Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 202
Filtrar
1.
J Neurochem ; 158(5): 1151-1171, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34287873

RESUMO

Individual differences in the effects of a chronic neuropathic injury on social behaviours characterize both the human experience and pre-clinical animal models. The impacts of these changes to the well-being of the individual are often underappreciated. Earlier work from our laboratory using GeneChip® microarrays identified increased cholecystokinin (CCK) gene expression in the periaqueductal gray (PAG) of rats that showed persistent changes in social interactions during a Resident-Intruder encounter following sciatic nerve chronic constriction injury (CCI). In this study, we confirmed these gene regulation patterns using RT-PCR and identified the anatomical location of the CCK-mRNA as well as the translated CCK peptides in the midbrains of rats with a CCI. We found that rats with persistent CCI-induced changes in social behaviours had increased CCK-mRNA in neurons of the ventrolateral PAG and dorsal raphe nuclei, as well as increased CCK-8 peptide expression in terminal boutons located in the lateral and ventrolateral PAG. The functional significance of these changes was explored by microinjecting small volumes of CCK-8 into the PAG of uninjured rats and observing their Resident-Intruder social interactions. Disturbances to social interactions identical to those observed in CCI rats were evoked when injection sites were located in the rostral lateral and ventrolateral PAG. We suggest that CCI-induced changes in CCK expression in these PAG regions contributes to the disruptions to social behaviours experienced by a subset of individuals with neuropathic injury.


Assuntos
Colecistocinina/biossíntese , Reação de Fuga/fisiologia , Substância Cinzenta Periaquedutal/metabolismo , Neuropatia Ciática/metabolismo , Interação Social , Animais , Reação de Fuga/efeitos dos fármacos , Masculino , Microinjeções/métodos , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/patologia , Traumatismos dos Nervos Periféricos/psicologia , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/patologia , Neuropatia Ciática/psicologia , Sincalida/administração & dosagem
2.
Exp Neurol ; 342: 113724, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33915166

RESUMO

Temporal lobe epilepsy (TLE) is characterized by recurrent spontaneous seizures and behavioral comorbidities. Reduced hippocampal theta oscillations and hyperexcitability that contribute to cognitive deficits and spontaneous seizures are present beyond the sclerotic hippocampus in TLE. However, the mechanisms underlying compromised network oscillations and hyperexcitability observed in circuits remote from the sclerotic hippocampus are largely unknown. Cholecystokinin (CCK)-expressing basket cells (CCKBCs) critically participate in hippocampal theta rhythmogenesis, and regulate neuronal excitability. Thus, we examined whether CCKBCs were vulnerable in nonsclerotic regions of the ventral hippocampus remote from dorsal sclerotic hippocampus using the intrahippocampal kainate (IHK) mouse model of TLE, targeting unilateral dorsal hippocampus. We found a decrease in the number of CCK+ interneurons in ipsilateral ventral CA1 regions from epileptic mice compared to those from sham controls. We also found that the number of boutons from CCK+ interneurons was reduced in the stratum pyramidale, but not in other CA1 layers, of ipsilateral hippocampus in epileptic mice, suggesting that CCKBCs are vulnerable. Electrical recordings showed that synaptic connectivity and strength from surviving CCKBCs to CA1 pyramidal cells (PCs) were similar between epileptic mice and sham controls. In agreement with reduced CCKBC number in TLE, electrical recordings revealed a significant reduction in amplitude and frequency of IPSCs in CA1 PCs evoked by carbachol (commonly used to excite CCK+ interneurons) in ventral CA1 regions from epileptic mice versus sham controls. These findings suggest that loss of CCKBCs beyond the hippocampal lesion may contribute to hyperexcitability and compromised network oscillations in TLE.


Assuntos
Região CA1 Hipocampal/metabolismo , Colecistocinina/biossíntese , Epilepsia do Lobo Temporal/metabolismo , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Ácido Caínico/toxicidade , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Colecistocinina/genética , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/genética , Feminino , Neurônios GABAérgicos/efeitos dos fármacos , Expressão Gênica , Interneurônios/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Peptides ; 138: 170492, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33422646

RESUMO

In 1973, Gibbs, Young, and Smith showed that exogenous cholecystokinin (CCK) administration reduces food intake in rats. This initial report has led to thousands of studies investigating the physiological role of CCK in regulating feeding behavior. CCK is released from enteroendocrine I cells present along the gastrointestinal (GI) tract. CCK binding to its receptor CCK1R leads to vagal afferent activation providing post-ingestive feedback to the hindbrain. Vagal afferent neurons' (VAN) sensitivity to CCK is modulated by energy status while CCK signaling regulates gene expression of other feeding related signals and receptors expressed by VAN. In addition to its satiation effects, CCK acts all along the GI tract to optimize digestion and nutrient absorption. Diet-induced obesity (DIO) is characterized by reduced sensitivity to CCK and every part of the CCK system is negatively affected by chronic intake of energy-dense foods. EEC have recently been shown to adapt to diet, CCK1R is affected by dietary fats consumption, and the VAN phenotypic flexibility is lost in DIO. Altered endocannabinoid tone, changes in gut microbiota composition, and chronic inflammation are currently being explored as potential mechanisms for diet driven loss in CCK signaling. This review discusses our current understanding of how CCK controls food intake in conditions of leanness and how control is lost in chronic energy excess and obesity, potentially perpetuating excessive intake.


Assuntos
Colecistocinina/genética , Ingestão de Alimentos/genética , Obesidade/genética , Receptor de Colecistocinina A/genética , Animais , Colecistocinina/biossíntese , Colecistocinina/farmacologia , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/efeitos adversos , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Trato Gastrointestinal/metabolismo , Humanos , Neurônios Aferentes/metabolismo , Neurônios Aferentes/patologia , Obesidade/etiologia , Obesidade/fisiopatologia , Obesidade/terapia , Ratos
4.
J Mol Endocrinol ; 66(1): 11-22, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33151898

RESUMO

Cholecystokinin (CCK) is secreted from enteroendocrine I cells in response to fat, carbohydrate, and protein ingestion. Gene expression of nutrient-sensing molecules in I cells remains unclear, primarily due to the difficulty in distinguishing I cells from intestinal epithelial cells in vivo. In this study, we generated CCK reporter male mice in which the red fluorescence protein tdTomato (Tomato) is produced by activation of the native murine Cck promoter. Fluorescence microscopy revealed the presence of Tomato-positive cells in upper small intestine (SI), lower SI, and colon. Flow cytometer analysis revealed that Tomato-positive cells among epithelial cells of upper SI, lower SI, and colon occurred at the rate of 0.95, 0.54, and 0.06%, respectively. In upper SI and lower SI, expression levels of Cck mRNA were higher in Tomato-positive cells than those in Tomato-negative cells. The fatty acid receptors Gpr120, Gpr40, and Gpr43 and the oleoylethanolamide receptor Gpr119 were highly expressed in Tomato-positive cells isolated from SI, but were not found in Tomato-positive cells from colon. The glucose and fructose transporters Sglt1, Glut2, and Glut5 were expressed in both Tomato-positive cells and -negative cells, but these expression levels tended to be decreased in Tomato-positive cells from upper SI to colon. The peptide transporter Pept1 and receptor Gpr93 were expressed in both Tomato-positive cells and -negative cells, whereas Casr was expressed only in Tomato-positive cells isolated from SI. Thus, this transgenic mouse reveals that I cell number and gene expression in I cells vary according to region in the gastrointestinal tract.


Assuntos
Colecistocinina/biossíntese , Células Enteroendócrinas/metabolismo , Expressão Gênica , Genes Reporter , Nutrientes/metabolismo , Animais , Ácidos Graxos/metabolismo , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Receptores Acoplados a Proteínas G/metabolismo
5.
PLoS One ; 15(3): e0225917, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32142555

RESUMO

Nutritional Programming (NP) has been shown to counteract the negative effects of dietary plant protein (PP) by introducing PP at an early age towards enhancement of PP utilization during later life stages. This study explored the effect of NP and its induction time on growth, expression of appetite-stimulating hormones, and any morphological changes in the gut possibly responsible for improved dietary PP utilization. At 3 days post-hatch (dph) zebrafish were distributed into 12 (3 L) tanks, 100 larvae per tank. This study included four groups: 1) The control (NP-FM) group received fishmeal (FM)-based diet from 13-36 dph and was challenged with PP-based diet during 36-66 dph; 2) The NP-PP group received NP with dietary PP in larval stage via live food enrichment during 3-13 dph followed by FM diet during 13-36 dph and PP diet during 36-66 dph; 3) The T-NP group received NP between 13-23 dph through PP diet followed by FM diet during 23-36 dph and PP diet during 36-66 dph; and 4) The PP group received PP diet from 13-66 dph. During the PP challenge the T-NP group achieved the highest weight gain compared to control and PP. Ghrelin expression in the brain was higher in T-NP compared to NP-FM and NP-PP, while in the gut it was reduced in both NP-PP and T-NP groups. Cholecystokinin expression showed an opposite trend to ghrelin. The brain neuropeptide Y expression was lower in NP-PP compared to PP but not different with NP-FM and T-NP groups. The highest villus length to width ratio in the middle intestine was found in T-NP compared to all other groups. The study suggests that NP induced during juvenile stages improves zebrafish growth and affects digestive hormone regulation and morphology of the intestinal lining-possible mechanisms behind the improved PP utilization in pre-adult zebrafish stages.


Assuntos
Ração Animal , Encéfalo/metabolismo , Colecistocinina/biossíntese , Grelina/biossíntese , Proteínas de Vegetais Comestíveis/farmacologia , Proteínas de Peixe-Zebra/biossíntese , Peixe-Zebra/metabolismo , Animais
6.
J Diabetes Investig ; 7 Suppl 1: 44-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27186355

RESUMO

Precise control of blood glucose is dependent on adequate ß-cell mass and function. Thus, reductions in ß-cell mass and function lead to insufficient insulin production to meet demand, and result in diabetes. Recent evidence suggests that paracrine signaling in the islet might be important in obesity, and disruption of this signaling could play a role in the pathogenesis of diabetes. For example, we recently discovered a novel islet incretin axis where glucagon-like peptide-1 regulates ß-cell production of another classic gut hormone, cholecystokinin. This axis is stimulated by obesity, and plays a role in enhancing ß-cell survival. In the present review, we place our observations in the wider context of the literature on incretin regulation in the islet, and discuss the potential for therapeutic targeting of these pathways.


Assuntos
Colecistocinina/biossíntese , Colecistocinina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Ilhotas Pancreáticas/metabolismo , Obesidade/metabolismo , Comunicação Parácrina , Animais , Humanos , Incretinas/metabolismo
7.
Bull Exp Biol Med ; 160(5): 628-31, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27021097

RESUMO

Experiments were performed on cultured slices of rat ventral hippocampus. Using extracellular stimulation and patch clamp recording from pyramidal neurons in the hippocampal CA1 area, we studied characteristics of GABAergic synapse formed on these neurons by cholecystokinin-expressing interneurons. This synapse was characterized by asynchronous release of GABA and depolarization-induced suppression of inhibitory response. It was observed that administration of corticosterone increased the amplitude of evoked inhibitory postsynaptic currents in 5 minutes, but the paired ratio did not significantly change. Obtained data reflect that corticosterone can induce rapid genome-independent effects on inhibitory neurotransmission in one of hippocampal synapses.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Corticosterona/farmacologia , Potenciais Evocados/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Hipocampo/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/metabolismo , Animais , Colecistocinina/biossíntese , Estimulação Elétrica , Hipocampo/fisiologia , Interneurônios/metabolismo , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Ratos , Ratos Wistar , Sinapses/efeitos dos fármacos
8.
Mol Endocrinol ; 29(7): 978-87, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25984632

RESUMO

Cholecystokinin (CCK) is a classic gut hormone that is also expressed in the pancreatic islet, where it is highly up-regulated with obesity. Loss of CCK results in increased ß-cell apoptosis in obese mice. Similarly, islet α-cells produce increased amounts of another gut peptide, glucagon-like peptide 1 (GLP-1), in response to cytokine and nutrient stimulation. GLP-1 also protects ß-cells from apoptosis via cAMP-mediated mechanisms. Therefore, we hypothesized that the activation of islet-derived CCK and GLP-1 may be linked. We show here that both human and mouse islets secrete active GLP-1 as a function of body mass index/obesity. Furthermore, GLP-1 can rapidly stimulate ß-cell CCK production and secretion through direct targeting by the cAMP-modulated transcription factor, cAMP response element binding protein (CREB). We find that cAMP-mediated signaling is required for Cck expression, but CCK regulation by cAMP does not require stimulatory levels of glucose or insulin secretion. We also show that CREB directly targets the Cck promoter in islets from obese (Leptin(ob/ob)) mice. Finally, we demonstrate that the ability of GLP-1 to protect ß-cells from cytokine-induced apoptosis is partially dependent on CCK receptor signaling. Taken together, our work suggests that in obesity, active GLP-1 produced in the islet stimulates CCK production and secretion in a paracrine manner via cAMP and CREB. This intraislet incretin loop may be one mechanism whereby GLP-1 protects ß-cells from apoptosis.


Assuntos
Apoptose , Colecistocinina/biossíntese , Citoproteção , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Sequência de Bases , Índice de Massa Corporal , Linhagem Celular Tumoral , Colecistocinina/metabolismo , AMP Cíclico , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Citoproteção/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Obesidade/genética , Obesidade/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Ratos , Receptores da Colecistocinina/metabolismo
9.
Invest Ophthalmol Vis Sci ; 55(3): 1965-75, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24576871

RESUMO

PURPOSE: Cholecystokinin (CCK) is a neuropeptide that has been identified in trigeminal ganglion neurons. Gastrin (GAST) is a related peptide never explored in the cornea. The presence and role of both gastrointestinal peptides in the cornea and corneal sensory neurons remain to be established. We explored here in mice whether CCK, GAST, and their receptors CCK1R and CCK2R are expressed in the corneal epithelium and trigeminal ganglion neurons innervating the cornea. METHODS: We used RT-PCR analysis to detect mRNAs of CCK, GAST, CCK1R, and CCK2R in mouse cornea epithelium, trigeminal ganglia, and primary cultured corneal epithelial cells. Immunofluorescence microscopy was used to localize these peptides and their receptors in the cornea, cultured corneal epithelial cells, and corneal nerves, as well as in the cell bodies of corneal trigeminal ganglion neurons identified by retrograde labeling with Fast Blue. RESULTS: Mouse corneal epithelial cells in the cornea in situ and in cell cultures expressed CCK and GAST. Only the receptor CCK2R was found in the corneal epithelium. In addition, mouse corneal afferent sensory neurons expressed CCK and GAST, and the CCK1R receptors. CONCLUSIONS: The presence of CCK, GAST, and their receptors in the mouse corneal epithelium, and in trigeminal ganglion neurons supplying sensory innervation to the cornea, opens the possibility that these neuropeptides are involved in corneal neurogenic inflammation and in the modulation of repairing/remodeling processes following corneal injury.


Assuntos
Colecistocinina/genética , Córnea/metabolismo , Gastrinas/genética , Nervo Trigêmeo/metabolismo , Animais , Células Cultivadas , Colecistocinina/biossíntese , Córnea/inervação , Epitélio Corneano/citologia , Epitélio Corneano/metabolismo , Gastrinas/biossíntese , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA/genética , Receptor de Colecistocinina B/biossíntese , Receptor de Colecistocinina B/genética , Receptores da Colecistocinina/biossíntese , Receptores da Colecistocinina/genética
10.
Mol Psychiatry ; 19(3): 342-50, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23399917

RESUMO

Mice with a mutation in the Clock gene (ClockΔ19) have been identified as a model of mania; however, the mechanisms that underlie this phenotype, and the changes in the brain that are necessary for lithium's effectiveness on these mice remain unclear. Here, we find that cholecystokinin (Cck) is a direct transcriptional target of CLOCK and levels of Cck are reduced in the ventral tegmental area (VTA) of ClockΔ19 mice. Selective knockdown of Cck expression via RNA interference in the VTA of wild-type mice produces a manic-like phenotype. Moreover, chronic treatment with lithium restores Cck expression to near wild-type and this increase is necessary for the therapeutic actions of lithium. The decrease in Cck expression in the ClockΔ19 mice appears to be due to a lack of interaction with the histone methyltransferase, MLL1, resulting in decreased histone H3K4me3 and gene transcription, an effect reversed by lithium. Human postmortem tissue from bipolar subjects reveals a similar increase in Cck expression in the VTA with mood stabilizer treatment. These studies identify a key role for Cck in the development and treatment of mania, and describe some of the molecular mechanisms by which lithium may act as an effective antimanic agent.


Assuntos
Transtorno Bipolar/tratamento farmacológico , Transtorno Bipolar/metabolismo , Transtorno Bipolar/fisiopatologia , Proteínas CLOCK/fisiologia , Colecistocinina/fisiologia , Cloreto de Lítio/uso terapêutico , Animais , Comportamento Animal/fisiologia , Proteínas CLOCK/genética , Colecistocinina/biossíntese , Técnicas de Silenciamento de Genes , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Cloreto de Lítio/farmacologia , Masculino , Camundongos , Mutação , Proteína de Leucina Linfoide-Mieloide/metabolismo , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
11.
Artigo em Inglês | MEDLINE | ID: mdl-23587878

RESUMO

Cobia (Rachycentron canadum, Actinopterygii, Perciformes;10.5±0.1g) were fed to satiation with three plant-based protein test diets with different lysine (L) to arginine (A) ratios (LL/A, 0.8; BL/A, 1.1; and HL/A, 1.8), using a commercial diet as control for six weeks. The test diets contained 730 g kg(-1) plant ingredients with 505-529 g protein, 90.2-93.9 g lipid kg(-1) dry matter; control diet contained 550 g protein and 95 g lipid kg(-1) dry matter. Periprandial expression of brain NPY and CCK (npy and cck) was measured twice (weeks 1 and 6). At week one, npy levels were higher in pre-feeding than postfeeding cobia for all diets, except LL/A. At week six, npy levels in pre-feeding were higher than in postfeeding cobia for all diets. cck in pre-feeding cobia did not differ from that in postfeeding for all diets, at either time point. Cobia fed LL/A had lower feed intake (FI) than cobia fed BL/A and control diet, but no clear correlations between dietary L/A ratio and FI, growth and expression of npy and cck were detected. The data suggest that NPY serves as an orexigenic factor, but further studies are necessary to describe links between dietary L/A and regulation of appetite and FI in cobia.


Assuntos
Arginina/administração & dosagem , Encéfalo/fisiologia , Colecistocinina/genética , Lisina/administração & dosagem , Neuropeptídeo Y/genética , Perciformes/genética , Proteínas de Vegetais Comestíveis/administração & dosagem , Sequência de Aminoácidos , Animais , Arginina/metabolismo , Encéfalo/metabolismo , Colecistocinina/biossíntese , Colecistocinina/metabolismo , Dieta , Ingestão de Alimentos/fisiologia , Expressão Gênica , Lisina/metabolismo , Dados de Sequência Molecular , Neuropeptídeo Y/biossíntese , Neuropeptídeo Y/metabolismo , Perciformes/crescimento & desenvolvimento , Perciformes/metabolismo , Proteínas de Vegetais Comestíveis/metabolismo , Alinhamento de Sequência
12.
Acta Biochim Biophys Sin (Shanghai) ; 45(5): 345-52, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23532063

RESUMO

Serotonin (5-HT) neurons synthesize a variety of peptides. How these peptides are controlled during development remains unclear. It has been reported that the co-localization of peptides and 5-HT varies by species. In contrast to the situations in the rostral 5-HT neurons of human and rat brains, several peptides do not coexist with 5-HT in the rostral 5-HT neurons of mouse brain. In this study, we found that the peptide substance P and peptide genes, including those encoding peptides thyrotropin-releasing hormone, enkephalin, and calcitonin gene-related peptide, were expressed in the caudal 5-HT neurons of mouse brain; these findings are in line with observations in rat and monkey 5-HT neurons. We also revealed that these peptides/peptide genes partially overlapped with the transcription factor Lmx1b that specifies the 5-HT cell fate. Furthermore, we found that the peptide cholecystokinin was expressed in developing dopaminergic neurons and greatly overlapped with Lmx1b that specifies the dopaminergic cell fate. By examining the phenotype of Lmx1b deletion mice, we found that Lmx1b was required for the expression of above peptides expressed in 5-HT or dopaminergic neurons. Together, our results indicate that Lmx1b, a key transcription factor for the specification of 5-HT and dopaminergic transmitter phenotypes during embryogenesis, determines some peptide phenotypes in these neurons as well.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Proteínas com Homeodomínio LIM/fisiologia , Neurônios/metabolismo , Neurônios Serotoninérgicos/metabolismo , Fatores de Transcrição/fisiologia , Animais , Calcitonina/biossíntese , Colecistocinina/biossíntese , Encefalinas/biossíntese , Camundongos , Substância Cinzenta Periaquedutal/embriologia , Substância Cinzenta Periaquedutal/metabolismo , Fenótipo , Precursores de Proteínas/biossíntese , Núcleos da Rafe/embriologia , Núcleos da Rafe/metabolismo , Substância P/biossíntese , Hormônio Liberador de Tireotropina/biossíntese
13.
Cell Tissue Res ; 352(2): 199-206, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23370601

RESUMO

Cholecystokinin (CCK) is an early marker of both neuronal and endocrine cell lineages in the developing gastrointestinal tract. To determine the quantitative properties and the spatial distribution of the CCK-expressing myenteric neurones in early postnatal life, a transgenic mouse strain with a CCK promoter-driven red fluorescent protein (DsRedT3/CCK) was established. The cell-specific expression of DsRedT3/CCK was validated by in situ hybridization with a CCK antisense riboprobe and by in situ hybridization coupled with immunohistochemistry involving a monoclonal antibody to CCK. A gradual increase in the DsRedT3/CCK-expressing enteric neurones with clear regional differences was documented from birth until the suckling to weaning transition, in parallel with the period of rapid intestinal growth and functional maturation. To evaluate the proportion of myenteric neurones in which DsRedT3/CCK transgene expression was colocalized with the enteric neuronal marker peripherin, immunofluorescence techniques were applied. All DsRedT3/CCK neurones were peripherin-immunoreactive and the proportion of DsRedT3/CCK-expressing myenteric neurones in the duodenum was the highest after the third week of life, when the number of peripherin-immunoreactive myenteric neurones in this region had decreased. Nearly all of the DsRedT3/CCK-expressing neurones also expressed 5-hydroxytryptophan (5-HT). Thus, by utilizing a new transgenic mouse strain, we have demonstrated a small number of CCK-expressing myenteric neurones with a developmentally regulated spatiotemporal distribution. The coexistence of CCK and 5-HT in the majority of these neurones suggests their possible regulatory role in feeding at the suckling to weaning transition.


Assuntos
Colecistocinina/biossíntese , Plexo Mientérico/crescimento & desenvolvimento , Plexo Mientérico/metabolismo , 5-Hidroxitriptofano/metabolismo , Animais , Colecistocinina/genética , Colecistocinina/metabolismo , Feminino , Corantes Fluorescentes/química , Perfilação da Expressão Gênica , Imuno-Histoquímica , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/química , Neurônios/citologia , Neurônios/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Plexo Submucoso/metabolismo , Proteína Vermelha Fluorescente
14.
Tissue Cell ; 45(2): 153-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23274134

RESUMO

The characteristics and distributions of cholecystokinin (CCK)-producing cells and mucus-secreting goblet cells were investigated in the digestive tract of the blacktip grouper (Epinephelus fasciatus). CCK-producing cells were scattered throughout the digestive tract. The highest frequency of CCK-producing cells was observed in the anterior intestine portion and pyloric ceca, with a very small number of cells distributed as far as the rectum. Mucus-secreting goblet cells were found to differ remarkably in their regional distributions and relative frequencies. High frequencies of mucus-secreting goblet cells were found in the digestive tract, mainly in the anterior intestine portion and pyloric ceca, but not the esophagus; the frequency decreased slightly toward the rectum. Our result suggests that food digested by gastric acid in the stomach moves on the anterior (including the pyloric ceca) and mid intestine portion, thereby ensuring effective stimulation of the CCK-producing cells. In addition, the distribution pattern of the CCK-producing cells closely resembled that of mucus-secreting goblet cells. In E. fasciatus, CCK-producing cells and mucus-secreting goblet cells seem to be well adapted to promoting optimal control of the digestive process.


Assuntos
Colecistocinina/biossíntese , Trato Gastrointestinal/metabolismo , Células Caliciformes/metabolismo , Muco/metabolismo , Animais , Trato Gastrointestinal/citologia , Trato Gastrointestinal/ultraestrutura , Células Caliciformes/citologia , Perciformes/metabolismo
15.
Neurogastroenterol Motil ; 25(1): e70-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23095091

RESUMO

BACKGROUND: Roux-en-Y gastric bypass (RYGB) surgery is very effective in reducing excess body weight and improving glucose homeostasis in obese subjects. Changes in the pattern of gut hormone secretion are thought to play a major role, but the mechanisms leading to both changed hormone secretion and beneficial effects remain unclear. Specifically, it is not clear whether changes in the number of hormone-secreting enteroendocrine cells, or changes in the releasing stimuli, or both, are important. METHODS: We estimated numbers of enteroendocrine cells after immunohistochemical staining in fixed tissue samples from rats at 10-11 months after RYGB. KEY RESULTS: Numbers of glucagon-like peptide-1 (GLP-1) (L-cells, co-expressing peptide YY (PYY)), cholecystokinin (CCK), neurotensin, and 5-HT-immunoreactive cells were significantly increased in the Roux and common limbs, but not the biliopancreatic limb in RYGB rats compared with sham-operated, obese rats fed high-fat diet, and chow-fed controls. This increase was mostly accounted for by general hyperplasia of all intestinal wall layers of the nutrient-perfused Roux and common limbs, and less to increased density of expression. The number of ghrelin cells in the bypassed stomach was not different among the three groups. CONCLUSIONS & INFERENCES: The findings suggest that the number of enteroendocrine cells increases passively as the gut adapts, and that the increased total number of L- and I-cells is likely to contribute to the higher circulating levels of GLP-1, PYY, and CCK, potentially leading to suppression of food intake and stimulation of insulin secretion. Whether changes in releasing stimuli also contribute to altered circulating levels will have to be determined in future studies.


Assuntos
Colecistocinina/biossíntese , Células Enteroendócrinas/citologia , Derivação Gástrica , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Neurotensina/biossíntese , Serotonina/biossíntese , Animais , Células Enteroendócrinas/metabolismo , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley
16.
Endocrinology ; 153(12): 5782-95, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23064014

RESUMO

Enteroendocrine cells such as duodenal cholecystokinin (CCK cells) are generally thought to be confined to certain segments of the gastrointestinal (GI) tract and to store and release peptides derived from only a single peptide precursor. In the current study, however, transgenic mice expressing enhanced green fluorescent protein (eGFP) under the control of the CCK promoter demonstrated a distribution pattern of CCK-eGFP positive cells that extended throughout the intestine. Quantitative PCR and liquid chromatography-mass spectrometry proteomic analyses of isolated, FACS-purified CCK-eGFP-positive cells demonstrated expression of not only CCK but also glucagon-like peptide 1 (GLP-1), gastric inhibitory peptide (GIP), peptide YY (PYY), neurotensin, and secretin, but not somatostatin. Immunohistochemistry confirmed this expression pattern. The broad coexpression phenomenon was observed both in crypts and villi as demonstrated by immunohistochemistry and FACS analysis of separated cell populations. Single-cell quantitative PCR indicated that approximately half of the duodenal CCK-eGFP cells express one peptide precursor in addition to CCK, whereas an additional smaller fraction expresses two peptide precursors in addition to CCK. The coexpression pattern was further confirmed through a cell ablation study based on expression of the human diphtheria toxin receptor under the control of the proglucagon promoter, in which activation of the receptor resulted in a marked reduction not only in GLP-1 cells, but also PYY, neurotensin, GIP, CCK, and secretin cells, whereas somatostatin cells were spared. Key elements of the coexpression pattern were confirmed by immunohistochemical double staining in human small intestine. It is concluded that a lineage of mature enteroendocrine cells have the ability to coexpress members of a group of functionally related peptides: CCK, secretin, GIP, GLP-1, PYY, and neurotensin, suggesting a potential therapeutic target for the treatment and prevention of diabetes and obesity.


Assuntos
Colecistocinina/biossíntese , Células Enteroendócrinas/citologia , Polipeptídeo Inibidor Gástrico/biossíntese , Regulação da Expressão Gênica , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Neurotensina/biossíntese , Peptídeo YY/metabolismo , Animais , Linhagem da Célula , Separação Celular , Diabetes Mellitus/prevenção & controle , Células Enteroendócrinas/metabolismo , Citometria de Fluxo , Grelina/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica/métodos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Transgênicos , Obesidade/prevenção & controle , Regiões Promotoras Genéticas
17.
Biosci Biotechnol Biochem ; 76(6): 1104-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22790930

RESUMO

We have recently reported that oral gavage of a potato extract (Potein®) suppressed the food intake in rats. The satiating effect of the potato extract was compared in the present study to other protein sources, and the involvement of endogenous cholecystokinin (CCK) secretion was examined. Food consumption was measured in 18-h fasted rats after oral gavage of the potato extract or other protein sources. The CCK-releasing activity of the potato extract was then examined in anesthetized rats with a portal cannula. Oral gavage of the potato extract reduced the food intake in the rats, the effect being greater than with casein and a soybean ß-conglycinin hydrolysate. The suppressive effect on appetite of the potato extract was attenuated by treating with a CCK-receptor antagonist (devazepide). The portal CCK concentration was increased after a duodenal administration of the potato extract to anesthetized rats. These results indicate that the potato extract suppressed the food intake in rats through CCK secretion.


Assuntos
Apetite/efeitos dos fármacos , Colecistocinina/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Extratos Vegetais/administração & dosagem , Solanum tuberosum/química , Administração Oral , Animais , Antígenos de Plantas/farmacologia , Apetite/fisiologia , Caseínas/farmacologia , Colecistocinina/biossíntese , Devazepida/farmacologia , Duodeno/efeitos dos fármacos , Duodeno/fisiologia , Ingestão de Alimentos/fisiologia , Jejum , Globulinas/farmacologia , Antagonistas de Hormônios/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores da Colecistocinina/antagonistas & inibidores , Receptores da Colecistocinina/metabolismo , Saciação/efeitos dos fármacos , Saciação/fisiologia , Proteínas de Armazenamento de Sementes/farmacologia , Proteínas de Soja/farmacologia
18.
J Comp Neurol ; 520(16): 3553-73, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22678985

RESUMO

Gene markers are useful tools to identify cell types for fine mapping of neuronal circuits. Here we report area-specific sublamina structure of the rat cerebral cortex using cholecystokinin (cck) and purkinje cell protein4 (pcp4) mRNAs as the markers for excitatory neuron subtypes in layers 5 and 6. We found a segregated expression, especially pronounced in layer 6, where corticothalamic and corticocortical projecting neurons reside. To examine the relationship between gene expression and projection target, we injected retrograde tracers into several thalamic subnuclei, ventral posterior (VP), posterior (PO), mediodorsal (MD), medial and lateral geniculate nuclei (MGN and LGN); as well as into two cortical areas (M1 and V1). This combination of tracer-in situ hybridization (ISH) experiments revealed that corticocortical neurons predominantly express cck and corticothalamic neurons predominantly express pcp4 mRNAs in all areas tested. In general, cck(+) and pcp4(+) cells occupied the upper and lower compartment of layer 6a, respectively. However, the sublaminar distribution and the relative abundance of cck(+) and pcp4(+) cells were quite distinctive across areas. For example, layer 6 of the prelimbic cortex was almost devoid of cck(+) neurons, and was occupied instead by corticothalamic pcp4(+) neurons. In the lateral areas, such as S2, there was an additional layer of cck(+) cells positioned below the pcp4(+) compartment. The claustrum, which has a tight relationship with the cortex, mostly consisted of cck(+)/pcp4(-) cells. In summary, the combination of gene markers and retrograde tracers revealed a distinct sublaminar organization, with conspicuous cross-area variation in the arrangement and relative density of corticothalamic connections.


Assuntos
Córtex Cerebral/citologia , Vias Neurais/citologia , Neurônios/citologia , Animais , Biomarcadores/análise , Proteínas de Ligação a Calmodulina/análise , Proteínas de Ligação a Calmodulina/biossíntese , Córtex Cerebral/metabolismo , Colecistocinina/análise , Colecistocinina/biossíntese , Hibridização In Situ , Masculino , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/biossíntese , Vias Neurais/metabolismo , Neurônios/metabolismo , Ratos , Ratos Wistar
19.
J Biomed Biotechnol ; 2012: 484869, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22619495

RESUMO

The aim of this paper was to investigate the effect of heat stress on the regulation of appetite-associated genes in laying hens. Forty eight laying hens were randomly divided into two circumstances: high (31 ± 1.5°C; relative humidity, 82.0 ± 2.2%) or normal (20 ± 2°C, control; relative humidity, 60.1 ± 4.5%) ambient environment. Heat stress decreased body weight gain (P < 0.01), feed intake (P < 0.01), laying rate (P < 0.05), average egg mass (P < 0.01), egg production (P < 0.01), shell thickness (P < 0.01), and feed efficiency (P < 0.05). High ambient temperature decreased plasma uric acid (P < 0.05). Heat stress significantly increased mRNA levels of ghrelin and cocaine- and amphetamine-regulated transcript (P < 0.05) and decreased mRNA levels of cholecystokinin (P < 0.05) in the hypothalamus. Heat stress significantly increased (P < 0.05) mRNA levels of ghrelin in the glandular stomach and jejunum but significantly decreased (P < 0.05) mRNA levels of cholecystokinin in the duodenum and jejunum. In conclusion, heat stress plays a unique role in some special neuropeptides (e.g., ghrelin, cocaine- and amphetamine-regulated transcript, and cholecystokinin), which might participate in the regulation of feed intake in laying hens under high ambient temperature.


Assuntos
Galinhas/fisiologia , Ingestão de Alimentos/fisiologia , Regulação da Expressão Gênica/fisiologia , Resposta ao Choque Térmico/fisiologia , Hormônios Peptídicos/genética , Animais , Regulação do Apetite/fisiologia , Peso Corporal/fisiologia , Colecistocinina/biossíntese , Colecistocinina/sangue , Colecistocinina/genética , Colecistocinina/metabolismo , Tamanho da Ninhada , Casca de Ovo/fisiologia , Feminino , Perfilação da Expressão Gênica , Hipotálamo/metabolismo , Neuropeptídeos/biossíntese , Neuropeptídeos/sangue , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Especificidade de Órgãos , Oviposição , Hormônios Peptídicos/biossíntese , Hormônios Peptídicos/sangue , Hormônios Peptídicos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
20.
Horm Behav ; 62(1): 18-26, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22561338

RESUMO

Feeding behavior and reproduction are coordinately regulated by the brain via neurotransmitters, circulating hormones, and neuropeptides. Reduced feeding allows animals to engage in other behaviors important for fitness, including mating and parental care. Some fishes cease feeding for weeks at a time in order to provide care to their young by brooding them inside the male or female parent's mouth. Maternal mouthbrooding is known to impact circulating hormones and subsequent reproductive cycles, but neither the full effects of food deprivation nor the neural mechanisms are known. Here we ask what effects mouthbrooding has on several physiological processes including gonad and body mass, brain neuropeptide and receptor gene expression, and circulating steroid hormones in a mouthbrooding cichlid species, Astatotilapia burtoni. We ask whether any observed changes can be explained by food deprivation, and show that during mouthbrooding, ovary size and circulating levels of androgens and estrogens match those seen during food deprivation. Levels of gonadotropin-releasing hormone 1 (GnRH1) mRNA in the brain were low in food-deprived females compared to controls and in mouthbrooding females compared to gravid females. Levels of mRNA encoding two peptides involved in regulating feeding, hypocretin and cholecystokinin, were increased in the brains of food-deprived females. Brain mRNA levels of two receptors, GnRH receptor 2 and NPY receptor Y8c, were elevated in mouthbrooding females compared to the fed condition, but NPY receptor Y8b mRNA was differently regulated by mouthbrooding. These results suggest that many, but not all, of the characteristic physiological changes that occur during mouthbrooding are consequences of food deprivation.


Assuntos
Androgênios/sangue , Ciclídeos/metabolismo , Estrogênios/sangue , Privação de Alimentos/fisiologia , Neuropeptídeos/metabolismo , Ovário/metabolismo , Animais , Peso Corporal , Encéfalo/metabolismo , Colecistocinina/biossíntese , Feminino , Hormônio Liberador de Gonadotropina/biossíntese , Hormônio Liberador de Gonadotropina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/biossíntese , Orexinas , Tamanho do Órgão , Ovário/anatomia & histologia , Precursores de Proteínas/biossíntese , RNA Mensageiro/biossíntese , Receptores LHRH/biossíntese , Receptores de Neuropeptídeo Y/biossíntese , Reprodução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA