Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Molecules ; 24(10)2019 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-31100979

RESUMO

In this work we summarize our understanding of melanocortin 4 receptor (MC4R) pathway activation, aiming to define a safe and effective therapeutic targeting strategy for the MC4R. Delineation of cellular MC4R pathways has provided evidence for distinct MC4R signaling events characterized by unique receptor activation kinetics. While these studies remain narrow in scope, and have largely been explored with peptidic agonists, the results provide a possible correlation between distinct ligand groups and differential MC4R activation kinetics. In addition, when a set of small-molecule and peptide MC4R agonists are compared, evidence of biased signaling has been reported. The results of such mechanistic studies are discussed.


Assuntos
Peptídeos/farmacocinética , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais , Animais , Peso Corporal , Sistema Cardiovascular/efeitos dos fármacos , AMP Cíclico/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Cinética , Ligantes , Peptídeos/química , Peptídeos/farmacologia , Primatas , Ligação Proteica , Transporte Proteico , Roedores , Transdução de Sinais/efeitos dos fármacos , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
2.
Proc Natl Acad Sci U S A ; 112(12): E1471-9, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25775521

RESUMO

Therapeutics developed and sold as racemates can exhibit a limited therapeutic index because of side effects resulting from the undesired enantiomer (distomer) and/or its metabolites, which at times, forces researchers to abandon valuable scaffolds. Therefore, most chiral drugs are developed as single enantiomers. Unfortunately, the development of some chirally pure drug molecules is hampered by rapid in vivo racemization. The class of compounds known as immunomodulatory drugs derived from thalidomide is developed and sold as racemates because of racemization at the chiral center of the 3-aminoglutarimide moiety. Herein, we show that replacement of the exchangeable hydrogen at the chiral center with deuterium allows the stabilization and testing of individual enantiomers for two thalidomide analogs, including CC-122, a compound currently in human clinical trials for hematological cancers and solid tumors. Using "deuterium-enabled chiral switching" (DECS), in vitro antiinflammatory differences of up to 20-fold are observed between the deuterium-stabilized enantiomers. In vivo, the exposure is dramatically increased for each enantiomer while they retain similar pharmacokinetics. Furthermore, the single deuterated enantiomers related to CC-122 exhibit profoundly different in vivo responses in an NCI-H929 myeloma xenograft model. The (-)-deuterated enantiomer is antitumorigenic, whereas the (+)-deuterated enantiomer has little to no effect on tumor growth. The ability to stabilize and differentiate enantiomers by DECS opens up a vast window of opportunity to characterize the class effects of thalidomide analogs and improve on the therapeutic promise of other racemic compounds, including the development of safer therapeutics and the discovery of new mechanisms and clinical applications for existing therapeutics.


Assuntos
Anti-Inflamatórios/química , Antineoplásicos/química , Inflamação/tratamento farmacológico , Neoplasias/tratamento farmacológico , Piperidonas/química , Quinazolinonas/química , Talidomida/análogos & derivados , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos , Leucócitos Mononucleares/citologia , Camundongos , Camundongos SCID , Modelos Químicos , Transplante de Neoplasias , Neoplasias/imunologia , Estereoisomerismo , Talidomida/química , Fator de Necrose Tumoral alfa/metabolismo
3.
J Neurol ; 270(3): 1615-1623, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36462055

RESUMO

OBJECTIVES: Friedreich ataxia is (FRDA) an autosomal recessive neurodegenerative disorder associated with intrinsic oxidative damage, suggesting that decreasing lipid peroxidation (LPO) might ameliorate disease progression. The present study tested the ability of RT001, a deuterated form of linoleic acid (D2-LA), to alter disease severity in patients with FRDA in a double-blind placebo-controlled trial. METHODS: Sixty-five subjects were recruited across six sites and received either placebo or active drug for an 11-month study. Subjects were evaluated at 0, 4, 9, and 11 months, with the primary outcome measure being maximum oxygen consumption (MVO2) during cardiopulmonary exercise testing (CPET). A key secondary outcome measure was a composite statistical test using results from the timed 1-min walk (T1MW), peak workload, and MVO2. RESULTS: Forty-five subjects completed the protocol. RT001 was well tolerated, with no serious adverse events related to drug. Plasma and red blood cell (RBC) membrane levels of D2-LA and its primary metabolite deuterated arachidonic acid (D2-AA) achieved steady-state concentrations by 4 months. No significant changes in MVO2 were observed for RT001 compared to placebo. Similarly, no differences between the groups were found in secondary or exploratory outcome measures. Post hoc evaluations also suggested minimal effects of RT001 at the dosages used in this study. INTERPRETATIONS: The results of this study provide no evidence for a significant benefit of RT001 at the dosages tested in this Friedreich ataxia patient population.


Assuntos
Ataxia de Friedreich , Ácido Linoleico , Humanos , Ataxia de Friedreich/tratamento farmacológico , Ácido Linoleico/uso terapêutico , Ácidos Linoleicos/uso terapêutico , Caminhada , Método Duplo-Cego
4.
Hepatol Commun ; 5(8): 1412-1425, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34430785

RESUMO

The antidiabetic drug pioglitazone is, to date, the most efficacious oral drug recommended off-label for the treatment of nondiabetic or diabetic patients with biopsy-proven nonalcoholic steatohepatitis (NASH). However, weight gain and edema side effects have limited its use for NASH. Pioglitazone is a mixture of two stereoisomers ((R)-pioglitazone and (S)-pioglitazone) that interconvert in vitro and in vivo. We aimed to characterize their individual pharmacology to develop a safer and potentially more potent drug for NASH. We stabilized the stereoisomers of pioglitazone with deuterium at the chiral center. Preclinical studies with deuterium-stabilized (R)-pioglitazone (PXL065) and (S)-pioglitazone demonstrated that (R)-pioglitazone retains the efficacy of pioglitazone in NASH, including reduced hepatic triglycerides, free fatty acids, cholesterol, steatosis, inflammation, hepatocyte enlargement, and fibrosis. Although both stereoisomers inhibit the mitochondrial pyruvate carrier, PXL065 shows limited to no peroxisome proliferator-activated receptor gamma (PPARγ) activity, whereas (S)-pioglitazone appears responsible for the PPARγ activity and associated weight gain. Nonetheless, in preclinical models, both stereoisomers reduce plasma glucose and hepatic fibrosis to the same extent as pioglitazone, suggesting that these benefits may also be mediated by altered mitochondrial metabolism. In a phase 1a clinical study, we demonstrated safety and tolerability of single 7.5-mg, 22.5-mg, and 30-mg doses of PXL065 as well as preferential exposure to the (R)-stereoisomer in comparison to 45-mg pioglitazone. Conclusion: PXL065 at a dose lower than 22.5 mg is predicted to exhibit efficacy for NASH equal to, or greater than, 45-mg pioglitazone without the potentially detrimental weight gain and edema. The development of PXL065 for NASH represents a unique opportunity to leverage the therapeutic benefits of pioglitazone, while reducing or eliminating PPARγ-related side effects.

5.
Mol Metab ; 48: 101206, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33684608

RESUMO

BACKGROUND: Over the past 20 years, insights from human and mouse genetics have illuminated the central role of the brain leptin-melanocortin pathway in controlling mammalian food intake, with genetic disruption resulting in extreme obesity, and more subtle polymorphic variations influencing the population distribution of body weight. At the end of 2020, the U.S. Food and Drug Administration (FDA) approved setmelanotide, a melanocortin 4 receptor agonist, for use in individuals with severe obesity due to either pro-opiomelanocortin (POMC), proprotein convertase subtilisin/kexin type 1 (PCSK1), or leptin receptor (LEPR) deficiency. SCOPE OF REVIEW: Herein, we chart the melanocortin pathway's history, explore its pharmacology, genetics, and physiology, and describe how a neuropeptidergic circuit became an important druggable obesity target. MAJOR CONCLUSIONS: Unravelling the genetics of the subset of severe obesity has revealed the importance of the melanocortin pathway in appetitive control; coupling this with studying the molecular pharmacology of compounds that bind melanocortin receptors has brought a new obesity drug to the market. This process provides a drug discovery template for complex disorders, which for setmelanotide took 25 years to transform from a single gene into an approved drug.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Metabolismo Energético/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Melanocortinas/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Receptor Tipo 4 de Melanocortina/agonistas , Transdução de Sinais/efeitos dos fármacos , alfa-MSH/análogos & derivados , Animais , Fármacos Antiobesidade/farmacologia , Aprovação de Drogas/história , Descoberta de Drogas/história , História do Século XX , História do Século XXI , Humanos , Camundongos , Obesidade/epidemiologia , Receptor Tipo 4 de Melanocortina/metabolismo , Estados Unidos/epidemiologia , alfa-MSH/farmacologia , alfa-MSH/uso terapêutico
6.
Bioorg Med Chem Lett ; 20(16): 4757-61, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20643546

RESUMO

The design, synthesis, and binding activity of ring constrained analogs of the acyclic cannabinoid-1 receptor (CB1R) inverse agonist taranabant 1 are described. The initial inspiration for these taranabant derivatives was its conformation 1a, determined by (1)H NMR, X-ray, and molecular modeling. The constrained analogs were all much less potent than their acyclic parent structure. The results obtained are discussed in the context of a predicted binding of 1 to a homology model of CB1R.


Assuntos
Amidas/química , Fármacos Antiobesidade/síntese química , Piridinas/química , Receptor CB1 de Canabinoide/química , Amidas/síntese química , Amidas/farmacologia , Fármacos Antiobesidade/química , Fármacos Antiobesidade/farmacologia , Simulação por Computador , Humanos , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Piridinas/síntese química , Piridinas/farmacologia , Receptor CB1 de Canabinoide/metabolismo
8.
J Pharm Sci ; 109(11): 3496-3503, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32871154

RESUMO

RT001 is the di-deutero isotopologue of linoleic acid ethyl ester (D2-LA). Resistance to oxidative damage at the carbon-deuterium bond depends upon the concentration of D2-LA as a percentage of total LA. We report here on the plasma and red cell (RBC) pharmacokinetics (PK) of D2-LA, and its metabolite 13,13-D2-arachidonic acid (D2-AA), in patients with multiple neurodegenerative diseases (total of 59 participants). In Friedreich's ataxia patients, D2-LA was absorbed and transported similarly to dietary LA, peaking at about 6 h after oral dosing. Plasma D2-LA concentrations approached steady state after 28 days of dosing. After 6 months of daily dosing in subjects with other disorders, D2-LA and D2-AA levels were at or above the 20% of total (D2-LA/total LA, or D2-AA/total AA) therapeutic targets for most subjects. We conclude that chronic dosing of RT001 and associated dietary guidance can be maintained over many months to achieve target plasma and RBC levels, forming a basis for therapeutic dosing across a broad range of conditions. RT001 has been safe and well-tolerated in 59 different participants treated across 10 different neurodegenerative diseases in multiple clinical trials for up to 36 months with no significant drug related adverse events limiting use.


Assuntos
Ácido Linoleico , Preparações Farmacêuticas , Membrana Celular , Ésteres , Humanos , Ácidos Linoleicos
9.
Int J Multiscale Comput Eng ; 18(3): 329-333, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32831809

RESUMO

We write to introduce our novel group formed to confront some of the issues raised by the COVID-19 pandemic. Information about the group, which we named "cure COVid for Ever and for All" (RxCOVEA), its dynamic membership (changing regularly), and some of its activities-described in more technical detail for expert perusal and commentary-are available upon request.

10.
Appl Clin Genet ; 12: 87-93, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31239751

RESUMO

Purpose: The hypothalamic melanocortin-4 receptor (MC4R) pathway, a component of the central melanocortin pathway, regulates energy balance and satiety. Rare genetic disorders of obesity may be characterized by impaired MC4R pathway signaling, which results in early-onset severe obesity and insatiable hunger (hyperphagia). The TEMPO registry (NCT03479437) is a voluntary, prospective, open-ended registry of individuals with rare genetic disorders of obesity due to mutations in genes within the MC4R pathway who have early-onset severe obesity. The objective of the TEMPO registry is to evaluate the burden of rare genetic disorders of obesity on individuals, their parents/caregivers, health care providers, and the health care system. Patients and methods: Individuals with rare genetic disorders of obesity (adults aged ≥18 years and children and adolescents aged from 2 to 17 years) will be referred by their health care providers or by a genetic screening study. Individuals must meet age- and sex-specific body mass index values that define the clinical criteria for severe obesity and carry selected variants in MC4R or in one of several genes upstream or downstream of the MC4R. Online surveys will be completed by the individual, parent/caregiver, and health care provider at baseline and annually thereafter and will collect data on demographics, results of genetic testing, medical/family history, disease characteristics, resource utilization, eating habits/hunger episodes, social and emotional impacts, and interest in future clinical trial participation. Conclusions: The TEMPO registry will provide insights into the overall course and disease burden for individuals with rare genetic disorders of obesity. Health care providers may use this resource to improve the identification, diagnosis, and treatment of individuals with rare forms of genetic obesity.

12.
J Clin Endocrinol Metab ; 103(7): 2601-2612, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29726959

RESUMO

Context: The hypothalamic melanocortin 4 receptor (MC4R) pathway serves a critical role in regulating body weight. Loss of function (LoF) mutations in the MC4R pathway, including mutations in the pro-opiomelanocortin (POMC), prohormone convertase 1 (PCSK1), leptin receptor (LEPR), or MC4R genes, have been shown to cause early-onset severe obesity. Methods: Through a comprehensive epidemiological analysis of known and predicted LoF variants in the POMC, PCSK1, and LEPR genes, we sought to estimate the number of US individuals with biallelic MC4R pathway LoF variants. Results: We predict ~650 α-melanocyte-stimulating hormone (MSH)/POMC, 8500 PCSK1, and 3600 LEPR homozygous and compound heterozygous individuals in the United States, cumulatively enumerating >12,800 MC4R pathway-deficient obese patients. Few of these variants have been genetically diagnosed to date. These estimates increase when we include a small subset of less rare variants: ß-MSH/POMC,PCSK1 N221D, and a PCSK1 LoF variant (T640A). To further define the MC4R pathway and its potential impact on obesity, we tested associations between body mass index (BMI) and LoF mutation burden in the POMC, PCSK1, and LEPR genes in various populations. We show that the cumulative allele burden in individuals with two or more LoF alleles in one or more genes in the MC4R pathway are predisposed to a higher BMI than noncarriers or heterozygous LoF carriers with a defect in only one gene. Conclusions: Our analysis represents a genetically rationalized study of the hypothalamic MC4R pathway aimed at genetic patient stratification to determine which obese subpopulations should be studied to elucidate MC4R agonist (e.g., setmelanotide) treatment responsiveness.


Assuntos
Mutação com Perda de Função/genética , Obesidade/epidemiologia , Obesidade/genética , Receptor Tipo 4 de Melanocortina/genética , Transdução de Sinais/genética , Alelos , Fármacos Antiobesidade/farmacologia , Índice de Massa Corporal , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Obesidade/tratamento farmacológico , Pró-Opiomelanocortina/genética , Pró-Proteína Convertase 1/genética , Receptor Tipo 4 de Melanocortina/agonistas , Receptores para Leptina/genética , Estados Unidos/epidemiologia , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
13.
Nat Med ; 24(5): 551-555, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29736023

RESUMO

Genetic defects underlying the melanocortin-4 receptor (MC4R) signaling pathway lead to severe obesity. Three severely obese LEPR-deficient individuals were administered the MC4R agonist setmelanotide, resulting in substantial and durable reductions in hyperphagia and body weight over an observation period of 45-61 weeks. Compared to formerly developed and tested MC4R agonists, setmelanotide has the unique capability of activating nuclear factor of activated T cell (NFAT) signaling and restoring function of this signaling pathway for selected MC4R variants. Our data demonstrate the potency of setmelanotide in treatment of individuals with diverse MC4R-related pathway deficiencies.


Assuntos
Receptor Tipo 4 de Melanocortina/agonistas , Receptores para Leptina/deficiência , Redução de Peso , Adolescente , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Peptídeos/farmacologia , Receptores para Leptina/genética , Fosfolipases Tipo C/metabolismo , Redução de Peso/efeitos dos fármacos , Adulto Jovem , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
14.
Mol Cell Biol ; 22(14): 5027-35, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12077332

RESUMO

Agouti-related protein (AgRP), a neuropeptide abundantly expressed in the arcuate nucleus of the hypothalamus, potently stimulates feeding and body weight gain in rodents. AgRP is believed to exert its effects through the blockade of signaling by alpha-melanocyte-stimulating hormone at central nervous system (CNS) melanocortin-3 receptor (Mc3r) and Mc4r. We generated AgRP-deficient (Agrp(-/-)) mice to examine the physiological role of AgRP. Agrp(-/-) mice are viable and exhibit normal locomotor activity, growth rates, body composition, and food intake. Additionally, Agrp(-/-) mice display normal responses to starvation, diet-induced obesity, and the administration of exogenous leptin or neuropeptide Y (NPY). In situ hybridization failed to detect altered CNS expression levels for proopiomelanocortin, Mc3r, Mc4r, or NPY mRNAs in Agrp(-/-) mice. As AgRP and the orexigenic peptide NPY are coexpressed in neurons of the arcuate nucleus, we generated AgRP and NPY double-knockout (Agrp(-/-);Npy(-/-)) mice to determine whether NPY or AgRP plays a compensatory role in Agrp(-/-) or NPY-deficient (Npy(-/-)) mice, respectively. Similarly to mice deficient in either AgRP or NPY, Agrp(-/-);Npy(-/-) mice suffer no obvious feeding or body weight deficits and maintain a normal response to starvation. Our results demonstrate that neither AgRP nor NPY is a critically required orexigenic factor, suggesting that other pathways capable of regulating energy homeostasis can compensate for the loss of both AgRP and NPY.


Assuntos
Metabolismo Energético , Neuropeptídeo Y/deficiência , Proteínas/metabolismo , Proteína Relacionada com Agouti , Animais , Sistema Nervoso Central/metabolismo , Homeostase , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Knockout , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Fenótipo , Pró-Opiomelanocortina/genética , Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 3 de Melanocortina , Receptor Tipo 4 de Melanocortina , Receptores da Corticotropina/genética , Receptores de Peptídeos/genética , Transdução de Sinais
15.
Atherosclerosis ; 264: 100-107, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28655430

RESUMO

BACKGROUND AND AIMS: Oxidative modification of lipoproteins is a crucial step in atherosclerosis development. Isotopic-reinforced polyunsaturated fatty acids (D-PUFAs) are more resistant to reactive oxygen species-initiated chain reaction of lipid peroxidation than regular hydrogenated (H-)PUFAs. We aimed at investigating the effect of D-PUFA treatment on lipid peroxidation, hypercholesterolemia and atherosclerosis development. METHODS: Transgenic APOE*3-Leiden.CETP mice, a well-established model for human-like lipoprotein metabolism, were pre-treated with D-PUFAs or control H-PUFAs-containing diet (1.2%, w/w) for 4 weeks. Thereafter, mice were fed a Western-type diet (containing 0.15% cholesterol, w/w) for another 12 weeks, while continuing the D-/H-PUFA treatment. RESULTS: D-PUFA treatment markedly decreased hepatic and plasma F2-isoprostanes (approx. -80%) and prostaglandin F2α (approx. -40%) as compared to H-PUFA treatment. Moreover, D-PUFAs reduced body weight gain during the study (-54%) by decreasing body fat mass gain (-87%) without altering lean mass. D-PUFAs consistently reduced plasma total cholesterol levels (approx. -25%), as reflected in reduced plasma non-HDL-cholesterol (-28%). Additional analyses of hepatic cholesterol metabolism indicated that D-PUFAs reduced the hepatic cholesterol content (-21%). Sterol markers of intestinal cholesterol absorption and cholesterol breakdown were decreased. Markers of cholesterol synthesis were increased. Finally, D-PUFAs reduced atherosclerotic lesion area formation throughout the aortic root of the heart (-26%). CONCLUSIONS: D-PUFAs reduce body weight gain, improve cholesterol handling and reduce atherosclerosis development by reducing lipid peroxidation and plasma cholesterol levels. D-PUFAs, therefore, represent a promising new strategy to broadly reduce rates of lipid peroxidation, and combat hypercholesterolemia and cardiovascular diseases.


Assuntos
Anticolesterolemiantes/farmacologia , Antioxidantes/farmacologia , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Colesterol/sangue , Ácidos Graxos Insaturados/farmacologia , Hipercolesterolemia/tratamento farmacológico , Peroxidação de Lipídeos/efeitos dos fármacos , Adiposidade/efeitos dos fármacos , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteína E3/genética , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Biomarcadores/sangue , Proteínas de Transferência de Ésteres de Colesterol/genética , Dinoprosta/sangue , Modelos Animais de Doenças , F2-Isoprostanos/sangue , Feminino , Predisposição Genética para Doença , Hipercolesterolemia/sangue , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Camundongos Knockout para ApoE , Fenótipo , Placa Aterosclerótica , Fatores de Tempo , Aumento de Peso/efeitos dos fármacos
16.
Mol Metab ; 6(10): 1321-1329, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29031731

RESUMO

OBJECTIVE: Pro-opiomelanocortin (POMC)-derived peptides act on neurons expressing the Melanocortin 4 receptor (MC4R) to reduce body weight. Setmelanotide is a highly potent MC4R agonist that leads to weight loss in diet-induced obese animals and in obese individuals with complete POMC deficiency. While POMC deficiency is very rare, 1-5% of severely obese individuals harbor heterozygous mutations in MC4R. We sought to assess the efficacy of Setmelanotide in human MC4R deficiency. METHODS: We studied the effects of Setmelanotide on mutant MC4Rs in cells and the weight loss response to Setmelanotide administration in rodent studies and a human clinical trial. We annotated the functional status of 369 published MC4R variants. RESULTS: In cells, we showed that Setmelanotide is significantly more potent at MC4R than the endogenous ligand alpha-melanocyte stimulating hormone and can disproportionally rescue signaling by a subset of severely impaired MC4R mutants. Wild-type rodents appear more sensitive to Setmelanotide when compared to MC4R heterozygous deficient mice, while MC4R knockout mice fail to respond. In a 28-day Phase 1b clinical trial, Setmelanotide led to weight loss in obese MC4R variant carriers. Patients with POMC defects upstream of MC4R show significantly more weight loss with Setmelanotide than MC4R deficient patients or obese controls. CONCLUSIONS: Setmelanotide led to weight loss in obese people with MC4R deficiency; however, further studies are justified to establish whether Setmelanotide can elicit clinically meaningful weight loss in a subset of the MC4R deficient obese population.


Assuntos
Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/deficiência , alfa-MSH/análogos & derivados , Insuficiência Adrenal/tratamento farmacológico , Insuficiência Adrenal/metabolismo , Adulto , Sequência de Aminoácidos , Animais , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Pró-Opiomelanocortina/deficiência , Pró-Opiomelanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , alfa-MSH/farmacologia
17.
J Med Chem ; 49(26): 7584-7, 2006 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-17181138

RESUMO

The discovery of novel acyclic amide cannabinoid-1 receptor inverse agonists is described. They are potent, selective, orally bioavailable, and active in rodent models of food intake and body weight reduction. A major focus of the optimization process was to increase in vivo efficacy and to reduce the potential for formation of reactive metabolites. These efforts led to the identification of compound 48 for development as a clinical candidate for the treatment of obesity.


Assuntos
Fármacos Antiobesidade/farmacologia , Canabinoides/farmacologia , Obesidade/tratamento farmacológico , Receptor CB1 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/agonistas , Animais , Fármacos Antiobesidade/síntese química , Fármacos Antiobesidade/química , Peso Corporal/efeitos dos fármacos , Canabinoides/síntese química , Canabinoides/química , AMP Cíclico/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Microssomos/efeitos dos fármacos , Microssomos/metabolismo , Ratos , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/metabolismo , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/metabolismo
18.
Prog Brain Res ; 153: 107-18, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16876570

RESUMO

Over the past decade, hypothalamic circuits have been described that impact energy homeostasis in rodents and humans. Our drug development efforts for the treatment of obesity and the metabolic syndrome have largely focused on selected genetic and/or pharmacologically validated pathways. The translation of these pathways into therapeutics for the treatment of obesity will find its first clinical successes over the coming decade. Initial efforts have focused on gaining a better understanding of the relevance of rodent pharmacological and genetic observations for the development of therapeutics for the treatment of human obesity. We pursue pathways defined by the expression of the ghrelin receptor, melanin-concentrating hormone receptors, melanocortin receptors, cannabinoid receptors and neuropeptide Y1 and Y5 receptors. In this review, we will discuss drug development efforts for the treatment of obesity, focused on selective melanocortin 4 receptor agonists and neuropeptide Y1 and Y5 receptor antagonists. These drug development efforts required an in-depth understanding of cell-based observations which drive the development of compound structure-activity relationships. These include understanding of receptor function in selected cell-based backgrounds and early evaluation and validation of ex vivo observations in appropriate in vivo models. In order to develop selective and safe anti-obesity drugs, diverse approaches are needed to increase the likelihood of clinical success, including: (i) developing a detailed understanding of the predictive value of rodent pathways for treatment of human disease; (ii) knowledge of the exact location of targeted receptor subtypes for the clinical indication under study in order to derive a suitable compound profile; (iii) predictive measures of in vivo and/or ex vivo receptor occupancy required to bring about a desired physiological effect; (iv) predictive parameters that outline that the drug-derived effects are safe and mechanism-based; and (v) the refinement of selected compound classes, aimed at their clinical use.


Assuntos
Adiposidade/efeitos dos fármacos , Fármacos Antiobesidade , Apetite/efeitos dos fármacos , Desenho de Fármacos , Animais , Fármacos Antiobesidade/síntese química , Fármacos Antiobesidade/química , Fármacos Antiobesidade/farmacologia , Interações Medicamentosas , Humanos , Neuropeptídeo Y/agonistas , Neuropeptídeo Y/antagonistas & inibidores , Receptor Tipo 4 de Melanocortina/agonistas , Receptor Tipo 4 de Melanocortina/antagonistas & inibidores
19.
Br J Pharmacol ; 173(17): 2614-21, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27339818

RESUMO

BACKGROUND AND PURPOSE: α- and ß-melanocyte-stimulating hormones (MSH) are derived from pro-opiomelanocortin (POMC) and are the natural agonist ligands of the melanocortin 4 receptor, a key regulator of energy homeostasis. Recent rodent and human data have implicated the MAGEL2 gene, which may regulate activation of POMC neurons, as a significant contributor to the metabolic symptoms observed in Prader-Willi Syndrome (PWS). Firstly, patients with protein truncating mutations in MAGEL2 exhibit numerous clinical characteristics of PWS. Secondly, Magel2-null mice may not normally activate MC4 receptors, as they are defective in the activation of their POMC neurons and hence may fail to normally release the POMC-derived MC4 receptor agonist ligands α- and ß-MSH. Magel2-null mice represent a tractable animal model for the metabolic and appetitive imbalance seen in patients with PWS. EXPERIMENTAL APPROACH: We tested a dose titration of the MC4 receptor agonist setmelanotide, in development for rare monogenic forms of obesity, in Magel2-null mice. KEY RESULTS: We show that Magel2-null mice are hypersensitive to the appetite suppressing and metabolic effects of setmelanotide. CONCLUSION AND IMPLICATIONS: Setmelanotide may be a useful investigational hormone/neuropeptide replacement therapy for PWS and rare monogenic forms of obesity exhibiting impaired function of POMC neurons.


Assuntos
Antígenos de Neoplasias/genética , Regulação do Apetite/efeitos dos fármacos , Metabolismo/efeitos dos fármacos , Proteínas/genética , Receptor Tipo 4 de Melanocortina/agonistas , alfa-MSH/análogos & derivados , Animais , Antígenos de Neoplasias/metabolismo , Relação Dose-Resposta a Droga , Feminino , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Síndrome de Prader-Willi/metabolismo , Pró-Opiomelanocortina/metabolismo , Proteínas/metabolismo , Relação Estrutura-Atividade , alfa-MSH/administração & dosagem , alfa-MSH/farmacologia
20.
J Neurosci ; 23(32): 10351-8, 2003 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-14614094

RESUMO

The peptides alpha-melanocyte stimulating hormone (alpha-MSH) and oxytocin, when administered centrally, produce similar behavioral effects. alpha-MSH induces Fos expression in supraoptic oxytocin neurons, and alpha-MSH melanocortin-4 receptors (MC4Rs) are highly expressed in the supraoptic nucleus, suggesting that alpha-MSH and oxytocin actions are not independent. Here we investigated the effects of alpha-MSH on the activity of supraoptic neurons. We confirmed that alpha-MSH induces Fos expression in the supraoptic nucleus when injected centrally and demonstrated that alpha-MSH also stimulates Fos expression in the nucleus when applied locally by retrodialysis. Thus alpha-MSH-induced Fos expression is not associated with electrophysiological excitation of supraoptic neurons because central injection of alpha-MSH or selective MC4 receptor agonists inhibited the electrical activity of oxytocin neurons in the supraoptic nucleus recorded in vivo. Consistent with these observations, oxytocin secretion into the bloodstream decreased after central injection of alpha-MSH. However, MC4R ligands induced substantial release of oxytocin from dendrites in isolated supraoptic nuclei. Because dendritic oxytocin release can be triggered by changes in [Ca2+]i, we measured [Ca2+]i responses in isolated supraoptic neurons and found that MC4R ligands induce a transient [Ca2+]i increase in oxytocin neurons. This response was still observed in low extracellular Ca2+ concentration and probably reflects mobilization of [Ca2+]i from intracellular stores rather than entry via voltage-gated channels. Taken together, these results show for the first time that a peptide, here alpha-MSH, can induce differential regulation of dendritic release and systemic secretion of oxytocin, accompanied by dissociation of Fos expression and electrical activity.


Assuntos
Dendritos/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Ocitocina/metabolismo , Neuro-Hipófise/metabolismo , alfa-MSH/farmacologia , Animais , Cálcio/metabolismo , Separação Celular , Dendritos/efeitos dos fármacos , Feminino , Hipotálamo/citologia , Técnicas In Vitro , Injeções Intraventriculares , Microdiálise , Neurônios/efeitos dos fármacos , Neuro-Hipófise/citologia , Neuro-Hipófise/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptor Tipo 4 de Melanocortina/agonistas , Núcleo Supraóptico/citologia , Núcleo Supraóptico/efeitos dos fármacos , Núcleo Supraóptico/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa