Thyronamine

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 219 Experts worldwide ranked by ideXlab platform

Josef Köhrle - One of the best experts on this subject based on the ideXlab platform.

  • Canonical TSH Regulation of Cathepsin-Mediated Thyroglobulin Processing in the Thyroid Gland of Male Mice Requires Taar1 Expression
    Frontiers Media S.A., 2018
    Co-Authors: Maria Qatato, Josef Köhrle, Joanna Szumska, Vladislav Skripnik, Eddy Rijntjes, Klaudia Brix
    Abstract:

    Trace amine-associated receptor 1 (Taar1) has been suggested as putative receptor of Thyronamines. These are aminergic messengers with potential metabolic and neurological effects countering their contingent precursors, the thyroid hormones (THs). Recently, we found Taar1 to be localized at the primary cilia of rodent thyroid epithelial cells in vitro and in situ. Thus, Taar1 is present in a location of thyroid follicles where it might be involved in regulation of cathepsin-mediated proteolytic processing of thyroglobulin, and consequently TH synthesis. In this study, taar1 knock-out male mice (taar1-/-) were used to determine whether Taar1 function would entail differential alterations in thyroid states of young and adult animals. Analyses of blood serum revealed unaltered T4 and T3 concentrations and unaltered T3-over-T4 ratios upon Taar1 deficiency accompanied, however, by elevated TSH concentrations. Interestingly, TSH receptors, typically localized at the basolateral plasma membrane domain of wild type controls, were located at vesicular membranes in thyrocytes of taar1-/- mice. In addition, determination of epithelial extensions in taar1-/- thyroids showed prismatic cells, which might indicate activation states higher than in the wild type. While gross degradation of thyroglobulin was comparable to controls, deregulated thyroglobulin turnover in taar1-/- mice was indicated by luminal accumulation of covalently cross-linked thyroglobulin storage forms. These findings were in line with decreased proteolytic activities of thyroglobulin-solubilizing and -processing proteases, due to upregulated cystatins acting as their endogenous inhibitors in situ. In conclusion, Taar1-deficient mice are hyperthyrotropinemic in the absence of respective signs of primary hypothyroidism such as changes in body weight or TH concentrations in blood serum. Thyrocytes of taar1-/- mice are characterized by non-canonical TSH receptor localization in intracellular compartments, which is accompanied by altered thyroglobulin turnover due to a disbalanced proteolytic network. These finding are of significance considering the rising popularity of using TAAR1 agonists or antagonists as neuromodulating pharmacological drugs. Our study highlights the importance of further evaluating potential off-target effects regarding TSH receptor mislocalization and the thyroglobulin processing machinery, which may not only affect the TH-generating thyroid gland, but may emanate to other TH target organs like the CNS dependent on their proper supply

  • avoiding the pitfalls when quantifying thyroid hormones and their metabolites using mass spectrometric methods the role of quality assurance
    Molecular and Cellular Endocrinology, 2017
    Co-Authors: Keith Richards, Eddy Rijntjes, Daniel Rathmann, Josef Köhrle
    Abstract:

    Abstract This short review aims to assess the application of basic quality assurance (QA) principles in published thyroid hormone bioanalytical methods using mass spectrometry (MS). The use of tandem MS, in particular linked to liquid chromatography has become an essential bioanalytical tool for the thyroid hormone research community. Although basic research laboratories do not usually work within the constraints of a quality management system and regulated environment, all of the reviewed publications, to a lesser or greater extent, document the application of QA principles to the MS methods described. After a brief description of the history of MS in thyroid hormone analysis, the article reviews the application of QA to published bioanalytical methods from the perspective of selectivity, accuracy, precision, recovery, instrument calibration, matrix effects, sensitivity and sample stability. During the last decade the emphasis has shifted from developing methods for the determination of L-thyroxine (T4) and 3,3′,5-triiodo-L-thyronine (T3), present in blood serum/plasma in the 1–100 nM concentration range, to metabolites such as 3-iodo-L-Thyronamine (3-T1AM), 3,5-diiodo-L-thyronine (3,5-T2) and 3,3’-diiodo-L-thyronine (3,3′-T2). These metabolites seem likely to be present in the low pM concentrations; consequently, QA parameters such as selectivity and sensitivity become more critical. The authors conclude that improvements, particularly in the areas of analyte selectivity, matrix effect measurement/documentation and analyte recovery would be beneficial.

  • trace amine associated receptor 1 localization at the apical plasma membrane domain of fisher rat thyroid epithelial cells is confined to cilia
    European thyroid journal, 2015
    Co-Authors: Joanna Szumska, David K. Grandy, Maren Rehders, Heike Biebermann, Josef Köhrle, Maria Qatato, Dagmar Fuhrer, Klaudia Brix
    Abstract:

    Background: The trace amine-associated receptor 1 (Taar1) is one member of the Taar family of G-protein-coupled receptors (GPCR) accepting various biogenic amines as ligands. It has been proposed that Taar1 mediates rapid, membrane-initiated effects of Thyronamines, the endogenous decarboxylated and deiodinated relatives of the classical thyroid hormones T4 and T3. Objectives: Although the physiological actions of Thyronamines in general and 3-iodoThyronamine (T1AM) in particular are incompletely understood, studies published to date suggest that synthetic T1AM-activated Taar1 signaling antagonizes thyromimetic effects exerted by T3. However, the location of Taar1 is currently unknown. Methods: To fill this gap in our knowledge we employed immunofluorescence microscopy and a polyclonal antibody to detect Taar1 protein expression in thyroid tissue from Fisher rats, wild-type and taar1-deficient mice, and in the polarized FRT cells. Results: With this approach we found that Taar1 is expressed in the membranes of subcellular compartments of the secretory pathway and on the apical plasma membrane of FRT cells. Three-dimensional analyses further revealed Taar1 immunoreactivity in cilial extensions of postconfluent FRT cell cultures that had formed follicle-like structures. Conclusions: The results suggest Taar1 transport along the secretory pathway and its accumulation in the primary cilium of thyrocytes. These findings are of significance considering the increasing interest in the role of cilia in harboring functional GPCR. We hypothesize that Thyronamines can reach and activate Taar1 in thyroid follicular epithelia by acting from within the thyroid follicle lumen, their potential site of synthesis, as part of a nonclassical mechanism of thyroid autoregulation.

  • detection of 3 5 diiodothyronine in sera of patients with altered thyroid status using a new monoclonal antibody based chemiluminescence immunoassay
    Thyroid, 2014
    Co-Authors: Ina Lehmphul, G Brabant, Henri Wallaschofski, Marek Ruchala, Christian J Strasburger, Josef Köhrle
    Abstract:

    Background: 3,5-Diiodo-L-thyronine (3,5-T2), a potential metabolite of 3,3′,5-triiodothyronine (T3), exerts marked metabolic actions without the undesirable cardiac and central side effects of T3. So far the lack of reliable quantification methods for endogenous 3,5-T2 in human serum has limited further insight into its physiological and pathophysiological roles in endocrine homeostasis and disease status. Methods: Monoclonal anti-3,5-T2 antibodies (3,5-T2 mAbs) were produced in mice. We developed a competitive chemiluminescence immunoassay (CLIA) with one selected mAb and optimized it for high sensitivity, linearity, recovery, and low cross-reactivity to structurally related thyroid hormones (THs) and Thyronamines. The CLIA was then used to investigate the origin and action of 3,5-T2 in humans under physiological and pathophysiological conditions in comparison with THs. Patient analysis included individuals with confirmed hypo- or hyperthyroidism and a separate population of thyroidectomized patients on ...

  • development of a validated liquid chromatography tandem mass spectrometry method for the distinction of thyronine and Thyronamine constitutional isomers and for the identification of new deiodinase substrates
    Rapid Communications in Mass Spectrometry, 2008
    Co-Authors: Susanne Piehl, Thomas S Scanlan, Thomas Heberer, Gabor Balizs, Josef Köhrle
    Abstract:

    Thyronines (THs) and Thyronamines (TAMs) are two groups of endogenous iodine-containing signaling molecules whose representatives differ from each other only regarding the number and/or the position of the iodine atoms. Both groups of compounds are substrates of three deiodinase isozymes, which catalyze the sequential reductive removal of iodine from the respective precursor molecule. In this study, a novel analytical method applying liquid chromatography/tandem mass spectrometry (LC-MS/MS) was developed. This method permitted the unequivocal, simultaneous identification and quantification of all THs and TAMs in the same biological sample. Furthermore, a liquid-liquid extraction procedure permitting the concurrent isolation of all THs and TAMs from biological matrices, namely deiodinase (Dio) reaction mixtures, was established. Method validation experiments with extracted TH and TAM analytes demonstrated that the method was selective, devoid of matrix effects, sensitive, linear over a wide range of analyte concentrations and robust in terms of reproducible recoveries, process efficiencies as well as intra-assay and inter-assay stability parameters. The method was applied to study the deiodination reactions of iodinated THs catalyzed by the three deiodinase isozymes. With the HPLC protocol developed herein, sufficient chromatographic separation of all constitutional TH and TAM isomers was achieved. Accordingly, the position of each iodine atom removed from a TH substrate in a Dio-catalyzed reaction was backtracked unequivocally. While several established deiodination reactions were verified, two as yet unknown reactions, namely the phenolic ring deiodination of 3',5'-diiodothyronine (3',5'-T2) by Dio2 and the tyrosyl ring deiodination of 3-monoiodothyronine (3-T1) by Dio3, were newly identified.

Riccardo Zucchi - One of the best experts on this subject based on the ideXlab platform.

  • Endogenous TH metabolite 3-iodoThyronamine (T1AM) and synthetic Thyronamine-like analogues SG-1 and SG-2 induce autophagy in human glioblastoma cells (U-87MG)
    Endocrine Abstracts, 2017
    Co-Authors: Lorenza Bellusci, Riccardo Zucchi, Giulia Nesi, Martina Sabatini, Paola Lenzi, Simona Rapposelli, Francesco Fornai, Leonardo Rossi, Grazia Chiellini
    Abstract:

    Endogenous TH metabolite 3-iodoThyronamine (T1AM) and recently developed Thyronamine-like synthetic analogues SG1 and SG2 have emerged as neuroprotective agents. Autophagy has proved to be an effective therapeutic approach for neurodegenerative diseases. Therefore we investigated whether these compounds can induce autophagy in human glioblastoma cells (U-87MG).\ud \ud Cultured U-87MG cells were initially treated with 1 μM T1AM, SG-1, SG-2 or vehicle for 30’, 4, 8 and 24 h and autophagy was monitored morphologically by assessing the presence of autophagic vacuoles and LC3-II puncta formation with transmission electron microscopy (TEM) and immunofluorescence (IF) microscopy, respectively. In addition, cellular lysates were subjected to western blotting to assess the accumulation of microtubule-associated protein 1 light chain 3 (LC3). This protein is widely used as a marker for autophagy because it is a structural protein vital in autophagosome formation. TEM and IF microscopy showed a significant time dependent increase of autophagy-like vacuoles density and LC3 punta formation in U-87MG cells exposed to the treatment with test compounds, with T1AM and SG-1 being the most effective. Along with extensive cytoplasmic vacuolization, western blotting analysis revealed significant up regulation of LC3-II expression (P

  • Cardiac actions of thyroid hormone metabolites
    Molecular and cellular endocrinology, 2017
    Co-Authors: Grazia Rutigliano, Riccardo Zucchi
    Abstract:

    Thyroid hormones (THs) have a major role in regulating cardiac function. Their classical mechanism of action is genomic. Recent findings have broadened our knowledge about the (patho)physiology of cardiac regulation by THs, to include non-genomic actions of THs and their metabolites (THM). This review provides an overview of classical and non-classical cardiac effects controlled by: i) iodothyronines (thyroxine, T4; 3,5,3’-triiodothyronine,T3; 3, 5-diiodothyronine, T2); ii) Thyronamines (Thyronamine, T0AM; 3-iodoThyronamine, T1AM); and iii) iodothyroacetic acids (3, 5, 3′, 5’-tetraiodothyroacetic acid, tetrac; 3, 5, 3’-triiodothyroacetic acid, triac; 3-iodothyroacetic acid, TA1). Whereas iodothyronines enhance both diastolic and systolic function and heart rate, Thyronamines were observed to have negative inotropic and chronotropic effects and might function as a brake with respect to THs, although their physiological role is unclear. Moreover, Thyronamines showed a cardioprotective effect at physiological concentrations. The cardiac effects of iodothyroacetic acids seem to be limited and need to be elucidated.

  • Thyronamines and Analogues - The Route from Rediscovery to Translational Research on Thyronergic Amines
    Molecular and cellular endocrinology, 2017
    Co-Authors: Grazia Chiellini, Martina Sabatini, Lorenza Bellusci, Riccardo Zucchi
    Abstract:

    Thyronamines are a novel class of endogenous signaling compounds, structurally related to thyroid hormones (THs). Specific Thyronamines, particularly 3-iodoThyronamine (T1AM), stimulate with nanomolar affinity trace amine-associated receptor 1 (TAAR1), a G protein-coupled membrane receptor, and may also interact with other TAAR subtypes (particularly TAAR5), adrenergic receptors (particularly α2 receptors), amine transporters, and mitochondrial proteins. In addition to its structural similarities with THs, T1AM also contains the arylethylamine scaffold as in monoamine neurotransmitters, implicating an intriguing role for T1AM as both a neuromodulator and a hormone-like molecule constituting a part of thyroid hormone signaling. A large number of T1AM derivatives have already been synthesized. We discuss the different chemical strategies followed to obtain Thyronamine analogues, their potency at TAAR1, and their structure-activity relationship. Preliminary characterization of the functional effects of these synthetic compounds is also provided.

  • Tissue thyroid hormones and Thyronamines
    Heart Failure Reviews, 2016
    Co-Authors: Alice Accorroni, Federica Saponaro, Riccardo Zucchi
    Abstract:

    It has been known for a long time that changes in cardiac function are a major component of the clinical presentation of thyroid disease. Increased heart rate and hyperdynamic circulation are hallmarks of hyperthyroidism, while bradycardia and decreased contractility characterize hypothyroidism. Recent findings have provided novel insights in the physiology and pathophysiology of heart regulation by thyroid hormones. In this review, we summarize the present knowledge on thyroxine (T_4) transport and metabolism and on the biochemical pathways leading to genomic and non-genomic effects produced by 3,5,3′-triiodothyronine (T_3) and by its active metabolites, particularly 3,5-diiodothyronine (T_2) and 3-iodoThyronamine (T_1AM). On this basis, specific issues of special interest for cardiology are discussed, namely (1) relevance of the regulation of proteins involved in the control of calcium homeostasis and in pacemaker cell activity, due to non-genomic as well as to classical genomic effects; (2) stimulation of fatty acid oxidation by T_2 and T_1AM, the latter also causing a negative inotropic and chronotropic action at micromolar concentrations; (3) induction of D3 deiodinase in heart failure, potentially causing selective cardiac hypothyroidism, whose clinical implications are still controversial; and (4) cardioprotective effect of T_1AM, possibly occurring at physiological concentrations, and relevance of T_3 and of thyroid hormone receptor α_1 in post-infarction repair.

  • tissue distribution and cardiac metabolism of 3 iodoThyronamine
    Endocrinology, 2010
    Co-Authors: Alessandro Saba, Sandra Ghelardoni, Grazia Chiellini, Maja Marchini, Sabina Frascarelli, Thomas S Scanlan, Andrea Raffaelli, Massimo Tonacchera, Paolo Vitti, Riccardo Zucchi
    Abstract:

    3-IodoThyronamine (T1AM) is a novel relative of thyroid hormone, able to interact with specific G protein-coupled receptors, known as trace amine-associated receptors. Significant functional effects are produced by exogenous T1AM, including a negative inotropic and chronotropic effect in cardiac preparations. This work was aimed at estimating endogenous T1AM concentration in different tissues and determining its cardiac metabolism. A novel HPLC tandem mass spectrometry assay was developed, allowing detection of T1AM, Thyronamine, 3-iodothyroacetic acid, and thyroacetic acid. T1AM was detected in rat serum, at the concentration of 0.3 ± 0.03 pmol/ml, and in all tested organs (heart, liver, kidney, skeletal muscle, stomach, lung, and brain), at concentrations significantly higher than the serum concentration, ranging from 5.6 ± 1.5 pmol/g in lung to 92.9 ± 28.5 pmol/g in liver. T1AM was also identified for the first time in human blood. In H9c2 cardiomyocytes and isolated perfused rat hearts, significant Na...

Thomas S Scanlan - One of the best experts on this subject based on the ideXlab platform.

  • Proof of concept of the electrochemical sensing of 3-iodoThyronamine (T1AM) and Thyronamine (T0AM)
    ChemElectroChem, 2014
    Co-Authors: Luís Moreira Gonçalves, Thomas S Scanlan, Manuela M. Moreira, Carla F. Azevedo, Inês M. Valente, João Sousa, Richard G. Compton, José A. Rodrigues
    Abstract:

    Recent studies have shown that, besides the well-recognized T3 and T4 hormones, there are other relevant thyroid hormones circulating in the human body. In particular, this is the case for 3-iodoThyronamine (T1AM) and Thyronamine (T0AM). One of the reasons for the lack of studies showing their precise importance is the absence of analytical methodologies available. Herein, for the first time, T1AM and T0AM are electrochemically characterized. T0AM was sensed by means of a glassy carbon electrode; furthermore, T1AM was sensed both with a graphitic surface (oxidatively) as well as with mercury (reductively). For both compounds, after oxidation, it was possible to observe the reversible redox reaction concerning the benzoquinone/hydroquinone couple, thus increasing the specificity of the electroanalysis. Therefore, this work provides the basis for an ‘at-point-of-use’ electrochemical strip test for T1AM and T0AM.

  • does the aromatic l amino acid decarboxylase contribute to Thyronamine biosynthesis
    Molecular and Cellular Endocrinology, 2012
    Co-Authors: Carolin S Hoefig, Thomas S Scanlan, Georg F Hoffmann, Susanne Piehl, Kostja Renko, Mariarita Bertoldi, Thomas Opladen, Jeannette Klein, Oliver Blankenstein, Ulrich Schweizer
    Abstract:

    Thyronamines (TAM), recently described endogenous signaling molecules, exert metabolic and pharmacological actions partly opposing those of the thyromimetic hormone T(3). TAM biosynthesis from thyroid hormone (TH) precursors requires decarboxylation of the L-alanine side chain and several deiodination steps to convert e.g. L-thyroxine (T(4)) into the most potent 3-T(1)AM. Aromatic L-amino acid decarboxylase (AADC) was proposed to mediate TAM biosynthesis via decarboxylation of TH. This hypothesis was tested by incubating recombinant human AADC, which actively catalyzes dopamine production from DOPA, with several TH. Under all reaction conditions tested, AADC failed to catalyze TH decarboxylation, thus challenging the initial hypothesis. These in vitro observations are supported by detection of 3-T(1)AM in plasma of patients with AADC-deficiency at levels (46 ± 18 nM, n=4) similar to those of healthy controls. Therefore, we propose that the enzymatic decarboxylation needed to form TAM from TH is catalyzed by another unique, perhaps TH-specific, decarboxylase.

  • tissue distribution and cardiac metabolism of 3 iodoThyronamine
    Endocrinology, 2010
    Co-Authors: Alessandro Saba, Sandra Ghelardoni, Grazia Chiellini, Maja Marchini, Sabina Frascarelli, Thomas S Scanlan, Andrea Raffaelli, Massimo Tonacchera, Paolo Vitti, Riccardo Zucchi
    Abstract:

    3-IodoThyronamine (T1AM) is a novel relative of thyroid hormone, able to interact with specific G protein-coupled receptors, known as trace amine-associated receptors. Significant functional effects are produced by exogenous T1AM, including a negative inotropic and chronotropic effect in cardiac preparations. This work was aimed at estimating endogenous T1AM concentration in different tissues and determining its cardiac metabolism. A novel HPLC tandem mass spectrometry assay was developed, allowing detection of T1AM, Thyronamine, 3-iodothyroacetic acid, and thyroacetic acid. T1AM was detected in rat serum, at the concentration of 0.3 ± 0.03 pmol/ml, and in all tested organs (heart, liver, kidney, skeletal muscle, stomach, lung, and brain), at concentrations significantly higher than the serum concentration, ranging from 5.6 ± 1.5 pmol/g in lung to 92.9 ± 28.5 pmol/g in liver. T1AM was also identified for the first time in human blood. In H9c2 cardiomyocytes and isolated perfused rat hearts, significant Na...

  • Identification and Characterization of 3-IodoThyronamine Intracellular Transport
    Endocrinology, 2008
    Co-Authors: Alexandra G Ianculescu, Kathleen M. Giacomini, Thomas S Scanlan
    Abstract:

    3-IodoThyronamine (T1AM) is a naturally occurring thyroid hormone metabolite with distinct biological effects that are opposite those of thyroid hormone. The known molecular targets of T1AM include both plasma membrane and intracellular proteins, suggesting that intracellular transport of T1AM may be an important component of its action, although no uptake mechanism has yet been described. Using various human cell lines, we show that, indeed, cellular uptake of T1AM occurs in multiple cell types and that this process involves specific, saturable, and inhibitable transport mechanisms. These mechanisms are sodium and chloride independent, pH dependent, Thyronamine specific, and do not involve the likely candidate transporters of other monoamines, organic cations, or thyroid hormones. A large-scale RNA interference screen targeting the entire solute carrier superfamily of transporter genes reveals that the transport of T1AM into cells involves multiple transporters, and we identify eight transporters that may contribute to the uptake of T1AM in HeLa cells. This type of transporter small interfering RNA screening approach can be used in general to identify the constellation of transporters that participate in the intracellular disposition of compounds.

  • development of a validated liquid chromatography tandem mass spectrometry method for the distinction of thyronine and Thyronamine constitutional isomers and for the identification of new deiodinase substrates
    Rapid Communications in Mass Spectrometry, 2008
    Co-Authors: Susanne Piehl, Thomas S Scanlan, Thomas Heberer, Gabor Balizs, Josef Köhrle
    Abstract:

    Thyronines (THs) and Thyronamines (TAMs) are two groups of endogenous iodine-containing signaling molecules whose representatives differ from each other only regarding the number and/or the position of the iodine atoms. Both groups of compounds are substrates of three deiodinase isozymes, which catalyze the sequential reductive removal of iodine from the respective precursor molecule. In this study, a novel analytical method applying liquid chromatography/tandem mass spectrometry (LC-MS/MS) was developed. This method permitted the unequivocal, simultaneous identification and quantification of all THs and TAMs in the same biological sample. Furthermore, a liquid-liquid extraction procedure permitting the concurrent isolation of all THs and TAMs from biological matrices, namely deiodinase (Dio) reaction mixtures, was established. Method validation experiments with extracted TH and TAM analytes demonstrated that the method was selective, devoid of matrix effects, sensitive, linear over a wide range of analyte concentrations and robust in terms of reproducible recoveries, process efficiencies as well as intra-assay and inter-assay stability parameters. The method was applied to study the deiodination reactions of iodinated THs catalyzed by the three deiodinase isozymes. With the HPLC protocol developed herein, sufficient chromatographic separation of all constitutional TH and TAM isomers was achieved. Accordingly, the position of each iodine atom removed from a TH substrate in a Dio-catalyzed reaction was backtracked unequivocally. While several established deiodination reactions were verified, two as yet unknown reactions, namely the phenolic ring deiodination of 3',5'-diiodothyronine (3',5'-T2) by Dio2 and the tyrosyl ring deiodination of 3-monoiodothyronine (3-T1) by Dio3, were newly identified.

Grazia Chiellini - One of the best experts on this subject based on the ideXlab platform.

  • Endogenous TH metabolite 3-iodoThyronamine (T1AM) and synthetic Thyronamine-like analogues SG-1 and SG-2 induce autophagy in human glioblastoma cells (U-87MG)
    Endocrine Abstracts, 2017
    Co-Authors: Lorenza Bellusci, Riccardo Zucchi, Giulia Nesi, Martina Sabatini, Paola Lenzi, Simona Rapposelli, Francesco Fornai, Leonardo Rossi, Grazia Chiellini
    Abstract:

    Endogenous TH metabolite 3-iodoThyronamine (T1AM) and recently developed Thyronamine-like synthetic analogues SG1 and SG2 have emerged as neuroprotective agents. Autophagy has proved to be an effective therapeutic approach for neurodegenerative diseases. Therefore we investigated whether these compounds can induce autophagy in human glioblastoma cells (U-87MG).\ud \ud Cultured U-87MG cells were initially treated with 1 μM T1AM, SG-1, SG-2 or vehicle for 30’, 4, 8 and 24 h and autophagy was monitored morphologically by assessing the presence of autophagic vacuoles and LC3-II puncta formation with transmission electron microscopy (TEM) and immunofluorescence (IF) microscopy, respectively. In addition, cellular lysates were subjected to western blotting to assess the accumulation of microtubule-associated protein 1 light chain 3 (LC3). This protein is widely used as a marker for autophagy because it is a structural protein vital in autophagosome formation. TEM and IF microscopy showed a significant time dependent increase of autophagy-like vacuoles density and LC3 punta formation in U-87MG cells exposed to the treatment with test compounds, with T1AM and SG-1 being the most effective. Along with extensive cytoplasmic vacuolization, western blotting analysis revealed significant up regulation of LC3-II expression (P

  • Thyronamines and Analogues - The Route from Rediscovery to Translational Research on Thyronergic Amines
    Molecular and cellular endocrinology, 2017
    Co-Authors: Grazia Chiellini, Martina Sabatini, Lorenza Bellusci, Riccardo Zucchi
    Abstract:

    Thyronamines are a novel class of endogenous signaling compounds, structurally related to thyroid hormones (THs). Specific Thyronamines, particularly 3-iodoThyronamine (T1AM), stimulate with nanomolar affinity trace amine-associated receptor 1 (TAAR1), a G protein-coupled membrane receptor, and may also interact with other TAAR subtypes (particularly TAAR5), adrenergic receptors (particularly α2 receptors), amine transporters, and mitochondrial proteins. In addition to its structural similarities with THs, T1AM also contains the arylethylamine scaffold as in monoamine neurotransmitters, implicating an intriguing role for T1AM as both a neuromodulator and a hormone-like molecule constituting a part of thyroid hormone signaling. A large number of T1AM derivatives have already been synthesized. We discuss the different chemical strategies followed to obtain Thyronamine analogues, their potency at TAAR1, and their structure-activity relationship. Preliminary characterization of the functional effects of these synthetic compounds is also provided.

  • Hit-to-Lead Optimization of Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists with a Diphenylmethane-Scaffold: Design, Synthesis, and Biological Study.
    Journal of Medicinal Chemistry, 2016
    Co-Authors: Grazia Chiellini, Giulia Nesi, Simona Sestito, Sara Chiarugi, Massimiliano Runfola, Stefano Espinoza, Martina Sabatini, Lorenza Bellusci, Annunziatina Laurino, Elena Cichero
    Abstract:

    The trace amine-associated receptor 1 (TAAR1) is a G-protein-coupled receptors (GPCR) potently activated by a variety of molecules besides trace amines (TAs), including thyroid hormone-derivatives like 3-iodoThyronamine (T1AM), catechol-O-methyltransferase products like 3-methoxytyramine, and amphetamine-related compounds. Accordingly, TAAR1 is considered a promising target for medicinal development. To gain more insights into TAAR1 physiological functions and validation of its therapeutic potential, we recently developed a new class of Thyronamine-like derivatives. Among them compound SG2 showed high affinity and potent agonist activity at mouse TAAR1. In the present work, we describe design, synthesis, and SAR study of a new series of compounds (1–16) obtained by introducing specific structural changes at key points of our lead compound SG2 skeleton. Five of the newly synthesized compounds displayed mTAAR1 agonist activity higher than both SG2 and T1AM. Selected diphenylmethane analogues, namely 1 and 2...

  • Hit-to-Lead Optimization of Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists with a Diphenylmethane-Scaffold: Design, Synthesis, and Biological Study
    2016
    Co-Authors: Grazia Chiellini, Giulia Nesi, Simona Sestito, Sara Chiarugi, Massimiliano Runfola, Stefano Espinoza, Martina Sabatini, Lorenza Bellusci, Annunziatina Laurino, Elena Cichero
    Abstract:

    The trace amine-associated receptor 1 (TAAR1) is a G-protein-coupled receptors (GPCR) potently activated by a variety of molecules besides trace amines (TAs), including thyroid hormone-derivatives like 3-iodoThyronamine (T1AM), catechol-O-methyltransferase products like 3-methoxytyramine, and amphetamine-related compounds. Accordingly, TAAR1 is considered a promising target for medicinal development. To gain more insights into TAAR1 physiological functions and validation of its therapeutic potential, we recently developed a new class of Thyronamine-like derivatives. Among them compound SG2 showed high affinity and potent agonist activity at mouse TAAR1. In the present work, we describe design, synthesis, and SAR study of a new series of compounds (1–16) obtained by introducing specific structural changes at key points of our lead compound SG2 skeleton. Five of the newly synthesized compounds displayed mTAAR1 agonist activity higher than both SG2 and T1AM. Selected diphenylmethane analogues, namely 1 and 2, showed potent functional activity in in vitro and in vivo models

  • design synthesis and evaluation of Thyronamine analogues as novel potent mouse trace amine associated receptor 1 mtaar1 agonists
    Journal of Medicinal Chemistry, 2015
    Co-Authors: Grazia Chiellini, Sabina Frascarelli, Giulia Nesi, Stefano Espinoza, Martina Sabatini, Annunziatina Laurino, Elena Cichero, Maria Digiacomo, Rossella Malvasi, Marco Macchia
    Abstract:

    Trace amine associated receptor 1 (TAAR1) is a G protein coupled receptor (GPCR) expressed in brain and periphery activated by a wide spectrum of agonists that include, but are not limited to, trace amines (TAs), amphetamine-like psychostimulants, and endogenous Thyronamines such as Thyronamine (T0AM) and 3-iodoThyronamine (T1AM). Such polypharmacology has made it challenging to understand the role and the biology of TAAR1. In an effort to understand the molecular basis of TAAR1 activation, we rationally designed and synthesized a small family of Thyronamine derivatives. Among them, compounds 2 and 3 appeared to be a good mimic of the parent endogenous Thyronamine, T0AM and T1AM, respectively, both in vitro and in vivo. Thus, these compounds offer suitable tools for studying the physiological roles of mouse TAAR1 and could represent the starting point for the development of more potent and selective TAAR1 ligands.

Martina Sabatini - One of the best experts on this subject based on the ideXlab platform.

  • New Insights into the Potential Roles of 3-IodoThyronamine (T1AM) and Newly Developed Thyronamine-Like TAAR1 Agonists in Neuroprotection.
    Frontiers in pharmacology, 2017
    Co-Authors: Lorenza Bellusci, Simona Sestito, Martina Sabatini, Annunziatina Laurino, Laura Raimondi, Paola Lenzi, Simona Rapposelli, Francesca Biagioni, Francesco Fornai, Alessandra Salvetti
    Abstract:

    3-IodoThyronamine (T1AM) is an endogenous high-affinity ligand of the trace amine-associated receptor 1 (TAAR1), detected in mammals in many organs, including the brain. Recent evidence indicates that pharmacological TAAR1 activation may offer a novel therapeutic option for the treatment of a wide range of neuropsychiatric and metabolic disorders. To assess potential neuroprotection by TAAR1 agonists, in the present work, we initially investigated whether T1AM and its corresponding 3-methylbiaryl-methane analog SG-2 can improve learning and memory when systemically administered to mice at submicromolar doses, and whether these effects are modified under conditions of MAO inhibition by clorgyline. Our results revealed that when i.p. injected to mice, both T1AM and SG-2 produced memory-enhancing and hyperalgesic effects, while increasing ERK1/2 phosphorylation and expression of transcription factor c-fos. Notably, both compounds appeared to rely on the action of ubiquitous enzymes MAO to produce the corresponding oxidative metabolites that were then able to activate the histaminergic system. Since autophagy is key for neuronal plasticity, in a second line of experiments we explored whether T1AM and synthetic TAAR1 agonists SG1 and SG2 were able to induce autophagy in human glioblastoma cell lines (U-87MG). After treatment of U-87MG cells with 1 μM T1AM, SG-1, SG-2 transmission electron microscopy (TEM) and immunofluorescence (IF) showed a significant time-dependent increase of autophagy vacuoles and microtubule-associated protein 1 light chain 3 (LC3). Consistently, Western blot analysis revealed a significant increase of the LC3II/LC3I ratio, with T1AM and SG-1 being the most effective agents. A decreased level of the p62 protein was also observed after treatment with T1AM and SG-1, which confirms the efficacy of these compounds as autophagy inducers in U-87MG cells. In the process to dissect which pathway induces ATG, the effects of these compounds were evaluated on the PI3K-AKT-mTOR pathway. We found that 1 μM T1AM, SG-1 and SG-2 decreased pAKT/AKT ratio at 0.5 and 4 h after treatment, suggesting that autophagy is induced by inhibiting mTOR phosphorylation by PI3K-AKT-mTOR pathway. In conclusion, our study shows that T1AM and Thyronamine-like derivatives SG-1 and SG-2 might represent valuable tools to therapeutically intervene with neurological disorders.

  • Endogenous TH metabolite 3-iodoThyronamine (T1AM) and synthetic Thyronamine-like analogues SG-1 and SG-2 induce autophagy in human glioblastoma cells (U-87MG)
    Endocrine Abstracts, 2017
    Co-Authors: Lorenza Bellusci, Riccardo Zucchi, Giulia Nesi, Martina Sabatini, Paola Lenzi, Simona Rapposelli, Francesco Fornai, Leonardo Rossi, Grazia Chiellini
    Abstract:

    Endogenous TH metabolite 3-iodoThyronamine (T1AM) and recently developed Thyronamine-like synthetic analogues SG1 and SG2 have emerged as neuroprotective agents. Autophagy has proved to be an effective therapeutic approach for neurodegenerative diseases. Therefore we investigated whether these compounds can induce autophagy in human glioblastoma cells (U-87MG).\ud \ud Cultured U-87MG cells were initially treated with 1 μM T1AM, SG-1, SG-2 or vehicle for 30’, 4, 8 and 24 h and autophagy was monitored morphologically by assessing the presence of autophagic vacuoles and LC3-II puncta formation with transmission electron microscopy (TEM) and immunofluorescence (IF) microscopy, respectively. In addition, cellular lysates were subjected to western blotting to assess the accumulation of microtubule-associated protein 1 light chain 3 (LC3). This protein is widely used as a marker for autophagy because it is a structural protein vital in autophagosome formation. TEM and IF microscopy showed a significant time dependent increase of autophagy-like vacuoles density and LC3 punta formation in U-87MG cells exposed to the treatment with test compounds, with T1AM and SG-1 being the most effective. Along with extensive cytoplasmic vacuolization, western blotting analysis revealed significant up regulation of LC3-II expression (P

  • Thyronamines and Analogues - The Route from Rediscovery to Translational Research on Thyronergic Amines
    Molecular and cellular endocrinology, 2017
    Co-Authors: Grazia Chiellini, Martina Sabatini, Lorenza Bellusci, Riccardo Zucchi
    Abstract:

    Thyronamines are a novel class of endogenous signaling compounds, structurally related to thyroid hormones (THs). Specific Thyronamines, particularly 3-iodoThyronamine (T1AM), stimulate with nanomolar affinity trace amine-associated receptor 1 (TAAR1), a G protein-coupled membrane receptor, and may also interact with other TAAR subtypes (particularly TAAR5), adrenergic receptors (particularly α2 receptors), amine transporters, and mitochondrial proteins. In addition to its structural similarities with THs, T1AM also contains the arylethylamine scaffold as in monoamine neurotransmitters, implicating an intriguing role for T1AM as both a neuromodulator and a hormone-like molecule constituting a part of thyroid hormone signaling. A large number of T1AM derivatives have already been synthesized. We discuss the different chemical strategies followed to obtain Thyronamine analogues, their potency at TAAR1, and their structure-activity relationship. Preliminary characterization of the functional effects of these synthetic compounds is also provided.

  • Hit-to-Lead Optimization of Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists with a Diphenylmethane-Scaffold: Design, Synthesis, and Biological Study.
    Journal of Medicinal Chemistry, 2016
    Co-Authors: Grazia Chiellini, Giulia Nesi, Simona Sestito, Sara Chiarugi, Massimiliano Runfola, Stefano Espinoza, Martina Sabatini, Lorenza Bellusci, Annunziatina Laurino, Elena Cichero
    Abstract:

    The trace amine-associated receptor 1 (TAAR1) is a G-protein-coupled receptors (GPCR) potently activated by a variety of molecules besides trace amines (TAs), including thyroid hormone-derivatives like 3-iodoThyronamine (T1AM), catechol-O-methyltransferase products like 3-methoxytyramine, and amphetamine-related compounds. Accordingly, TAAR1 is considered a promising target for medicinal development. To gain more insights into TAAR1 physiological functions and validation of its therapeutic potential, we recently developed a new class of Thyronamine-like derivatives. Among them compound SG2 showed high affinity and potent agonist activity at mouse TAAR1. In the present work, we describe design, synthesis, and SAR study of a new series of compounds (1–16) obtained by introducing specific structural changes at key points of our lead compound SG2 skeleton. Five of the newly synthesized compounds displayed mTAAR1 agonist activity higher than both SG2 and T1AM. Selected diphenylmethane analogues, namely 1 and 2...

  • Hit-to-Lead Optimization of Mouse Trace Amine Associated Receptor 1 (mTAAR1) Agonists with a Diphenylmethane-Scaffold: Design, Synthesis, and Biological Study
    2016
    Co-Authors: Grazia Chiellini, Giulia Nesi, Simona Sestito, Sara Chiarugi, Massimiliano Runfola, Stefano Espinoza, Martina Sabatini, Lorenza Bellusci, Annunziatina Laurino, Elena Cichero
    Abstract:

    The trace amine-associated receptor 1 (TAAR1) is a G-protein-coupled receptors (GPCR) potently activated by a variety of molecules besides trace amines (TAs), including thyroid hormone-derivatives like 3-iodoThyronamine (T1AM), catechol-O-methyltransferase products like 3-methoxytyramine, and amphetamine-related compounds. Accordingly, TAAR1 is considered a promising target for medicinal development. To gain more insights into TAAR1 physiological functions and validation of its therapeutic potential, we recently developed a new class of Thyronamine-like derivatives. Among them compound SG2 showed high affinity and potent agonist activity at mouse TAAR1. In the present work, we describe design, synthesis, and SAR study of a new series of compounds (1–16) obtained by introducing specific structural changes at key points of our lead compound SG2 skeleton. Five of the newly synthesized compounds displayed mTAAR1 agonist activity higher than both SG2 and T1AM. Selected diphenylmethane analogues, namely 1 and 2, showed potent functional activity in in vitro and in vivo models