Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Eur J Neurosci ; 59(2): 308-315, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38086536

RESUMO

Multiple system atrophy (MSA) is a rare and rapidly progressive atypical parkinsonian disorder characterized by oligodendroglial cytoplasmic inclusions containing α-synuclein (α-syn), demyelination, inflammation and neuronal loss. To date, no disease-modifying therapy is available. Targeting α-syn-driven oligodendroglial dysfunction and demyelination presents a potential therapeutic approach for restricting axonal dysfunction, neuronal loss and disease progression. The present study investigated the promyelinogenic potential of sobetirome, a blood-brain barrier permeable and central nervous system selective thyromimetic in the context of an in vitro MSA model. Oligodendrocyte precursor cells (OPCs) were obtained from transgenic mice overexpressing human α-syn specifically in oligodendrocytes (MBP29 mouse line), a well-described MSA model, and non-transgenic littermates. mRNA and protein expression analyses revealed a substantial rescue effect of sobetirome on myelin-specific proteins in control and α-syn overexpressing oligodendrocytes. Furthermore, myelination analysis using nanofibres confirmed that sobetirome increases both the length and number of myelinated segments per oligodendrocyte in primary murine α-syn overexpressing oligodendrocytes and their respective control. These results suggest that sobetirome may be a promising thyromimetic compound targeting an important neuropathological hallmark of MSA.


Assuntos
Doenças Desmielinizantes , Atrofia de Múltiplos Sistemas , Fenóis , Camundongos , Humanos , Animais , Atrofia de Múltiplos Sistemas/tratamento farmacológico , Atrofia de Múltiplos Sistemas/genética , Atrofia de Múltiplos Sistemas/metabolismo , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Acetatos/metabolismo , Camundongos Transgênicos , Oligodendroglia/metabolismo , Doenças Desmielinizantes/metabolismo , Modelos Animais de Doenças
2.
Brain ; 146(8): 3331-3346, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37068912

RESUMO

Pitt-Hopkins syndrome is an autism spectrum disorder caused by autosomal dominant mutations in the human transcription factor 4 gene (TCF4). One pathobiological process caused by murine Tcf4 mutation is a cell autonomous reduction in oligodendrocytes and myelination. In this study, we show that the promyelinating compounds, clemastine, sobetirome and Sob-AM2 are effective at restoring myelination defects in a Pitt-Hopkins syndrome mouse model. In vitro, clemastine treatment reduced excess oligodendrocyte precursor cells and normalized oligodendrocyte density. In vivo, 2-week intraperitoneal administration of clemastine also normalized oligodendrocyte precursor cell and oligodendrocyte density in the cortex of Tcf4 mutant mice and appeared to increase the number of axons undergoing myelination, as EM imaging of the corpus callosum showed a significant increase in the proportion of uncompacted myelin and an overall reduction in the g-ratio. Importantly, this treatment paradigm resulted in functional rescue by improving electrophysiology and behaviour. To confirm behavioural rescue was achieved via enhancing myelination, we show that treatment with the thyroid hormone receptor agonist sobetirome or its brain penetrating prodrug Sob-AM2, was also effective at normalizing oligodendrocyte precursor cell and oligodendrocyte densities and behaviour in the Pitt-Hopkins syndrome mouse model. Together, these results provide preclinical evidence that promyelinating therapies may be beneficial in Pitt-Hopkins syndrome and potentially other neurodevelopmental disorders characterized by dysmyelination.


Assuntos
Transtorno do Espectro Autista , Deficiência Intelectual , Humanos , Animais , Camundongos , Clemastina , Transtorno do Espectro Autista/tratamento farmacológico , Transtorno do Espectro Autista/genética , Preparações Farmacêuticas , Deficiência Intelectual/tratamento farmacológico , Deficiência Intelectual/genética
3.
Int J Mol Sci ; 22(7)2021 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-33800546

RESUMO

Thyromimetics, whose physicochemical characteristics are analog to thyroid hormones (THs) and their derivatives, are promising candidates as novel therapeutics for neurodegenerative and metabolic pathologies. In particular, sobetirome (GC-1), one of the initial halogen-free thyromimetics, and newly synthesized IS25 and TG68, with optimized ADME-Tox profile, have recently attracted attention owing to their superior therapeutic benefits, selectivity, and enhanced permeability. Here, we further explored the functional capabilities of these thyromimetics to inhibit transthyretin (TTR) amyloidosis. TTR is a homotetrameric transporter protein for THs, yet it is also responsible for severe amyloid fibril formation, which is facilitated by tetramer dissociation into non-native monomers. By combining nuclear magnetic resonance (NMR) spectroscopy, computational simulation, and biochemical assays, we found that GC-1 and newly designed diphenyl-methane-based thyromimetics, namely IS25 and TG68, are TTR stabilizers and efficient suppressors of TTR aggregation. Based on these observations, we propose the novel potential of thyromimetics as a multi-functional therapeutic molecule for TTR-related pathologies, including neurodegenerative diseases.


Assuntos
Neuropatias Amiloides Familiares/tratamento farmacológico , Compostos de Bifenilo/química , Metano/química , Hormônios Tireóideos/farmacologia , Acetatos/farmacologia , Amiloide/metabolismo , Benzotiazóis/química , Desenho de Fármacos , Humanos , Espectroscopia de Ressonância Magnética , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Permeabilidade , Fenóis/farmacologia , Pré-Albumina/metabolismo , Ligação Proteica , Dobramento de Proteína , Proteínas Recombinantes/química , Hormônios Tireóideos/química
4.
Biomed Microdevices ; 20(2): 49, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29916059

RESUMO

With nearly 40% of U.S. adults obese, and childhood and adolescent rates rising, obesity and associated comorbidities are serious public health concerns with massive societal costs. Often, lifestyle interventions do not offer sufficient weight loss to improve health, requiring surgery and medications as adjunct management strategies. Here, we present a 4-month case study in which the sustained, low-dose, and constant administration of the thyroid receptor ß selective agonist GC-1 (sobetirome) from a novel nanochannel membrane implant was assessed in an obese, pre-diabetic rhesus macaque. Dramatic loss of white adipose tissue in the abdomen from 36 to 18% was observed via magnetic resonance imaging in conjunction with normalized serum insulin and glycemia, with no signs of cardiotoxicity shown. The non-human primate study highlights sustained low-dose delivery of GC-1 from our minimally invasive subcutaneous implant as a valuable approach to induce weight loss and manage obesity and comorbidities, including type 2 diabetes.


Assuntos
Acetatos/metabolismo , Sistemas de Liberação de Medicamentos/instrumentação , Nanotecnologia/instrumentação , Obesidade/metabolismo , Fenóis/metabolismo , Animais , Macaca mulatta
5.
Nanomedicine ; 13(5): 1739-1744, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28259802

RESUMO

This study demonstrated a nanochannel membrane device (NMD) for controlled and sustained release of GC-1 in rats, in the context of the treatment of metabolic syndrome. Release profiles were established in vitro both with and without 5% labrasol for over 2 months. In vivo pharmacokinetic evaluation showed effective GC-1 plasma concentrations, which resulted in significant reductions in body weight after just one week of treatment when compared to the NMD releasing vehicle only (PBS). We also provided evidence that rats treated with NMD-GC-1 present sub-active thyroids and clear differences in the morphology of the epithelium and follicles as compared to the controls, while the heart showed changes in weight. Moreover, body temperatures remained stable throughout treatment, and glucose, pancreatic islet size, and liver histology appeared similar between the treated and control groups. Prolonged constant administration of GC-1 from the NMD proved to be a valid strategy to facilitate weight loss.


Assuntos
Acetatos/farmacocinética , Nanotecnologia , Fenóis/farmacocinética , Acetatos/administração & dosagem , Animais , Peso Corporal , Fígado , Fenóis/administração & dosagem , Ratos , Ratos Endogâmicos F344
6.
Tetrahedron ; 71(35): 5946-5951, 2015 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-28316349

RESUMO

New synthetic routes for the preparation of isotopically labeled versions of thyroid hormone agonist sobetirome were developed using Knochel's iodine-magnesium exchange. A more efficient synthesis of the thyroid hormone antagonist NH-3 was developed from a common intermediate in the sobetirome route. Using the new synthetic routes, d6- and 3H-sobetirome were prepared for their use in studying biodistribution and the cellular uptake of sobetirome. The new route to NH-3 allows for a more rapid and efficient synthesis and provides access to an advanced intermediate to facilitate antagonist analog production in the final bond-forming synthetic step.

7.
Endocrinology ; 164(10)2023 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-37702560

RESUMO

Thyroid hormone receptor beta (TRß) is a recognized tumor suppressor in numerous solid cancers. The molecular signaling of TRß has been elucidated in several cancer types through re-expression models. Remarkably, the potential impact of selective activation of endogenous TRß on tumor progression remains largely unexplored. We used cell-based and in vivo assays to evaluate the effects of the TRß agonist sobetirome (GC-1) on a particularly aggressive and dedifferentiated cancer, anaplastic thyroid cancer (ATC). Here we report that GC-1 reduced the tumorigenic phenotype, decreased cancer stem-like cell populations, and induced redifferentiation of the ATC cell lines with different mutational backgrounds. Of note, this selective activation of TRß amplified the effects of therapeutic agents in blunting the aggressive cell phenotype and stem cell growth. In xenograft assays, GC-1 alone inhibited tumor growth and was as effective as the kinase inhibitor, sorafenib. These results indicate that selective activation of TRß not only induces a tumor suppression program de novo but enhances the effectiveness of anticancer agents, revealing potential novel combination therapies for ATC and other aggressive solid tumors.


Assuntos
Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide , Feminino , Humanos , Animais , Camundongos , Carcinoma Anaplásico da Tireoide/tratamento farmacológico , Receptores beta dos Hormônios Tireóideos , Agressão , Neoplasias da Glândula Tireoide/tratamento farmacológico
8.
Front Neurosci ; 14: 380, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32410949

RESUMO

Allan-Herndon-Dudley syndrome is a rare disease caused by inactivating mutations in the SLC16A2 gene, which encodes the monocarboxylate transporter 8 (MCT8), a transmembrane transporter specific for thyroid hormones (T3 and T4). Lack of MCT8 function produces serious neurological disturbances, most likely due to impaired transport of thyroid hormones across brain barriers during development resulting in severe brain hypothyroidism. Patients also suffer from thyrotoxicity in other organs due to the presence of a high concentration of T3 in the serum. An effective therapeutic strategy should restore thyroid hormone serum levels (both T3 and T4) and should address MCT8 transporter deficiency in brain barriers and neural cells, to enable the access of thyroid hormones to target neural cells. Unfortunately, targeted therapeutic options are currently scarce and their effect is limited to an improvement in the thyrotoxic state, with no sign of any neurological improvement. The use of thyroid hormone analogs such as TRIAC, DITPA, or sobetirome, that do not require MCT8 to cross cell membranes and whose controlled thyromimetic activity could potentially restore the normal function of the affected organs, are being explored to improve the cerebral availability of these analogs. Other strategies aiming to restore the transport of THs through MCT8 at the brain barriers and the cellular membranes include gene replacement therapy and the use of pharmacological chaperones. The design of an appropriate therapeutic strategy in combination with an early diagnosis (at prenatal stages), will be key aspects to improve the devastating alterations present in these patients.

9.
Thyroid ; 30(8): 1099-1105, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32098589

RESUMO

The development of thyroid hormone (TH) analogues was prompted by the attempt to exploit the effects of TH on lipid metabolism, avoiding cardiac thyrotoxicosis. Analysis of the relative distribution of the α and ß subtypes of nuclear TH receptors (TRα and TRß) showed that TRα and TRß are responsible for cardiac and metabolic responses, respectively. Therefore, analogues with TRß selectivity were developed, and four different compounds have been used in clinical trials: GC-1 (sobetirome), KB-2115 (eprotirome), MB07344/VK2809, and MGL-3196 (resmetirom). Each of these compounds was able to reduce low-density lipoprotein cholesterol, but a phase 3 trial with eprotirome was interrupted because of a significant increase in liver enzymes and the contemporary report of cartilage side effects in animals. As a consequence, the other projects were terminated as well. However, in recent years, TRß agonists have raised new interest for the treatment of nonalcoholic fatty liver disease (NAFLD). After obtaining excellent results in experimental models, clinical trials have been started with MGL-3196 and VK2809, and the initial reports are encouraging. Sobetirome turned out to be effective also in experimental models of demyelinating disease. Aside TRß agonists, TH analogues include some TH metabolites that are biologically active on their own, and their synthetic analogues. 3,5,3'-triiodothyroacetic acid has already found clinical use in the treatment of some cases of TH resistance due to TRß mutations, and interesting results have recently been reported in patients with the Allan-Herndon-Dudley syndrome, a rare disease caused by mutations in the TH transporter MCT8. 3,5-diiodothyronine (T2) has been used with success in rat models of dyslipidemia and NAFLD, but the outcome of a clinical trial with a synthetic T2 analogue was disappointing. 3-iodothyronamine (T1AM) is the last entry in the group of active TH metabolites. Promising results have been obtained in animal models of neurological injury induced by ß-amyloid or by convulsive agents, but no clinical data are available so far.


Assuntos
Hormônios Tireóideos/metabolismo , Acetatos/farmacologia , Anilidas/farmacologia , Animais , Doenças do Sistema Nervoso Central/sangue , Ensaios Clínicos como Assunto , Di-Iodotironinas/sangue , Desenho de Fármacos , Dislipidemias/sangue , Humanos , Hepatopatias/sangue , Masculino , Camundongos , Mutação , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fenóis/farmacologia , Piridazinas/farmacologia , Ratos , Transdução de Sinais , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Hormônios Tireóideos/química , Tironinas/sangue , Tri-Iodotironina/análogos & derivados , Tri-Iodotironina/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia
10.
Methods Mol Biol ; 1801: 193-206, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29892826

RESUMO

Thyroid hormone is a principal regulator of essential processes in vertebrate physiology and homeostasis. Synthetic derivatives of thyroid hormone, known as thyromimetics, display desirable therapeutic properties. Thoroughly understanding how thyromimetics distribute throughout the body is crucial for their development and this requires appropriate bioanalytical techniques to quantify drug levels in different tissues. Here, we describe a detailed protocol for the quantification of the thyromimetic sobetirome using liquid chromatography tandem-mass spectrometry (LC-MS/MS).


Assuntos
Acetatos/farmacocinética , Mimetismo Molecular , Fenóis/farmacocinética , Hormônios Tireóideos/farmacocinética , Acetatos/química , Animais , Cromatografia Líquida , Humanos , Redes e Vias Metabólicas , Camundongos , Estrutura Molecular , Fenóis/química , Espectrometria de Massas em Tandem , Hormônios Tireóideos/química , Distribuição Tecidual
11.
Thyroid ; 28(9): 1211-1220, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29845892

RESUMO

BACKGROUND: Loss of function mutations in the thyroid hormone (TH)-specific cell membrane transporter, the monocarboxylate transporter 8 (MCT8), lead to profound psychomotor retardation and abnormal TH serum levels, with low thyroxine (T4) and high triiodothyronine (T3). Several studies point to impaired TH transport across brain barriers as a crucial pathophysiological mechanism resulting in cerebral hypothyroidism. Treatment options for MCT8-deficient patients are limited and are focused on overcoming the brain barriers. The aim of this study was to evaluate the ability of the TH analog sobetirome and its prodrug Sob-AM2 to access the brain and exert thyromimetic actions in the absence of Mct8. METHODS: Juvenile wild-type (Wt) mice and mice lacking Mct8 and deiodinase type 2 (Mct8/Dio2KO) were treated systemically with daily injections of vehicle, 1 mg of sobetirome/kg body weight/day, or 0.3 mg of Sob-AM2/kg body weight/day for seven days. Sobetirome content was measured using liquid chromatography-tandem mass spectrometry, and T4 and T3 levels by specific radioimmunoassays. The effect of sobetirome treatment in the expression of T3-dependent genes was measured in the heart, liver, and cerebral cortex by real-time polymerase chain reaction. RESULTS: Sob-AM2 treatment in Mct8/Dio2KO animals led to 1.8-fold more sobetirome content in the brain and 2.5-fold less in plasma in comparison to the treatment with the parent drug sobetirome. Both sobetirome and Sob-AM2 treatments in Mct8/Dio2KO mice greatly decreased plasma T4 and T3 levels. Dio1 and Ucp2 gene expression was altered in the liver of Mct8/Dio2KO mice and was not affected by the treatments. In the heart, Hcn2 but not Atp2a2 expression was increased after treatment with the analogs. Interestingly, both sobetirome and Sob-AM2 treatments increased the expression of several T3-dependent genes in the brain such as Hr, Abcd2, Mme, and Flywch2 in Mct8/Dio2KO mice. CONCLUSIONS: Sobetirome and its amide prodrug Sob-AM2 can access the brain in the absence of Mct8 and exert thyromimetic actions modulating the expression of T3-dependent genes. At the peripheral level, the administration of these TH analogs results in the depletion of circulating T4 and T3. Therefore, sobetirome and Sob-AM2 have the potential to address the cerebral hypothyroidism and the peripheral hyperthyroidism characteristic of MCT8 deficiency.


Assuntos
Acetatos/farmacologia , Encéfalo/efeitos dos fármacos , Proteínas de Membrana Transportadoras/genética , Fenóis/farmacologia , Pró-Fármacos/farmacologia , Animais , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos , Simportadores , Tiroxina/sangue , Tri-Iodotironina/sangue , Proteína Desacopladora 2/genética , Proteína Desacopladora 2/metabolismo
12.
ACS Chem Neurosci ; 8(11): 2468-2476, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28756656

RESUMO

The blood-brain barrier (BBB) can be a substantial impediment to achieving therapeutic levels of drugs in the CNS. Certain chemical functionality such as the carboxylic acid is a general liability for BBB permeability preventing significant CNS distribution of a drug from a systemic dose. Here, we report a strategy for CNS-selective distribution of the carboxylic acid containing thyromimetic sobetirome using prodrugs targeted to fatty-acid amide hydrolase (FAAH), which is expressed in the brain. Two amide prodrugs of sobetirome were shown to be efficient substrates of FAAH with Vmax/KM values comparable to the natural endocannabinoid FAAH substrate anandamide. In mice, a systemic dose of sobetirome prodrug leads to a substantial ∼60-fold increase in brain distribution (Kp) of sobetirome compared to an equimolar systemic dose of the parent drug. The increased delivery of sobetirome to the brain from the prodrug was diminished by both pharmacological inhibition and genetic deletion of FAAH in vivo. The increased brain exposure of sobetirome arising from the prodrug corresponds to ∼30-fold increased potency in brain target engagement compared to the parent drug. These results suggest that FAAH-targeted prodrugs can considerably increase drug exposure to the CNS with a concomitant decrease in systemic drug levels generating a desirable distribution profile for CNS acting drugs.


Assuntos
Acetatos/farmacocinética , Amidoidrolases/metabolismo , Fenóis/farmacocinética , Pró-Fármacos/farmacocinética , Ativação Metabólica , Amidas/farmacocinética , Amidoidrolases/antagonistas & inibidores , Amidoidrolases/deficiência , Amidoidrolases/genética , Animais , Ácidos Araquidônicos/metabolismo , Barreira Hematoencefálica , Química Encefálica , Endocanabinoides/metabolismo , Humanos , Hidrólise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Molecular , Ácidos Oleicos/metabolismo , Especificidade de Órgãos , Alcamidas Poli-Insaturadas/metabolismo , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Hormônios Tireóideos/fisiologia , Distribuição Tecidual
13.
Expert Opin Ther Targets ; 20(2): 145-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26565124

RESUMO

Sobetirome binds selectively to the main hepatic form of thyroid hormone (TH) receptor, TRß1, compared to TRα1, which is principally responsible for thyrotoxic effects on heart, muscle and bone. Sobetirome also preferentially accumulates in liver. It was originally envisaged that sobetirome could be used to stimulate hepatic pathways that lower cholesterol without harmful side effects and might be used in conjunction with statins. Indeed, sobetirome progressed through preclinical animal studies and Phase I human clinical trials with excellent results and without obvious harmful side effects. Despite the fact that cardiovascular disease remains a major cause of mortality and that new therapies are desperately needed, it is unlikely that sobetirome will progress in further human clinical trials in the near future. The emergence of alternative cholesterol-lowering therapeutics may render selective thyromimetics redundant. Further, fears of thyrotoxic effects in the heart and emergence of cartilage defects in dogs after long-term use of eprotirome, a similar though not identical compound, has reduced enthusiasm for this strategy. We argue that it is nevertheless important to explore uses of sobetirome in humans; more treatment strategies would help patients with hard-to-treat dyslipidemias. Sobetirome may also have additional applications in orphan indications and short-term controlled weight loss.


Assuntos
Acetatos/farmacologia , Doenças Cardiovasculares/tratamento farmacológico , Dislipidemias/tratamento farmacológico , Fenóis/farmacologia , Acetatos/efeitos adversos , Animais , Colesterol/sangue , Ensaios Clínicos como Assunto , Cães , Humanos , Fígado/metabolismo , Fenóis/efeitos adversos , Receptores beta dos Hormônios Tireóideos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA