Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
Physiol Behav ; 276: 114474, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38272107

ABSTRACT

Nausea is an uncomfortable sensation that accompanies many therapeutics, especially diabetes treatments involving glucagon-like peptide-1 receptor (GLP1R) agonists. Recent studies in mice have revealed that GLP1R-expressing neurons in the area postrema play critical roles in nausea. Here, we characterized a ligand-conjugated saporin that can efficiently ablate GLP1R+ cells from humans, mice, and the Suncus murinus, a small animal model capable of emesis. This new tool provides a strategy to manipulate specific neural pathways in the area postrema in the Suncus murinus and may help elucidate roles of area postrema GLP1R+ neurons in emesis during therapeutics involving GLP1R agonists.


Subject(s)
Area Postrema , Glucagon-Like Peptide-1 Receptor , Animals , Humans , Mice , Glucagon-Like Peptide-1 Receptor/metabolism , Nausea , Neurons/metabolism , Vomiting/metabolism , Shrews
2.
Nature ; 625(7996): 760-767, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38092039

ABSTRACT

GDF15, a hormone acting on the brainstem, has been implicated in the nausea and vomiting of pregnancy, including its most severe form, hyperemesis gravidarum (HG), but a full mechanistic understanding is lacking1-4. Here we report that fetal production of GDF15 and maternal sensitivity to it both contribute substantially to the risk of HG. We confirmed that higher GDF15 levels in maternal blood are associated with vomiting in pregnancy and HG. Using mass spectrometry to detect a naturally labelled GDF15 variant, we demonstrate that the vast majority of GDF15 in the maternal plasma is derived from the feto-placental unit. By studying carriers of rare and common genetic variants, we found that low levels of GDF15 in the non-pregnant state increase the risk of developing HG. Conversely, women with ß-thalassaemia, a condition in which GDF15 levels are chronically high5, report very low levels of nausea and vomiting of pregnancy. In mice, the acute food intake response to a bolus of GDF15 is influenced bi-directionally by prior levels of circulating GDF15 in a manner suggesting that this system is susceptible to desensitization. Our findings support a putative causal role for fetally derived GDF15 in the nausea and vomiting of human pregnancy, with maternal sensitivity, at least partly determined by prepregnancy exposure to the hormone, being a major influence on its severity. They also suggest mechanism-based approaches to the treatment and prevention of HG.


Subject(s)
Growth Differentiation Factor 15 , Hyperemesis Gravidarum , Nausea , Vomiting , Animals , Female , Humans , Mice , Pregnancy , beta-Thalassemia/blood , beta-Thalassemia/metabolism , Fetus/metabolism , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/metabolism , Hormones/blood , Hormones/metabolism , Hyperemesis Gravidarum/complications , Hyperemesis Gravidarum/metabolism , Hyperemesis Gravidarum/prevention & control , Hyperemesis Gravidarum/therapy , Nausea/blood , Nausea/complications , Nausea/metabolism , Placenta/metabolism , Vomiting/blood , Vomiting/complications , Vomiting/metabolism
3.
Curr Mol Med ; 23(4): 341-349, 2023.
Article in English | MEDLINE | ID: mdl-35549869

ABSTRACT

5-HT3 receptor antagonists corresponding to ondansetron, granisetron, tropisetron, and palonosetron are clinically accustomed to treating nausea and emesis in chemotherapy patients. However, current and previous studies reveal novel potentials of those ligands in other diseases involving the nervous system, such as addiction, pruritus, and neurological disorders, such as anxiety, psychosis, nociception, and cognitive function. This review gathers existing studies to support the role of 5-HT3 receptors in CIPN modulation. It has been reported that chemotherapy drugs increase the 5-HT content that binds with the 5-HT3 receptor, which later induces pain. As also shown in pre-clinical and clinical studies that various neuropathic pains could be blocked by the 5-HT3 receptor antagonists, we proposed that 5-HT3 receptor antagonists via 5- HT3 receptors may also inhibit neuropathic pain induced by chemotherapy. Our review suggests that future studies focus more on the 5-HT3 receptor antagonists and their modulation in CIPN to reduce the gap in the current pharmacotherapy for cancer-related pain.


Subject(s)
Antineoplastic Agents , Peripheral Nervous System Diseases , Humans , Vomiting/chemically induced , Vomiting/drug therapy , Vomiting/metabolism , Serotonin/adverse effects , Serotonin 5-HT3 Receptor Antagonists/adverse effects , Antineoplastic Agents/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/prevention & control
4.
Nihon Saikingaku Zasshi ; 76(2): 139-148, 2021.
Article in Japanese | MEDLINE | ID: mdl-34039783

ABSTRACT

Staphylococcus aureus food poisoning was shown by Dack et al. in 1930 to be caused by staphylococcal enterotoxin (SE) produced by S. aureus, rather than by the bacterial infection. However, the emetic mechanism of SE has remained unclear. In this study, we analyzed the emetic activity of SE in several emetic animal models and tried to elucidate the mechanism of emesis. We established a small primate, common marmoset, as a novel emetic model for SE. We also analyzed the immunofluorescence analysis of the gastrointestinal tract of the common marmoset and found that SE binds to submucosal mast cells in the gastrointestinal tract and SE induces degranulation of the mast cells. Furthermore, we showed that SE induces histamine releases, which is inhibited by mast cell stabilizer. In addition, treatment of common marmosets with either mast cell stabilizer or histamine H1 receptor antagonists suppressed the emetic response induced by SE. These results indicate that orally administered SE binds to submucosal mast cells in the gastrointestinal tract and causes degranulation, resulting in the release of histamine, which in turn causes emesis.


Subject(s)
Enterotoxins/adverse effects , Histamine Release/drug effects , Vomiting/chemically induced , Vomiting/drug therapy , Amino Acid Sequence , Animals , Callithrix , Cell Degranulation/drug effects , Disease Models, Animal , Enterotoxins/chemistry , Enterotoxins/metabolism , Histamine Antagonists/pharmacology , Histamine Antagonists/therapeutic use , Intestinal Mucosa/cytology , Mast Cells/metabolism , Mast Cells/physiology , Protein Conformation , Shrews , Vomiting/metabolism
5.
Eur J Gastroenterol Hepatol ; 33(5): 691-694, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33787540

ABSTRACT

BACKGROUND: Infection due to severe acute respiratory syndrome coronavirus 2 is typically associated with a respiratory syndrome, but gastrointestinal symptoms have been described in early reports from China. However, data from European centres are scarce. OBJECTIVES: We aimed to characterise the gastrointestinal manifestations of patients with coronavirus disease 2019 (COVID-19) and their disease course. METHODS: Patients admitted at our centre between March and April 2020 with diagnosis of COVID-19 were included. Asymptomatic patients or those without symptom information were excluded. Clinical features, laboratory data and disease severity (mechanical ventilation, intensive care admission or death) were analysed. RESULTS: Two-hundred one patients were included (median age 71 years; 56.2% male). Digestive symptoms were reported by 60 (29.9%) patients during the disease course, being part of the disease presentation in 34 (16.9%). The most frequent were diarrhoea in 36 patients (17.9%). Patients with gastrointestinal symptoms were younger (P = 0.032), had higher haemoglobin levels (P = 0.002) and lower C-reactive protein (P = 0.045) and potassium levels (P = 0.004). Patients with digestive symptoms had less severe disease (28.3 vs. 44.0%; P = 0.038). Regarding liver damage, aspartate aminotransferase (AST) was elevated in 65.2% of patients and alanine aminotransferase (ALT) in 62.7%, but these patients did not present a more severe disease (elevated AST P = 0.062; elevated ALT P = 0.276). CONCLUSION: A significant portion of COVID-19 patients have digestive symptoms, mostly at presentation. This should be taken into account in order to keep a high level of suspicion to reach an early diagnosis and setup infection control measures to control the transmission rate. This subgroup of patients appears to have a less severe disease course.


Subject(s)
COVID-19/physiopathology , Diarrhea/physiopathology , Vomiting/physiopathology , Abdominal Pain/epidemiology , Abdominal Pain/metabolism , Abdominal Pain/physiopathology , Adolescent , Adult , Age Distribution , Aged , Aged, 80 and over , Ageusia/epidemiology , Ageusia/metabolism , Ageusia/physiopathology , Alanine Transaminase/metabolism , Aspartate Aminotransferases/metabolism , C-Reactive Protein/metabolism , COVID-19/metabolism , Diarrhea/epidemiology , Diarrhea/metabolism , Female , Hemoglobins/metabolism , Humans , Male , Middle Aged , Nausea/epidemiology , Nausea/metabolism , Nausea/physiopathology , Portugal/epidemiology , Retrospective Studies , SARS-CoV-2 , Severity of Illness Index , Vomiting/epidemiology , Vomiting/metabolism , Young Adult
6.
Toxicology ; 452: 152718, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33581213

ABSTRACT

Type A trichothecene neosolaniol (NEO) is considered a potential risk to human and animal health by the European Food Safety Authority (EFSA). To date, available data do not allow making conclusions about the toxicological properties of this toxin. Trichothecenes have been previously demonstrated to induce emetic responses in mink, and this response has been associated with neurotransmitter peptide YY (PYY) and serotonin (5-hydroxytryptamine, 5-HT). The goal of this study was to compare emetic effects of NEO administered by intraperitoneal and oral routes and relate these effects to PYY and 5-HT. The effective doses resulting in emetic events in 50% of the animals following intraperitoneal and oral exposure to NEO were 0.4 and 0.09 mg/kg bw, respectively. This emetic response corresponded to elevated PYY and 5-HT levels. Blocking the neuropeptide Y2 receptor diminished emesis induction by PYY and NEO. The 5-HT3 receptor inhibitor granisetron completely restrained the induction of emesis by 5-HT and NEO. To summarize, our findings demonstrate that PYY and 5-HT play important roles in the NEO-induced emetic response.


Subject(s)
Emetics/toxicity , Receptors, Neuropeptide Y/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Trichothecenes/toxicity , Vomiting/chemically induced , Vomiting/metabolism , Animals , Dose-Response Relationship, Drug , Female , Mink , Receptors, Neuropeptide Y/antagonists & inhibitors , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Vomiting/prevention & control
7.
Curr Treat Options Oncol ; 22(2): 17, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33443705

ABSTRACT

OPINION STATEMENT: In cancer patients, the management of nausea and vomiting that is not directly related to treatment is challenging. Much current practice is based on expert opinion and anecdote. Fortunately, over recent years, a number of quality trials have been undertaken to strengthen the evidence base that guides the care of our patients with these distressing symptoms. Much is still unknown however. In this article, we present the latest literature that addresses some of the outstanding issues.


Subject(s)
Disease Susceptibility , Nausea/etiology , Nausea/therapy , Neoplasms/complications , Vomiting/etiology , Vomiting/therapy , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Antiemetics/pharmacology , Antiemetics/therapeutic use , Biomarkers , Disease Management , Drug Therapy, Combination , Humans , Intestinal Obstruction/etiology , Medical Marijuana/pharmacology , Medical Marijuana/therapeutic use , Molecular Targeted Therapy , Nausea/diagnosis , Nausea/metabolism , Prognosis , Serotonin Antagonists/pharmacology , Serotonin Antagonists/therapeutic use , Treatment Outcome , Vomiting/diagnosis , Vomiting/metabolism
8.
BMC Med Genet ; 21(1): 239, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33261563

ABSTRACT

BACKGROUND: Early-onset chronic diarrhoea often indicates a congenital disorder. Mutation in diacylglycerol o-acyltransferase 1 (DGAT1) has recently been linked to early-onset chronic diarrhoea. To date, only a few cases of DGAT1 deficiency have been reported. Diarrhoea in those cases was severe and developed in the neonatal period or within 2 months after birth. CASE PRESENTATION: Here, we report a female patient with DGAT1 mutations with delayed-onset chronic diarrhoea. The patient had vomiting, hypoalbuminemia, hypertriglyceridemia, and failure to thrive at early infancy. Her intractable chronic diarrhoea occurred until she was 8 months of age. A compound heterozygous DGAT1 mutation was found in the patient, which was first found in the Chinese population. Her symptoms and nutrition status improved after nutritional therapy, including a fat restriction diet. CONCLUSIONS: This case expanded our knowledge of the clinical features of patients with DGAT1 mutations. Intractable diarrhoea with delayed onset could also be a congenital disorder.


Subject(s)
Diacylglycerol O-Acyltransferase/genetics , Diarrhea/genetics , Failure to Thrive/genetics , Hypertriglyceridemia/genetics , Hypoalbuminemia/genetics , Mutation , Vomiting/genetics , Age of Onset , Base Sequence , Diacylglycerol O-Acyltransferase/deficiency , Diarrhea/diet therapy , Diarrhea/metabolism , Diarrhea/physiopathology , Diet, Fat-Restricted , Failure to Thrive/diet therapy , Failure to Thrive/metabolism , Failure to Thrive/physiopathology , Female , Gene Expression , Heterozygote , Humans , Hypertriglyceridemia/diet therapy , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/physiopathology , Hypoalbuminemia/diet therapy , Hypoalbuminemia/metabolism , Hypoalbuminemia/physiopathology , Infant , Severity of Illness Index , Vomiting/diet therapy , Vomiting/metabolism , Vomiting/physiopathology
9.
Pak J Pharm Sci ; 33(3): 1169-1172, 2020 May.
Article in English | MEDLINE | ID: mdl-33191244

ABSTRACT

Chemotherapy, radiotherapy, surgery and depression are the conditions that run in parallel fashions. All these conditions cause the release of an increased amount of serotonin in the body. Serotonin acts on these 5HT3 receptors and causes nausea and vomiting. Ondansetron acts by blocking serotonin from acting on the receptors and thus is useful in decreasing episodes of nausea and vomiting but when used concomitantly with SSRIs (selective serotonin reuptake inhibitors) as cancer patient also suffered from depression. This combination tends to decrease the efficacy of ondansetron. The present study was carried out to observe the modulatory role of ondansetron on ileal smooth muscle motility in vitro. Experiments were performed in four groups (n=6) and ileal smooth muscle activity was recorded on the power lab (USA). The effects of increasing concentrations of serotonin, ondansetron and paroxetine alone were observed. In the fourth group effects of paroxetine in the presence of fixed concentration (1ml) of ondansetron (10-6M) was observed. The maximum response obtained by serotonin served as a control for our study (100%). Paroxetine response on intestinal motility was completely blocked in the presence of ondansetron. Our findings hence, reinforce the hypothesis that paroxetine decreases the antiemetic activity of serotonin antagonist ondansetron, by super sensitization of serotonergic receptors resulting in an increased incidence of nausea and vomiting in cancer patient despite adequate antiemetic prophylaxis.


Subject(s)
Antiemetics/pharmacology , Gastrointestinal Motility/drug effects , Ileum/drug effects , Muscle, Smooth/drug effects , Ondansetron/pharmacology , Paroxetine/pharmacology , Receptors, Serotonin/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Antagonists/pharmacology , Animals , Drug Interactions , Female , Ileum/metabolism , Male , Muscle, Smooth/metabolism , Nausea/chemically induced , Nausea/metabolism , Nausea/physiopathology , Paroxetine/toxicity , Rabbits , Receptors, Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/toxicity , Vomiting/chemically induced , Vomiting/metabolism , Vomiting/physiopathology
10.
Eur J Pharmacol ; 888: 173528, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-32871177

ABSTRACT

GLP-1 receptor agonists are used for the treatment of type 2 diabetes but they may reduce appetite and cause nausea and emesis. We investigated if GLP-1 (7-36) amide can modulate glucose homoeostasis, emesis and feeding via an exendin (9-39)-sensitive mechanism in Suncus murinus. The effect of GLP-1 (7-36) amide on glucose homeostasis was examined using an intraperitoneal glucose tolerance test. In conscious fasted animals, food and water consumption and behavior were measured for 1 h following drug administration. c-Fos expression in the brain was measured using immunohistochemistry. GLP-1 (7-36) amide reduced blood glucose levels dose-dependently. Exendin (9-39) did not modify blood glucose levels but suppressed the glucose-lowering effect of GLP-1 (7-36) amide. GLP-1 (7-36) amide inhibited food and water intake, induced emesis and elevated c-Fos expression in the brainstem and hypothalamic nuclei in the brain. Exendin (9-39) antagonised the inhibition of food and water intake and emesis induced by GLP-1 (7-36) amide and the effects on c-Fos expression in the hypothalamus and brainstem, excepting for the bed nucleus of the stria terminalis. These data suggest that the action of GLP-1 (7-36) amide to modulate blood glucose, suppress food and water intake and induce emesis involve GLP-1 receptors in the hypothalamus and brainstem.


Subject(s)
Blood Glucose/metabolism , Feeding Behavior/physiology , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide-1 Receptor/metabolism , Homeostasis/physiology , Peptide Fragments/administration & dosage , Vomiting/metabolism , Animals , Blood Glucose/drug effects , Dose-Response Relationship, Drug , Feeding Behavior/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Homeostasis/drug effects , Injections, Intraventricular , Male , Shrews
11.
Pharmacol Res ; 161: 105124, 2020 11.
Article in English | MEDLINE | ID: mdl-32814171

ABSTRACT

Dopamine is a member of the catecholamine family and is associated with multiple physiological functions. Together with its five receptor subtypes, dopamine is closely linked to neurological disorders such as schizophrenia, Parkinson's disease, depression, attention deficit-hyperactivity, and restless leg syndrome. Unfortunately, several dopamine receptor-based agonists used to treat some of these diseases cause nausea and vomiting as impending side-effects. The high degree of cross interactions of dopamine receptor ligands with many other targets including G-protein coupled receptors, transporters, enzymes, and ion-channels, add to the complexity of discovering new targets for the treatment of nausea and vomiting. Using activation status of signaling cascades as mechanism-based biomarkers to foresee drug sensitivity combined with the development of dopamine receptor-based biased agonists may hold great promise and seems as the next step in drug development for the treatment of such multifactorial diseases. In this review, we update the present knowledge on dopamine and dopamine receptors and their potential roles in nausea and vomiting. The pre- and clinical evidence provided in this review supports the implication of both dopamine and dopamine receptor agonists in the incidence of emesis. Besides the conventional dopaminergic antiemetic drugs, potential novel antiemetic targeting emetic protein signaling cascades may offer superior selectivity profile and potency.


Subject(s)
Dopamine/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Vomiting/metabolism , Animals , Antiemetics/therapeutic use , Dopamine Agonists/adverse effects , Dopamine D2 Receptor Antagonists/therapeutic use , Humans , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D3/antagonists & inhibitors , Signal Transduction , Vomiting/chemically induced , Vomiting/physiopathology
12.
Dig Liver Dis ; 52(10): 1076-1079, 2020 10.
Article in English | MEDLINE | ID: mdl-32507692

ABSTRACT

Objective: To explore the clinical characteristics of Coronavirus Disease (COVID-19) patients with gastrointestinal symptoms. Methods: The clinical data of 164 COVID-19 patients with gastrointestinal symptoms were extracted and analysed retrospectively. Results: In total, 505 COVID-19 patients were divided into two groups: those with gastrointestinal symptoms (G group) and those without gastrointestinal symptoms (NG group). Common gastrointestinal symptoms included inappetence, diarrhoea, nausea, abdominal pain, and vomiting. Significantly higher proportions of patients with fever, dizziness, myalgia, and fatigue were noted in group G than in group NG. Compared with patients without fever, there was a significant difference between G group and NG group in moderate fever or above, while there was no significant difference between the two groups in low fever. The laboratory results showed that patients in the G group had significantly higher C-reactive protein, lactate dehydrogenase, and α-hydroxybutyrate dehydrogenase levels than those in the NG group. Moreover, the proportion of patients with severe pneumonia was significantly higher in the G group than in the NG group. Conclusion: In Wuhan, the proportion of COVID-19 patients who experience gastrointestinal symptoms is relatively high. Patients who experience gastrointestinal symptoms are more likely to suffer from severe pneumonia, which may help clinicians identify patients at high risk of COVID-19 and thus reduce the incidence of this condition.


Subject(s)
Abdominal Pain/physiopathology , Anorexia/physiopathology , Coronavirus Infections/physiopathology , Diarrhea/physiopathology , Nausea/physiopathology , Pneumonia, Viral/physiopathology , Vomiting/physiopathology , Abdominal Pain/etiology , Abdominal Pain/metabolism , Adult , Aged , Anorexia/etiology , Anorexia/metabolism , Betacoronavirus , C-Reactive Protein/metabolism , COVID-19 , Case-Control Studies , Coronavirus Infections/complications , Coronavirus Infections/metabolism , Diarrhea/etiology , Diarrhea/metabolism , Dizziness/etiology , Dizziness/physiopathology , Fatigue/etiology , Fatigue/physiopathology , Female , Fever/etiology , Fever/physiopathology , Humans , Hydroxybutyrate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/metabolism , Male , Middle Aged , Myalgia/etiology , Myalgia/physiopathology , Nausea/etiology , Nausea/metabolism , Pandemics , Pneumonia, Viral/complications , Pneumonia, Viral/metabolism , Retrospective Studies , SARS-CoV-2 , Severity of Illness Index , Vomiting/etiology , Vomiting/metabolism
13.
Neurocrit Care ; 32(1): 353-356, 2020 02.
Article in English | MEDLINE | ID: mdl-31342448

Subject(s)
Blood Flow Velocity , Brain Edema/physiopathology , Brain Injuries, Traumatic/physiopathology , Kidney Failure, Chronic/therapy , Middle Cerebral Artery/diagnostic imaging , Renal Dialysis/adverse effects , Status Epilepticus/physiopathology , Vascular Resistance , Aged , Blood-Brain Barrier/metabolism , Brain Contusion/complications , Brain Contusion/diagnostic imaging , Brain Contusion/metabolism , Brain Contusion/physiopathology , Brain Edema/diagnostic imaging , Brain Edema/etiology , Brain Edema/metabolism , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/metabolism , Cerebral Hemorrhage, Traumatic/complications , Cerebral Hemorrhage, Traumatic/diagnostic imaging , Cerebral Hemorrhage, Traumatic/metabolism , Cerebral Hemorrhage, Traumatic/physiopathology , Consciousness Disorders/etiology , Consciousness Disorders/metabolism , Consciousness Disorders/physiopathology , Headache/etiology , Headache/metabolism , Headache/physiopathology , Hematoma, Subdural, Acute/complications , Hematoma, Subdural, Acute/diagnostic imaging , Hematoma, Subdural, Acute/metabolism , Hematoma, Subdural, Acute/physiopathology , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/metabolism , Male , Middle Cerebral Artery/physiopathology , Monitoring, Physiologic , Nausea/etiology , Nausea/metabolism , Nausea/physiopathology , Pulsatile Flow , Status Epilepticus/etiology , Status Epilepticus/metabolism , Ultrasonography, Doppler, Transcranial , Vomiting/etiology , Vomiting/metabolism , Vomiting/physiopathology
14.
Support Care Cancer ; 28(7): 3279-3286, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31745695

ABSTRACT

BACKGROUND: Anorexia-cachexia syndrome (ACS) is a complex condition in advanced cancer patients, defined by disproportionate loss of skeletal muscle mass, and a lack or loss of appetite. This condition greatly lowers the quality of life and limits the treatment options. ACS is commonly associated with gastrointestinal symptoms such as nausea and vomiting. Ginger has been successful in treating these symptoms but has not yet been tested on patients with advanced cancer. Electrogastrography is a technology that allows the direct recording of the gastric myoelectrical activity (GMA). PURPOSE: The aim of this study is to (1) determine the effects of ginger on the GMA in these patients, (2) evaluate the subjective symptoms using 3 validated scales, and (3) correlate the level of inflammatory factors and ghrelin in this patient population. METHODS: Patients with ACS and advanced cancer were recruited from the Palliative Rehabilitation outpatient program at Elisabeth Bruyère Hospital. Patients were instructed to take a daily capsule of 1650 mg of ginger for 14 days and outcome measures were recorded at pre- and post-intervention, which included a blood test for analysis of CRP, albumin and ghrelin levels, 3 self-administered surveys (DSSI, PG-SGA, ESAS), patient-reported symptoms, and an EGG diagnosis. RESULTS: Fifteen patients with a median age of 58 and varying cancer diagnoses were enrolled. EGG diagnosis showed that 9 of the 15 patients had a direct improvement in their GMA, and all patients showed improvement in reported symptoms, most notably nausea, dysmotility- and reflux-like symptoms. There was no correlation found for ginger administration and inflammatory factors. CONCLUSION: These findings suggest that ginger may improve GMA as measured by EGG and may have a notable effect on symptom improvement.


Subject(s)
Anorexia/drug therapy , Cachexia/drug therapy , Neoplasms/metabolism , Zingiber officinale , Adult , Anorexia/metabolism , Cachexia/metabolism , Female , Ghrelin/metabolism , Humans , Male , Nausea/drug therapy , Nausea/metabolism , Phytotherapy/methods , Quality of Life , Vomiting/drug therapy , Vomiting/metabolism
15.
Invest New Drugs ; 37(1): 139-146, 2019 02.
Article in English | MEDLINE | ID: mdl-30032410

ABSTRACT

Rolapitant is a neurokinin-1 receptor antagonist that is approved in combination with other antiemetic agents in adults for the prevention of delayed nausea and vomiting (CINV) associated with initial and repeat courses of emetogenic cancer chemotherapy, including but not limited to highly emetogenic chemotherapy. Here, we assessed the absorption, metabolism, and excretion of 14C-labeled rolapitant in healthy male subjects. Rolapitant was administered as a single 180-mg oral dose containing approximately 100 µCi of total radioactivity, with plasma, urine, and fecal samples collected at defined intervals after dosing. Rolapitant had a large apparent volume of distribution, indicating that it is widely distributed into body tissues. Rolapitant was slowly metabolized and eliminated with a mean half-life of 186 h. Exposure to the major metabolite of rolapitant, C4-pyrrolidinyl hydroxylated rolapitant or M19, was approximately 50% of rolapitant exposure in plasma. Renal clearance was not a significant elimination route for rolapitant-related entities. Total radioactivity recovered in urine accounted for 14.2% of the dose, compared to 72.7% recovery in feces. Adverse events (AEs) were generally mild; there were no serious AEs, and no clinically significant changes in laboratory or electrocardiogram parameters were observed. The combination of rolapitant safety, its long half-life, extensive tissue distribution, and slow elimination via the hepatobiliary route (rather than renal excretion) suggest suitability that a single dose of rolapitant may provide protection against CINV beyond the first 24 h after chemotherapy administration.


Subject(s)
Antiemetics/administration & dosage , Nausea/metabolism , Neurokinin-1 Receptor Antagonists/administration & dosage , Spiro Compounds/administration & dosage , Vomiting/metabolism , Adult , Antiemetics/pharmacokinetics , Healthy Volunteers , Humans , Male , Middle Aged , Nausea/drug therapy , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Spiro Compounds/pharmacokinetics , Tissue Distribution , Vomiting/drug therapy
16.
Molecules ; 23(12)2018 Dec 03.
Article in English | MEDLINE | ID: mdl-30513973

ABSTRACT

Chemotherapy-induced nausea and vomiting (CINV) remain the most common and devastating side-effects associated with cancer chemotherapy. In recent decades, several lines of research emphasize the importance of 5-hydroxytryptamine3 (5-HT3; serotonin) receptors in the pathogenesis and treatment of CINV. 5-HT3 receptors are members of ligand-gated ion channels that mediate the rapid and transient membrane-depolarizing effect of 5-HT in the central and peripheral nervous system. These receptors play important roles in nausea and vomiting, as well as regulation of peristalsis and pain transmission. The development of antagonists for 5-HT3 receptor dramatically improved the treatment of CINV in cancer patients. In fact, the most common use of 5-HT3 receptor antagonists to date is the treatment of nausea and vomiting. In recent years, there has been an increasing tendency to use natural plant products as important therapeutic entities in the treatment of various diseases. In this article, we examined the results of earlier studies on the actions of natural compounds on the functional properties of 5-HT3 receptors. It is likely that these natural modulators of 5-HT3 receptors can be employed as lead structures for the synthesis of therapeutic agents for treating CINV in future clinical studies.


Subject(s)
Nausea/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Vomiting/metabolism , Allosteric Regulation , Antineoplastic Agents/adverse effects , Binding Sites , Cannabidiol/pharmacology , Zingiber officinale/chemistry , Humans , Nausea/chemically induced , Serotonin 5-HT3 Receptor Antagonists/chemistry , Terpenes/pharmacology , Vomiting/chemically induced
17.
Clin Nucl Med ; 43(10): e363-e365, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30004938

ABSTRACT

A 22-year-old woman with acute lymphoblastic leukemia underwent FDG PET/CT to evaluate possible extramedullary disease. The patient experienced severe nausea and vomiting due to ongoing chemotherapy. The image demonstrated increased FDG uptake in multiple muscle groups, including intercostal, bilateral external oblique, internal oblique, transverse abdominal muscles, and psoas major. One week after the patient stopped vomiting, a repeated PET/CT showed much less muscle uptake.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Muscles/metabolism , Positron Emission Tomography Computed Tomography , Vomiting/diagnostic imaging , Vomiting/metabolism , Biological Transport , Female , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Vomiting/chemically induced , Young Adult
18.
Eur J Pharmacol ; 834: 157-168, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-29966616

ABSTRACT

Ca2+ plays a major role in maintaining cellular homeostasis and regulates processes including apoptotic cell death and side-effects of cancer chemotherapy including vomiting. Currently we explored the emetic mechanisms of FPL64176, an L-type Ca2+ channel (LTCC) agonist with maximal emetogenic effect at its 10 mg/kg dose. FPL64176 evoked c-Fos immunoreactivity in shrew brainstem sections containing the vomit-associated nuclei, nucleus tractus solitarius (NTS) and dorsal motor nucleus of the vagus. FPL64176 also increased phosphorylation of proteins ERK1/2, PKCα/ßII and Akt in the brainstem. Moreover, their corresponding inhibitors (PD98059, GF 109203X and LY294002, respectively) reduced FPL64176-evoked vomiting. A 30 min subcutaneous (s.c.) pretreatment with the LTCC antagonist nifedipine (10 mg/kg) abolished FPL64176-elicited vomiting, c-Fos expression, and emetic effector phosphorylation. Ryanodine receptors (RyRs) and inositol trisphosphate receptors (IP3Rs) mediate intracellular Ca2+ release from the sarcoplasmic/endoplasmic reticulum. The RyR antagonist dantrolene (i.p.), or a combination of low doses of nifedipine and dantrolene, but not the IP3R antagonist 2-APB, significantly attenuated FPL64176-induced vomiting. The serotonin type 3 receptor (5-HT3R) antagonist palonosetron (s.c.), the neurokinin 1 receptor (NK1R) antagonist netupitant (i.p.) or a combination of non-effective doses of netupitant and palonosetron showed antiemetic potential against FPL64176-evoked vomiting. Serotonin (5-HT) and substance P immunostaining revealed FPL64176-induced emesis was accompanied by an increase in 5-HT but not SP-immunoreactivity in the dorsomedial subdivision of the NTS. These findings demonstrate that Ca2+ mobilization through LTCCs and RyRs, and subsequent emetic effector phosphorylation and 5-HT release play important roles in FPL64176-induced emesis which can be prevented by 5-HT3R and NK1R antagonists.


Subject(s)
Calcium Channels, L-Type/metabolism , Emetics/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Pyrroles/pharmacology , Signal Transduction/drug effects , Vomiting/chemically induced , Animals , Calcium/metabolism , Shrews , Vomiting/metabolism , Vomiting/pathology
19.
Nat Commun ; 9(1): 1178, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29563502

ABSTRACT

Hyperemesis gravidarum (HG), severe nausea and vomiting of pregnancy, occurs in 0.3-2% of pregnancies and is associated with maternal and fetal morbidity. The cause of HG remains unknown, but familial aggregation and results of twin studies suggest that understanding the genetic contribution is essential for comprehending the disease etiology. Here, we conduct a genome-wide association study (GWAS) for binary (HG) and ordinal (severity of nausea and vomiting) phenotypes of pregnancy complications. Two loci, chr19p13.11 and chr4q12, are genome-wide significant (p < 5 × 10-8) in both association scans and are replicated in an independent cohort. The genes implicated at these two loci are GDF15 and IGFBP7 respectively, both known to be involved in placentation, appetite, and cachexia. While proving the casual roles of GDF15 and IGFBP7 in nausea and vomiting of pregnancy requires further study, this GWAS provides insights into the genetic risk factors contributing to the disease.


Subject(s)
Growth Differentiation Factor 15/genetics , Hyperemesis Gravidarum/genetics , Insulin-Like Growth Factor Binding Proteins/genetics , Nausea/genetics , Placenta/metabolism , Pregnancy Complications/genetics , Vomiting/genetics , Adult , Appetite/genetics , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 4 , Cohort Studies , Female , Gene Expression , Genome, Human , Genome-Wide Association Study , Growth Differentiation Factor 15/metabolism , Humans , Hyperemesis Gravidarum/metabolism , Hyperemesis Gravidarum/physiopathology , Insulin-Like Growth Factor Binding Proteins/metabolism , Nausea/etiology , Nausea/metabolism , Nausea/physiopathology , Phenotype , Placenta/pathology , Pregnancy , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology , Quantitative Trait Loci , Risk Factors , Severity of Illness Index , Vomiting/metabolism , Vomiting/physiopathology
20.
Eur J Pharmacol ; 809: 20-31, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28501575

ABSTRACT

The introduction of second generation serotonin 5-HT3 receptor (5-HT3) antagonist palonosetron combined with long-acting substance P neurokinin NK1 receptor (NK1) antagonists (e.g. netupitant) has substantially improved antiemetic therapy against early- and delayed-phases of emesis caused by highly emetogenic chemotherapeutics such as cisplatin. However, the improved efficacy comes at a cost that many patients cannot afford. We introduce a new class of antiemetic, the antiasthmatic leukotriene CysLT1 receptor antagonist pranlukast for the suppression of cisplatin-evoked vomiting. Pranlukast (10mg/kg) by itself significantly reduced the mean frequency of vomits (70%) and fully protected least shrews from vomiting (46%) during the delayed-phase of cisplatin (10mg/kg)-evoked vomiting. Although, pranlukast tended to substantially reduce both the mean frequency of vomits and the number of shrews vomiting during the early-phase, these reductions failed to attain significance. When combined with a first (tropisetron)- or a second (palonosetron)-generation 5-HT3 receptor antagonist, pranlukast potentiated their antiemetic efficacy during both phases of vomiting. In addition, pranlukast by itself prevented several intracellular signal markers of cisplatin-evoked delayed-vomiting such as phosphorylation of ERK1/2 and PKA. When pranlukast was combined with either palonosetron or tropisetron, these combinations suppressed the evoked phosphorylation of: i) ERK1/2 during both acute- and delayed-phase, ii) PKCα/ß at the peak acute-phase, and iii) PKA at the peak delayed-phase. The current and our published findings suggest that overall behavioral and intracellular signaling effects of pranlukast via blockade of CysLT1 receptors generally appear to be similar to the NK1 receptor antagonist netupitant with some differences.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Chromones/pharmacology , Cisplatin/adverse effects , Intracellular Space/drug effects , Vomiting/chemically induced , Vomiting/prevention & control , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Synergism , Female , Intracellular Space/metabolism , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Protein Kinase C/metabolism , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Shrews , Vomiting/metabolism , Vomiting/pathology
SELECTION OF CITATIONS
SEARCH DETAIL