Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.621
Filter
1.
Elife ; 132024 May 03.
Article in English | MEDLINE | ID: mdl-38700136

ABSTRACT

Cholecystokinin (CCK) is an essential modulator for neuroplasticity in sensory and emotional domains. Here, we investigated the role of CCK in motor learning using a single pellet reaching task in mice. Mice with a knockout of Cck gene (Cck-/-) or blockade of CCK-B receptor (CCKBR) showed defective motor learning ability; the success rate of retrieving reward remained at the baseline level compared to the wildtype mice with significantly increased success rate. We observed no long-term potentiation upon high-frequency stimulation in the motor cortex of Cck-/- mice, indicating a possible association between motor learning deficiency and neuroplasticity in the motor cortex. In vivo calcium imaging demonstrated that the deficiency of CCK signaling disrupted the refinement of population neuronal activity in the motor cortex during motor skill training. Anatomical tracing revealed direct projections from CCK-expressing neurons in the rhinal cortex to the motor cortex. Inactivation of the CCK neurons in the rhinal cortex that project to the motor cortex bilaterally using chemogenetic methods significantly suppressed motor learning, and intraperitoneal application of CCK4, a tetrapeptide CCK agonist, rescued the motor learning deficits of Cck-/- mice. In summary, our results suggest that CCK, which could be provided from the rhinal cortex, may surpport motor skill learning by modulating neuroplasticity in the motor cortex.


Subject(s)
Cholecystokinin , Learning , Mice, Knockout , Motor Cortex , Motor Skills , Neuronal Plasticity , Animals , Male , Mice , Cholecystokinin/metabolism , Learning/physiology , Motor Cortex/physiology , Motor Cortex/metabolism , Motor Cortex/drug effects , Motor Skills/physiology , Neuronal Plasticity/physiology , Neuronal Plasticity/drug effects
2.
Alzheimers Res Ther ; 16(1): 109, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750512

ABSTRACT

BACKGROUND: As one major symptom of Alzheimer's disease (AD), anterograde amnesia describes patients with an inability in new memory formation. The crucial role of the entorhinal cortex in forming new memories has been well established, and the neuropeptide cholecystokinin (CCK) is reported to be released from the entorhinal cortex to enable neocortical associated memory and long-term potentiation. Though several studies reveal that the entorhinal cortex and CCK are related to AD, it is less well studied. It is unclear whether CCK is a good biomarker or further a great drug candidate for AD. METHODS: mRNA expressions of CCK and CCK-B receptor (CCKBR) were examined in two mouse models, 3xTg AD and CCK knock-out (CCK-/-) mice. Animals' cognition was investigated with Morris water maze, novel object recognition test and neuroplasticity with in-vitro electrophysiological recording. Drugs were given intraperitoneally to animals to investigate the rescue effects on cognitive deficits, or applied to brain slices directly to explore the influence in inducement of long-term potentiation. RESULTS: Aged 3xTg AD mice exhibited reduced CCK mRNA expression in the entorhinal cortex but reduced CCKBR expression in the neocortex and hippocampus, and impaired cognition and neuroplasticity comparable with CCK-/- mice. Importantly, the animals displayed improved performance and enhanced long-term potentiation after the treatment of CCKBR agonists. CONCLUSIONS: Here we provide more evidence to support the role of CCK in learning and memory and its potential to treat AD. We elaborated on the rescue effect of a promising novel drug, HT-267, on aged 3xTg AD mice. Although the physiological etiology of CCK in AD still needs to be further investigated, this study sheds light on a potential pharmaceutical candidate for AD and dementia.


Subject(s)
Alzheimer Disease , Amnesia, Anterograde , Cholecystokinin , Disease Models, Animal , Mice, Transgenic , Receptor, Cholecystokinin B , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Mice , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/deficiency , Amnesia, Anterograde/drug therapy , Cholecystokinin/metabolism , Entorhinal Cortex/drug effects , Entorhinal Cortex/metabolism , Male , Mice, Knockout , Mice, Inbred C57BL , Long-Term Potentiation/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Aging/drug effects
3.
Reprod Domest Anim ; 59(5): e14586, 2024 May.
Article in English | MEDLINE | ID: mdl-38757644

ABSTRACT

The current study aimed to explore the molecular mechanism by which the cholecystokinin (CCK)-mediated CCKAR and CCKBR, as well as the molecular mechanisms of CCK-mediated insulin signalling pathway, regulate oestrogen in the granulosa cells. Also, the expression of CCK in ovaries, uterus, hypothalamus and pituitary gland was investigated in Camelus bactrianus. Ovaries, uterus, hypothalamus and pituitary gland were collected from six, three before ovulation (control) and three after ovulation, slaughtered Camelus bactrianus. Ovulation was induced by IM injection of seminal plasma before slaughtering in the ovulated group. The results showed that there were differences in the transcription and protein levels of CCK in various tissues before and after ovulation (p < .05, p < .01). After transfection with p-IRES2-EGFP-CCK, the mRNA and protein levels of CCK, CCKAR, CCKBR and ER in follicular granulosa cells were significantly upregulated (p < .05, p < .01), and the content of E2 was significantly upregulated (p < .01); On the contrary, after transfection with si-CCK, the mRNA and protein levels of CCK, CCKAR, CCKBR and ER in follicular granulosa cells were significantly downregulated (p < .05, p < .01), and the content of E2 was significantly downregulated (p < .01). Regulating CCK can affect the mRNA levels of INS, INSR, IGF and IGF-R. In summary, regulating the expression level of CCK can activate insulin-related signalling pathways by CCKR, thereby regulating the steroidogenic activity of granulosa cells.


Subject(s)
Cholecystokinin , Granulosa Cells , Insulin , Signal Transduction , Animals , Female , Granulosa Cells/metabolism , Cholecystokinin/metabolism , Cholecystokinin/genetics , Insulin/metabolism , Ovulation , Uterus/metabolism , Ovary/metabolism , Pituitary Gland/metabolism , Hypothalamus/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics
4.
Food Chem ; 452: 139466, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38735106

ABSTRACT

γ-Glutamylation of beef protein hydrolysate (BPH) by L-glutaminase was carried out to improve the taste, as well as enhance the stimulating effect of gastrointestinal hormone (CCK and GLP-1) secretion and the anti-inflammatory property. Results of sensory evaluation showed that the kokumi taste, umaminess, saltiness of the γ-glutamylated product (γ-GBPH) were significantly higher (p < 0.05), whilst the bitterness was remarkably decreased (p < 0.05) than that of BPH. γ-GBPH had a better promoting effect (p < 0.05) on CCK and GLP-1 secretion and a higher inhibition (p < 0.05) on TNF-α and IL-8 production than BPH in vitro cell experiments. In γ-GBPH, 15 γ-Glutamylated amino acids (γ-[Glu](n =1/2)-AAs) and 10 γ-Glutamyl-tripeptide (γ-Glu-AA-AAs) were synthesized from the bitter amino acids and bitter peptides, respectively, and their total production yield was 140.01-170.46 mg/g and 149.06 mg/g, respectively. The synthesized γ-Glu-AA-AAs entered the binding pocket of the calcium-sensitive receptor (CaSR), and they all interacted with three reported amino acid residues (Ser147, Ala168, and Ser170) of CaSR.


Subject(s)
Anti-Inflammatory Agents , Glucagon-Like Peptide 1 , Protein Hydrolysates , Taste , Protein Hydrolysates/chemistry , Protein Hydrolysates/metabolism , Protein Hydrolysates/pharmacology , Animals , Humans , Cattle , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/chemistry , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Cholecystokinin/metabolism , Cholecystokinin/chemistry
5.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38697841

ABSTRACT

Interneurons in the medial prefrontal cortex (PFC) regulate local neural activity to influence cognitive, motivated, and emotional behaviors. Parvalbumin-expressing (PV+) interneurons are the primary mediators of thalamus-evoked feed-forward inhibition across the mouse cortex, including the anterior cingulate cortex, where they are engaged by inputs from the mediodorsal (MD) thalamus. In contrast, in the adjacent prelimbic (PL) cortex, we find that PV+ interneurons are scarce in the principal thalamorecipient layer 3 (L3), suggesting distinct mechanisms of inhibition. To identify the interneurons that mediate MD-evoked inhibition in PL, we combine slice physiology, optogenetics, and intersectional genetic tools in mice of both sexes. We find interneurons expressing cholecystokinin (CCK+) are abundant in L3 of PL, with cells exhibiting fast-spiking (fs) or non-fast-spiking (nfs) properties. MD inputs make stronger connections onto fs-CCK+ interneurons, driving them to fire more readily than nearby L3 pyramidal cells and other interneurons. CCK+ interneurons in turn make inhibitory, perisomatic connections onto L3 pyramidal cells, where they exhibit cannabinoid 1 receptor (CB1R) mediated modulation. Moreover, MD-evoked feed-forward inhibition, but not direct excitation, is also sensitive to CB1R modulation. Our findings indicate that CCK+ interneurons contribute to MD-evoked inhibition in PL, revealing a mechanism by which cannabinoids can modulate MD-PFC communication.


Subject(s)
Cholecystokinin , Interneurons , Neural Inhibition , Prefrontal Cortex , Animals , Interneurons/physiology , Cholecystokinin/metabolism , Prefrontal Cortex/physiology , Mice , Male , Female , Neural Inhibition/physiology , Thalamus/physiology , Mice, Inbred C57BL , Parvalbumins/metabolism , Mice, Transgenic , Neural Pathways/physiology , Optogenetics
6.
Article in English | MEDLINE | ID: mdl-38703991

ABSTRACT

The pond loach (Misgurnus anguillicaudatus) is an important aquaculture freshwater species, used as an ornamental fish, food source for humans and angling bait. Pond loaches are resistant to fasting and extreme environmental conditions, including temperature and low oxygen levels. Little is known about how these factors affect the feeding physiology and the endocrine regulation of feeding of loaches. In this study, we examined the effects of fasting, as well as increased temperature and decreased oxygen levels on food intake and transcript levels of appetite regulators. Fasted fish had lower blood glucose levels, and lower expression levels of intestine CCK and PYY, and brain CART1, but had higher levels of brain orexin and ghrelin than fed fish. Fish held at 30 °C had higher food intake, glucose levels, and mRNA levels of intestine CCK and PYY, and brain CART2, but lower brain orexin levels than fish at 20 °C. Fish held at low oxygen levels had a lower food intake, higher intestine CCKa and ghrelin, and brain orexin, CART2 and ghrelin mRNA expression levels than fish held at high O2 levels. Our results suggest that fasting and high temperatures increase the expression of orexigenic and anorexigenic factors respectively, whereas the increase in expression of both orexigenic and anorexigenic factors in low O2 environments might not be related to their role in feeding, but possibly to protection from tissue damage. The results of our study might shed new light on how pond loaches are able to cope with extreme environmental conditions such as low food availability, extreme temperatures and hypoxia.


Subject(s)
Cypriniformes , Fasting , Ghrelin , Animals , Fasting/physiology , Cypriniformes/physiology , Cypriniformes/genetics , Cypriniformes/metabolism , Ghrelin/metabolism , Orexins/metabolism , Brain/metabolism , Brain/physiology , Cholecystokinin/metabolism , Appetite Regulation/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Blood Glucose/metabolism , Oxygen/metabolism , Peptide YY/metabolism , Peptide YY/blood , Eating/physiology , Temperature , Feeding Behavior/physiology
7.
Sci Rep ; 14(1): 6277, 2024 03 15.
Article in English | MEDLINE | ID: mdl-38491056

ABSTRACT

The cholecystokinin (CCK)/gastrin family peptides are involved in regulation of feeding and digestion in vertebrates. In the ascidian Ciona intestinalis type A (Ciona robusta), cionin, a CCK/gastrin family peptide, has been identified. Cionin is expressed exclusively in the central nervous system (CNS). In contrast, cionin receptor expression has been detected in the CNS, digestive tract, and ovary. Although cionin has been reported to be involved in ovulation, its physiological function in the CNS remains to be investigated. To elucidate its neural function, in the present study, we analyzed the expression of cionin and cionin receptors in the CNS. Cionin was expressed mainly in neurons residing in the anterior region of the cerebral ganglion. In contrast, the gene expressin of the cionin receptor gene CioR1, was detected in the middle part of the cerebral ganglion and showed a similar expression pattern to that of VACHT, a cholinergic neuron marker gene. Moreover, CioR1 was found to be expressed in cholinergic neurons. Consequently, these results suggest that cionin interacts with cholinergic neurons as a neurotransmitter or neuromodulator via CioR1. This study provides insights into a biological role of a CCK/gastrin family peptide in the CNS of ascidians.


Subject(s)
Cholecystokinin , Ciona intestinalis , Neuropeptides , Animals , Female , Cholecystokinin/genetics , Cholecystokinin/metabolism , Gastrins , Ciona intestinalis/genetics , Ciona intestinalis/metabolism , Amino Acid Sequence , Central Nervous System
8.
J Neurosci ; 44(17)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38438258

ABSTRACT

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


Subject(s)
Acetylcholine , Cholecystokinin , Mice, Inbred C57BL , Theta Rhythm , Theta Rhythm/drug effects , Theta Rhythm/physiology , Animals , Male , Mice , Female , Acetylcholine/pharmacology , Acetylcholine/metabolism , Cholecystokinin/pharmacology , Cholecystokinin/metabolism , Interneurons/physiology , Interneurons/drug effects , Somatostatin/metabolism , Somatostatin/pharmacology , Amygdala/physiology , Amygdala/drug effects , Basolateral Nuclear Complex/physiology , Basolateral Nuclear Complex/drug effects , Nerve Net/physiology , Nerve Net/drug effects , Receptor, Muscarinic M3/physiology , Receptor, Muscarinic M3/metabolism , Parvalbumins/metabolism
9.
Curr Opin Endocrinol Diabetes Obes ; 31(3): 115-121, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38511400

ABSTRACT

PURPOSE OF REVIEW: Various gut hormones interact with the brain through delicate communication, thereby influencing appetite and subsequent changes in body weight. This review summarizes the effects of gut hormones on appetite, with a focus on recent research. RECENT FINDINGS: Ghrelin is known as an orexigenic hormone, whereas glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), postprandial peptide YY (PYY), and oxyntomodulin (OXM) are known as anorexigenic hormones. Recent human studies have revealed that gut hormones act differently in various systems, including adipose tissue, beyond appetite and energy intake, and even involve in high-order thinking. Environmental factors including meal schedule, food contents and quality, type of exercise, and sleep deprivation also play a role in the influence of gut hormone on appetite, weight change, and obesity. Recently published studies have shown that retatrutide, a triple-agonist of GLP-1, GIP, and glucagon receptor, and orforglipron, a GLP-1 receptor partial agonist, are effective in weight loss and improving various metabolic parameters associated with obesity. SUMMARY: Various gut hormones influence appetite, and several drugs targeting these receptors have been reported to exert positive effects on weight loss in humans. Given that diverse dietary and environmental factors affect the actions of gut hormones and appetite, there is a need for integrated and largescale long-term studies in this field.


Subject(s)
Appetite Regulation , Gastrointestinal Hormones , Obesity , Humans , Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/physiology , Appetite Regulation/physiology , Obesity/metabolism , Obesity/physiopathology , Cholecystokinin/physiology , Cholecystokinin/metabolism , Gastric Inhibitory Polypeptide/physiology , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/physiology , Peptide YY/metabolism , Peptide YY/physiology , Oxyntomodulin , Animals , Ghrelin/physiology , Ghrelin/metabolism , Appetite/physiology , Appetite/drug effects
10.
Elife ; 132024 Mar 04.
Article in English | MEDLINE | ID: mdl-38436304

ABSTRACT

The entorhinal cortex is involved in establishing enduring visuo-auditory associative memory in the neocortex. Here we explored the mechanisms underlying this synaptic plasticity related to projections from the visual and entorhinal cortices to the auditory cortex in mice using optogenetics of dual pathways. High-frequency laser stimulation (HFS laser) of the visuo-auditory projection did not induce long-term potentiation. However, after pairing with sound stimulus, the visuo-auditory inputs were potentiated following either infusion of cholecystokinin (CCK) or HFS laser of the entorhino-auditory CCK-expressing projection. Combining retrograde tracing and RNAscope in situ hybridization, we show that Cck expression is higher in entorhinal cortex neurons projecting to the auditory cortex than in those originating from the visual cortex. In the presence of CCK, potentiation in the neocortex occurred when the presynaptic input arrived 200 ms before postsynaptic firing, even after just five trials of pairing. Behaviorally, inactivation of the CCK+ projection from the entorhinal cortex to the auditory cortex blocked the formation of visuo-auditory associative memory. Our results indicate that neocortical visuo-auditory association is formed through heterosynaptic plasticity, which depends on release of CCK in the neocortex mostly from entorhinal afferents.


Subject(s)
Cholecystokinin , Entorhinal Cortex , Mice , Animals , Entorhinal Cortex/physiology , Cholecystokinin/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Neurons/metabolism
11.
J Physiol ; 602(6): 1065-1083, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38389307

ABSTRACT

Type 1 diabetes is a disease of the endocrine pancreas; however, it also affects exocrine function. Although most studies have examined the effects of diabetes on acinar cells, much less is known regarding ductal cells, despite their important protective function in the pancreas. Therefore, we investigated the effect of diabetes on ductal function. Diabetes was induced in wild-type and cystic fibrosis transmembrane conductance regulator (CFTR) knockout mice following an i.p. administration of streptozotocin. Pancreatic ductal fluid and HCO3 - secretion were determined using fluid secretion measurements and fluorescence microscopy, respectively. The expression of ion transporters was measured by real-time PCR and immunohistochemistry. Transmission electron microscopy was used for the morphological characterization of the pancreas. Serum secretin and cholecystokinin levels were measured by an enzyme-linked immunosorbent assay. Ductal fluid and HCO3 - secretion, CFTR activity, and the expression of CFTR, Na+ /H+ exchanger-1, anoctamine-1 and aquaporin-1 were significantly elevated in diabetic mice. Acute or chronic glucose treatment did not affect HCO3 - secretion, but increased alkalizing transporter activity. Inhibition of CFTR significantly reduced HCO3 - secretion in both normal and diabetic mice. Serum levels of secretin and cholecystokinin were unchanged, but the expression of secretin receptors significantly increased in diabetic mice. Diabetes increases fluid and HCO3 - secretion in pancreatic ductal cells, which is associated with the increased function of ion and water transporters, particularly CFTR. KEY POINTS: There is a lively interaction between the exocrine and endocrine pancreas not only under physiological conditions, but also under pathophysiological conditions The most common disease affecting the endocrine part is type-1 diabetes mellitus (T1DM), which is often associated with pancreatic exocrine insufficiency Compared with acinar cells, there is considerably less information regarding the effect of diabetes on pancreatic ductal epithelial cells, despite the fact that the large amount of fluid and HCO3 - produced by ductal cells is essential for maintaining normal pancreatic functions Ductal fluid and HCO3 - secretion increase in T1DM, in which increased cystic fibrosis transmembrane conductance regulator activation plays a central role. We have identified a novel interaction between T1DM and ductal cells. Presumably, the increased ductal secretion represents a defence mechanism in the prevention of diabetes, but further studies are needed to clarify this issue.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Animals , Mice , Bicarbonates/metabolism , Cholecystokinin/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Pancreatic Ducts/metabolism , Secretin/metabolism
12.
J Physiol Sci ; 74(1): 11, 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-38368346

ABSTRACT

Many hormones act on the hypothalamus to control hunger and satiety through various pathways closely associated with several factors. When food is present in the gastro intestinal (GI) tract, enteroendocrine cells (EECs) emit satiety signals such as cholecystokinin (CCK), glucagon like peptide-1 (GLP-1) and peptide YY (PYY), which can then communicate with the vagus nerve to control food intake. More specifically, satiety has been shown to be particularly affected by the GLP-1 hormone and its receptor agonists that have lately been acknowledged as a promising way to reduce weight. In addition, there is increasing evidence that normal flora is also involved in the peripheral, central, and reward system that impact satiety. Moreover, neurologic pathways control satiety through neurotransmitters. In this review, we discuss the different roles of each of the GLP-1 hormone and its agonist, gut microbiomes, as well as neurotransmitters and their interconnected relation in the regulation of body's satiety homeostasis.


Subject(s)
Cholecystokinin , Glucagon-Like Peptide 1 , Cholecystokinin/metabolism , Peptide YY/metabolism , Brain/metabolism , Neurotransmitter Agents
13.
Front Neuroendocrinol ; 73: 101122, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38346453

ABSTRACT

Cholecystokinin (CCK) is a neuropeptide modulating digestion, glucose levels, neurotransmitters and memory. Recent studies suggest that CCK exhibits neuroprotective effects in Alzheimer's disease (AD) and Parkinson's disease (PD). Thus, we review the physiological function and therapeutic potential of CCK. The neuropeptide facilitates hippocampal glutamate release and gates GABAergic basket cell activity, which improves declarative memory acquisition, but inhibits consolidation. Cortical CCK alters recognition memory and enhances audio-visual processing. By stimulating CCK-1 receptors (CCK-1Rs), sulphated CCK-8 elicits dopamine release in the substantia nigra and striatum. In the mesolimbic pathway, CCK release is triggered by dopamine and terminates reward responses via CCK-2Rs. Importantly, activation of hippocampal and nigral CCK-2Rs is neuroprotective by evoking AMPK activation, expression of mitochondrial fusion modulators and autophagy. Other benefits include vagus nerve/CCK-1R-mediated expression of brain-derived neurotrophic factor, intestinal protection and suppression of inflammation. We also discuss caveats and the therapeutic combination of CCK with other peptide hormones.


Subject(s)
Alzheimer Disease , Cholecystokinin , Parkinson Disease , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Cholecystokinin/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Animals , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
14.
Am J Physiol Gastrointest Liver Physiol ; 326(3): G291-G309, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38252699

ABSTRACT

Hepatocellular carcinoma (HCC) is the fastest-growing cause of cancer-related deaths worldwide. Chronic inflammation and fibrosis are the greatest risk factors for the development of HCC. Although the cell of origin for HCC is uncertain, many theories believe this cancer may arise from liver progenitor cells or stem cells. Here, we describe the activation of hepatic stem cells that overexpress the cholecystokinin-B receptor (CCK-BR) after liver injury with either a DDC diet (0.1% 3, 5-diethoxy-carbonyl 1,4-dihydrocollidine) or a NASH-inducing CDE diet (choline-deficient ethionine) in murine models. Pharmacologic blockade of the CCK-BR with a receptor antagonist proglumide or knockout of the CCK-BR in genetically engineered mice during the injury diet reduces the expression of hepatic stem cells and prevents the formation of three-dimensional tumorspheres in culture. RNA sequencing of livers from DDC-fed mice treated with proglumide or DDC-fed CCK-BR knockout mice showed downregulation of differentially expressed genes involved in cell proliferation and oncogenesis and upregulation of tumor suppressor genes compared with controls. Inhibition of the CCK-BR decreases hepatic transaminases, fibrosis, cytokine expression, and alters the hepatic immune cell signature rendering the liver microenvironment less oncogenic. Furthermore, proglumide hastened recovery after liver injury by reversing fibrosis and improving markers of synthetic function. Proglumide is an older drug that is orally bioavailable and being repurposed for liver conditions. These findings support a promising therapeutic intervention applicable to patients to prevent the development of HCC and decrease hepatic fibrosis.NEW & NOTEWORTHY This investigation identified a novel pathway involving the activation of hepatic stem cells and liver oncogenesis. Receptor blockade or genetic disruption of the cholecystokinin-B receptor (CCK-BR) signaling pathway decreased the activation and proliferation of hepatic stem cells after liver injury without eliminating the regenerative capacity of healthy hepatocytes.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Mice , Animals , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Carcinoma, Hepatocellular/pathology , Proglumide/pharmacology , Liver Neoplasms/metabolism , Liver/metabolism , Fibrosis , Stem Cells/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Cholecystokinin/metabolism , Tumor Microenvironment
15.
CNS Neurosci Ther ; 30(2): e14389, 2024 02.
Article in English | MEDLINE | ID: mdl-37545429

ABSTRACT

AIM: The nocebo effect, such as nausea and vomiting, is one of the major reasons patients discontinue therapy. The underlying mechanisms remain unknown due to a lack of reliable experimental models. The goal of this study was to develop a new animal model of nocebo-related nausea by combining observational learning and Pavlovian conditioning paradigms. METHODS: Male Sprague-Dawley rats with nitroglycerin-induced migraine were given 0.9% saline (a placebo) or LiCl (a nausea inducer) following headache relief, according to different paradigms. RESULTS: Both strategies provoked nocebo nausea responses, with the conditioning paradigm having a greater induction impact. The superposition of two mechanisms led to a further increase in nausea responses. A preliminary investigation of the underlying mechanism revealed clearly raised peripheral and central cholecystokinin (CCK) levels, as well as specific changes in the 5-hydroxytryptamine and cannabinoid systems. Brain networks related to emotion, cognition, and visceral sense expressed higher c-Fos-positive neurons, including the anterior cingulate cortex (ACC), insula, basolateral amygdala (BLA), thalamic paraventricular nucleus (PVT), hypothalamic paraventricular nucleus (PVN), nucleus tractus solitarius (NTS), periaqueductal gray (PAG), and dorsal raphe nucleus-dorsal part (DRD). We also found that nausea expectances in the model could last for at least 12 days. CONCLUSION: The present study provides a useful experimental model of nocebo nausea that might be used to develop potential molecular pathways and therapeutic strategies for nocebo.


Subject(s)
Nocebo Effect , Solitary Nucleus , Humans , Rats , Male , Animals , Rats, Sprague-Dawley , Solitary Nucleus/metabolism , Cholecystokinin/metabolism , Cholecystokinin/pharmacology , Nausea/chemically induced , Nausea/metabolism , Proto-Oncogene Proteins c-fos/metabolism
16.
Eur Neuropsychopharmacol ; 78: 54-63, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37931511

ABSTRACT

Major depressive disorder (MDD) is a serious disease and a burden to patients, families and society. Rodent experiments and human studies suggest that several neuropeptide systems are involved in mood regulation. The aim of this study is two-fold: (i) to monitor, with qPCR, transcript levels of the substance P/tachykinin (TAC), NPY and CCK systems in bulk samples from control and suicide subjects, targeting five postmortem brain regions including locus coeruleus (LC); and (ii) to analyse expression of neuropeptide family transcripts in LC neurons of 'normal' postmortem brains by using laser capture microdissection with Smart-Seq2 RNA sequencing. qPCR revealed distinct regional expression patterns in male and female controls with higher levels for the TAC system in the dorsal raphe nucleus and LC, versus higher transcripts levels of the NPY and CCK systems in prefrontal cortex. In suicide patients, TAC, TAC receptors and a few NPY family transcript levels were increased mainly in prefrontal cortex and LC. The second study on 'normal' noradrenergic LC neurons revealed expression of transcripts for GAL, NPY, TAC1, CCK, and TACR1 and many other peptides (e.g. Cerebellin4 and CARTPT) and receptors (e.g. Adcyap1R1 and GPR173). These data and our previous results on suicide brains indicates that the tachykinin and galanin systems may be valid targets for developing antidepressant medicines. Moreover, the perturbation of neuropeptide systems in MDD patients, and the detection of further neuropeptide and receptor transcripts in LC, shed new light on signalling in noradrenergic LC neurons and on mechanisms possibly associated with mood disorders.


Subject(s)
Depressive Disorder, Major , Neuropeptides , Female , Humans , Male , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Dorsal Raphe Nucleus , Gene Expression Profiling , Locus Coeruleus/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Substance P/metabolism , Cholecystokinin/metabolism
17.
Peptides ; 172: 171114, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37926186

ABSTRACT

Small intestinal satiation pathways involve nutrient-induced stimulation of chemoreceptors leading to release of satiety hormones from intestinal enteroendocrine cells (ECCs). Whether adaptations in these pathways contribute to increased maternal food intake during pregnancy is unknown. To determine the expression of intestinal nutrient-sensors and satiety hormone transcripts and proteins across pregnancy in mice. Female C57BL/6J mice (10-12 weeks old) were randomized to mating and then tissue collection at early- (6.5 d), mid- (12.5 d) or late-pregnancy (17.5 d), or to an unmated age matched control group. Relative transcript expression of intestinal fatty acid, peptide and amino acid and carbohydrate chemoreceptors, as well as gut hormones was determined across pregnancy. The density of G-protein coupled receptor 93 (GPR93), free fatty acid receptor (FFAR) 4, cholecystokinin (CCK) and glucagon-like peptide1 (GLP-1) immunopositive cells was then compared between non-pregnant and late-pregnant mice. Duodenal GPR93 expression was lower in late pregnant than non-pregnant mice (P < 0.05). Ileal FFAR1 expression was higher at mid- than at early- or late-pregnancy. Ileal FFAR2 expression was higher at mid-pregnancy than in early pregnancy. Although FFAR4 expression was consistently lower in late-pregnant than non-pregnant mice (P < 0.001), the density of FFAR4 immunopositive cells was higher in the jejunum of late-pregnant than non-pregnant mice. A subset of protein and fatty acid chemoreceptor transcripts undergo region-specific change during murine pregnancy, which could augment hormone release and contribute to increased food intake. Further investigations are needed to determine the functional relevance of these changes.


Subject(s)
Gastrointestinal Hormones , Satiation , Animals , Female , Mice , Pregnancy , Cholecystokinin/metabolism , Fatty Acids , Gastrointestinal Hormones/metabolism , Mice, Inbred C57BL , Nutrients , Satiation/physiology
18.
J Sci Food Agric ; 104(1): 295-302, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37563097

ABSTRACT

BACKGROUND: Wheat protein intake leads to improved appetite control. However, the active components causing appetite in wheat have not been fully clarified. Gut cholecystokinin (CCK) plays a vital role in appetite control. This study aimed to investigate the ability of wheat protein digest (WPD) to stimulate CCK secretion and clarify the active components and target of action. RESULTS: WPD was prepared by a simulated gastrointestinal digestion model. WPD treatment with a concentration of 5 mg mL-1 significantly stimulated CCK secretion in enteroendocrine STC-1 cells (P < 0.05). Furthermore, oral gavage with WPD in mice significantly increased plasma CCK level at 60 min (P < 0.01). Preparative C18 column separation was used to isolate peptide fractions associated with CCK secretion and peptide sequences were identified by liquid chromatography-tandem mass spectrometry. A new CCK-releasing peptide, RYIVPL, that potently stimulated CCK secretion was successfully identified. After pretreatment with a specific calcium-sensing receptor (CaSR) antagonist, NPS 2143, CCK secretion induced by WPD or RYIVPL was greatly suppressed, suggesting that CaSR was involved in WPD- or RYIVPL-induced CCK secretion. CONCLUSION: The present study demonstrated that WPD has an ability to stimulate CCK secretion in vitro and in vivo, and determined that peptide RYIVPL in WPD could stimulate CCK secretion through CaSR. © 2023 Society of Chemical Industry.


Subject(s)
Cholecystokinin , Triticum , Mice , Animals , Cholecystokinin/metabolism , Triticum/metabolism , Cell Line , Peptides/pharmacology , Receptors, Calcium-Sensing/metabolism , Digestion
19.
Cell Rep ; 42(12): 113467, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37979171

ABSTRACT

The hippocampus is broadly impacted by neuromodulations. However, how neuropeptides shape the function of the hippocampus and the related spatial learning and memory remains unclear. Here, we discover the crucial role of cholecystokinin (CCK) in heterosynaptic neuromodulation from the medial entorhinal cortex (MEC) to the hippocampus. Systematic knockout of the CCK gene impairs CA3-CA1 LTP and space-related performance. The MEC provides most of the CCK-positive neurons projecting to the hippocampal region, which potentiates CA3-CA1 long-term plasticity heterosynaptically in a frequency- and NMDA receptor (NMDAR)-dependent manner. Selective inhibition of MEC CCKergic neurons or downregulation of their CCK mRNA levels also impairs CA3-CA1 LTP formation and animals' performance in the water maze. This excitatory extrahippocampal projection releases CCK upon high-frequency excitation and is active during animal exploration. Our results reveal the critical role of entorhinal CCKergic projections in bridging intra- and extrahippocampal circuitry at electrophysiological and behavioral levels.


Subject(s)
CA1 Region, Hippocampal , CA2 Region, Hippocampal , CA3 Region, Hippocampal , Cholecystokinin , Entorhinal Cortex , Neuronal Plasticity , Spatial Learning , Cholecystokinin/genetics , Cholecystokinin/metabolism , Entorhinal Cortex/metabolism , CA3 Region, Hippocampal/physiology , CA1 Region, Hippocampal/physiology , CA2 Region, Hippocampal/physiology , Synapses/physiology , Spatial Learning/physiology , Animals , Mice , Mice, Knockout , Long-Term Potentiation
20.
World J Gastroenterol ; 29(38): 5374-5382, 2023 Oct 14.
Article in English | MEDLINE | ID: mdl-37900582

ABSTRACT

BACKGROUND: Many studies have shown that interstitial Cajal-like cell (ICLC) abnormalities are closely related to a variety of dynamic gastrointestinal disorders. ICLCs are pacemaker cells for gastrointestinal movement and are involved in the transmission of nerve impulses. AIM: To elucidate the expression profile and significance of cholecystokinin-A (CCK-A) receptors in ICLCs in the common bile duct (CBD), as well as the role of CCK in regulating CBD motility through CCK-A receptors on CBD ICLCs. METHODS: The levels of tyrosine kinase receptor (c-kit) and CCK-A receptors in CBD tissues and isolated CBD cells were quantified using the double immunofluorescence labeling technique. The CCK-mediated enhancement of the movement of CBD muscle strips through CBD ICLCs was observed by a muscle strip contraction test. RESULTS: Immunofluorescence showed co-expression of c-kit and CCK-A receptors in the CBD muscularis layer. Observations of isolated CBD cells showed that c-kit was expressed on the surface of ICLCs, the cell body and synapse were colored and polygonal, and some cells presented protrusions and formed networks adjacent to the CBD while others formed filaments at the synaptic terminals of local cells. CCK-A receptors were also expressed on CBD ICLCs. At concentrations ranging from 10-6 mol/L to 10-10 mol/L, CCK promoted CBD smooth muscle contractility in a dose-dependent manner. In contrast, after ICLC removal, the contractility mediated by CCK in CBD smooth muscle decreased. CONCLUSION: CCK-A receptors are highly expressed on CBD ICLCs, and CCK may regulate CBD motility through the CCK-A receptors on ICLCs.


Subject(s)
Gallbladder , Telocytes , Guinea Pigs , Animals , Receptor, Cholecystokinin A/metabolism , Common Bile Duct , Telocytes/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Cholecystokinin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...