Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
bioRxiv ; 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38853855

RESUMO

Psychological states can regulate intestinal mucosal immunity by altering the gut microbiome. However, the link between the brain and microbiome composition remains elusive. We show that Brunner's glands in the duodenal submucosa couple brain activity to intestinal bacterial homeostasis. Brunner's glands mediated the enrichment of gut probiotic species in response to stimulation of abdominal vagal fibers. Cell-specific ablation of the glands triggered transmissible dysbiosis associated with an immunodeficiency syndrome that led to mortality upon gut infection with pathogens. The syndrome could be largely prevented by oral or intra-intestinal administration of probiotics. In the forebrain, we identified a vagally-mediated, polysynaptic circuit connecting the glands of Brunner to the central nucleus of the amygdala. Intra-vital imaging revealed that excitation of central amygdala neurons activated Brunner's glands and promoted the growth of probiotic populations. Our findings unveil a vagal-glandular neuroimmune circuitry that may be targeted for the modulation of the gut microbiome. The glands of Brunner may be the critical cells that regulate the levels of Lactobacilli species in the intestine.

2.
Res Sq ; 2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37961128

RESUMO

Chronic stress underlies the etiology of both major depressive disorder (MDD) and irritable bowel syndrome (IBS), two highly prevalent and debilitating conditions with high rates of co-morbidity. However, it is not fully understood how the brain and gut bi-directionally communicate during stress to impact intestinal homeostasis and stress-relevant behaviours. Using the chronic social defeat stress (CSDS) model, we find that stressed mice display greater intestinal permeability and circulating levels of the endotoxin lipopolysaccharide (LPS) compared to unstressed control (CON) mice. Interestingly, the microbiota in the colon also exhibit elevated LPS biosynthesis gene expression following CSDS. Additionally, CSDS triggers an increase in pro-inflammatory colonic IFNγ+ Th1 cells and a decrease in IL4+ Th2 cells compared to CON mice, and this gut inflammation contributes to stress-induced intestinal barrier permeability and social avoidance behaviour. We next investigated the role of enteric neurons and identified that noradrenergic dopamine beta-hydroxylase (DBH)+ neurons in the colon are activated by CSDS, and that their ablation protects against gut pathophysiology and disturbances in social behaviour. Retrograde tracing from the colon identified a population of corticotropin-releasing hormone-expressing (CRH+) neurons in the paraventricular nucleus of the hypothalamus (PVH) that innervate the colon and are activated by stress. Chemogenetically activating these PVH CRH+ neurons is sufficient to induce gut inflammation, barrier permeability, and social avoidance behaviour, while inhibiting these cells prevents these effects following exposure to CSDS. Thus, we define a stress-activated brain-to-gut circuit that confers colonic inflammation, leading to impaired intestinal barrier function, and consequent behavioural deficits.

3.
BMC Pulm Med ; 23(1): 482, 2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38031067

RESUMO

BACKGROUND: The efficacy of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC) patients harboring neurotrophin receptor kinase (NTRK) family mutations remains obscure. METHODS: The Zehir cohort from cBioPortal was used to analyze the mutations (MT) frequency of NTRK family in patients with NSCLC, and their correlation with clinical characteristics and patient survival. The influence of NTRK MT on ICIs efficacy was evaluated in ICIs-treated patients from Samstein cohort and further validated by use of data from OAK/POPLAR cohort. RESULTS: In the Zehir cohort, a significant difference was observed in median overall survival (mOS) between patients with NTRK MT and wild-type (WT) (mOS: 18.97 vs. 21.27 months, HR = 1.34, 95%CI 1.00-1.78; log-rank P = 0.047). In Samstein cohort, the mOS of NTRK mutant patients receiving ICIs has improved compared to WT patients (mOS: 21.00 vs. 11.00 months, log-rank P = 0.103). Notably, in subgroup analysis, ICIs significantly prolonged mOS in patients with NTRK3 MT than in WT patients (mOS: not available vs. 11.00 months, HR = 0.36, 95%CI 0.16-0.81; log-rank P = 0.009). Identical mOS between NTRK MT and WT patients receiving ICIs treatment (mOS: 13.24 vs. 13.50 months, log-rank P = 0.775) was observed in OAK/POPLAR cohort. Moreover, a similar programmed death ligand 1 (PD-L1) expression, but higher tumor mutational burden (TMB), blood TMB (bTMB) and enriched anti-tumor immunity were observed in NTRK MT compared to WT (P < 0.05). CONCLUSION: Taking high TMB or bTMB into consideration, patients with NTRK mutant NSCLC could benefit from ICIs treatment.


Assuntos
Antineoplásicos Imunológicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Mutação , Biomarcadores Tumorais/genética
4.
Cell Rep ; 42(3): 112190, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36857179

RESUMO

Although the consumption of carbohydrates is needed for survival, their potent reinforcing properties drive obesity worldwide. In turn, sugar overconsumption reveals a major role for brain reward systems in regulating sugar intake. However, it remains elusive how different cell types within the reward circuitries control the initiation and termination of sugary meals. Here, we identified the distinct nucleus accumbens cell types that mediate the chemosensory versus postprandial properties of sweet sugars. Specifically, D1 neurons enhance sugar intake via specialized connections to taste ganglia, whereas D2 neurons mediate the termination of sugary meals via anatomical connections to circuits involved in appetite suppression. Consistently, D2, but not D1, neurons partially mediate the satiating effects of glucagon-like peptide 1 (GLP-1) agonists. Thus, these nucleus accumbens cell types function as a behavioral switch, enabling positive versus negative control over sugar intake. Our study contributes to unveiling the cellular and circuit substrates of sugar overconsumption.


Assuntos
Neurônios , Núcleo Accumbens , Camundongos , Animais , Núcleo Accumbens/metabolismo , Neurônios/metabolismo , Encéfalo/metabolismo , Açúcares/metabolismo , Receptores de Dopamina D1/metabolismo
5.
Front Immunol ; 13: 931718, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35990690

RESUMO

Background: Although immune checkpoint inhibitors (ICIs) generally show poor therapeutic efficacy in patients with epidermal growth factor receptor (EGFR) mutations, certain research indicate that a small proportion of these patients do respond to ICIs. The present study sought to identify the features of patients with EGFR mutations who might benefit from ICIs from multiple studies and discussed the optimal treatment paradigm for advanced non-small cell lung cancer (NSCLC) patients with EGFR mutations. Methods: The profiles of 114 advanced NSCLC patients with EGFR mutations who received ICIs treatment were retrospectively reviewed. EGFR subtypes, programmed cell death ligand 1 (PD-L1) expression, and clinical characteristics regarding their impact on the efficacy of ICIs were investigated. Results: Patients with major EGFR mutations (L858R or 19Del) had a shorter progression-free survival (PFS) and a lower objective response rate (ORR) as compared to patients with rare (20ins or G719X) and other EGFR mutations. Although not statistically significant, median overall survival (OS) tended to be longer in patients with negative (<1%) PD-L1 expression than with positive (≥1%) PD-L1 expression (15.61 vs. 7.40 months, p = 0.138). Median PFS and OS were significantly shorter in heavily treated patients (prior lines of therapy ≥3 lines vs. <3 lines: mPFS, 1.80 vs. 2.50 months, p = 0.003; mOS, 6.70 vs. 14.00 months, p = 0.031). ORR was also lower in patients who had received ≥3 prior lines of therapy compared to in those <3 prior lines of therapy (0.00% vs. 21.67%, p = 0.002). Conclusion: Patients with major EGFR mutations showed poorer responses to ICIs than those with rare EGFR mutations. EGFR-mutated patients with lower PD-L1 expression showed a trend towards a longer OS after receiving ICIs. ICIs should be administered as early as possible to previously treated EGFR-mutated NSCLC patients. ICI-based combined therapies may be a direction for treatment of these patient subtypes in the future.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antígeno B7-H1 , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Estudos Retrospectivos
6.
Cell ; 185(14): 2478-2494.e28, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35662413

RESUMO

Glucagon-like peptide-1 (GLP-1) is a signal peptide released from enteroendocrine cells of the lower intestine. GLP-1 exerts anorectic and antimotility actions that protect the body against nutrient malabsorption. However, little is known about how intestinal GLP-1 affects distant organs despite rapid enzymatic inactivation. We show that intestinal GLP-1 inhibits gastric emptying and eating via intestinofugal neurons, a subclass of myenteric neurons that project to abdominal sympathetic ganglia. Remarkably, cell-specific ablation of intestinofugal neurons eliminated intestinal GLP-1 effects, and their chemical activation functioned as a GLP-1 mimetic. GLP-1 sensing by intestinofugal neurons then engaged a sympatho-gastro-spinal-reticular-hypothalamic pathway that links abnormal stomach distension to craniofacial programs for food rejection. Within this pathway, cell-specific activation of discrete neuronal populations caused systemic GLP-1-like effects. These molecularly identified, delimited enteric circuits may be targeted to ameliorate the abdominal bloating and loss of appetite typical of gastric motility disorders.


Assuntos
Apetite , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Íleo , Neurônios , Estômago , Abdome , Animais , Comunicação Celular , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Íleo/inervação , Íleo/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais , Estômago/inervação , Estômago/metabolismo
7.
Front Oncol ; 12: 867788, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35574406

RESUMO

The application of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer has significantly improved patient survival. However, most patients fail to respond to ICIs or develop drug resistance during treatment. Therefore, novel biomarkers are needed to predict the efficacy of ICIs or provide clues on how to overcome drug resistance. Here, it was revealed that cell division cycle 25C (CDC25C) expression was upregulated in lung adenocarcinoma (LUAD) compared to that of normal lung tissue in multiple databases. This was further verified by q-PCR. Furthermore, higher CDC25C expression was associated with shorter overall survival and worse pathological stage. Most importantly, a higher CDC25C expression was associated with shorter progression-free survival in LUAD patients treated with nivolumab, suggesting the role of the cell cycle in immunotherapy. In addition, CDC25C expression was significantly associated with immune cell infiltration and immune-related signatures in the LUAD tumor microenvironment. Moreover, CDC25C was differentially expressed and correlated with overall survival in multiple tumors, indicating that CDC25C is a broad-spectrum biomarker. Taken together, our study demonstrates that CDC25C is a prognostic biomarker for LUAD patients, especially for patients treated with ICIs. Our study also provides strong evidence for the role of the cell cycle in ICIs therapy and tumor microenvironment.

8.
PeerJ ; 10: e12802, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35186455

RESUMO

Pancreatic cancer (PC) often correlates with high mortality due to late diagnosis, rapid metastasis, and resistance to chemotherapy. miR-128-3p has been validated as a tumor suppressor in PC. This study explored the functional mechanism of miR-128-3p in epithelial-mesenchymal transition (EMT) of PC cells. Four PC cancer cell lines with different degrees of malignancy and normal pancreatic cells were selected to detect expressions of hsa-miR-128-3p and ZEB1 by RT-qPCR and Western blot. miR-128-3p mimic or si-ZEB1 was delivered into PANC-1 cells and miR-128-3p inhibitor or oe-ZEB1 was delivered into AsPC-1 cells. Expressions of epithelial and mesenchymal markers were analyzed by Western blot and cell fluorescence staining. The binding relationship between miR-128-3p and ZEB1 was examined by bioinformatics analysis and dual-luciferase assay, and verified by RT-qPCR and Western blot. PC cell invasion and migration were assessed by Transwell assays. Generally, hsa-miR-128-3p was poorly-expressed in PC cells. However, it was relatively more expressed in AsPC-1 cells with epithelial phenotypes relative to PANC-1 cells with mesenchymal phenotype, whereas ZEB1 expression showed opposite tendencies. PANC-1 cells transfected with miR-128-3p mimic or si-ZEB1 showed upregulated E-cadherin and downregulated N-cadherin, and transformed from mesenchymal phenotypes to epithelial phenotypes, with decreased invasion and migration, while opposite results occurred in AsPC-1 cells transfected with miR-128-3p inhibitor or oe-ZEB1. miR-128-3p targeted ZEB1. oe-ZEB1 antagonized the inhibition of miR-128-3p mimic on PANC-1 cell EMT, invasion, and migration, while si-ZEB1 reversed the facilitation of miR-128-3p inhibitor in AsPC-1 cells. In conclusion, miR-128-3p inhibited PC cell EMT, invasion, and migration by targeting ZEB1.


Assuntos
MicroRNAs , Neoplasias Pancreáticas , Humanos , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , MicroRNAs/genética , Hormônios Pancreáticos , Neoplasias Pancreáticas/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Neoplasias Pancreáticas
9.
Thorac Cancer ; 12(22): 3053-3061, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34617400

RESUMO

The application of chemotherapy in non-small cell lung cancer (NSCLC) is limited by the toxicity to normal cells and the development of multi-drug resistance. Targeted chemotherapy using cytotoxic analogs against specific receptors on cancer cells could be a less toxic and more efficacious approach. We identified that the expressions of somatostatin receptor (SSTR) 2 and 5 in tumor tissues from NSCLC patients were higher than those in the adjacent normal tissues by immunohistochemistry, and therefore, cytotoxic somatostatin analogues might be applied for SSTRs-mediated targeted therapy against NSCLC. Two cytotoxic analogs, paclitaxel-octreotide (PTX-OCT) and 2paclitaxel-octreotide (2PTX-OCT), were synthesized by linking one or two molecules of paclitaxel to one molecule of somatostatin analog octreotide. PTX-OCT and 2PTX-OCT significantly inhibited the growth and induced apoptosis of SSTR2- and SSTR5-positive A549 cells, compared with the control (p < 0.01), and had less inhibitory effect on SSTR2- and SSTR5-negative H157 cells than paclitaxel (p < 0.01). Moreover, compared with paclitaxel, PTX-OCT conjugates induced lower expression of MDR-1 gene both in vitro and in vivo. Three A549 paclitaxel-resistant cell lines were established through different approaches, and the paclitaxel-resistant cell showed higher sensitivity to PTX-OCT conjugates than to paclitaxel, which might be because of the differential MDR-related gene expressions and cell-cycle distribution in paclitaxel-resistant A549 cells. Our results suggested that PTX-OCT conjugates could be potentially used for SSTRs-mediated targeted therapy for NSCLC, especially for those with paclitaxel resistance and induced less multidrug resistance.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Octreotida/farmacologia , Paclitaxel/farmacologia , Receptores de Somatostatina/metabolismo , Células A549 , Adulto , Idoso , Animais , Antineoplásicos Hormonais/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade
10.
STAR Protoc ; 2(2): 100474, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33997807

RESUMO

The jugular-nodose ganglia contain the sensory peripheral neurons of the vagus nerve, linking visceral organs to the medulla oblongata. Accessing these ganglia in smaller animals without damaging the vascular and neural structures may be challenging, as ganglionic fibers imbed deeply into the carotid sheath, and vagal parasympathetic fibers cross through the interior of the ganglia. We describe a practical protocol for locating and accessing the mouse jugular-nodose ganglia in vivo, including instructions for intraganglionic injections and postperfusion dissection. For complete details on the use and execution of this protocol, please refer to Han et al. (2018).


Assuntos
Dissecação/métodos , Gânglio Nodoso , Animais , Feminino , Forâmen Jugular/inervação , Masculino , Camundongos , Gânglio Nodoso/anatomia & histologia , Gânglio Nodoso/cirurgia
11.
Neuron ; 109(3): 391-393, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33539771

RESUMO

Area postrema in brainstem has long been known to trigger emesis by detecting blood-borne toxins and pathogens. In this issue, Zhang and colleagues provide a single-cell molecular atlas of this region, opening new possibilities for harnessing its neurons in vivo.


Assuntos
Área Postrema , Náusea , Tronco Encefálico , Humanos , Neurônios , Vômito
13.
Nature ; 583(7816): 441-446, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32641826

RESUMO

Connections between the gut and brain monitor the intestinal tissue and its microbial and dietary content1, regulating both physiological intestinal functions such as nutrient absorption and motility2,3, and brain-wired feeding behaviour2. It is therefore plausible that circuits exist to detect gut microorganisms and relay this information to areas of the central nervous system that, in turn, regulate gut physiology4. Here we characterize the influence of the microbiota on enteric-associated neurons by combining gnotobiotic mouse models with transcriptomics, circuit-tracing methods and functional manipulations. We find that the gut microbiome modulates gut-extrinsic sympathetic neurons: microbiota depletion leads to increased expression of the neuronal transcription factor cFos, and colonization of germ-free mice with bacteria that produce short-chain fatty acids suppresses cFos expression in the gut sympathetic ganglia. Chemogenetic manipulations, translational profiling and anterograde tracing identify a subset of distal intestine-projecting vagal neurons that are positioned to have an afferent role in microbiota-mediated modulation of gut sympathetic neurons. Retrograde polysynaptic neuronal tracing from the intestinal wall identifies brainstem sensory nuclei that are activated during microbial depletion, as well as efferent sympathetic premotor glutamatergic neurons that regulate gastrointestinal transit. These results reveal microbiota-dependent control of gut-extrinsic sympathetic activation through a gut-brain circuit.


Assuntos
Microbioma Gastrointestinal/fisiologia , Intestinos/inervação , Neurônios/fisiologia , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/fisiologia , Animais , Disbiose/fisiopatologia , Feminino , Gânglios Simpáticos/citologia , Gânglios Simpáticos/fisiologia , Motilidade Gastrointestinal , Vida Livre de Germes , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Vias Neurais/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Transcriptoma
14.
J Clin Invest ; 130(9): 4985-4998, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32516139

RESUMO

The brain has evolved in an environment where food sources are scarce, and foraging for food is one of the major challenges for survival of the individual and species. Basic and clinical studies show that obesity or overnutrition leads to overwhelming changes in the brain in animals and humans. However, the exact mechanisms underlying the consequences of excessive energy intake are not well understood. Neurons expressing the neuropeptide hypocretin/orexin (Hcrt) in the lateral/perifonical hypothalamus (LH) are critical for homeostatic regulation, reward seeking, stress response, and cognitive functions. In this study, we examined adaptations in Hcrt cells regulating behavioral responses to salient stimuli in diet-induced obese mice. Our results demonstrated changes in primary cilia, synaptic transmission and plasticity, cellular responses to neurotransmitters necessary for reward seeking, and stress responses in Hcrt neurons from obese mice. Activities of neuronal networks in the LH and hippocampus were impaired as a result of decreased hypocretinergic function. The weakened Hcrt system decreased reward seeking while altering responses to acute stress (stress-coping strategy), which were reversed by selectively activating Hcrt cells with chemogenetics. Taken together, our data suggest that a deficiency in Hcrt signaling may be a common cause of behavioral changes (such as lowered arousal, weakened reward seeking, and altered stress response) in obese animals.


Assuntos
Comportamento Alimentar , Hipotálamo , Rede Nervosa , Neurônios , Obesidade , Orexinas , Animais , Hipotálamo/metabolismo , Hipotálamo/patologia , Hipotálamo/fisiopatologia , Masculino , Camundongos , Camundongos Transgênicos , Rede Nervosa/metabolismo , Rede Nervosa/patologia , Rede Nervosa/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Orexinas/genética , Orexinas/metabolismo , Estresse Psicológico/genética , Estresse Psicológico/metabolismo , Estresse Psicológico/patologia , Estresse Psicológico/fisiopatologia
15.
Appetite ; 139: 145-151, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31029689

RESUMO

BACKGROUND AND AIM: In most species, including humans, food preference is primarily controlled by nutrient value. However, the gut-brain pathways involved in preference learning remain elusive. The aim of the present study, performed in C57BL6/J mice, was to characterize the roles in nutrient preference of two critical elements of gut-brain pathways, i.e. the duodenum and vagal gut innervation. METHODS: Adult wild-type C57BL6/J mice from a normal-weight cohort sustained one of the following three procedures: duodenal-jejunal bypass intestinal rerouting (DJB), total subdiaphragmatic vagotomy (SDV), or sham surgery. Mice were assessed in short-term two-bottle preference tests before and after 24 h s exposures to solutions containing one of glutamate, lipids, sodium, or glucose. RESULTS: DJB and SDV interfered in preference formation in a nutrient-specific manner: whereas normal preference learning for lipids and glutamate was disrupted by both DJB and SDV, these interventions did not alter the formation of preferences for glucose. Interestingly, sodium preferences were abrogated by DJB but not by SDV. CONCLUSIONS: Different macronutrients make use of distinct gut-brain pathways to influence food preferences, thereby mirroring nutrient-specific processes of food digestion. Specifically, whereas both vagal innervation and duodenal sensing appear critical for generating responses to fats and protein, glucose preferences recruit post-duodenal, vagal-independent pathways in pair with the control of glucose homeostasis. Overall, our data suggest that the physiological processes involved in digesting and absorbing fats, amino acids, and glucose overlap with those mediating learned preferences for each of these nutrients.


Assuntos
Encéfalo/fisiologia , Duodeno/inervação , Preferências Alimentares/fisiologia , Nutrientes/fisiologia , Nervo Vago/fisiologia , Animais , Digestão/fisiologia , Duodeno/cirurgia , Derivação Gástrica , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nervo Vago/cirurgia
16.
Front Integr Neurosci ; 12: 57, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30519164

RESUMO

In most species, including humans, food preference is primarily controlled by nutrient value. In particular, glucose-containing sugars exert exquisitely strong effects on food choice via gut-generated signals. However, the identity of the visceral signals underlying glucose's rewarding effects remains uncertain. In particular, it is unknown whether sugar metabolism mediates the formation of preferences for glucose-containing sugars. Using the mouse as a model organism, we made use of a combination of conditioning schedules, gastrointestinal nutrient administration, and chromatographic/electrochemical methods to assess the behavioral and neural effects of activating the gut with either metabolizable glucose or a non-metabolizable glucose analog. We show that mice display much superior preferences for flavors associated with intra-gastric infusions of glucose compared to flavors associated with intra-gastric infusions of the non-metabolizable glucose analog α-methyl-D-glucopyranoside ("MDG," an activator of intestinal sodium/glucose co-transporters). These effects were unaffected by surgical bypassing of the duodenum, suggesting glucose-specific post-absorptive sensing mechanisms. Consistently, intra-portal infusions of glucose, but not of MDG, induced significant rises in dopamine (DA) levels within brain reward circuits. Our data reveal that the unmatched rewarding effects of glucose-containing sugars cannot be accounted for by metabolism-independent activation of sodium/glucose cotransporters; rather, they point to glucose metabolism as the physiological mechanism underlying the potent reward value of sugar-sweetened flavored beverages. In particular, no circulating "gut factors" need to be invoked to explain the reward value of ingested glucose. Thus, instead of circulating gut hormones, portal-mesenteric sensing of glucose emerges as the preferential physiological pathway for sugar reward.

18.
Cell ; 175(3): 665-678.e23, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30245012

RESUMO

The gut is now recognized as a major regulator of motivational and emotional states. However, the relevant gut-brain neuronal circuitry remains unknown. We show that optical activation of gut-innervating vagal sensory neurons recapitulates the hallmark effects of stimulating brain reward neurons. Specifically, right, but not left, vagal sensory ganglion activation sustained self-stimulation behavior, conditioned both flavor and place preferences, and induced dopamine release from Substantia nigra. Cell-specific transneuronal tracing revealed asymmetric ascending pathways of vagal origin throughout the CNS. In particular, transneuronal labeling identified the glutamatergic neurons of the dorsolateral parabrachial region as the obligatory relay linking the right vagal sensory ganglion to dopamine cells in Substantia nigra. Consistently, optical activation of parabrachio-nigral projections replicated the rewarding effects of right vagus excitation. Our findings establish the vagal gut-to-brain axis as an integral component of the neuronal reward pathway. They also suggest novel vagal stimulation approaches to affective disorders.


Assuntos
Intestinos/fisiologia , Recompensa , Substância Negra/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/metabolismo , Vias Aferentes/fisiologia , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Ácido Glutâmico/metabolismo , Intestinos/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Optogenética
19.
Cell ; 168(1-2): 311-324.e18, 2017 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-28086095

RESUMO

Superior predatory skills led to the evolutionary triumph of jawed vertebrates. However, the mechanisms by which the vertebrate brain controls predation remain largely unknown. Here, we reveal a critical role for the central nucleus of the amygdala in predatory hunting. Both optogenetic and chemogenetic stimulation of central amygdala of mice elicited predatory-like attacks upon both insect and artificial prey. Coordinated control of cervical and mandibular musculatures, which is necessary for accurately positioning lethal bites on prey, was mediated by a central amygdala projection to the reticular formation in the brainstem. In contrast, prey pursuit was mediated by projections to the midbrain periaqueductal gray matter. Targeted lesions to these two pathways separately disrupted biting attacks upon prey versus the initiation of prey pursuit. Our findings delineate a neural network that integrates distinct behavioral modules and suggest that central amygdala neurons instruct predatory hunting across jawed vertebrates.


Assuntos
Núcleo Central da Amígdala/fisiologia , Comportamento Predatório , Animais , Ansiedade/metabolismo , Núcleo Central da Amígdala/anatomia & histologia , Eletromiografia , Interneurônios/metabolismo , Mandíbula/anatomia & histologia , Mandíbula/inervação , Mandíbula/fisiologia , Camundongos , Pescoço/anatomia & histologia , Pescoço/inervação , Pescoço/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Substância Cinzenta Periaquedutal/fisiologia
20.
Nat Neurosci ; 19(3): 465-70, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26807950

RESUMO

Sugar exerts its potent reinforcing effects via both gustatory and post-ingestive pathways. It is, however, unknown whether sweetness and nutritional signals engage segregated brain networks to motivate ingestion. We found in mice that separate basal ganglia circuitries mediated the hedonic and nutritional actions of sugar. During sugar intake, suppressing hedonic value inhibited dopamine release in ventral, but not dorsal, striatum, whereas suppressing nutritional value inhibited dopamine release in dorsal, but not ventral, striatum. Consistently, cell-specific ablation of dopamine-excitable cells in dorsal, but not ventral, striatum inhibited sugar's ability to drive the ingestion of unpalatable solutions. Conversely, optogenetic stimulation of dopamine-excitable cells in dorsal, but not ventral, striatum substituted for sugar in its ability to drive the ingestion of unpalatable solutions. Our data indicate that sugar recruits a distributed dopamine-excitable striatal circuitry that acts to prioritize energy-seeking over taste quality.


Assuntos
Corpo Estriado/fisiologia , Glucose/farmacologia , Valor Nutritivo/fisiologia , Prazer/fisiologia , Percepção Gustatória/fisiologia , Animais , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Camundongos , Optogenética , Prazer/efeitos dos fármacos , Sacarose/análogos & derivados , Sacarose/farmacologia , Percepção Gustatória/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA