Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Aging Cell ; 22(4): e13787, 2023 04.
Article in English | MEDLINE | ID: mdl-36734122

ABSTRACT

Interventions for animal lifespan extension like caloric restriction (CR) have identified physiologic and biochemical pathways related to hunger and energy-sensing status as possible contributors, but mechanisms have not been fully elucidated. Prior studies using ghrelin agonists show greater food intake but no effect on lifespan in rodent models. This experiment in male C57BL/6J mice tested the influence of ghrelin agonism for perceived hunger, in the absence of CR, on longevity. Mice aged 4 weeks were allowed to acclimate for 2 weeks prior to being assigned (N = 60/group). Prior to lights off daily (12:12 cycle), animals were fed a ghrelin agonist pill (LY444711; Eli Lilly) or a placebo control (Ctrl) until death. Treatment (GhrAg) animals were pair-fed daily based on the group mean food intake consumed by Ctrl (ad libitum feeding) the prior week. Results indicate an increased lifespan effect (log-rank p = 0.0032) for GhrAg versus placebo Ctrl, which weighed significantly more than GhrAg (adjusted for baseline weight). Further studies are needed to determine the full scope of effects of this ghrelin agonist, either directly via increased ghrelin receptor signaling or indirectly via other hypothalamic, systemic, or tissue-specific mechanisms.


Subject(s)
Ghrelin , Longevity , Animals , Male , Mice , Caloric Restriction , Ghrelin/agonists , Mice, Inbred C57BL
2.
Biomolecules ; 12(12)2022 12 05.
Article in English | MEDLINE | ID: mdl-36551241

ABSTRACT

The growth hormone secretagogue receptor-1a (GHSR1a) is the endogenous receptor for ghrelin. Activation of GHSR1a participates in many physiological processes including energy homeostasis and eating behavior. Due to its transitory half-life, the efficacy of ghrelin treatment in patients is restricted; hence the development of new adjuvant therapy is an urgent need. This study aimed to establish a cell line stably expressing GHSR1a, which could be employed to screen potential ghrelin agonists from natural compounds. First, by means of lentiviral transduction, the genome of a human HEK293T cell was modified, and a cell platform stably overexpressing GHSR1a was successfully established. In this platform, GHSR1a was expressed as a fusion protein tagged with mCherry, which allowed the monitoring of the dynamic cellular distribution of GHSR1a by fluorescent microscopy. Subsequently, the authenticity of the GHSR1a mediated signaling was further characterized by using ghrelin and teaghrelin, two molecules known to stimulate GHSR1a. The results indicated that both ghrelin and teaghrelin readily activated GHSR1a mediated signaling pathways, presumably via increasing phosphorylation levels of ERK. The specific GHSR1a signaling was further validated by using SP-analog, an antagonist of GHSR1a as well as using a cell model with the knockdown expression of GHSR1a. Molecular modeling predicted that crocin might be a potential ghrelin agonist, and this prediction was further confirmed by the established platform.


Subject(s)
Carotenoids , Ghrelin , Receptors, Ghrelin , Humans , Ghrelin/agonists , HEK293 Cells , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Signal Transduction , Carotenoids/pharmacology
3.
J Med Chem ; 65(4): 3098-3118, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35157454

ABSTRACT

Ghrelin is an octanoylated peptide acting by the activation of the growth hormone secretagogue receptor, namely, GHS-R1a. The involvement of ghrelin in several physiological processes, including stimulation of food intake, gastric emptying, body energy balance, glucose homeostasis, reduction of insulin secretion, and lipogenesis validates the considerable interest in GHS-R1a as a promising target for the treatment of numerous disorders. Over the years, several GHS-R1a ligands have been identified and some of them have been extensively studied in clinical trials. The recently resolved structures of GHS-R1a bound to ghrelin or potent ligands have provided useful information for the design of new GHS-R1a drugs. This perspective is focused on the development of recent nonpeptide small molecules acting as GHS-R1a agonists, antagonists, and inverse agonists, bearing classical or new molecular scaffolds, as well as on radiolabeled GHS-R1a ligands developed for imaging. Moreover, the pharmacological effects of the most studied ligands have been discussed.


Subject(s)
Drug Design , Ghrelin/agonists , Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/agonists , Receptors, Ghrelin/antagonists & inhibitors , Small Molecule Libraries , Animals , Ghrelin/genetics , Homeostasis , Humans , Ligands , Receptors, Ghrelin/genetics
4.
Nat Commun ; 13(1): 300, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35027551

ABSTRACT

Much effort has been invested in the investigation of the structural basis of G protein-coupled receptors (GPCRs) activation. Inverse agonists, which can inhibit GPCRs with constitutive activity, are considered useful therapeutic agents, but the molecular mechanism of such ligands remains insufficiently understood. Here, we report a crystal structure of the ghrelin receptor bound to the inverse agonist PF-05190457 and a cryo-electron microscopy structure of the active ghrelin receptor-Go complex bound to the endogenous agonist ghrelin. Our structures reveal a distinct binding mode of the inverse agonist PF-05190457 in the ghrelin receptor, different from the binding mode of agonists and neutral antagonists. Combining the structural comparisons and cellular function assays, we find that a polar network and a notable hydrophobic cluster are required for receptor activation and constitutive activity. Together, our study provides insights into the detailed mechanism of ghrelin receptor binding to agonists and inverse agonists, and paves the way to design specific ligands targeting ghrelin receptors.


Subject(s)
Receptors, Ghrelin/agonists , Receptors, Ghrelin/antagonists & inhibitors , Cryoelectron Microscopy , Crystallography, X-Ray , Ghrelin/agonists , Ghrelin/genetics , Humans , Ligands , Mutation , Protein Binding , Receptors, Ghrelin/genetics
5.
Nutrients ; 12(12)2020 Nov 28.
Article in English | MEDLINE | ID: mdl-33260513

ABSTRACT

The prevalence and incidence of Parkinson's disease (PD), an age-related neurodegenerative disease, are higher among elderly people. Independent of etiology, dysfunction and loss of dopaminergic neurons are common pathophysiological changes in PD patients with impaired motor and non-motor function. Currently, preventive or therapeutic treatment for combating PD is limited. The ghrelin axis and ghrelin receptor have been implicated in the preservation of dopaminergic neurons and have potential implications in PD treatment. Teaghrelin, a compound originating from Chin-Shin Oolong tea, exhibits ghrelin agonist activity. In this study, the neuroprotective potential of teaghrelin against PD was explored in a cell model in which human neuroblastoma SH-SY5Y cells were treated with the mitochondrial toxin 1-methyl-4-phenylpyridinium (MPP+). Upon MPP+ exposure, SH-SY5Y cells exhibited decreased mitochondrial complex I activity and apoptotic cell death. Teaghrelin activated AMP-activated protein kinase (AMPK)/sirtuin 1(SIRT1)/peroxisome proliferator-activated receptor gamma (PPARγ) coactivator-1α (PGC-1α) and extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathways to antagonize MPP+-induced cell death. Herein, we propose that teaghrelin is a potential candidate for the therapeutic treatment of PD.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Adenylate Kinase/metabolism , Camellia sinensis/chemistry , Cell Survival/drug effects , Ghrelin/agonists , Sirtuin 1/metabolism , Adenylate Kinase/genetics , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/genetics
6.
Gastroenterology ; 156(6): 1650-1660, 2019 05.
Article in English | MEDLINE | ID: mdl-30711628

ABSTRACT

BACKGROUND & AIMS: Studies have reported a lack of association between improvements in gastric emptying (GE) and upper gastrointestinal (UGI) symptoms with promotility drugs. However, GE test methods were suboptimal in some studies. We assessed improvements in GE and UGI symptoms in patients given promotility agents in studies with optimal or moderate test methods (scintigraphy or breath test, solid meal, >2 hours duration) compared to studies with suboptimal GE test methods. METHODS: With an expert librarian, we completed an extensive search of publications in the Ovid MEDLINE (1946 to present), EMBASE (1988 to January 2018), and EBM Reviews Cochrane Central Register of Controlled Trials, without restrictions on language or year. Two independent reviewers evaluated the following inclusion criteria: randomized, blinded, parallel, or crossover trials of 5HT4 agonists, D2 receptor antagonist, or ghrelin agonists; trials that measured change in GE (T1/2) or composite UGI symptoms; trials of patients with functional dyspepsia and gastroparesis; and trials of GE test methods. Standardized mean differences (units expressed as SD) were used to standardize symptom assessments that were not uniform across studies. Random effects model was used to analyze data and meta-regression was used to evaluate the association between change in GE and UGI symptoms. RESULTS: Of 899 studies considered, 22 studies assessed change in GE; 23 evaluated UGI symptoms; and 14 evaluated GE and UGI symptoms. Promotility agents significantly accelerated GE (T1/2) in all studies (mean reduction in T1/2, 16.3 minutes; 95% confidence interval, -22.1 to -10.6 minutes) and in studies that used optimal GE test methods (mean reduction in T1/2, 23.6 minutes; 95% confidence interval, -32.3 to -14.9 minutes). Promotility agents also significantly reduced UGI symptoms (mean reduction, 0.25 SD; 95% confidence interval, -0.37 to -0.13 SD). Meta-regression found no significant association between change in GE and UGI symptoms. However, when only studies with optimal GE test methods were evaluated, there was a significant positive association between improvement in GE and UGI symptoms (P = .02). CONCLUSIONS: In a meta-analysis of published trials, we found promotility agents to significantly accelerate GE (when optimal test methods were used) and to produce significant improvements in UGI symptoms.


Subject(s)
Dopamine D2 Receptor Antagonists/pharmacology , Gastric Emptying/drug effects , Ghrelin/agonists , Serotonin 5-HT4 Receptor Agonists/pharmacology , Breath Tests , Cisapride/pharmacology , Domperidone/pharmacology , Dopamine D2 Receptor Antagonists/therapeutic use , Dyspepsia/drug therapy , Gastroparesis/drug therapy , Ghrelin/pharmacology , Humans , Macrocyclic Compounds/pharmacology , Oligopeptides/pharmacology , Radionuclide Imaging , Randomized Controlled Trials as Topic , Serotonin 5-HT4 Receptor Agonists/therapeutic use , Symptom Assessment
7.
J Pharmacol Exp Ther ; 368(1): 116-124, 2019 01.
Article in English | MEDLINE | ID: mdl-30377215

ABSTRACT

The gastrointestinal (GI) prokinetic effects of ghrelin occur through direct peripheral effects on ghrelin receptors within the enteric nervous system and via the ghrelin receptor on the vagus nerve, which activate a centrally mediated mechanism. However, the relative contribution of peripheral versus central effects to the overall prokinetic effect of ghrelin agonists requires further investigation. Here, we investigated the central versus peripheral prokinetic effect of ghrelin by using two novel ghrelin agonists: HM01 (N'-[(1S)-1-(2,3-dichloro-4-methoxyphenyl)ethyl]-N-methyl-N-[1,3,3-trimethyl-(4R)-piperidyl]-urea HCL) with high brain penetration compared with HM02 (N'-[(1S)-1-(2,3-dichloro-4-methoxyphenyl)ethyl]-N-hydroxy-N-(1-methyl-4-piperidinyl)-urea), a more peripherally acting ghrelin agonist. The pharmacokinetic profiles of both ghrelin agonists were evaluated after intravenous and oral administration in rats. The efficacy of HM01 and HM02 was assessed in a rat model of postoperative ileus (POI) induced by abdominal surgery and in a rodent defecation assay. Pharmacokinetic results in our models confirmed that HM01, but not HM02, was a brain-penetrant ghrelin agonist. Administration of either HM01 or HM02 reversed the delayed upper and lower gastrointestinal transit induced by abdominal surgery to levels resembling the non-POI controls. In the defecation test, HM01, but not HM02, significantly increased the weight of fecal pellets. Our findings suggest that, in a rodent model of POI, synthetic ghrelin agonists stimulate GI transit through a peripheral site of action. However, in the defecation assay, our data suggest that a ghrelin-mediated mechanism is located at a central site. Taken together, a ghrelin agonist with both central and peripheral prokinetic activity may show therapeutic potential to treat delayed GI transit disorders.


Subject(s)
Biomimetic Materials/administration & dosage , Gastrointestinal Transit/physiology , Ghrelin/administration & dosage , Ghrelin/agonists , Piperidines/administration & dosage , Administration, Intravenous , Administration, Oral , Animals , Dose-Response Relationship, Drug , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gastrointestinal Transit/drug effects , Male , Rats , Rats, Sprague-Dawley
8.
Eur J Pharmacol ; 840: 89-103, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30268665

ABSTRACT

Chemotherapy-Induced Peripheral Neurotoxicity (CIPN) is often dose-limiting and impacts life quality and survival of cancer patients. Ghrelin agonists have neuroprotectant effects and may have a role in treating or preventing CIPN. We evaluated the CNS-penetrant ghrelin agonist HM01 in three experimental models of CIPN at doses of 3-30 mg/kg p.o. daily monitoring orexigenic properties, nerve conduction, mechanical allodynia, and intra-epidermal nerve fiber density (IENFD). In a cisplatin-based study, rats were dosed daily for 3 days (0.5 mg/kg i.p.) + HM01. Cisplatin treatment induced mechanical hypersensitivity which was significantly reduced by HM01. In a second study, oxaliplatin was administered to mice (6 mg/kg i.p. 3 times/week for 4 weeks) resulting in significant digital nerve conduction velocity (NCV) deficits and reduction of IENFD. Concurrent HM01 dose dependently prevented the decline in NCV and attenuated the reduction in IENFD. Pharmacokinetic studies showed HM01 accumulation in the dorsal root ganglia and sciatic nerves which reached concentrations > 10 fold that of plasma. In a third model, HM01 was tested in preventive and therapeutic paradigms in a bortezomib-based rat model (0.2 mg/kg i.v., 3 times/week for 8 weeks). In the preventive setting, HM01 blocked bortezomib-induced hyperalgesia and IENFD reduction at all doses tested. In the therapeutic setting, significant effect was observed, but only at the highest dose. Altogether, the robust peripheral nervous system penetration of HM01 and its ability to improve multiple oxaliplatin-, cisplatin-, and bortezomib-induced neurotoxicities suggest that HM01 may be a useful neuroprotective adjuvant for CIPN.


Subject(s)
Antineoplastic Agents/adverse effects , Benzene Derivatives/pharmacology , Ghrelin/agonists , Nervous System/drug effects , Neuroprotective Agents/pharmacology , Animals , Body Weight/drug effects , Cisplatin/adverse effects , Dose-Response Relationship, Drug , Eating/drug effects , Female , Male , Mice , Neural Conduction/drug effects , Piperidines , Rats
9.
Curr Opin Support Palliat Care ; 12(4): 439-444, 2018 12.
Article in English | MEDLINE | ID: mdl-30138131

ABSTRACT

PURPOSE OF REVIEW: Randomized clinical trials of cancer cachexia interventions are based on the premise that an increase in the muscle mass of patients is associated with consequent improvements in muscle function, and ultimately, quality of life. However, recent trials that have succeeded in demonstrating increases in lean body mass have been unable to show associated increases in patient physical function. In this review, we examine the potential causes for this lack of association between muscle mass and function in cancer cachexia, paying particular attention to those factors that may be at play when using body composition analysis techniques involving cross-sectional imaging. Moreover, we propose a new population-specific model for the relationship between muscle mass and physical function in patients with cancer cachexia. RECENT FINDINGS: The ROMANA 1 and 2 trials of anamorelin (a novel ghrelin agonist) and the POWER 1 and 2 trials of enobosarm (a selective androgen receptor modulator) were able to demonstrate improvements in patient lean body mass, but not the functional co-primary endpoints of handgrip strength and stair climb power, respectively. We report similar confirmatory findings in other studies, and describe potential reasons for these observations. SUMMARY: The relationship between muscle mass and muscle function is complex and unlikely to be linear. Furthermore, the relationship is influenced by the techniques used to assess nutritional endpoints [e.g. computed tomography (CT)]; the nature of the chosen physical function outcome measures; and the sex and severity of the recruited cachectic patients. Such factors need to be considered when designing intervention trials for cancer cachexia with functional endpoints.


Subject(s)
Cachexia/etiology , Cachexia/physiopathology , Muscle Strength/physiology , Muscle, Skeletal/physiopathology , Neoplasms/complications , Body Composition/physiology , Cachexia/drug therapy , Ghrelin/agonists , Muscle, Skeletal/diagnostic imaging , Nutrition Assessment , Quality of Life , Randomized Controlled Trials as Topic , Severity of Illness Index , Tomography, X-Ray Computed
10.
Int J Mol Sci ; 19(6)2018 06 18.
Article in English | MEDLINE | ID: mdl-29912176

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive deficits, neuroinflammation, and neuronal death. The primary pathogenic cause is believed to be the accumulation of pathogenic amyloid beta (Aß) assemblies in the brain. Ghrelin, which is a peptide hormone predominantly secreted from the stomach, is an endogenous ligand for the growth hormone secretagogue-receptor type 1a (GHS-R1a). MK-0677 is a ghrelin agonist that potently stimulates the GHS-R1a ghrelin receptor. Interestingly, previous studies have shown that ghrelin improves cognitive impairments and attenuates neuronal death and neuroinflammation in several neurological disorders. However, it is unknown whether MK-0677 can affect Aß accumulation or Aß-mediated pathology in the brains of patients with AD. Therefore, we examined the effects of MK-0677 administration on AD-related pathology in 5XFAD mice, an Aß-overexpressing transgenic mouse model of AD. MK-0677 was intraperitoneally administered to three-month-old 5XFAD mice. To visualize Aß accumulation, neuroinflammation, and neurodegeneration, thioflavin-S staining and immunostaining with antibodies against Aß (4G8), ionized calcium-binding adaptor molecule 1 (Iba-1), glial fibrillary acidic protein (GFAP), neuronal nuclear antigen (NeuN), and synaptophysin were conducted in the neocortex of 5XFAD and wild-type mice, and to evaluate changes of phosphorylated cyclic adenosine monophosphate (cAMP) response element binding protein (pCREB) levels, immunostaining with antibody against pCREB was performed in dentate gyrus of the hippocampus of 5XFAD and wild-type mice. The histological analyses indicated that MK-0677-treated 5XFAD mice showed reduced Aß deposition, gliosis, and neuronal and synaptic loss in the deep cortical layers, and inhibited the decrement of pCREB levels in dentate gyrus of the hippocampus compared to vehicle-treated 5XFAD mice. Our results showed that activation of the ghrelin receptor with MK-0677 inhibited the Aß burden, neuroinflammation, and neurodegeneration, which suggested that MK-0677 might have potential as a treatment of the early phase of AD.


Subject(s)
Alzheimer Disease/drug therapy , Ghrelin/agonists , Indoles/therapeutic use , Neuroprotective Agents/therapeutic use , Spiro Compounds/therapeutic use , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Ghrelin/metabolism , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Neocortex/drug effects , Neocortex/metabolism , Neuroprotective Agents/pharmacology , Spiro Compounds/pharmacology
11.
Int J Pharm ; 536(1): 63-72, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29175643

ABSTRACT

There is an impetus to provide appropriate sustained release oral delivery vehicles to protect biofunctional peptide loads from gastric degradation in vivo. This study describes the generation of a high load capacity pellet formulation for sustained release of a freely water-soluble dairy-derived hydrolysate, FHI-2571. The activity of this novel peptidic ghrelin receptor agonist is reported using in vitro calcium mobilization assays. Conventional extrusion spheronization was then used to prepare peptide-loaded pellets which were subsequently coated with ethylcellulose (EC) film coats using a fluid bed coating system in bottom spray (Wurster) mode. Aqueous-based EC coating dispersions produced mechanically brittle coats which fractured due to osmotic pressure build-up within pellets in simulated media. In contrast, an ethanolic-based EC coating solution provided robust, near zero-order release in both USP Type 1 and Type 4 dissolution studies. Interestingly, the functionality of aqueous-based EC film coats was restored by first layering pellets with a methacrylic acid copolymer (MA) subcoat, thereby hindering pellet core swelling in acidic media. Broadband Acoustic Resonance Dissolution Spectroscopy (BARDS) was utilised as a complementary technique to confirm the results seen in USP dissolution studies. Retention of activity of the ghrelinergic peptide hydrolysate in the final encapsulated product was confirmed as being greater than 80%. The described pellet formulation is amenable to oral dosing in small animal studies in order to assess in vivo efficacy of the whey-derived ghrelinergic hydrolysate. In more general terms, it is also suitable as a delivery vehicle for peptide-based bioactives to special population groups e.g paediatric and geriatric.


Subject(s)
Delayed-Action Preparations/chemistry , Ghrelin/agonists , Peptides/administration & dosage , Peptides/antagonists & inhibitors , Administration, Oral , Animals , Cellulose/analogs & derivatives , Cellulose/chemistry , Chemistry, Pharmaceutical/methods , Drug Delivery Systems/methods , Excipients/chemistry , HEK293 Cells , Humans , Polymers/chemistry , Solubility/drug effects
12.
Endocr Pract ; 23(9): 1140-1149, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28704095

ABSTRACT

Sarcopenia is defined as low muscle function (walking speed or grip strength) in the presence of low muscle mass. A simple screening test-the SARC-F-is available to identify persons with sarcopenia. The major endocrine causes of sarcopenia are diabetes mellitus and male hypogonadism. Other causes are decreased physical activity, loss of motor neuron units, weight loss, inflammatory cytokines, reduced blood flow to muscles, very low 25(OH) vitamin D levels, and decreased growth hormone and insulin-like growth factor 1. Treatment for sarcopenia includes resistance and aerobic exercise, leucine-enriched essential amino acids, and vitamin D. In hypogonadal males, testosterone improves muscle mass, strength, and function. Selective androgen receptor molecules and anti-myostatin activin II receptor molecules are under development as possible treatments for sarcopenia. ABBREVIATIONS: COPD = chronic obstructive pulmonary disease DHEA = dehydroepiandrosterone IGF-1 = insulin-like growth factor 1 GH = growth hormone mTOR = mammalian target of rapamycin SARM = selective androgen receptor molecule.


Subject(s)
Endocrine System Diseases/complications , Sarcopenia/etiology , Diabetes Complications/etiology , Ghrelin/agonists , Humans , Hypogonadism/complications , Sarcopenia/therapy , Testosterone/blood , Testosterone/therapeutic use
13.
Biochem J ; 474(16): 2663-2678, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28751550

ABSTRACT

Anorexia and metabolic alterations are the main components of the cachectic syndrome. Glucose intolerance, fat depletion, muscle protein catabolism and other alterations are involved in the development of cancer cachexia, a multi-organ syndrome. Nutritional approach strategies are not satisfactory in reversing the cachectic syndrome. The aim of the present review is to deal with the recent therapeutic targeted approaches that have been designed to fight and counteract wasting in cancer patients. Indeed, some promising targeted therapeutic approaches include ghrelin agonists, selective androgen receptor agonists, ß-blockers and antimyostatin peptides. However, a multi-targeted approach seems absolutely essential to treat patients affected by cancer cachexia. This approach should not only involve combinations of drugs but also nutrition and an adequate program of physical exercise, factors that may lead to a synergy, essential to overcome the syndrome. This may efficiently reverse the metabolic changes described above and, at the same time, ameliorate the anorexia. Defining this therapeutic combination of drugs/nutrients/exercise is an exciting project that will stimulate many scientific efforts. Other aspects that will, no doubt, be very important for successful treatment of cancer wasting will be an optimized design of future clinical trials, together with a protocol for staging cancer patients in relation to their degree of cachexia. This will permit that nutritional/metabolic/pharmacological support can be started early in the course of the disease, before severe weight loss occurs. Indeed, timing is crucial and has to be taken very seriously when applying the therapeutic approach.


Subject(s)
Cachexia/therapy , Neoplasms/therapy , Adrenergic beta-Antagonists/therapeutic use , Androgen Receptor Antagonists/therapeutic use , Anorexia/metabolism , Anorexia/pathology , Anorexia/therapy , Cachexia/metabolism , Cachexia/pathology , Diet Therapy/methods , Exercise Therapy/methods , Ghrelin/agonists , Humans , Myostatin/antagonists & inhibitors , Myostatin/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Peptides/therapeutic use
14.
Eur J Pharmacol ; 811: 30-37, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28529141

ABSTRACT

Cancer cachexia is a progressive wasting syndrome characterized by anorexia and weight loss, specifically muscle wasting and fat depletion. There is no therapeutic agent for treatment of this syndrome. We investigated the anti-cachexia effects of Z-505 hydrochloride (Z-505), a new oral growth hormone secretagogue receptor 1a (GHSR1a) agonist, using a mouse model of cancer cachexia. We performed a calcium flux assay in Chinese hamster ovary (CHO-K1) cells stably expressing human GHSR1a to quantify the agonistic activity of Z-505. In Colon 26 tumor-bearing mice, Z-505 (300mg/kg, p.o., twice daily) was administered for 7 days to assess its anti-cachexia effects. Body weight and food intake were monitored during the period, and the skeletal muscle and epididymal fat weights were measured. Serum levels of insulin, insulin-like growth factor 1 (IGF-1), interleukin-6 (IL-6), and corticosterone were measured to confirm the mechanism of the anti-cachexia action of Z-505. Z-505 showed strong agonistic activity similar to that of human ghrelin, with a half maximal effective concentration (EC50) value of 0.45nM. Z-505 treatment significantly increased food intake and inhibited the progression of weight loss. Z-505 also significantly attenuated muscle wasting and fat loss, and increased circulating levels of anabolic factors such as insulin and IGF-1, but not catabolic factors such as IL-6 and corticosterone. These findings suggest that Z-505 might be effective in the treatment of cachexia via the increased anabolic hormone levels stimulated by the activation of the ghrelin receptor, GHSR1a.


Subject(s)
Cachexia/drug therapy , Cachexia/metabolism , Colonic Neoplasms/complications , Ghrelin/agonists , Hormones/metabolism , Quinolines/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/pathology , Administration, Oral , Animals , BALB 3T3 Cells , Body Weight/drug effects , CHO Cells , Cachexia/complications , Cachexia/physiopathology , Cell Line, Tumor , Cricetulus , Disease Models, Animal , Disease Progression , Eating/drug effects , Ghrelin/metabolism , Humans , Male , Mice , Quinolines/administration & dosage , Quinolines/metabolism , Quinolines/therapeutic use , Rats , Receptors, Ghrelin/metabolism
15.
Diabetes Obes Metab ; 19(9): 1276-1288, 2017 09.
Article in English | MEDLINE | ID: mdl-28345814

ABSTRACT

AIMS: Dapagliflozin and exenatide reduce body weight by differing mechanisms. Dual therapy with these agents reduces body weight, adipose tissue volume, glycaemia and systolic blood pressure (SBP) over 24 weeks. Here, we examined these effects over 1 year in obese adults without diabetes. MATERIALS AND METHODS: Obese adults without diabetes (N = 50; aged 18-70 years; body mass index, 30-45 kg/m2 ) were initially randomized to double-blind oral dapagliflozin 10 mg once daily plus subcutaneous long-acting exenatide 2 mg once weekly or to placebo. They entered an open-label extension from 24 to 52 weeks during which all participants received active treatment. RESULTS: Of the original 25 dapagliflozin + exenatide-treated and 25 placebo-treated participants, respectively, 21 (84%) and 17 (68%) entered the open-label period and 16 (64%) and 17 (68%) completed 52 weeks of treatment. At baseline, mean body weight was 104.6 kg, and 73.5% of participants had prediabetes (impaired fasting glucose or impaired glucose tolerance). Reductions with dapagliflozin + exenatide at 24 weeks were sustained at 52 weeks, respectively, for body weight (-4.5 and -5.7 kg), total adipose tissue volume (-3.8 and -5.3 L), proportion with prediabetes (34.8% and 35.3%), and SBP (-9.8 and -12.0 mm Hg). Effects on body weight, SBP and glycaemia at 52 weeks with placebo → dapagliflozin + exenatide were similar to those observed with continuation of dapagliflozin + exenatide. Nausea and injection-site reactions were more frequent with dapagliflozin + exenatide than with placebo and diminished over time. Safety and tolerability were similar to that in previous diabetes trials with these agents. No clear difference in adverse event-related withdrawals between placebo and active treatment periods was observed. CONCLUSIONS: Dapagliflozin + exenatide dual therapy produced sustained reductions in body weight, prediabetes and SBP over 52 weeks and was well tolerated in obese adults without diabetes.


Subject(s)
Anti-Obesity Agents/therapeutic use , Benzhydryl Compounds/therapeutic use , Ghrelin/agonists , Glucosides/therapeutic use , Membrane Transport Modulators/therapeutic use , Obesity/drug therapy , Peptides/therapeutic use , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Venoms/therapeutic use , Adiposity/drug effects , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/adverse effects , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/adverse effects , Body Mass Index , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination/adverse effects , Exenatide , Female , Follow-Up Studies , Ghrelin/metabolism , Glucosides/administration & dosage , Glucosides/adverse effects , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/therapeutic use , Male , Membrane Transport Modulators/administration & dosage , Membrane Transport Modulators/adverse effects , Middle Aged , Obesity/metabolism , Obesity/physiopathology , Peptides/administration & dosage , Peptides/adverse effects , Prediabetic State/epidemiology , Prediabetic State/etiology , Prediabetic State/prevention & control , Proof of Concept Study , Risk Factors , Sodium-Glucose Transport Proteins/metabolism , Sweden/epidemiology , Venoms/administration & dosage , Venoms/adverse effects , Weight Loss/drug effects
16.
Article in English | MEDLINE | ID: mdl-28089858

ABSTRACT

Ghrelin and nesfatin-1 are two peptidyl hormones primarily involved in food intake regulation. We previously reported that the amount of dietary carbohydrates, protein and lipids modulates the expression of these peptides in goldfish in vivo. In the present work, we aimed to characterize the effects of single nutrients on ghrelin and nesfatin-1 in the intestine and hepatopancreas. First, immunolocalization of ghrelin and NUCB2/nesfatin-1 in goldfish hepatopancreas cells was studied by immunohistochemistry. Second, the effects of 2 and 4hour-long exposures of cultured intestine and hepatopancreas sections to glucose, l-tryptophan, oleic acid, linolenic acid (LNA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on ghrelin and nesfatin-1 gene and protein expression were studied. Co-localization of ghrelin and NUCB2/nesfatin-1 in the cytoplasm of goldfish hepatocytes was found. Exposure to glucose led to an upregulation of preproghrelin and a downregulation of nucb2/nesfatin-1 in the intestine. l-Tryptophan mainly decreased the expression of both peptides in the intestine and hepatopancreas. Fatty acids, in general, downregulated NUCB2/nesfatin-1 in the intestine, but only the longer and highly unsaturated fatty acids inhibited preproghrelin. EPA exposure led to a decrease in preproghrelin, and an increase in nucb2/nesfatin-1 expression in hepatopancreas after 2h. These results show that macronutrients exert a dose- and time-dependent, direct regulation of ghrelin and nesfatin-1 in the intestine and hepatopancreas, and suggest a role for these hormones in the digestive process and nutrient metabolism.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Ghrelin/metabolism , Goldfish/physiology , Hepatopancreas/metabolism , Intestinal Mucosa/metabolism , Nerve Tissue Proteins/metabolism , Animals , Calcium-Binding Proteins/agonists , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cytoplasm/metabolism , DNA-Binding Proteins/agonists , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Fatty Acids, Nonesterified/metabolism , Fish Proteins/agonists , Fish Proteins/antagonists & inhibitors , Fish Proteins/genetics , Fish Proteins/metabolism , Ghrelin/agonists , Ghrelin/antagonists & inhibitors , Ghrelin/genetics , Glucose/metabolism , Hepatopancreas/cytology , Immunohistochemistry/veterinary , Intestinal Mucosa/cytology , Intestines/cytology , Kinetics , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nucleobindins , Organ Specificity , Protein Precursors/agonists , Protein Precursors/antagonists & inhibitors , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Transport , RNA, Messenger/metabolism , Tissue Culture Techniques/veterinary , Tryptophan/metabolism
17.
J Biosci ; 42(3): 439-448, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29358557

ABSTRACT

Diabetes patients associated with liver disease carry a significant risk of morbidity and mortality. Cinnamon has been reported to reduce fructose-induced oxidative stress in the rat liver. However, the mechanism by which cinnamon protects the liver in a high-saccharide environment remains to be investigated. HepG2 cells were cultured with 30 mM D-ribose to mimic the high-oxidative-stress environment, typical of a liver in a diabetic patient. Three different chemical types of C. osmophloeum ethanol extracts (CEEs) were added in HepG2 culture media and the administration of all three CEEs protected HepG2 cells from D-ribose damage and increased cell survival by approximately 20 percent. Exclusively, the transcript variant 1 of the ghrelin gene, but not variant 3, was 2-3 times induced by the addition of these CEEs. Moreover, the mRNAs of ghrelin processing enzyme, furin, and mboat4 were detected in HepG2 cells. The ghrelin hormones in the culture media were increased 4-9 times by the addition of CEEs. The protective effects of ghrelin on HepG2 cells in D-ribose environment were further confirmed by recombinant ghrelin transfection. We conclude that the CEEs induce ghrelin gene expression and protect HepG2 cells from D-ribose-induced oxidative damage through ghrelin signalling.


Subject(s)
Antioxidants/pharmacology , Cinnamomum/chemistry , Ghrelin/agonists , Plant Extracts/pharmacology , RNA, Messenger/agonists , Acyltransferases/genetics , Acyltransferases/metabolism , Antioxidants/chemistry , Cell Survival/drug effects , Ethanol/chemistry , Furin/genetics , Furin/metabolism , Gene Expression Regulation , Ghrelin/genetics , Ghrelin/metabolism , Hep G2 Cells , Humans , Oxidative Stress/drug effects , Plant Extracts/chemistry , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribose/antagonists & inhibitors , Ribose/pharmacology , Signal Transduction , Solvents/chemistry
18.
Expert Opin Biol Ther ; 16(10): 1239-44, 2016 10.
Article in English | MEDLINE | ID: mdl-27382931

ABSTRACT

INTRODUCTION: Cachexia and sarcopenia are conditions phenotypically characterized by muscle loss and represent a factor of poor prognosis, increasing patients' morbidity and mortality. Cachectic and sarcopenic patients often suffer from low quality of life, presenting lower muscle strength and appetite loss, which makes research on novel treatment strategies to ameliorate clinical response including patient's symptoms, the objective of scientific interest. AREAS COVERED: This article covers recent developments in the area of cachexia and sarcopenia treatment and therapeutic interventions, targeting central nervous system involvement, key inflammatory and muscle-specific metabolic pathways. EXPERT OPINION: A number of promising agents have being evaluated, such as enobosarm, a selected androgen receptor modulator, and anamorelin, a ghrelin agonist which have been recently studied in phase III trials. These and other agents (i.e., infliximab, tocilizumab, MABp1, bimagrumab) have shown significant impact on reversal of skeletal muscle loss, but limited effect on physical function. In the last few years advancement in the number and type of potential treatments for cachexia and sarcopenia have been obtained and we have now available more data on measurable effects of several drugs on patients' nutritional and metabolic parameters and outcomes.


Subject(s)
Anilides/therapeutic use , Cachexia/drug therapy , Ghrelin/agonists , Sarcopenia/drug therapy , Anilides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Cachexia/metabolism , Ghrelin/therapeutic use , Humans , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Quality of Life , Receptors, Androgen/metabolism , Sarcopenia/metabolism
19.
Curr Pharm Des ; 21(25): 3596-605, 2015.
Article in English | MEDLINE | ID: mdl-26166609

ABSTRACT

Metabolic homeostasis requires a tight balance between energy intake and energy expenditure; hence, the physiological circuits implicated in the regulation of energy metabolism must be able to quickly adjust to changes in either side of the equation. Circulating orexigenic and anorexigenic factors, including ghrelin and leptin, are produced in the gastrointestinal tract and adipose tissue, respectively, in relation to an individual's nutritional status. These signals interact with central metabolic circuits to regulate the production and secretion of neuropeptides implicated in the control of appetite and energy expenditure. However, this physiological equilibrium can be perturbed by diverse processes, with weight gain occurring due to a positive energy balance and weight loss taking place if there is a negative energy balance. If a situation of positive energy balance continues for an extended period of time, excess weight is accumulated and this can eventually result in obesity. Obesity has become one of the most important health problems facing the industrialized world, indicating that metabolic equilibrium is frequently disrupted. Understanding how and why this occurs will allow new therapeutical targets to be identified.


Subject(s)
Drug Discovery , Ghrelin/metabolism , Hypothalamus/drug effects , Leptin/metabolism , Obesity/drug therapy , Signal Transduction/drug effects , Animals , Appetite Regulation/drug effects , Exercise , Feeding Behavior/drug effects , Ghrelin/agonists , Ghrelin/antagonists & inhibitors , Ghrelin/genetics , Humans , Hypothalamus/metabolism , Leptin/agonists , Leptin/antagonists & inhibitors , Leptin/genetics , Obesity/genetics , Obesity/metabolism , Obesity/prevention & control
20.
Sci Rep ; 5: 11452, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26090621

ABSTRACT

The orexigenic hormone ghrelin, a potential antagonist of the insulin system, ensures sufficient serum glucose in times of fasting. In the race for new therapeutics for diabetes, one focus of study has been antagonizing the ghrelin system in order to improve glucose tolerance. We provide evidence for a differential role of a ghrelin agonist on glucose homeostasis in an Alzheimer's disease mouse model fed a high-glycemic index diet as a constant challenge for glucose homeostasis. The ghrelin agonist impaired glucose tolerance immediately after administration but not in the long term. At the same time, the ghrelin agonist improved spatial learning in the mice, raised their activity levels, and reduced their body weight and fat mass. Immunoassay results showed a beneficial impact of long-term treatment on insulin signaling pathways in hippocampal tissue. The present results suggest that ghrelin might improve cognition in Alzheimer's disease via a central nervous system mechanism involving insulin signaling.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Cognition , Ghrelin/agonists , Insulin Resistance , Insulin/metabolism , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Cognition/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Disease Models, Animal , Glucose Intolerance/metabolism , Homeostasis , Mice , Microglia/drug effects , Microglia/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Signal Transduction/drug effects , Spatial Learning/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL