Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Int J Mol Sci ; 23(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35269859

RESUMEN

3-iodothyronamine (T1AM) and 3-iodothyroacetic acid (TA1) are thyroid-hormone-related compounds endowed with pharmacological activity through mechanisms that remain elusive. Some evidence suggests that they may have redox features. We assessed the chemical activity of T1AM and TA1 at pro-oxidant conditions. Further, in the cell model consisting of brown adipocytes (BAs) differentiated for 6 days in the absence (M cells) or in the presence of 20 nM T1AM (M + T1AM cells), characterized by pro-oxidant metabolism, or TA1 (M + TA1 cells), we investigated the expression/activity levels of pro- and anti-oxidant proteins, including UCP-1, sirtuin-1 (SIRT1), mitochondrial monoamine (MAO-A and MAO-B), semicarbazide-sensitive amine oxidase (SSAO), and reactive oxygen species (ROS)-dependent lipoperoxidation. T1AM and TA1 showed in-vitro antioxidant and superoxide scavenging properties, while only TA1 acted as a hydroxyl radical scavenger. M + T1AM cells showed higher lipoperoxidation levels and reduced SIRT1 expression and activity, similar MAO-A, but higher MAO-B activity in terms of M cells. Instead, the M + TA1 cells exhibited increased levels of SIRT1 protein and activity and significantly lower UCP-1, MAO-A, MAO-B, and SSAO in comparison with the M cells, and did not show signs of lipoperoxidation. Our results suggest that SIRT1 is the mediator of T1AM and TA1 pro-or anti-oxidant effects as a result of ROS intracellular levels, including the hydroxyl radical. Here, we provide evidence indicating that T1AM and TA1 administration impacts on the redox status of a biological system, a feature that indicates the novel mechanism of action of these two thyroid-hormone-related compounds.


Asunto(s)
Radical Hidroxilo , Sirtuina 1 , Monoaminooxidasa/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno , Sirtuina 1/metabolismo , Hormonas Tiroideas/metabolismo , Tironinas/metabolismo , Tironinas/farmacología
2.
Eur J Pharmacol ; 912: 174606, 2021 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-34717926

RESUMEN

The 3-iodothyronamine (T1AM) and 3-iodothryoacetic acid (TA1), are endogenous occurring compounds structurally related with thyroid hormones (THs, the pro-hormone T4 and the active hormone T3) initially proposed as possible mediators of the rapid effects of T3. However, after years from their identification, the physio-pathological meaning of T1AM and TA1 tissue levels remains an unsolved issue while pharmacological evidence indicates both compounds promote in rodents central and peripheral effects with mechanisms which remain mostly elusive. Pharmacodynamics of T1AM includes the recognition of G-coupled receptors, ion channels but also biotransformation into an active metabolite, i.e. the TA1. Furthermore, long term T1AM treatment associates with post-translational modifications of cell proteins. Such array of signaling may represent an added value, rather than a limit, equipping T1AM to play different functions depending on local expression of targets and enzymes involved in its biotransformation. Up to date, no information regarding TA1 mechanistic is available. We here review some of the main findings describing effects of T1AM (and TA1) which suggest these compounds interplay with the histaminergic system. These data reveal T1AM and TA1 are part of a network of signals involved in neuronal plasticity including neuroprotection and suggest T1AM and TA1 as lead compounds for a novel class of atypical psychoactive drugs.


Asunto(s)
Histamina/metabolismo , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Tironinas/farmacología , Animales , Humanos , Fármacos Neuroprotectores/uso terapéutico , Receptores Histamínicos/metabolismo , Tironinas/uso terapéutico
3.
Neurobiol Dis ; 151: 105271, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33482355

RESUMEN

Abnormalities in thyroid hormones (TH) availability and/or metabolism have been hypothesized to contribute to Alzheimer's disease (AD) and to be a risk factor for stroke. Recently, 3-iodothyronamine (T1AM), an endogenous amine putatively derived from TH metabolism, gained interest for its ability to promote learning and memory in the mouse. Moreover, T1AM has been demonstrated to rescue the ß-Amyloid dependent LTP impairment in the entorhinal cortex (EC), a brain area crucially involved in learning and memory and early affected during AD. In the present work, we have investigated the effect of T1AM on ischemia-induced EC synaptic dysfunction. In EC brain slices exposed to oxygen-glucose deprivation (OGD), we demonstrated that the acute perfusion of T1AM (5 µM) was capable of preventing ischemia-induced synaptic depression and that this protective effect was mediated by the trace amine-associated receptor 1 (TAAR1). Moreover, we demonstrated that activation of the BDNF-TrkB signalling is required for T1AM action during ischemia. The protective effect of T1AM was more evident when using EC slices from transgenic mutant human APP (mhAPP mice) that are more vulnerable to the effect of OGD. Our results confirm that the TH derivative T1AM can rescue synaptic function after transient ischemia, an effect that was also observed in a Aß-enriched environment.


Asunto(s)
Isquemia Encefálica/patología , Corteza Entorrinal/patología , Receptores Acoplados a Proteínas G/metabolismo , Tironinas/farmacología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Isquemia Encefálica/metabolismo , Corteza Entorrinal/efectos de los fármacos , Humanos , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
4.
J Physiol Pharmacol ; 71(3)2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33077693

RESUMEN

Obesity is a major health concern in modern societies as it is linked to diverse chronic diseases, such as diabetes, cancer, stroke, and skeletomuscular disorders. This study aimed to investigate the lipolytic potency of the metabolic suppressor 3-iodothyronamine (T1AM) and its molecular mechanism in differentiated 3T3-L1 adipocytes. Cells stained with Oil Red O showed a remarkable accumulation of lipid droplets by 20-d post-differentiation and a plateau at 26 - 30 day. Treatment with 100 µM T1AM for 6 h increased the liberation of free fatty acids (FFAs) and glycerol (P < 0.05) detected in the culture media. However, this stimulatory effect was significantly suppressed by ATGListatin, an inhibitor of adipose triglyceride lipase (ATGL), suggesting that ATGL plays a rate-limiting role in triglyceride (TG) turnover. To understand the lipolytic mechanism, immunoblotting and confocal image analyses of the T1AM-treated and control groups were conducted. The elevated lipolysis was accompanied by increases in the phosphorylation of adenosine monophosphate-activated protein kinase (p-AMPK), nuclear localization of forkhead box O1 (FoxO1), and expression of monoacylglycerol lipase (MGL) protein (P < 0.05). Finally, the treated cells exhibited downregulated expression of acetyl-CoA carboxylase (ACC) relative to p-ACC and increased protein expression of carnitine palmitoyltransferase 1 (CPT1) (P < 0.05). Taken together, T1AM showed lipolytic potency via activation of the AMPK/FoxO1/ATGL/MGL axis for decomposing TGs to FFAs and glycerol and of the AMPK/ACC/CPT1 pathway in facilitating the mobilization of FFAs into the mitochondria, highlighting its therapeutic potential for the treatment of obesity.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Proteína Forkhead Box O1/metabolismo , Lipólisis/efectos de los fármacos , Tironinas/farmacología , Células 3T3-L1 , Acetil-CoA Carboxilasa/metabolismo , Adipocitos/enzimología , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Glicerol/metabolismo , Lipasa/metabolismo , Ratones , Monoacilglicerol Lipasas/metabolismo , Fosforilación , Transducción de Señal , Triglicéridos/metabolismo
5.
Biomed Res Int ; 2020: 1302453, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32685439

RESUMEN

BACKGROUND: The thyroid hormone metabolite 3-iodothyronamine (3-T1AM) is rapidly emerging as a promising compound in decreasing the heart rate and lowering the cardiac output. The aim of our study was to fully understand the molecular mechanism of 3-T1AM on cardiomyocytes and its potential targets in cardiovascular diseases. MATERIALS AND METHODS: In our study, we utilized RNA-Seq to characterize the gene expression in H9C2 cells after 3-T1AM treatment. Comparative transcriptome analysis, including gene ontology, signaling pathways, disease connectivity analysis, and protein-protein interaction networks (PPI), was presented to find the critical gene function, hub genes, and related pathways. RESULTS: A total of 1494 differently expressed genes (DEGs) were identified (192 upregulated and 1302 downregulated genes) in H9C2 cells for 3-T1AM treatment. Of these, 90 genes were associated with cardiovascular diseases. The PPI analysis indicated that 5 hub genes might be the targets of 3-T1AM. Subsequently, eight DEGs characterized using RNA-Seq were confirmed by RT-qPCR assays. CONCLUSIONS: Our study provides a comprehensive analysis of 3-T1AM on H9C2 cells and delineates a new insight into the therapeutic intervention of 3-T1AM for the cardiovascular diseases.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Sustancias Protectoras/farmacología , Tironinas/farmacología , Transcriptoma/efectos de los fármacos , Animales , Biomarcadores/análisis , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/patología , Células Cultivadas , Perfilación de la Expresión Génica , Ontología de Genes , Terapia Molecular Dirigida , Miocitos Cardíacos/metabolismo , Mapas de Interacción de Proteínas/efectos de los fármacos , Ratas , Transducción de Señal
6.
Aquat Toxicol ; 225: 105547, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32623180

RESUMEN

Some chemicals in the environment disrupt thyroid hormone (TH) systems leading to alterations in organism development, but their effect mechanisms are poorly understood. In fish, this has been limited by a lack of fundamental knowledge on thyroid gene ontogeny and tissue expression in early life stages. Here we established detailed expression profiles for a suite of genes in the hypothalamic-pituitary-thyroid (HPT) axis of zebrafish (Danio rerio) between 24-120 h post fertilisation (hpf) and quantified their responses following exposure to 3,3',5-triiodo-L-thyronine (T3) using whole mount in situ hybridisation (WISH) and qRT-PCR (using whole-body extracts). All of the selected genes in the HPT axis demonstrated dynamic transcript expression profiles across the developmental stages examined. The expression of thyroid receptor alpha (thraa) was observed in the brain, gastrointestinal tract, craniofacial tissues and pectoral fins, while thyroid receptor beta (thrb) expression occurred in the brain, otic vesicles, liver and lower jaw. The TH deiodinases (dio1, dio2 and dio3b) were expressed in the liver, pronephric ducts and brain and the patterns differed depending on life stage. Both dio1 and dio2 were also expressed in the intestinal bulb (96-120 hpf), and dio2 expression occurred also in the pituitary (48-120 hpf). Exposure of zebrafish embryo-larvae to T3 (30 and 100 µg L-1) for periods of 48, 96 or 120 hpf resulted in the up-regulation of thraa, thrb, dio3b, thyroid follicle synthesis proteins (pax8) and corticotropin-releasing hormone (crhb) and down-regulation of dio1, dio2, glucuronidation enzymes (ugt1ab) and thyroid stimulating hormone (tshb) (assessed via qRT-PCR) and responses differed across life stage and tissues. T3 induced thraa expression in the pineal gland, pectoral fins, brain, somites, gastrointestinal tract, craniofacial tissues, liver and pronephric ducts. T3 enhanced thrb expression in the brain, jaw cartilage and intestine, while thrb expression was suppressed in the liver. T3 exposure suppressed the transcript levels of dio1 and dio2 in the liver, brain, gastrointestinal tract and craniofacial tissues, while dio2 signalling was also suppressed in the pituitary gland. Dio3b expression was induced by T3 exposure in the jaw cartilage, pectoral fins and brain. The involvement of THs in the development of numerous body tissues and the responsiveness of these tissues to T3 in zebrafish highlights their potential vulnerability to exposure to environmental thyroid-disrupting chemicals.


Asunto(s)
Triyodotironina/toxicidad , Contaminantes Químicos del Agua/toxicidad , Animales , Hormona Liberadora de Corticotropina , Hipotálamo/efectos de los fármacos , Larva/efectos de los fármacos , Hipófisis/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Tironinas/metabolismo , Tironinas/farmacología , Tirotropina , Pez Cebra/fisiología , Proteínas de Pez Cebra/genética
7.
Horm Mol Biol Clin Investig ; 41(1)2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32114521

RESUMEN

Background 3-Iodothyronamine (T1AM) is an endogenous messenger chemically related to thyroid hormone. Recent results indicate significant transcriptional effects of chronic T1AM administration involving the protein family of sirtuins, which regulate important metabolic pathways and tumor progression. Therefore, the aim of this work was to compare the effect of exogenous T1AM and 3,5,3'-triiodo-L-thyronine (T3) chronic treatment on mammalian sirtuin expression in hepatocellular carcinoma cells (HepG2) and in primary rat hepatocytes at micromolar concentrations. Materials and methods Sirtuin (SIRT) activity and expression were determined using a colorimetric assay and Western blot analysis, respectively, in cells treated for 24 h with 1-20 µM T1AM or T3. In addition, cell viability was evaluated by the MTTtest upon 24 h of treatment with 0.1-20 µM T1AM or T3. Results In HepG2, T1AM significantly reduced SIRT 1 (20 µM) and SIRT4 (10-20 µM) protein expression, while T3 strongly decreased the expression of SIRT1 (20 µM) and SIRT2 (any tested concentration). In primary rat hepatocytes, T3 decreased SIRT2 expression and cellular nicotinamide adenine dinucleotide (NAD) concentration, while on sirtuin activity it showed opposite effects, depending on the evaluated cell fraction. The extent of MTT staining was moderately but significantly reduced by T1AM, particularly in HepG2 cells, whereas T3 reduced cell viability only in the tumor cell line. Conclusions T1AM and T3 downregulated the expression of sirtuins, mainly SIRT1, in hepatocytes, albeit in different ways. Differences in mechanisms are only observational, and further investigations are required to highlight the potential role of T1AM and T3 in modulating sirtuin expression and, therefore, in regulating cell cycle or tumorigenesis.


Asunto(s)
Sirtuina 1/metabolismo , Tironinas/farmacología , Triyodotironina/análogos & derivados , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Células Hep G2 , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Ratas , Ratas Wistar , Sirtuina 1/genética , Sirtuinas/genética , Sirtuinas/metabolismo , Triyodotironina/farmacología
8.
Med Sci Monit ; 26: e923195, 2020 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-32162616

RESUMEN

BACKGROUND The thyroid hormone metabolite 3-iodothyronamine (T1AM) is rapidly emerging as promising compound of decreasing heart rate and lowering cardiac output. The aim of our study was to fully understand the molecular mechanism of T1AM on cardiomyocytes and its potential targets in cardiovascular diseases. MATERIAL AND METHODS We developed an in vitro myocardial ischemia-reperfusion injury model of AC-16 cells by hypoxia-reoxygenation injury. Cell viability of AC-16 cells was detected using CCK-8 assay and apoptosis was detected by flow cytometry. RNA-seq was used to characterize the gene expression in H/R-induced AC-16 cells after T1AM treatment. The mRNA levels of FoxO1, PPARalpha, Akt, and GCK and the protein levels of PPARalpha, GCK, and components of the Akt/FoxO1 pathway were detected by qRT-PCR and Western blotting, respectively. RESULTS Exogenous T1AM increased the H/R-induced AC-16 cell viability in a relatively low concentration. A total of 210 DEGs, including 142 upregulated and 68 downregulated genes, were determined in H/R-induced AC-16 cells treated with or without T1AM. A Venn diagram showed 135 common DEGs. The FoxO signaling pathway was identified via KEGG enrichment analysis of these 135 DEGs. Moreover, T1AM mediated hypometabolism and reduced the apoptosis of H/R-induced AC-16 cells via the Akt/FoxO1 pathway. CONCLUSIONS Exogenous T1AM protects against cell injury induced by H/R in AC-16 cells via regulation of the FoxO signaling pathway. Our results suggest that T1AM can play a preventive role in myocardial H/R injury and also provide new insight for clinical management of AMI patients.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Tironinas/farmacología , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Humanos , Infarto del Miocardio/complicaciones , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Tironinas/uso terapéutico
9.
Molecules ; 25(5)2020 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-32110992

RESUMEN

3-iodothyronamine (T1AM) and the recently developed analog SG-2 are rapidly emerging as promising multi-target neuroprotective ligands able to reprogram lipid metabolism and to produce memory enhancement in mice. To elucidate the molecular mechanisms underlying the multi-target effects of these novel drug candidates, here we investigated whether the modulation of SIRT6, known to play a key role in reprogramming energy metabolism, might also drive the activation of clearing pathways, such as autophagy and ubiquitine-proteasome (UP), as further mechanisms against neurodegeneration. We show that both T1AM and SG-2 increase autophagy in U87MG cells by inducing the expression of SIRT6, which suppresses Akt activity thus leading to mTOR inhibition. This effect was concomitant with down-regulation of autophagy-related genes, including Hif1α, p53 and mTOR. Remarkably, when mTOR was inhibited a concomitant activation of autophagy and UP took place in U87MG cells. Since both compounds activate autophagy, which is known to sustain long term potentiation (LTP) in the entorhinal cortex (EC) and counteracting AD pathology, further electrophysiological studies were carried out in a transgenic mouse model of AD. We found that SG-2 was able to rescue LTP with an efficacy comparable to T1AM, further underlying its potential as a novel pleiotropic agent for neurodegenerative disorders treatment.


Asunto(s)
Gangliósidos/farmacología , Fármacos Neuroprotectores/farmacología , Sirtuinas/metabolismo , Tironinas/farmacología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/ultraestructura , Autofagia/efectos de los fármacos , Autofagia/genética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Corteza Entorrinal/patología , Gangliósidos/química , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Potenciación a Largo Plazo/efectos de los fármacos , Ratones Transgénicos , Fármacos Neuroprotectores/química , Serina-Treonina Quinasas TOR/metabolismo , Tironinas/química
10.
Thyroid ; 30(1): 147-160, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31709926

RESUMEN

Background: A novel form of thyroid hormone (TH) signaling is represented by 3-iodothyronamine (T1AM), an endogenous TH derivative that interacts with specific molecular targets, including trace amine-associated receptor 1 (TAAR1), and induces pro-learning and anti-amnestic effects in mice. Dysregulation of TH signaling has long been hypothesized to play a role in Alzheimer's disease (AD). In the present investigation, we explored the neuroprotective role of T1AM in beta amyloid (Aß)-induced synaptic and behavioral impairment, focusing on the entorhinal cortex (EC), an area that is affected early by AD pathology. Methods: Field potentials were evoked in EC layer II, and long-term potentiation (LTP) was elicited by high frequency stimulation (HFS). T1AM (5 µM) and/or Aß(1-42) (200 nM), were administered for 10 minutes, starting 5 minutes before HFS. Selective TAAR1 agonist RO5166017 (250 nM) and TAAR1 antagonist EPPTB (5 nM) were also used. The electrophysiological experiments were repeated in EC-slices taken from a mouse model of AD (mutant human amyloid precursor protein [mhAPP], J20 line). We also assessed the in vivo effects of T1AM on EC-dependent associative memory deficits, which were detected in mhAPP mice by behavioral evaluations based on the novel-object recognition paradigm. TAAR1 expression was determined by Western blot, whereas T1AM and its metabolite 3-iodothyroacetic acid (TA1) were assayed by high-performance liquid chromatography coupled to mass spectrometry. Results: We demonstrate the presence of endogenous T1AM and TAAR1 in the EC of wild-type and mhAPP mice. Exposure to Aß(1-42) inhibited LTP, and T1AM perfusion (at a concentration of 5 µM, leading to an actual concentration in the perfusion buffer ranging from 44 to 298 nM) restored it, whereas equimolar amounts of 3,5,3'-triiodo-L-thyronine (T3) and TA1 were ineffective. The response to T1AM was abolished by the TAAR1 antagonist EPPTB, whereas it was mimicked by the TAAR1 agonist RO5166017. In the EC of APPJ20 mice, LTP could not be elicited, but it was rescued by T1AM. The intra-cerebro-ventricular administration of T1AM (0.89 µg/kg) also restored recognition memory that was impaired in mhAPP mice. Conclusions: Our results suggest that T1AM and TAAR1 are part of an endogenous system that can be modulated to prevent synaptic and behavioral deficits associated with Aß-related toxicity.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Corteza Entorrinal/efectos de los fármacos , Potenciales Evocados/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Tironinas/farmacología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Corteza Entorrinal/fisiología , Potenciales Evocados/fisiología , Ratones , Ratones Transgénicos
11.
Int J Mol Sci ; 20(16)2019 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-31434215

RESUMEN

3-Iodothyronamine (T1AM) and its synthetic analog SG-2 are rapidly emerging as promising drivers of cellular metabolic reprogramming. Our recent research indicates that in obese mice a sub-chronic low dose T1AM treatment increased lipolysis, associated with significant weight loss independent of food consumption. The specific cellular mechanism of T1AM's lipolytic effect and its site of action remains unknown. First, to study the mechanism used by T1AM to gain entry into cells, we synthesized a fluoro-labeled version of T1AM (FL-T1AM) by conjugating it to rhodamine (TRITC) and analyzed its cellular uptake and localization in 3T3-L1 mouse adipocytes. Cell imaging using confocal microscopy revealed a rapid intercellular uptake of FL-T1AM into mitochondria without localization to the lipid droplet or nucleus of mature adipocytes. Treatment of 3T3-L1 adipocytes with T1AM and SG-2 resulted in decreased lipid accumulation, the latter showing a significantly higher potency than T1AM (10 µM vs. 20 µM, respectively). We further examined the effects of T1AM and SG-2 on liver HepG2 cells. A significant decrease in lipid accumulation was observed in HepG2 cells treated with T1AM or SG-2, due to increased lipolytic activity. This was confirmed by accumulation of glycerol in the culture media and through activation of the AMPK/ACC signaling pathways.


Asunto(s)
Tironinas/farmacología , Células 3T3-L1 , Animales , Reprogramación Celular/efectos de los fármacos , Glicerol/metabolismo , Células Hep G2 , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
12.
Mol Cell Endocrinol ; 493: 110448, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31100496

RESUMEN

Thyroid hormone receptors (TRs) are ligand-dependent transcription factors that activate or repress gene transcription, resulting in the regulation of numerous physiological programs. While 3,3',5-L-triiodothyronine is the TR cognate ligand, these receptors can also be activated by various alternative ligands, including endogenous and synthetic molecules capable of inducing diverse active receptor conformations that influence thyroid hormone-dependent signaling pathways. This review mainly discusses current knowledge on 3,5-diiodo-L-thyronine and 3,5,3'-triiodothyroacetic acid, two endogenous molecules that bind to TRs and regulate gene expression; and the molecular interactions between TRs and ligands, like synthetic thyromimetics developed to target specific TR isoforms for tissue-specific regulation of thyroid-related disorders, or endocrine disruptors that have allowed the design of new analogues and revealed essential amino acids for thyroid hormone binding.


Asunto(s)
Diyodotironinas/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Tironinas/síntesis química , Triyodotironina/análogos & derivados , Animales , Mimetismo Biológico , Diyodotironinas/química , Diseño de Fármacos , Regulación de la Expresión Génica , Humanos , Ligandos , Especificidad de Órganos , Receptores de Hormona Tiroidea/química , Transducción de Señal/efectos de los fármacos , Tironinas/química , Tironinas/farmacología , Triyodotironina/química , Triyodotironina/metabolismo
13.
Neurochem Int ; 129: 104460, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31075293

RESUMEN

Thyroid hormone and thyroid hormone metabolites, including 3-iodothyronamine (T1AM) and 3-iodothyroacetic acid (TA1), activate AKT signaling in hippocampal neurons affording protection from excitotoxic damage. We aim to explore whether the mechanism of T1AM neuroprotection against kainic acid (KA)-induced excitotoxicity included the activation of the trace amine associated receptor isoform 1 (TAAR1), one of T1AM targets. Rat organotypic hippocampal slices were exposed to vehicle (Veh) or to 5 µM kA for 24 h in the absence or presence of 0.1, 1 and 10 µM T1AM or to 0.1, 1 and 10 µM T1AM and 1 µM N-(3-Ethoxy-phenyl)-4-pyrrolidin-1-yl-3-trifluoromethyl-benzamide (EPPTB), the only available TAAR1 antagonist, or to 1 µM T1AM in the absence or in the presence of 10 µM LY294002, an inhibitor of phosphoinositide 3-kinases (PI3Ks). Cell death was evaluated by measuring propidium iodide (PI) levels of fluorescence 24 h after treatment. In parallel, the expression levels of p-AKT and p-PKA were evaluated by Western blot analysis of slice lysates. The activity of mitochondrial monoamine oxidases (MAO) was assayed fluorimetrically. 24 h exposure of slices to T1AM resulted in the activation of AKT and PKA. KA exposure induced cell death in the CA3 region and significantly reduced p-AKT and p-PKA levels. The presence of 1 and 10 µM T1AM significantly protected neurons from death and conserved both kinase levels with the essential role of AKT in neuroprotection. Furthermore, EPPTB prevented T1AM-induced neuroprotection, activation of PKA and AKT. Of note, in the presence of EPPTB T1AM degradation by MAO was reduced. Our results indicate that the neuroprotection offered by T1AM depends, as for TA1, on AKT activation but do not allow to conclusively indicate TAAR1 as the target implicated.


Asunto(s)
Benzamidas/farmacología , Ácido Kaínico/toxicidad , Neuronas/efectos de los fármacos , Pirrolidinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Neuronas/metabolismo , Neuroprotección/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas Wistar , Receptores Acoplados a Proteínas G , Tironinas/farmacología
14.
Int J Mol Sci ; 19(5)2018 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-29786646

RESUMEN

Obesity is a complex disease associated with environmental and genetic factors. 3-Iodothyronamine (T1AM) has revealed great potential as an effective weight loss drug. We used metabolomics and associated transcriptional gene and protein expression analysis to investigate the tissue specific metabolic reprogramming effects of subchronic T1AM treatment at two pharmacological daily doses (10 and 25 mg/kg) on targeted metabolic pathways. Multi-analytical results indicated that T1AM at 25 mg/kg can act as a novel master regulator of both glucose and lipid metabolism in mice through sirtuin-mediated pathways. In liver, we observed an increased gene and protein expression of Sirt6 (a master gene regulator of glucose) and Gck (glucose kinase) and a decreased expression of Sirt4 (a negative regulator of fatty acids oxidation (FAO)), whereas in white adipose tissue only Sirt6 was increased. Metabolomics analysis supported physiological changes at both doses with most increases in FAO, glycolysis indicators and the mitochondrial substrate, at the highest dose of T1AM. Together our results suggest that T1AM acts through sirtuin-mediated pathways to metabolically reprogram fatty acid and glucose metabolism possibly through small molecules signaling. Our novel mechanistic findings indicate that T1AM has a great potential as a drug for the treatment of obesity and possibly diabetes.


Asunto(s)
Fármacos Antiobesidad/farmacología , Proteínas Mitocondriales/genética , Obesidad/metabolismo , Sirtuinas/genética , Tironinas/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Fármacos Antiobesidad/uso terapéutico , Ácidos Grasos/metabolismo , Femenino , Quinasas del Centro Germinal , Glucosa/metabolismo , Glucólisis , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , Obesidad/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Sirtuinas/metabolismo , Tironinas/uso terapéutico
15.
Mol Cell Endocrinol ; 460: 219-228, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28754352

RESUMEN

PURPOSE: 3-iodothyronamine (3-T1AM), a decarboxylated and deiodinated thyroid hormone metabolite, leads at pharmacological doses to hypoinsulinemia, hyperglucagonemia and hyperglycemia in vivo. As the pancreatic Langerhans islets express thyroid hormone transmembrane transporters (THTT), we tested the hypothesis that not only plasma membrane-mediated 3-T1AM binding to and activation of G-protein coupled receptors, but also 3-T1AM metabolite(s) generated by 3-T1AM uptake and metabolism might decrease glucose-stimulated insulin secretion (GSIS). METHODS: Murine pancreatic ß-cells MIN6 were characterized for gene expression of THTT, deiodinases and monoamine oxidases. 3-T1AM uptake and intracellular metabolism to the corresponding 3-iodothyroacetic acid were analysed by liquid-chromatography tandem mass spectrometry (LC-MS/MS) at different time points in cells as well as the conditioned medium. Mitochondrial activity, especially ATP-production, was monitored real-time after 3-T1AM application using Seahorse Bioanalyzer technique. Effect of 3-T1AM on GSIS into the culture medium was assayed by ELISA. RESULTS: MIN6 cells express classical THTT, proposed to transport 3-T1AM, as well as 3-T1AM metabolizing enzymes comparable to murine primary pancreatic islets. 3-T1AM accumulates in MIN6 cells and is metabolized by intracellular MaoB to 3-iodothyroacetic, which in turn is rapidly exported. 3-T1AM decreases mitochondrial ATP-production concentration dependently. GSIS is diminished by 3-T1AM treatment. Using LC-MS/MS, no further 3-T1AM metabolites except 3-iodothyroacetic were detectable. CONCLUSIONS: This data provides a first link between cellular 3-T1AM uptake and regulation of mitochondrial energy metabolism in ß-cells, resulting in reduced insulin secretion. We conclude that MIN6 is an appropriate cell model to study 3-T1AM-dependent (intra-)cellular biochemical mechanisms affecting insulin production in vitro.


Asunto(s)
Secreción de Insulina/efectos de los fármacos , Mitocondrias/metabolismo , Tironinas/farmacología , Animales , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Metaboloma , Ratones , Mitocondrias/efectos de los fármacos , Tironinas/química
16.
Neurochem Int ; 115: 31-36, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29032008

RESUMEN

3-iodothyroacetic acid (TA1) is among the by-products of thyroid hormone metabolism suspected to mediate the non-genomic effects of the hormone (T3). We aim to investigate whether TA1 systemically administered to mice stimulated mice wakefulness, an effect already described for T3 and for another T3 metabolite (i.e. 3-iodothryonamine; T1AM), and whether TA1 interacted at GABA-A receptors (GABA-AR). Mice were pre-treated with either saline (vehicle) or TA1 (1.32, 4 and 11 µg/kg) and, after 10 min, they received ethanol (3.5 g/kg, i.p.). In another set of experiments, TA1 was administered 5 min after ethanol. The latency of sleep onset and the time of sleep duration were recorded. Voltage-clamp experiments to evaluate the effect of 1 µM TA1 on bicuculline-sensitive currents in acute rat hippocampal slice neurons and binding experiments evaluating the capacity of 1, 10, 100 µM TA1 to displace [3H]flumazenil from mice brain membranes were also performed. 4 µg/kg TA1 increases the latency of onset and at 1.32 and 4 µg/kg it reduces the duration of ethanol-induced sleep only if administered before ethanol. TA1 does not functionally interact at GABA-AR. Overall these results indicate a further similarity between the pharmacological profile of TA1 and that of T1AM.


Asunto(s)
Antitiroideos/farmacología , Hipocampo/efectos de los fármacos , Receptores de GABA-A/efectos de los fármacos , Tironinas/farmacología , Animales , Etanol/farmacología , Hipocampo/metabolismo , Hipnóticos y Sedantes/farmacología , Masculino , Ratones , Ratas Wistar , Receptores de GABA-A/metabolismo , Hormonas Tiroideas/metabolismo , Tironinas/metabolismo
17.
Horm Behav ; 94: 93-96, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28711308

RESUMEN

We previously demonstrated that 3-iodothyronamine (T1AM), a by-product of thyroid hormone metabolism, pharmacologically administered to mice acutely stimulated learning and memory acquisition and provided hyperalgesia with a mechanism which remains to be defined. We now aimed to investigate whether the T1AM effect on memory and pain was maintained in mice pre-treated with scopolamine, a non-selective muscarinic antagonist expected to induce amnesia and, possibly, hyperalgesia. Mice were pre-treated with scopolamine and, after 20min, injected intracerebroventricularly (i.c.v.) with T1AM (0.13, 0.4, 1.32µg/kg). 15min after T1AM injection, the mice learning capacity or their pain threshold were evaluated by the light/dark box and by the hot plate test (51.5°C) respectively. Experiments in the light/dark box were repeated in mice receiving clorgyline (2.5mg/kg, i.p.), a monoamine oxidase (MAO) inhibitor administered 10min before scopolamine (0.3mg/kg). Our results demonstrated that 0.3mg/kg scopolamine induced amnesia without modifying the murine pain threshold. T1AM fully reversed scopolamine-induced amnesia and produced hyperalgesia at a dose as low as 0.13µg/kg. The T1AM anti-amnestic effect was lost in mice pre-treated with clorgyline. We report that the removal of muscarinic signalling increases T1AM pro learning and hyperalgesic effectiveness suggesting T1AM as a potential treatment as a "pro-drug" for memory dysfunction in neurodegenerative diseases.


Asunto(s)
Memoria/efectos de los fármacos , Dolor/inducido químicamente , Escopolamina/efectos adversos , Tironinas/farmacología , Amnesia/inducido químicamente , Amnesia/prevención & control , Animales , Hiperalgesia/inducido químicamente , Hiperalgesia/prevención & control , Aprendizaje/efectos de los fármacos , Masculino , Ratones , Inhibidores de la Monoaminooxidasa/farmacología , Dolor/prevención & control , Umbral del Dolor/efectos de los fármacos , Profármacos/administración & dosificación , Profármacos/farmacología , Escopolamina/administración & dosificación , Tironinas/administración & dosificación , Factores de Tiempo
18.
Endocrinology ; 158(6): 1977-1984, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368510

RESUMEN

3-Iodothyronamine (3-T1AM) is an endogenous thyroid hormone (TH)-derived metabolite that induces severe hypothermia in mice after systemic administration; however, the underlying mechanisms have remained enigmatic. We show here that the rapid 3-T1AM-induced loss in body temperature is a consequence of peripheral vasodilation and subsequent heat loss (e.g., over the tail surface). The condition is subsequently intensified by hypomotility and a lack of brown adipose tissue activation. Although the possible 3-T1AM targets trace amine-associated receptor 1 or α2a-adrenergic receptor were detected in tail artery and aorta respectively, myograph studies did not show any direct effect of 3-T1AM on vasodilation, suggesting that its actions are likely indirect. Intracerebroventricular application of 3-T1AM, however, replicated the phenotype of tail vasodilation and body temperature decline and led to neuronal activation in the hypothalamus, suggesting that the metabolite causes tail vasodilation through a hypothalamic signaling pathway. Consequently, the 3-T1AM response constitutes anapyrexia rather than hypothermia and closely resembles the heat-stress response mediated by hypothalamic temperature-sensitive neurons. Our results thus underline the well-known role of the hypothalamus as the body's thermostat and suggest an additional molecular link between TH signaling and the central control of body temperature.


Asunto(s)
Encéfalo/fisiología , Cola (estructura animal)/irrigación sanguínea , Tironinas/farmacología , Vasodilatación/efectos de los fármacos , Animales , Regulación de la Temperatura Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Infusiones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de los fármacos , Cola (estructura animal)/efectos de los fármacos , Tironinas/administración & dosificación
19.
Biol Pharm Bull ; 40(5): 576-582, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28163294

RESUMEN

The homeostasis of muscle properties depends on both physical and metabolic stresses. Whereas physical stress entails metabolic response for muscle homeostasis, the latter does not necessarily involve the former and may thus solely affect the homeostasis. We here report that metabolic suppression by the hypometabolic agent 3-iodothyronamine (T1AM) induced muscle cell atrophy without physical stress. We observed that the oxygen consumption rate of C2C12 myotubes decreased 40% upon treatment with 75 µM T1AM for 6 h versus 10% in the vehicle (dimethyl sulfoxide) control. The T1AM treatment reduced cell diameter of myotubes by 15% compared to the control (p<0.05). The cell diameter was reversed completely by 9 h after T1AM was removed. The T1AM treatment also significantly suppressed the expression levels of heat shock protein 72 and αB-crystallin as well as the phosphorylation levels of Akt1, mammalian target of rapamycin (mTOR), S6K, forkhead box O1 (FoxO1) and FoxO3. In contrast, the levels of ubiquitin E3 ligase MuRF1 and chymotrypsin-like activity of proteasome were significantly elevated by T1AM treatment. These results suggest that T1AM-mediated metabolic suppression induced muscle cell atrophy via activation of catabolic signaling and inhibition of anabolic signaling.


Asunto(s)
Proteína Forkhead Box O1/fisiología , Atrofia Muscular/inducido químicamente , Complejo de la Endopetidasa Proteasomal/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Quinasas S6 Ribosómicas/fisiología , Transducción de Señal/fisiología , Tironinas/farmacología , Animales , Células Cultivadas , Regulación hacia Abajo , Ratones , Fibras Musculares Esqueléticas , Serina-Treonina Quinasas TOR/fisiología
20.
Physiol Rep ; 5(1)2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28082426

RESUMEN

Complex diseases such as polycystic ovary syndrome (PCOS) are associated with intricate pathophysiological, hormonal, and metabolic feedbacks that make their early diagnosis challenging, thus increasing the prevalence risks for obesity, cardiovascular, and fatty liver diseases. To explore the crosstalk between endocrine and lipid metabolic pathways, we administered 3-iodothyronamine (T1AM), a natural analog of thyroid hormone, in a mouse model of PCOS and analyzed plasma and tissue extracts using multidisciplinary omics and biochemical approaches. T1AM administration induces a profound tissue-specific antilipogenic effect in liver and muscle by lowering gene expression of key regulators of lipid metabolism, PTP1B and PLIN2, significantly increasing metabolites (glucogenic, amino acids, carnitine, and citrate) levels, while enhancing protection against oxidative stress. In contrast, T1AM has an opposing effect on the regulation of estrogenic pathways in the ovary by upregulating STAR, CYP11A1, and CYP17A1. Biochemical measurements provide further evidence of significant reduction in liver cholesterol and triglycerides in post-T1AM treatment. Our results shed light onto tissue-specific metabolic vs. hormonal pathway interactions, thus illuminating the intricacies within the pathophysiology of PCOS This study opens up new avenues to design drugs for targeted therapeutics to improve quality of life in complex metabolic diseases.


Asunto(s)
Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/genética , Redes y Vías Metabólicas/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Tironinas/administración & dosificación , Animales , Colesterol/metabolismo , Femenino , Expresión Génica/genética , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Redes y Vías Metabólicas/genética , Metabolómica/métodos , Ratones , Músculos/metabolismo , Obesidad/metabolismo , Ovario/metabolismo , Estrés Oxidativo/efectos de los fármacos , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/fisiopatología , Calidad de Vida , Tironinas/metabolismo , Tironinas/farmacología , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...