Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 122
1.
Clin Nutr ; 42(1): 9-17, 2023 01.
Article En | MEDLINE | ID: mdl-36473427

BACKGROUND & AIMS: Anorexia is a disabling symptom in cancer and we aimed at investigating the role of changes in gene expression in lung cancer patients presenting with anorexia. METHODS: Genome-wide transcriptomic profiling was assessed in PBMCs RNA from newly diagnosed lung cancer patients and in a control group. RT-qPCR was used for selected genes. RESULTS: RNA-Seq analysis revealed among groups a large number of differentially expressed genes mainly implicated in immune system regulation, oxidative stress and cytokine-mediated inflammation signaling pathways. In particular, we identified a total of 983 DEGs (843 up-regulated; 140 down-regulated) in anorexic cancer compared to controls. A selected number of DEGs including ADAM8, SMAD4, CCR4 and CLU were differentially expressed within cancer group according to the presence/absence of anorexia. In terms of RT-qPCR, ADAM8 was less expressed in cancer patients than controls (p < 0.001), and in anorexic patients vs controls (p = 0.001). The expression of SMAD4 was lower in cancer vs controls (p = 0.005), and in anorexic patients vs controls (p = 0.009). We observed lower CCR4 expression in both anorexic and non-anorexic vs control (p = 0.004, p = 0.011, respectively) and a similar trend was present for CLU. CONCLUSIONS: Our data shed new light on the role of specific genes and their associated molecular pathways as potential key mechanisms for the development of anorexia and may represent a novel landmark for understanding the complex pathophysiology of impaired appetite in cancer.


Anorexia , Lung Neoplasms , Humans , Anorexia/genetics , Leukocytes, Mononuclear , Lung Neoplasms/genetics , Gene Expression Profiling , Gene Expression , Membrane Proteins , ADAM Proteins
2.
J Pharmacol Sci ; 148(1): 6-13, 2022 Jan.
Article En | MEDLINE | ID: mdl-34924131

This prospective study examined the impact of genetic polymorphisms on the pharmacokinetics and clinical efficacy and safety of lenvatinib, a substrate of ATP-binding transporters, in a cohort of 48 Japanese patients with hepatocellular carcinoma (HCC). Pharmacokinetic studies were performed at the start of lenvatinib therapy (day 1) and on day 15. The coefficients of variation in AUC0-24h of lenvatinib on days 1 and 15 were 44.0% and 52.4%, respectively. Although the ABCB1 3435C > A, 1236C > T, and 2677G>T/A polymorphisms did not influence pharmacokinetic parameters, the AUC0-24h values on days 1 and 15 of the ABCG2 C/A or A/A group were approximately 1.1-fold and 1.4-fold that in the ABCG2 C/C group (P = 0.164 and 0.024). There were no significant differences in AUC0-24h on days 1 and 15 between the responders (complete or partial response) and non-responders (stable or progressive disease). The AUC0-24h on day 15 in those developing anorexia of any grade was significantly higher than that without such development (P = 0.017). In multivariate analysis, ABCG2 421C > A C/A or A/A was significantly associated with the development of anorexia (odds ratio 9.009, P = 0.009). ABCG2 421C > A polymorphism could affect exposure to lenvatinib and the development of anorexia.


ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Neoplasm Proteins/genetics , Phenylurea Compounds/pharmacokinetics , Polymorphism, Genetic , Quinolines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Aged , Aged, 80 and over , Anorexia/chemically induced , Anorexia/genetics , Asian People , Carcinoma, Hepatocellular/drug therapy , Cohort Studies , Female , Humans , Liver Neoplasms/drug therapy , Male , Middle Aged , Phenylurea Compounds/adverse effects , Phenylurea Compounds/therapeutic use , Quinolines/adverse effects , Quinolines/therapeutic use , Time Factors
3.
Endocrinology ; 162(9)2021 09 01.
Article En | MEDLINE | ID: mdl-34190328

Neurotensin (NT) is an anorexic gut hormone and neuropeptide that increases in circulation following bariatric surgery in humans and rodents. We sought to determine the contribution of NT to the metabolic efficacy of vertical sleeve gastrectomy (VSG). To explore a potential mechanistic role of NT in VSG, we performed sham or VSG surgeries in diet-induced obese NT receptor 1 (NTSR1) wild-type and knockout (ko) mice and compared their weight and fat mass loss, glucose tolerance, food intake, and food preference after surgery. NTSR1 ko mice had reduced initial anorexia and body fat loss. Additionally, NTSR1 ko mice had an attenuated reduction in fat preference following VSG. Results from this study suggest that NTSR1 signaling contributes to the potent effect of VSG to initially reduce food intake following VSG surgeries and potentially also on the effects on macronutrient selection induced by VSG. However, maintenance of long-term weight loss after VSG requires signals in addition to NT.


Anorexia/etiology , Avoidant Restrictive Food Intake Disorder , Gastrectomy/adverse effects , Postoperative Complications/genetics , Receptors, Neurotensin/genetics , Animals , Anorexia/genetics , Dietary Fats , Gastrectomy/methods , Male , Mice , Mice, Knockout , Phobic Disorders/etiology , Phobic Disorders/genetics , Postoperative Complications/psychology
4.
Int J Mol Sci ; 22(6)2021 Mar 12.
Article En | MEDLINE | ID: mdl-33809200

During its evolution, cancer induces changes in patients' energy metabolism that strongly affect the overall clinical state and are responsible for cancer-related cachexia syndrome. To better understand the mechanisms underlying cachexia and its metabolic derangements, research efforts should focus on the events that are driven by the immune system activation during the evolution of neoplastic disease and on the phenomena of "resistance" and "tolerance" typically involved in the human body response against stress, pathogens, or cancer. Indeed, in the case where resistance is not able to eliminate the cancer, tolerance mechanisms can utilize the symptoms of cachexia (anemia, anorexia, and fatigue) to counteract unregulated cancer growth. These notions are also sustained by the evidence that cancer cachexia may be reversible if the resistance and tolerance phases are supported by appropriate antineoplastic treatments. Accordingly, there is no doubt that anticachectic therapies have an irreplaceable role in cases of reversible cancer cachexia where, if harmoniously associated with effective antineoplastic therapies, they can contribute to preserve the quality of life and improve prognosis. Such anticachectic treatments should be based on targeting the complex immunological, inflammatory, and metabolic pathways involved in the complex pathogenesis of cachexia. Meanwhile, the role of the anticachectic therapies is very different in the stage of irreversible cachexia when the available antineoplastic treatments are not able to control the disease and the resistance mechanisms fail with the prevalence of the tolerance phenomena. At this stage, they can be useful only to improve the quality of life, allowing the patient and their family to get a better awareness of the final phases of life, thereby opening to the best spiritual remodulation of the final event, death.


Cachexia/genetics , Energy Metabolism/genetics , Immune Tolerance/genetics , Neoplasms/genetics , Anorexia/genetics , Anorexia/metabolism , Anorexia/pathology , Cachexia/complications , Cachexia/metabolism , Cachexia/pathology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neoplasms/complications , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Quality of Life
5.
Int J Mol Sci ; 22(4)2021 Feb 20.
Article En | MEDLINE | ID: mdl-33672695

Circadian desynchrony induced by a long period of irregular feeding leads to metabolic diseases, such as obesity and diabetes mellitus. The recently identified neurosecretory protein GL (NPGL) and neurosecretory protein GM (NPGM) are hypothalamic small proteins that stimulate food intake and fat accumulation in several animals. To clarify the mechanisms that evoke feeding behavior and induce energy metabolism at the appropriate times in accordance with a circadian rhythm, diurnal fluctuations in Npgl and Npgm mRNA expression were investigated in mice. Quantitative RT-PCR analysis revealed that the mRNAs of these two genes were highly expressed in the mediobasal hypothalamus during the active dark phase under ad libitum feeding. In mice restricted to 3 h of feeding during the inactive light phase, the Npgl mRNA level was augmented in the moment prior to the feeding period and the midnight peak of Npgm mRNA was attenuated. Moreover, the mRNA expression levels of clock genes, feeding regulatory neuropeptides, and lipid metabolic enzymes in the central and peripheral tissues were comparable to those of central Npgl and Npgm. These data suggest that Npgl and Npgm transcription fluctuates daily and likely mediates feeding behavior and/or energy metabolism at an appropriate time according to the meal timing.


Feeding Behavior/physiology , Gene Expression Regulation , Hypothalamus/metabolism , Nerve Tissue Proteins/metabolism , Analysis of Variance , Animals , Anorexia/blood , Anorexia/genetics , Blood Glucose/metabolism , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Gene Expression Profiling , Insulin/blood , Lipid Metabolism/genetics , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Orexins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
6.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article En | MEDLINE | ID: mdl-33255553

Obesity-associated low-grade inflammation favors weight gain, whereas systemic infection frequently leads to anorexia. Thus, inflammatory signals can either induce positive or negative energy balance. In this study, we used whole-cell patch-clamp to investigate the acute effects of three important proinflammatory cytokines, tumor necrosis factor α (TNF-α), interleukin-6, and interleukin-1ß (IL-1ß) on the membrane excitability of agouti-related peptide (AgRP)- or proopiomelanocortin (POMC)-producing neurons. We found that both TNF-α and IL-1ß acutely inhibited the activity of 35-42% of AgRP-producing neurons, whereas very few POMC neurons were depolarized by TNF-α. Interleukin-6 induced no acute changes in the activity of AgRP or POMC neurons. Our findings indicate that the effect of TNF-α and IL-1ß, especially on the activity of AgRP-producing neurons, may contribute to inflammation-induced anorexia observed during acute inflammatory conditions.


Agouti-Related Protein/genetics , Inflammation/genetics , Interleukin-1beta/genetics , Obesity/genetics , Tumor Necrosis Factor-alpha/genetics , Animals , Anorexia/genetics , Anorexia/metabolism , Anorexia/pathology , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Energy Metabolism , Humans , Hypothalamus/metabolism , Hypothalamus/pathology , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/genetics , Mice , Neurons/metabolism , Neurons/pathology , Neuropeptide Y/genetics , Obesity/metabolism , Obesity/pathology , Patch-Clamp Techniques , Pro-Opiomelanocortin/genetics
7.
Neuropharmacology ; 180: 108289, 2020 12 01.
Article En | MEDLINE | ID: mdl-32890590

Up to 80% of cancer patients are affected by the cancer anorexia-cachexia syndrome (CACS), which leads to excessive body weight loss, reduced treatment success and increased lethality. The area postrema/nucleus of the solitary tract (AP/NTS) region emerged as a central nervous key structure in this multi-factorial process. Neurons in this area are targeted by cytokines and signal to downstream sites involved in energy homeostasis. NTS neurons expressing prolactin-releasing peptide (PrRP) are implicated in the control of energy intake and hypothalamus-pituitary-adrenal (HPA) axis activation, which contributes to muscle wasting. To explore if brainstem PrRP neurons contribute to CACS, we selectively knocked down PrRP expression in the NTS of hepatoma tumor-bearing rats by an AAV/shRNA gene silencing approach. PrRP knockdown reduced body weight loss and anorexia compared to tumor-bearing controls treated with a non-silencing AAV. Gastrocnemius and total hind limb muscle weight was higher in PrPR knockdown rats. Corticosterone levels were increased in the early phase after tumor induction at day 6 in both groups but returned to baseline levels at day 21 in the PrRP knockdown group. While we did not detect significant changes in gene expression of markers for muscle protein metabolism (MuRF-1, myostatin, mTOR and REDD1), mTOR and REDD1 tended to be lower after disruption PrRP signalling. In conclusion, we identified brainstem PrRP as a possible neuropeptide mediator of CACS in hepatoma tumor-bearing rats. The central and peripheral downstream mechanisms require further investigation and might involve HPA axis activation.


Anorexia/metabolism , Brain Stem/metabolism , Cachexia/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Prolactin-Releasing Hormone/metabolism , Animals , Anorexia/genetics , Cachexia/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Gene Knockdown Techniques/methods , Liver Neoplasms/genetics , Male , Prolactin-Releasing Hormone/deficiency , Prolactin-Releasing Hormone/genetics , Rats , Rats, Inbred BUF
8.
Adipocyte ; 9(1): 472-483, 2020 12.
Article En | MEDLINE | ID: mdl-32772766

Although adipose tissue metabolism in obesity has been widely studied, there is limited research on the anorexic state, where the endocrine system is disrupted by reduced adipose tissue mass and there are depot-specific changes in adipocyte type and function. Stress exposure at different stages of life can alter the balance between energy intake and expenditure and thereby contribute to the pathogenesis of anorexia nervosa. This review integrates information from human clinical trials to describe endocrine, genetic and epigenetic aspects of adipose tissue physiology in the anorexic condition. Changes in the hypothalamus-pituitary-thyroid, -adrenal, and -gonadal axes and their relationships to appetite regulation and adipocyte function are discussed. Because of the role of stress in triggering or magnifying anorexia, and the dynamic but also persistent nature of environmentally-induced epigenetic modifications, epigenetics is likely the link between stress and long-term changes in the endocrine system that disrupt homoeostatic food intake and adipose tissue metabolism. Herein, we focus on the adipocyte and changes in its function, including alterations reinforced by endocrine disturbance and dysfunctional adipokine regulation. This information is critical because of the poor understanding of anorexic pathophysiology, due to the lack of suitable research models, and the complexity of genetic and environmental interactions.


Adipose Tissue/metabolism , Anorexia/genetics , Anorexia/metabolism , Stress, Physiological , Adipocytes/metabolism , Animals , Disease Susceptibility , Endocrine System/metabolism , Energy Metabolism , Epigenesis, Genetic , Gene Expression Regulation , Humans , Lipid Metabolism , Organ Size
9.
Elife ; 92020 07 29.
Article En | MEDLINE | ID: mdl-32723474

The cytokine, GDF15, is produced in pathological states which cause cellular stress, including cancer. When over expressed, it causes dramatic weight reduction, suggesting a role in disease-related anorexia. Here, we demonstrate that the GDF15 receptor, GFRAL, is located in a subset of cholecystokinin neurons which span the area postrema and the nucleus of the tractus solitarius of the mouse. GDF15 activates GFRALAP/NTS neurons and supports conditioned taste and place aversions, while the anorexia it causes can be blocked by a monoclonal antibody directed at GFRAL or by disrupting CCK neuronal signalling. The cancer-therapeutic drug, cisplatin, induces the release of GDF15 and activates GFRALAP/NTS neurons, as well as causing significant reductions in food intake and body weight in mice. These metabolic effects of cisplatin are abolished by pre-treatment with the GFRAL monoclonal antibody. Our results suggest that GFRAL neutralising antibodies or antagonists may provide a co-treatment opportunity for patients undergoing chemotherapy.


Anorexia/genetics , Brain Stem/physiology , Growth Differentiation Factor 15/genetics , Neurons/physiology , Pica/genetics , Signal Transduction , Animals , Cholecystokinin/metabolism , Growth Differentiation Factor 15/administration & dosage , Growth Differentiation Factor 15/metabolism , Male , Mice , Mice, Inbred C57BL , Random Allocation , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage
10.
Sci Rep ; 10(1): 11475, 2020 07 10.
Article En | MEDLINE | ID: mdl-32651433

The increased awareness of obstructive sleep apnoea's (OSA) links to Alzheimer's disease and major psychiatric disorders has recently directed an intensified search for their potential shared mechanisms. We hypothesised that neuroinflammation and the microglial TLR2-system may act as a core process at the intersection of their pathophysiology. Moreover, we postulated that inflammatory-response might underlie development of key behavioural and neurostructural changes in OSA. Henceforth, we set out to investigate effects of 3 weeks' exposure to chronic intermittent hypoxia in mice with or without functional TRL2 (TLR2+/+, C57BL/6-Tyrc-Brd-Tg(Tlr2-luc/gfp)Kri/Gaj;TLR2-/-,C57BL/6-Tlr2tm1Kir). By utilising multimodal imaging in this established model of OSA, a discernible neuroinflammatory response was demonstrated for the first time. The septal nuclei and forebrain were shown as the initial key seed-sites of the inflammatory cascade that led to wider structural changes in the associated neurocircuitry. Finally, the modulatory role for the functional TLR2-system was suggested in aetiology of depressive, anxious and anorexiolytic symptoms in OSA.


Immunity, Innate/genetics , Inflammation/genetics , Sleep Apnea, Obstructive/genetics , Toll-Like Receptor 2/genetics , Animals , Anorexia/genetics , Anorexia/immunology , Anxiety/genetics , Anxiety/immunology , Depression/genetics , Depression/immunology , Humans , Hypoxia/genetics , Hypoxia/immunology , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Prosencephalon/metabolism , Prosencephalon/pathology , Septal Nuclei , Sleep Apnea, Obstructive/immunology , Sleep Apnea, Obstructive/pathology
12.
EMBO Rep ; 21(3): e48804, 2020 03 04.
Article En | MEDLINE | ID: mdl-32026535

Mitochondrial dysfunction promotes metabolic stress responses in a cell-autonomous as well as organismal manner. The wasting hormone growth differentiation factor 15 (GDF15) is recognized as a biomarker of mitochondrial disorders, but its pathophysiological function remains elusive. To test the hypothesis that GDF15 is fundamental to the metabolic stress response during mitochondrial dysfunction, we investigated transgenic mice (Ucp1-TG) with compromised muscle-specific mitochondrial OXPHOS capacity via respiratory uncoupling. Ucp1-TG mice show a skeletal muscle-specific induction and diurnal variation of GDF15 as a myokine. Remarkably, genetic loss of GDF15 in Ucp1-TG mice does not affect muscle wasting or transcriptional cell-autonomous stress response but promotes a progressive increase in body fat mass. Furthermore, muscle mitochondrial stress-induced systemic metabolic flexibility, insulin sensitivity, and white adipose tissue browning are fully abolished in the absence of GDF15. Mechanistically, we uncovered a GDF15-dependent daytime-restricted anorexia, whereas GDF15 is unable to suppress food intake at night. Altogether, our evidence suggests a novel diurnal action and key pathophysiological role of mitochondrial stress-induced GDF15 in the regulation of systemic energy metabolism.


Anorexia , Growth Differentiation Factor 15 , Adipose Tissue, White/metabolism , Animals , Anorexia/genetics , Anorexia/metabolism , Energy Metabolism , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Mice , Mitochondria/metabolism
13.
Peptides ; 122: 170157, 2019 12.
Article En | MEDLINE | ID: mdl-31550523

Neuropeptide K (NPK) induces satiety in birds and mammals. We demonstrated that in birds this effect was associated with the hypothalamus, but beyond this little is known in any species regarding the central mechanism of action. Thus, this study was designed to identify hypothalamic molecular mechanisms associated with the food intake-inhibiting effects of NPK in chicks. In Experiment 1, intracerebroventricular (ICV) injection of 1.0 and 3.0 nmol of NPK reduced food intake and we identified an effective dose for microinjection. In Experiment 2, food intake was reduced when NPK was microinjected into the PVN. In Experiment 3, whole hypothalamus was collected from chicks at 1 h post-ICV NPK injection. The abundance of corticotropin-releasing factor (CRF) and agouti-related peptide (AgRP) mRNA was reduced in NPK-injected chicks. In Experiment 4, within the isolated paraventricular nucleus (PVN) there was less CRF mRNA, and within the arcuate nucleus (ARC) there was less AgRP mRNA, in NPK- than vehicle-treated chicks at 1 h post-injection. We conclude that there are first order neurons for NPK that reside within the PVN, and the anorexigenic effect of NPK is associated with a decrease in AgRP in the ARC.


Anorexia/drug therapy , Appetite Depressants/pharmacology , Eating/genetics , Tachykinins/pharmacology , Agouti-Related Protein/genetics , Animals , Anorexia/genetics , Anorexia/pathology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Chickens , Corticotropin-Releasing Hormone/genetics , Disease Models, Animal , Drinking/drug effects , Gene Expression Regulation/drug effects , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Infusions, Intraventricular , Neurons/drug effects , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/genetics , Tachykinins/genetics
14.
Article En | MEDLINE | ID: mdl-31446069

The body weight-selected lines of chickens are a model for understanding factors that predispose an individual to anorexia or obesity. The high body weight-selected (HWS) individuals are compulsive eaters that become obese whereas the low body weight-selected (LWS) are relatively lean and hypophagic. The objective of this study was to measure gene expression of various preadipocyte, proliferation, metabolic, and apoptotic markers in the stromal-vascular fraction and adipocytes from LWS and HWS adipose tissue. Although preadipocyte and proliferation markers were more highly expressed in the stromal-vascular fraction of LWS than HWS chicks, greater expression of granzyme-A and the presence of more annexin V-positive cells suggests that apoptosis may limit the adipogenic potential of adipocyte precursor cells and represent a novel mechanism that regulates the expansion of adipose tissue. Results provide insights on cellular mechanisms associated with adipose tissue development in the lean and obese state.


Adipogenesis/genetics , Adipose Tissue/metabolism , Anorexia/genetics , Apoptosis/genetics , Biomarkers/metabolism , Chickens/genetics , Genetic Predisposition to Disease , Obesity/genetics , Animals , Caspase 3/genetics , Caspase 3/metabolism , Granzymes/genetics , Granzymes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
15.
Article En | MEDLINE | ID: mdl-31404649

Central administration of neuropeptide K (NPK), a 36-amino acid peptide, is associated with anorexigenic effects in rodents and chickens. The mechanisms underlying the potent anorexigenic effects of NPK are still poorly understood. Thus, the aim of the present study was to identify the hypothalamic nuclei and neuropeptides that mediate anorexic effects of NPK in 7 day-old Japanese quail (Coturnix japonica). After a 6 h fast, intracerebroventricular (ICV) injection of NPK decreased food and water intake for 180 min post-injection. Quail injected with NPK had more c-Fos immunoreactive cells in the arcuate nucleus (ARC), lateral hypothalamus, and paraventricular nucleus (PVN) compared to the birds that were injected with the vehicle. In the ARC of NPK-injected quail, there was decreased neuropeptide Y (NPY), NPY receptor sub-type 1, and agouti-related peptide mRNA, and increased CART, POMC, and neurokinin receptor 1 mRNA. NPK-injected quail expressed greater amounts of corticotropin-releasing factor (CRF), CRF receptor sub-type 2, melanocortin receptors 3 and 4, and urocortin 3 mRNA in the PVN. In conclusion, results provide insights into understanding NPK-induced changes in hypothalamic physiology and feeding behavior, and suggest that the anorexigenic effects of NPK involve the ARC and PVN, with increased CRF and melanocortin and reduced NPY signaling.


Anorexia/genetics , Coturnix/metabolism , Hypothalamus/metabolism , Tachykinins/pharmacology , Animals , Anorexia/chemically induced , Anorexia/metabolism , Anorexia/pathology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Corticotropin-Releasing Hormone/metabolism , Coturnix/genetics , Drinking/drug effects , Eating/drug effects , Gene Expression Regulation/drug effects , Humans , Hypothalamus/drug effects , Infusions, Intraventricular , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , Pro-Opiomelanocortin/genetics , Proto-Oncogene Proteins c-fos/genetics , Receptors, Melanocortin/genetics , Tachykinins/metabolism , Urocortins/genetics
16.
FEBS Lett ; 593(19): 2762-2770, 2019 10.
Article En | MEDLINE | ID: mdl-31281956

Tonicity-responsive enhancer binding protein (TonEBP) is a widely expressed transcription factor and is important in the regulation of inflammatory cytokines. Here, we have identified TonEBP expression in the hypothalamus, which is particularly high in proopiomelanocortin (POMC) neurons. TonEBP overexpression stimulates POMC transcription, and TonEBP haploinsufficiency in TonEBP (+/-) mice results in a decrease in hypothalamic POMC expression. TonEBP (+/-) mice show reduced sickness responses, which include anorexia and hyperthermia, that are initially induced by tumor necrosis factor (TNF)-α. TonEBP (+/-) mice also show lower levels of TNF-α-induced hypothalamic expression of POMC and pro-inflammatory cytokines. These results suggest that TonEBP is an important molecular regulator in the development of inflammatory sickness responses through the control of POMC and pro-inflammatory cytokine expression in the hypothalamus.


Anorexia/metabolism , Fever/metabolism , Hypothalamus/metabolism , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Anorexia/genetics , Cell Line , Fever/genetics , Hypothalamus/pathology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Transcription Factors/metabolism
17.
Sci Rep ; 9(1): 6011, 2019 04 12.
Article En | MEDLINE | ID: mdl-30979915

Helicobacter pylori (Hp) infection is related to the pathogenesis of chronic gastric disorders and extragastric diseases. Here, we examined the anorexigenic and anxiogenic effects of Hp vacuolating cytotoxin A (VacA) through activation of hypothalamic urocortin1 (Ucn1). VacA was detected in the hypothalamus after peripheral administration and increased Ucn1 mRNA expression and c-Fos-positive cells in the hypothalamus but not in the nucleus tractus solitarius. c-Fos and Ucn1-double positive cells were detected. CRF1 and CRF2 receptor antagonists suppressed VacA-induced anxiety and anorexia, respectively. VacA activated single paraventricular nucleus neurons and A7r5 cells; this activation was inhibited by phospholipase C (PLC) and protein kinase C (PKC) inhibitors. VacA causes anorexia and anxiety through the intracellular PLC-PKC pathway, migrates across the blood-brain barrier, and activates the Ucn1-CRF receptor axis.


Anorexia/chemically induced , Anxiety/chemically induced , Cytotoxins/toxicity , Helicobacter pylori/cytology , Hypothalamus/drug effects , Urocortins/metabolism , Vacuoles/metabolism , Animals , Anorexia/genetics , Anxiety/genetics , Body Weight/drug effects , Eating/drug effects , Gerbillinae , Helicobacter pylori/physiology , Hypothalamus/cytology , Hypothalamus/metabolism , Mice , RNA, Messenger/genetics , Signal Transduction/drug effects , Urocortins/genetics
18.
Am J Physiol Regul Integr Comp Physiol ; 316(6): R802-R818, 2019 06 01.
Article En | MEDLINE | ID: mdl-30969842

Chickens from lines selected for low (LWS) or high (HWS) body weight (BW) differ in appetite and adiposity. Mechanisms associated with the predisposition to becoming obese are unclear. The objective of the experiment was to evaluate developmental changes in depot-specific adipose tissue during the first 2 wk posthatch. Subcutaneous (SQ), clavicular (CL), and abdominal (AB) depots were collected at hatch (DOH) and days 4 (D4) and 14 (D14) posthatch for histological and mRNA measurements. LWS chicks had decreased SQ fat mass on a BW basis with reduced adipocyte size from DOH to D4 and increased BW and fat mass with unchanged adipocyte size from D4 to D14. HWS chicks increased in BW from DOH to D14 and increased in fat mass in all three depots with enlarged adipocytes in the AB depot from D4 to D14. Meanwhile, CCAAT/enhancer-binding protein-α, neuropeptide Y, peroxisome proliferator-activated receptor-γ, and acyl-CoA dehydrogenase mRNAs differed among depots between lines at different ages. Plasma nonesterified fatty acids were greater in LWS than HWS at D4 and D14. From DOH to D4, LWS chicks mobilized SQ fat and replenished the reservoir through hyperplasia, whereas HWS chicks were dependent on hyperplasia and hypertrophy to maintain adipocyte size and depot mass. From D4 to D14, adipose tissue catabolism and adipogenesis slowed. Whereas LWS fat depots and adipocyte sizes remained stable, HWS chicks rapidly accumulated fat in CL and AB depots. Chicks predisposed to be anorexic or obese have different fat development patterns during the first 2 wk posthatch.


Adipogenesis , Adipose Tissue/growth & development , Adiposity , Chickens/growth & development , Weight Gain , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Animals, Newborn , Anorexia/genetics , Anorexia/metabolism , Anorexia/physiopathology , Anorexia/veterinary , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Chickens/genetics , Chickens/metabolism , Female , Gene Expression Regulation , Male , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Obesity/veterinary , PPAR gamma/genetics , PPAR gamma/metabolism , Poultry Diseases/genetics , Poultry Diseases/metabolism , Poultry Diseases/physiopathology , Time Factors
19.
Yakugaku Zasshi ; 138(8): 1017-1024, 2018.
Article Ja | MEDLINE | ID: mdl-30068841

Various neuropeptides play an essential role in the nutrient sensing mechanism and related homeostasis. Nesfatin-1 is a newly identified neuropeptide having anorectic activity, and nesfatin-1-containing neurons are widely distributed in the brain, including the hypothalamus and brain stem. Our previous study showed that dehydration-induced anorectic effects are mediated via the central nesfatin-1 pathway in rats. Our recent studies have also shown that peripheral anorectic peptides (cholecystokinin-8, glucagon-like peptide-1, and leptin) and an antineoplastic agent (cisplatin) caused inhibition of feeding via the central nesfatin-1 pathway in rats. Nesfatin-1-containing neurons in the central nervous system, in particular the hypothalamus and the brain stem, may mediate peripheral nutrient signals and regulate feeding behavior.


Anorexia/etiology , Anorexia/genetics , Cholecystokinin/physiology , Eating/drug effects , Eating/genetics , Food , Glucagon-Like Peptide 1/physiology , Leptin/physiology , Nutritional Physiological Phenomena/genetics , Nutritional Physiological Phenomena/physiology , Signal Transduction/physiology , Animals , Antineoplastic Agents/adverse effects , Brain Stem/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/physiology , Cholecystokinin/metabolism , Cisplatin/adverse effects , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Glucagon-Like Peptide 1/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Male , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Nucleobindins , Rats, Wistar
20.
BMJ Case Rep ; 20182018 Jul 09.
Article En | MEDLINE | ID: mdl-29991577

The SHANK2 gene codes for a protein involved in organising the postsynaptic density and disruptions have been associated with autism spectrum disorders (ASDs). ASDs are frequently comorbid with intellectual disability and anxiety disorders and emerging evidence suggests potentially common aetiologies. Here, we report the case of an 18-year-old man with ASD who presented with severe anorexia due to fear of food contamination, food avoidance and stereotypies attributable to underlying obsessive compulsive disorder (OCD). The patient was found to be heterozygous for c.2518C>T (p.Pro840Ser), a likely damaging coding variant in the proline rich region of SHANK2 Interestingly, the patient's disordered eating behaviour began to improve only after high-dose fluoxetine was initiated to target OCD symptoms. Overall, this case highlights the utility of molecular genetic testing in clinical psychiatry and provides an example of how genetic information can inform clinicians in the treatment of complex neuropsychiatric syndromes.


Anorexia/genetics , Autistic Disorder/genetics , Fluoxetine/administration & dosage , Nerve Tissue Proteins/genetics , Obsessive-Compulsive Disorder/drug therapy , Obsessive-Compulsive Disorder/genetics , Selective Serotonin Reuptake Inhibitors/administration & dosage , Adolescent , Anorexia/complications , Autistic Disorder/complications , Humans , Male , Obsessive-Compulsive Disorder/complications
...