatin-expressing cells displayed a topographic-like innervation according to the location of their somata, whereas parvalbumin-expressing cells mostly innervated CA1. In the dentate gyrus molecular layer, commissural interneuron post-synaptic targets were predominantly putative granule cell apical dendrites. In the hilus, varicosities in close vicinity to various interneuron subtypes, as well as mossy cells, were observed, but most contralateral axon varicosities had no adjacent immunolabeled structure. Due to the relative sparsity of the connection and the likely distal dendritic location of their synapses, commissural projections made by interneurons were found to be weak. We postulate that these projections may become functionally active upon intense network activity during tasks requiring increased memory processing.


Assuntos
Axônios/metabolismo , Hipocampo/metabolismo , Interneurônios/metabolismo , Somatostatina/biossíntese , Animais , Feminino , Expressão Gênica , Hipocampo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Somatostatina/genética
12.
Cell Rep ; 26(12): 3173-3182.e5, 2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893591

RESUMO

Synchronized activity is a universal characteristic of immature neural circuits that is essential for their developmental refinement and strongly depends on GABAergic neurotransmission. A major subpopulation of GABA-releasing interneurons (INs) expresses somatostatin (SOM) and proved critical for rhythm generation in adulthood. Here, we report a mechanism whereby SOM INs promote neuronal synchrony in the neonatal CA1 region. Combining imaging and electrophysiological approaches, we demonstrate that SOM INs and pyramidal cells (PCs) coactivate during spontaneous activity. Bidirectional optogenetic manipulations reveal excitatory GABAergic outputs to PCs that evoke correlated network events in an NKCC1-dependent manner and contribute to spontaneous synchrony. Using a dynamic systems modeling approach, we show that SOM INs affect network dynamics through a modulation of network instability and amplification threshold. Our study identifies a network function of SOM INs with implications for the activity-dependent construction of developing brain circuits.


Assuntos
Hipocampo/metabolismo , Interneurônios/metabolismo , Células Piramidais/metabolismo , Somatostatina/biossíntese , Transmissão Sináptica , Animais , Hipocampo/citologia , Interneurônios/citologia , Camundongos , Camundongos Transgênicos , Optogenética , Células Piramidais/citologia
13.
Biol Psychiatry ; 85(6): 517-526, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30449530

RESUMO

BACKGROUND: A parallel downregulation of brain-derived neurotrophic factor (BDNF) and somatostatin (SST), a marker of inhibitory gamma-aminobutyric acid interneurons that target pyramidal cell dendrites, has been reported in several brain areas of subjects with major depressive disorder (MDD). Rodent genetic studies suggest that they are linked and that both contribute to the illness. However, the mechanism by which they contribute to the pathophysiology of the illness has remained elusive. METHODS: With quantitative polymerase chain reaction, we determined the expression level of BDNF transcript variants and synaptic markers in the prefrontal cortex of patients with MDD and matched control subjects (n = 19/group) and of C57BL/6J mice exposed to chronic stress or control conditions (n = 12/group). We next suppressed Bdnf transcripts with long 3' untranslated region (L-3'-UTR) using short hairpin RNA and investigated changes in cell morphology, gene expression, and behavior. RESULTS: L-3'-UTRs containing BDNF messenger RNAs, which migrate to distal dendrites of pyramidal neurons, are selectively reduced, and their expression was highly correlated with SST expression in the prefrontal cortex of subjects with MDD. A similar downregulation occurs in mice submitted to chronic stress. We next show that Bdnf L-3'-UTR knockdown is sufficient to induce 1) dendritic shrinkage in cortical neurons, 2) cell-specific MDD-like gene changes (including Sst downregulation), and 3) depressive- and anxiety-like behaviors. The translational validity of the Bdnf L-3'-UTR short hairpin RNA-treated mice was confirmed by significant cross-species correlation of changes in MDD-associated gene expression. CONCLUSIONS: These findings provide evidence for a novel MDD-related pathological mechanism linking local neurotrophic support, pyramidal cell structure, dendritic inhibition, and mood regulation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Dendritos/metabolismo , Transtorno Depressivo Maior/metabolismo , Somatostatina/biossíntese , Animais , Atrofia/patologia , Comportamento Animal , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Estudos de Casos e Controles , Dendritos/patologia , Regulação para Baixo , Feminino , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Córtex Pré-Frontal/metabolismo , Cultura Primária de Células , Células Piramidais/metabolismo , RNA Interferente Pequeno/farmacologia , Estresse Psicológico/metabolismo
14.
Stem Cell Res Ther ; 9(1): 277, 2018 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-30359326

RESUMO

BACKGROUND: Transplantation of pancreatic ß cells generated in vitro from pluripotent stem cells (hPSCs) such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) has been proposed as an alternative therapy for diabetes. Though many differentiation protocols have been developed for this purpose, lentivirus-mediated forced expression of transcription factors (TF)-PDX1 and NKX6.1-has been at the forefront for its relatively fast and straightforward approach. However, considering that such cells will be used for therapeutic purposes in the future, it is desirable to develop a procedure that does not leave any footprint on the genome, as any changes of DNAs could potentially be a source of unintended, concerning effects such as tumorigenicity. In this study, we attempted to establish a novel protocol for rapid and footprint-free hESC differentiation into a pancreatic endocrine lineage by using synthetic mRNAs (synRNAs) encoding PDX1 and NKX6.1. We also tested whether siPOU5F1, which reduces the expression of pluripotency gene POU5F1 (also known as OCT4), can enhance differentiation as reported previously for mesoderm and endoderm lineages. METHODS: synRNA-PDX1 and synRNA-NKX6.1 were synthesized in vitro and were transfected five times to hESCs with a lipofection reagent in a modified differentiation culture condition. siPOU5F1 was included only in the first transfection. Subsequently, cells were seeded onto a low attachment plate and aggregated by an orbital shaker. At day 13, the degree of differentiation was assessed by quantitative RT-PCR (qRT-PCR) and immunohistochemistry for endocrine hormones such as insulin, glucagon, and somatostatin. RESULTS: Both PDX1 and NKX6.1 expression were detected in cells co-transfected with synRNA-PDX1 and synRNA-NKX6.1 at day 3. Expression levels of insulin in the transfected cells at day 13 were 450 times and 14 times higher by qRT-PCR compared to the levels at day 0 and in cells cultured without synRNA transfection, respectively. Immunohistochemically, pancreatic endocrine hormones were not detected in cells cultured without synRNA transfection but were highly expressed in cells transfected with synRNA-PDX1, synRNA-NKX6.1, and siPOU5F1 at as early as day 13. CONCLUSIONS: In this study, we report a novel protocol for rapid and footprint-free differentiation of hESCs to endocrine cells.


Assuntos
Proteínas de Homeodomínio/genética , Células-Tronco Embrionárias Humanas/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , RNA Mensageiro/síntese química , Transativadores/genética , Diferenciação Celular , Engenharia Celular/métodos , Células Cultivadas , Regulação da Expressão Gênica , Glucagon/biossíntese , Proteínas de Homeodomínio/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Células Secretoras de Insulina/citologia , Lipídeos/química , Fator 3 de Transcrição de Octâmero/antagonistas & inibidores , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Somatostatina/biossíntese , Transativadores/metabolismo , Transfecção
15.
Neuron ; 100(4): 926-939.e3, 2018 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-30318409

RESUMO

Decreased hippocampal-prefrontal synchrony may mediate cognitive deficits in schizophrenia, but it remains unclear which cells orchestrate this long-range synchrony. Parvalbumin (PV)- and somatostatin (SOM)-expressing interneurons show histological abnormalities in individuals with schizophrenia and are hypothesized to regulate oscillatory synchrony within the prefrontal cortex. To examine the relationship between interneuron function, long-range hippocampal-prefrontal synchrony, and cognition, we optogenetically inhibited SOM and PV neurons in the medial prefrontal cortex (mPFC) of mice performing a spatial working memory task while simultaneously recording neural activity in the mPFC and the hippocampus (HPC). We found that inhibiting SOM, but not PV, interneurons during the encoding phase of the task impaired working memory accuracy. This behavioral impairment was associated with decreased hippocampal-prefrontal synchrony and impaired spatial encoding in mPFC neurons. These findings suggest that interneuron dysfunction may contribute to cognitive deficits associated with schizophrenia by disrupting long-range synchrony between the HPC and PFC.


Assuntos
Hipocampo/metabolismo , Interneurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Somatostatina/biossíntese , Animais , Hipocampo/química , Interneurônios/química , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Vias Neurais/química , Vias Neurais/metabolismo , Optogenética/métodos , Parvalbuminas/análise , Parvalbuminas/biossíntese , Córtex Pré-Frontal/química , Somatostatina/análise
16.
PLoS One ; 13(7): e0200567, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30001424

RESUMO

Somatostatin-expressing (SOM+), inhibitory interneurons represent a heterogeneous group of cells and given their remarkable diversity, classification of SOM+ interneurons remains a challenging task. Electrophysiological, morphological and neurochemical classes of SOM+ interneurons have been proposed in the past but it remains unclear as to what extent these classes are congruent. We performed whole-cell patch-clamp recordings from 127 GFP-labeled SOM+ interneurons ('GIN') of the superficial cingulate cortex with subsequent biocytin-filling and immunocytochemical labeling. Principal component analysis followed by k-means clustering predicted two putative subtypes of SOM+ interneurons, which we designated as group I and group II GIN. A key finding of our study is the fact that these electrophysiologically and morphologically distinct groups of SOM+ interneurons can be correlated with two neurochemical subtypes of SOM+ interneurons described recently in our laboratory. In particular, all SOM+ interneurons expressing calbindin but no calretinin could be classified as group I GIN, whereas all but one neuropeptide Y- and calretinin-positive interneurons were found in group II.


Assuntos
Neurônios GABAérgicos , Giro do Cíngulo , Interneurônios , Somatostatina/biossíntese , Animais , Calbindinas/biossíntese , Neurônios GABAérgicos/classificação , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Giro do Cíngulo/citologia , Giro do Cíngulo/metabolismo , Interneurônios/classificação , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Neuropeptídeo Y/biossíntese
17.
Nat Neurosci ; 21(5): 707-716, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29556030

RESUMO

Stimuli that elicit itch are detected by sensory neurons that innervate the skin. This information is processed by the spinal cord; however, the way in which this occurs is still poorly understood. Here we investigated the neuronal pathways for itch neurotransmission, particularly the contribution of the neuropeptide somatostatin. We find that in the periphery, somatostatin is exclusively expressed in Nppb+ neurons, and we demonstrate that Nppb+somatostatin+ cells function as pruriceptors. Employing chemogenetics, pharmacology and cell-specific ablation methods, we demonstrate that somatostatin potentiates itch by inhibiting inhibitory dynorphin neurons, which results in disinhibition of GRPR+ neurons. Furthermore, elimination of somatostatin from primary afferents and/or from spinal interneurons demonstrates differential involvement of the peptide released from these sources in itch and pain. Our results define the neural circuit underlying somatostatin-induced itch and characterize a contrasting antinociceptive role for the peptide.


Assuntos
Vias Neurais/fisiopatologia , Dor/fisiopatologia , Prurido/fisiopatologia , Somatostatina/metabolismo , Animais , Dinorfinas/metabolismo , Feminino , Gânglios Espinais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Optogenética , Dor/metabolismo , Prurido/metabolismo , Receptores do Fator Natriurético Atrial/biossíntese , Receptores do Fator Natriurético Atrial/metabolismo , Receptores Purinérgicos/metabolismo , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/genética , Células Receptoras Sensoriais , Somatostatina/biossíntese , Medula Espinal/citologia , Medula Espinal/fisiopatologia
18.
Future Oncol ; 14(12): 1155-1164, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29350062

RESUMO

Carcinoid syndrome (CS), characterized by diarrhea and flushing, is present in 20% of patients with neuroendocrine tumors at diagnosis and becomes more frequent with progression. The diarrhea of CS is caused mainly by tumoral secretion of serotonin. It may not be fully controlled by somatostatin analogs, the currently indicated drugs for symptomatic relief. Telotristat ethyl is a novel inhibitor of tryptophan hydroxylase, the rate-limiting enzyme in serotonin biosynthesis. Administration of the drug decreases diarrhea in patients with CS. Telotristat ethyl was approved in February 2017 (USA) and September 2017 (European Commission) for the treatment of CS diarrhea in adults inadequately controlled by somatostatin analog alone. This drug is expected to greatly improve the health and quality of life of patients with CS diarrhea.


Assuntos
Diarreia/tratamento farmacológico , Síndrome do Carcinoide Maligno/tratamento farmacológico , Fenilalanina/análogos & derivados , Pirimidinas/uso terapêutico , Qualidade de Vida , Triptofano Hidroxilase/antagonistas & inibidores , Diarreia/etiologia , Humanos , Síndrome do Carcinoide Maligno/complicações , Síndrome do Carcinoide Maligno/epidemiologia , Síndrome do Carcinoide Maligno/patologia , Fenilalanina/farmacologia , Fenilalanina/uso terapêutico , Prevalência , Pirimidinas/farmacologia , Somatostatina/biossíntese , Resultado do Tratamento , Triptofano Hidroxilase/metabolismo
19.
J Neurosci ; 38(7): 1622-1633, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29326172

RESUMO

Inhibitory interneurons represent 10-15% of the neurons in the somatosensory cortex, and their activity powerfully shapes sensory processing. Three major groups of GABAergic interneurons have been defined according to developmental, molecular, morphological, electrophysiological, and synaptic features. Dendritic-targeting somatostatin-expressing interneurons (SST-INs) have been shown to display diverse morphological, electrophysiological, and molecular properties and activity patterns in vivo However, the correlation between these properties and SST-IN subtype is unclear. In this study, we aimed to correlate the morphological diversity of layer 5 (L5) SST-INs with their electrophysiological and molecular diversity in mice of either sex. Our morphological analysis demonstrated the existence of three subtypes of L5 SST-INs with distinct electrophysiological properties: T-shaped Martinotti cells innervate L1, and are low-threshold spiking; fanning-out Martinotti cells innervate L2/3 and the lower half of L1, and show adapting firing patterns; non-Martinotti cells innervate L4, and show a quasi-fast spiking firing pattern. We estimated the proportion of each subtype in L5 and found that T-shaped Martinotti, fanning-out Martinotti, and Non-Martinotti cells represent ∼10, ∼50, and ∼40% of L5 SST-INs, respectively. Last, we examined the connectivity between the three SST-IN subtypes and L5 pyramidal cells (PCs). We found that L5 T-shaped Martinotti cells inhibit the L1 apical tuft of nearby PCs; L5 fanning-out Martinotti cells also inhibit nearby PCs but they target the dendrite mainly in L2/3. On the other hand, non-Martinotti cells inhibit the dendrites of L4 neurons while avoiding L5 PCs. Our data suggest that morphologically distinct SST-INs gate different excitatory inputs in the barrel cortex.SIGNIFICANCE STATEMENT Morphologically diverse layer 5 SST-INs show different patterns of activity in behaving animals. However, little is known about the abundance and connectivity of each morphological type and the correlation between morphological subtype and spiking properties. We demonstrate a correlation between the morphological and electrophysiological diversity of layer 5 SST-INs. Based on these findings we built a classifier to infer the abundance of each morphological subtype. Last, using paired recordings combined with morphological analysis, we investigated the connectivity of each morphological subtype. Our data suggest that, by targeting different cell types and cellular compartments, morphologically diverse SST-INs might gate different excitatory inputs in the mouse barrel cortex.


Assuntos
Interneurônios/fisiologia , Vias Neurais/fisiologia , Córtex Somatossensorial/fisiologia , Somatostatina/biossíntese , Animais , Dendritos/fisiologia , Dendritos/ultraestrutura , Fenômenos Eletrofisiológicos/fisiologia , Feminino , Técnicas In Vitro , Interneurônios/ultraestrutura , Masculino , Camundongos , Inibição Neural/fisiologia , Vias Neurais/citologia , Vias Neurais/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Células Piramidais/fisiologia , Células Piramidais/ultraestrutura , Córtex Somatossensorial/citologia , Somatostatina/genética
20.
J Cell Biol ; 216(12): 4299-4311, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29025873

RESUMO

The recent demonstration that pancreatic α cells can be continuously regenerated and converted into ß-like cells upon ectopic expression of Pax4 opened new avenues of research in the endocrine cell differentiation and diabetes fields. To determine whether such plasticity was also shared by δ cells, we generated and characterized transgenic animals that express Pax4 specifically in somatostatin-expressing cells. We demonstrate that the ectopic expression of Pax4 in δ cells is sufficient to induce their conversion into functional ß-like cells. Importantly, this conversion induces compensatory mechanisms involving the reactivation of endocrine developmental processes that result in dramatic ß-like cell hyperplasia. Importantly, these ß-like cells are functional and can partly reverse the consequences of chemically induced diabetes.


Assuntos
Diabetes Mellitus Experimental/genética , Expressão Ectópica do Gene , Proteínas de Homeodomínio/genética , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Box Pareados/genética , Células Secretoras de Somatostatina/metabolismo , Animais , Proliferação de Células , Transdiferenciação Celular/genética , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/terapia , Terapia Genética/métodos , Glucagon/biossíntese , Glucagon/genética , Proteínas de Homeodomínio/metabolismo , Insulina/biossíntese , Insulina/genética , Células Secretoras de Insulina/citologia , Masculino , Camundongos , Camundongos Transgênicos , Fatores de Transcrição Box Pareados/metabolismo , Somatostatina/biossíntese , Somatostatina/genética , Células Secretoras de Somatostatina/citologia , Estreptozocina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...