Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 262
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Biol Chem ; 300(8): 107495, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38925327

RESUMEN

Transthyretin (TTR) is an homotetrameric protein involved in the transport of thyroxine. More than 150 different mutations have been described in the TTR gene, several of them associated with familial amyloid cardiomyopathy. Recently, our group described a new variant of TTR in Brazil, namely A39D-TTR, which causes a severe cardiac condition. Position 39 is in the AB loop, a region of the protein that is located within the thyroxine-binding channels and is involved in tetramer formation. In the present study, we solved the structure and characterize the thermodynamic stability of this new variant of TTR using urea and high hydrostatic pressure. Interestingly, during the process of purification, A39D-TTR turned out to be a dimer and not a tetramer, a variation that might be explained by the close contact of the four aspartic acids at position 39, where they face each other inside the thyroxine channel. In the presence of subdenaturing concentrations of urea, bis-ANS binding and dynamic light scattering revealed A39D-TTR in the form of a molten-globule dimer. Co-expression of A39D and WT isoforms in the same bacterial cell did not produce heterodimers or heterotetramers, suggesting that somehow a negative charge at the AB loop precludes tetramer formation. A39D-TTR proved to be highly amyloidogenic, even at mildly acidic pH values where WT-TTR does not aggregate. Interestingly, despite being a dimer, aggregation of A39D-TTR was inhibited by diclofenac, which binds to the thyroxine channel in the tetramer, suggesting the existence of other pockets in A39D-TTR able to accommodate this molecule.


Asunto(s)
Cardiomiopatías , Prealbúmina , Multimerización de Proteína , Termodinámica , Prealbúmina/genética , Prealbúmina/química , Prealbúmina/metabolismo , Humanos , Cardiomiopatías/metabolismo , Cardiomiopatías/genética , Tiroxina/metabolismo , Tiroxina/química , Mutación Missense , Amiloide/metabolismo , Amiloide/química , Amiloide/genética , Sustitución de Aminoácidos , Urea/química , Urea/metabolismo
2.
Chemistry ; 29(3): e202202387, 2023 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-36254793

RESUMEN

Type-1 iodothyronine deiodinase (ID-1) catalyzes the reductive elimination of 5'-I and 5-I on the phenolic and tyrosyl rings of thyroxine (T4), respectively. Chemically verifying whether I atoms with different chemical properties undergo deiodination through a common mechanism is challenging. Herein, we report the modeling of ID-1 using aliphatic diselenide (Se-Se) and selenenylsulfide (Se-S) compounds. Mechanistic investigations of deiodination using the ID-1-like reagents suggested that the 5'-I and 5-I deiodinations proceed via the same mechanism through an unstable intermediate containing a Se⋅⋅⋅I halogen bond between a selenolate anion, reductively produced from Se-Se (or Se-S) in the compound, and an I atom in T4. Moreover, imidazolium and thiol groups, which may act as general acid catalysts, promoted the heterolytic cleavage of the C-I bond in the Se⋅⋅⋅I intermediate, which is the rate-determining step, by donating a proton to the C atom.


Asunto(s)
Yoduro Peroxidasa , Tiroxina , Yoduro Peroxidasa/química , Tiroxina/química , Halógenos/química , Catálisis , Fenoles , Triyodotironina/química
3.
Chemistry ; 29(9): e202203111, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36380701

RESUMEN

The regioselective deiodinations of L-thyroxine (T4) play key roles in the thyroid hormone homeostasis. These reactions are catalyzed by three isoforms of the selenoenzymes, iodothyronine deiodinases (Dio1, Dio2 and Dio3), which are highly homologous in nature. Dio1 mediates 5'- or 5-deiodinations of T4 to produce T3 and rT3, respectively. In contrast, Dio2 and Dio3 are selective to 5'- or 5-deiodination to produce T3 and rT3, respectively. Understanding of the regioselectivity of deiodination at the molecular level is important as abnormal levels of thyroid hormone have been implicated in various clinical conditions, such as hypoxia, myocardial infarction, neuronal ischemia and cancer. In this paper, we report that the electronic properties of the iodine atoms in thyroxine (T4) can be modulated through a simple substitution in the 4'-phenolic moiety. This leads to the change in the regioselectivity of deiodination by different small molecule mimics of Dio enzymes. By using this chemical approach, we also show that the substitution of a strong electron withdrawing group facilitates the removal of all four iodine atoms in the T4 derivative. Theoretical investigations on the hydrogen bonded adducts of T4 with imidazole indicate that the charge on the iodine atoms depend on the nature of hydrogen bond between the -OH group of T4 and the imidazole moiety. While the imidazole can act as either hydrogen bond acceptor (HBA) or hydrogen bond donor (HBD), the protonated imidazole acts exclusively as HBD in T4-imidazole complex. These studies support the earlier observations that the histidine residue at the active sites of the deiodinases play an important role not only in the substrate binding, but also in altering the regioselectivity of the deiodination reactions.


Asunto(s)
Yoduro Peroxidasa , Yodo , Yoduro Peroxidasa/metabolismo , Hormonas Tiroideas/química , Tiroxina/química , Tiroxina/metabolismo , Imidazoles , Triyodotironina/química , Triyodotironina/metabolismo
4.
Cell Mol Life Sci ; 78(17-18): 6105-6117, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34297165

RESUMEN

Transthyretin (TTR) is an extracellular protein mainly produced in the liver and choroid plexus, with a well-stablished role in the transport of thyroxin and retinol throughout the body and brain. TTR is prone to aggregation, as both wild-type and mutated forms of the protein can lead to the accumulation of amyloid deposits, resulting in a disease called TTR amyloidosis. Recently, novel activities for TTR in cell biology have emerged, ranging from neuronal health preservation in both central and peripheral nervous systems, to cellular fate determination, regulation of proliferation and metabolism. Here, we review the novel literature regarding TTR new cellular effects. We pinpoint TTR as major player on brain health and nerve biology, activities that might impact on nervous systems pathologies, and assign a new link between TTR and angiogenesis and cancer. We also explore the molecular mechanisms underlying TTR activities at the cellular level, and suggest that these might go beyond its most acknowledged carrier functions and include interaction with receptors and activation of intracellular signaling pathways.


Asunto(s)
Amiloidosis/etiología , Prealbúmina/metabolismo , Amiloidosis/metabolismo , Sistema Nervioso Central/metabolismo , Humanos , Neuronas/citología , Neuronas/metabolismo , Prealbúmina/química , Prealbúmina/genética , Agregado de Proteínas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Tiroxina/química , Tiroxina/metabolismo , Vitamina A/química , Vitamina A/metabolismo
5.
Mol Pharm ; 18(7): 2683-2693, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34061524

RESUMEN

A range of tablet excipients were evaluated for their influence on the physical form and chemical stability of levothyroxine sodium pentahydrate (LSP; C15H10I4NNaO4·5H2O). LSP-excipient binary powder blends were stored under two conditions: (a) in hermetically sealed containers at 40 °C and (b) at 40 °C/75% RH. By use of synchrotron X-ray diffractometry, the disappearance of LSP could be quantified and the appearance of crystalline levothyroxine (free acid) could be identified. Under hermetically sealed conditions (40 °C) hygroscopic excipients such as povidone induced partial dehydration of LSP to form levothyroxine sodium monohydrate. When stored at 40 °C/75% RH, acidic excipients induced measurable disproportionation of LSP resulting in the formation of levothyroxine (free acid). HPLC analyses of drug-excipient mixtures revealed that lactose monohydrate, microcrystalline cellulose, and croscarmellose sodium caused pronounced chemical decomposition of LSP. On the other hand, magnesium stearate, sodium stearyl fumarate, and alkaline pH modifiers did not affect the physical and chemical stability of the API following storage at 40 °C/75% RH. HPLC, being a solution based technique, revealed chemical decomposition of the API, but the technique was insensitive to physical transformations. Excipient properties such as hygroscopicity and microenvironmental acidity were identified to be critical determinants of both physical and chemical stability of LSP in a drug product. For drugs exhibiting both physical and chemical transformations, simultaneous solid-state and solution based analyses will enable comprehensive stability evaluation.


Asunto(s)
Composición de Medicamentos/métodos , Estabilidad de Medicamentos , Excipientes/química , Polvos/química , Comprimidos/química , Tiroxina/química , Agua/química , Química Farmacéutica , Humectabilidad
6.
Bioorg Med Chem ; 42: 116250, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34118788

RESUMEN

Receptor-mediated cancer therapy has received much attention in the last few decades. Neuroblastoma and other cancers of the sympathetic nervous system highly express norepinephrine transporter (NET) and cell plasma membrane integrin αvß3. Dual targeting of the NET and integrin αvß3 receptors using a Drug-Drug Conjugate (DDC) might provide effective treatment strategy in the fight against neuroblastoma and other neuroendocrine tumors. In this work, we synthesized three dual-targeting BG-P400-TAT derivatives, dI-BG-P400-TAT, dM-BG-P400-TAT, and BG-P400-PAT containing di-iodobenzene, di-methoxybenzene, and piperazine groups, respectively. These derivatives utilize to norepinephrine transporter (NET) and the integrin αvß3 receptor to simultaneously modulate both targets based on evaluation in a neuroblastoma animal model using the neuroblastoma SK-N-F1 cell line. Among the three synthesized agents, the piperazine substituted BG-P400-PAT exhibited potent integrin αvß3 antagonism and reduced neuroblastoma tumor growth and cancer cell viability by >90%. In conclusion, BG-P400-PAT and derivatives represent a potential therapeutic approach in the management of neuroblastoma.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Neuroblastoma/tratamiento farmacológico , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/antagonistas & inhibidores , Tiroxina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Relación Estructura-Actividad , Tiroxina/análogos & derivados , Tiroxina/química , Células Tumorales Cultivadas
7.
Chembiochem ; 21(7): 911-923, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31773854

RESUMEN

Thyroid hormones (THs) are key players in the endocrine system and play pivotal roles in carbohydrate and fat metabolism, protein synthesis, overall growth, and brain development. The thyroid gland predominantly produces thyroxine or 3,5,3',5'-tetraiodothyronine (T4) as a prohormone; three isoforms of a mammalian selenoenzyme-iodothyronine deiodinase (DIO1, DIO2 and DIO3)-catalyze the regioselective deiodination of T4 to produce biologically active and inactive metabolites. Whereas DIO1 catalyzes both 5- and 5'-deiodination of T4, DIO2 and DIO3 selectively mediate 5- and 5'-deiodination, respectively. In this review we discuss the regioselective deiodination of THs in the presence of organochalcogen compounds. Naphthalene-based compounds containing sulfur and/or selenium at the peri positions mediate regioselective 5-deiodination of THs, detailed mechanistic studies having revealed that the heterolytic cleavage of the C-I bond is facilitated by the formation of cooperative Se/S⋅⋅⋅I halogen bonds and Se/S⋅⋅⋅Se chalcogen bonds. We also discuss the biomimetic deiodination of several TH metabolites, including sulfated THs, iodothyronamines, and iodotyrosines. A brief discussion on the dehalogenation of halogenated nucleosides and nucleobases in the presence of organochalcogen compounds is also included.


Asunto(s)
Halógenos/metabolismo , Nucleósidos/metabolismo , Hormonas Tiroideas/metabolismo , Biomimética , Halógenos/química , Yoduro Peroxidasa/metabolismo , Nucleósidos/química , Isoformas de Proteínas/metabolismo , Estereoisomerismo , Hormonas Tiroideas/química , Tiroxina/química , Tiroxina/metabolismo
8.
Mol Pharm ; 17(10): 3915-3929, 2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32960611

RESUMEN

Levothyroxine sodium pentahydrate (LSP; C15H10I4NNaO4·5H2O) gradually loses one molecule of water of crystallization as the water vapor pressure is decreased from 90% to 15% RH (40 °C), a behavior characteristic of nonstoichiometric hydrates. LSP loses four molecules of water of crystallization to form levothyroxine sodium monohydrate (LSM; C15H10I4NNaO4·H2O) under realistic storage conditions (40 °C/0% RH for 3 h). The crystal structure of LSP was determined following which the specimen was partially dehydrated in situ to form LSM. The crystal structure of LSM provided insight into its potential for high reactivity. Thus, its presence in a drug product is undesirable. In LSP-oxalic acid mixtures stored in a hermetic container at 40 °C, there was moisture transfer from drug to excipient. Synchrotron X-ray diffractometry revealed dehydration of LSP resulting in LSM, while anhydrous oxalic acid transformed to its dihydrate. In formulations of LSP, chemical degradation of levothyroxine sodium may be preceded by its partial dehydration.


Asunto(s)
Desecación , Composición de Medicamentos/métodos , Tiroxina/química , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Excipientes/química , Humedad , Hipotiroidismo/tratamiento farmacológico , Comprimidos , Tiroxina/uso terapéutico , Difracción de Rayos X
9.
Bioorg Med Chem ; 28(1): 115212, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31761727

RESUMEN

It was previously reported that tetraiodothyroacetic acid (tetrac) inhibits angiogenesis by binding to the cell surface receptor for thyroid hormone on integrin αVß3. Therefore, we synthesized and evaluated two 64Cu-labeled tetrac derivatives and a Cy5.5-labeled tetrac derivative for tumor angiogenesis imaging. Tetrac was structurally modified to conjugate with 1,4,7,10-tetraazacyclododecane-N,N',N″,N″'-tetraacetic acid (DOTA) via its hydroxy or carboxylic acid end, and the resulting DOTA-conjugated tetrac derivatives were then labeled with 64Cu. Tetrac was also conjugated with Cy5.5 via its carboxylic acid end. All three tetrac derivatives (1-3) exhibited greater inhibitory activity than tetrac against endothelial cell tube formation. The U87MG cell binding of [64Cu]2 showed a time-dependent increase over 24 h and it was inhibited by 38% at 4 h in the presence of tetrac, indicating specificity of [64Cu]2 to the thyroid hormone receptor site on integrin αVß3. Positron emission tomography (PET) images of U87MG tumor-bearing mice injected with [64Cu]1 and [64Cu]2 revealed that high radioactivity accumulated in the tumors, and that the tumor uptake and tumor-to-nontarget uptake ratio were higher in small tumors than in large tumors. In addition, the Cy5.5-labeled tetrac derivative (3) displayed a strong near-infrared (NIR) signal in the tumors. Taken together, these results suggest that these ligands hold promise as imaging agents for visualization of tumor angiogenesis.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Carbocianinas/química , Neovascularización Patológica/diagnóstico por imagen , Tomografía de Emisión de Positrones , Tiroxina/análogos & derivados , Animales , Células Cultivadas , Radioisótopos de Cobre , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/diagnóstico por imagen , Tiroxina/síntesis química , Tiroxina/química
10.
Int J Mol Sci ; 21(19)2020 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998442

RESUMEN

Transthyretin (TTR) is a homotetrameric protein involved in human amyloidosis, including familial amyloid polyneuropathy (FAP). Discovering small-molecule stabilizers of the TTR tetramer is a therapeutic strategy for these diseases. Tafamidis, the only approved drug for FAP treatment, is not effective for all patients. Herein, we discovered that benzbromarone (BBM), a uricosuric drug, is an effective TTR stabilizer and inhibitor against TTR amyloid fibril formation. BBM rendered TTR more resistant to urea denaturation, similarly to iododiflunisal (IDIF), a very potent TTR stabilizer. BBM competes with thyroxine for binding in the TTR central channel, with an IC50 similar to IDIF and tafamidis. Results obtained by isothermal titration calorimetry (ITC) demonstrated that BBM binds TTR with an affinity similar to IDIF, tolcapone and tafamidis, confirming BBM as a potent binder of TTR. The crystal structure of the BBM-TTR complex shows two molecules binding deeply in the thyroxine binding channel, forming strong intermonomer hydrogen bonds and increasing the stability of the TTR tetramer. Finally, kinetic analysis of the ability of BBM to inhibit TTR fibrillogenesis at acidic pH and comparison with other stabilizers revealed that benzbromarone is a potent inhibitor of TTR amyloidogenesis, adding a new interesting scaffold for drug design of TTR stabilizers.


Asunto(s)
Benzbromarona/química , Reposicionamiento de Medicamentos , Fármacos Neuroprotectores/química , Prealbúmina/química , Tiroxina/química , Amiloide/antagonistas & inhibidores , Benzbromarona/metabolismo , Benzoxazoles/química , Benzoxazoles/metabolismo , Sitios de Unión , Unión Competitiva , Cristalografía por Rayos X , Diflunisal/análogos & derivados , Diflunisal/química , Diflunisal/metabolismo , Expresión Génica , Humanos , Enlace de Hidrógeno , Cinética , Simulación del Acoplamiento Molecular , Fármacos Neuroprotectores/metabolismo , Prealbúmina/agonistas , Prealbúmina/genética , Prealbúmina/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Termodinámica , Tiroxina/metabolismo , Tolcapona/química , Tolcapona/metabolismo
11.
Bioconjug Chem ; 30(12): 3087-3097, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31714064

RESUMEN

Discovery of bioactive molecules that target integrins has implicated their role in tumor angiogenesis, tumor growth, metastasis, and other pathological angiogenesis processes. Integrins are members of a family of cell surface receptors that play a critical role in the angiogenesis process. Tetraiodothyroacetic acid (tetrac), a deaminated derivative of l-thyroxine (T4), is a "thyrointegrin" antagonist that blocks the actions of l-triiodothyronine (T3) and T4 with an interaction site that is located at or near the RGD recognition site identified on integrin αvß3's binding pocket (thyrointegrin αvß3 receptors). We have enhanced the biological activity of a tetrac-based inhibitor via significantly improving its αvß3 receptor binding affinity by introducing a triazole ring on the outer ring of tetrac and covalently conjugating to polymer to increase the product's hydrophilicity via PEGylation. The product, P-bi-TAT, was restricted from nuclear translocation and demonstrated high blood brain barrier permeability and retention in contrast to the non-PEG conjugated derivative. Results of biological activity indicated that this macromolecule new chemical entity P-bi-TAT has greater than 400-fold potent integrin αvß3 affinity versus the parent compound tetrac and has potent anticancer/anti-angiogenesis efficacy against glioblastoma multiforme (GBM). P-bi-TAT administered subcutaneously once daily for 21 days at 1-10 mg/kg mouse body weight resulted in a dose-dependent suppression of GBM tumor growth and viability as monitored with IVIS imaging (P < 0.001). GBM tumors had >95% volume loss and maximal loss of GBM cell viability during the 21 days ON-treatment experiment as well as in the 21 days ON followed by 21 days OFF-treatment experiment (P < 0.001). In conclusion, P-bi-TAT is a promising lead clinical candidate effective in the treatment of human GBM.


Asunto(s)
Inhibidores de la Angiogénesis/química , Antineoplásicos/química , Glioblastoma/tratamiento farmacológico , Polietilenglicoles/química , Tiroxina/análogos & derivados , Triazoles/química , Animales , Barrera Hematoencefálica/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Glioblastoma/patología , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Ratones , Tiroxina/química , Triazoles/farmacología
12.
J Clin Lab Anal ; 33(2): e22667, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30221397

RESUMEN

BACKGROUND: Although biotin interferences in TSH, FT3, FT4, and other biotinylated antibody-based assays manufactured by Roche Diagnostics have been well studied, there are relatively few reports on biotin interference in biotin-based assays manufactured by other companies. We investigated biotin interferences in TSH, FT4, and FT3 assays based on the LOCI (luminescent oxygen channeling assay) technology using the Dimension Vista 1500 analyzer (Siemens). METHODS: We prepared four serum pools using leftover specimens. Three serum pools were prepared initially for the original study but the 4th pool was prepared three months later. The aliquots of serum pool one and two were supplemented with various amounts of biotin (50 -1200 ng/mL) followed by determination of TSH, FT4, and FT3 concentrations. The aliquots of third pool were also supplemented with biotin to investigate whether 1:3 dilution could identify biotin interference. Aliquots of serum pool four were supplemented with biotin in order to study reproducibility of our original data. RESULTS: We observed significantly elevated FT3 levels at biotin concentration of 100 ng/mL. In contrast, FT4 levels were falsely elevated but TSH levels were falsely decreased at a biotin level of 500 ng/mL. We also observed nonlinearity in dilution experiment. CONCLUSIONS: We conclude that FT3 assay is most susceptible to biotin interference (threshold: 100 ng/mL) while the FT4 and TSH assays are less affected (threshold: 500 ng/mL). In addition, we also observed nonlinearity upon 1:3 dilution, which may indicate biotin interference (or interference from other compounds).


Asunto(s)
Biotina/química , Análisis Químico de la Sangre/normas , Mediciones Luminiscentes/normas , Tirotropina/sangre , Tiroxina/sangre , Triyodotironina/sangre , Biotina/sangre , Humanos , Reproducibilidad de los Resultados , Tirotropina/química , Tiroxina/química , Triyodotironina/química
13.
AAPS PharmSciTech ; 20(1): 39, 2019 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-30604134

RESUMEN

Levothyroxine sodium pentahydrate (LEVO) tablets have been on the US market since the mid-twentieth century and remain the most highly prescribed product. Unfortunately, levothyroxine sodium tablets have also been one of the most highly recalled products due to potency and dissolution failures on stability. In 2008, the assay limits were tightened, yet the recalls did not decline, which highlights the serious quality concerns remaining to be elucidated. The aim of the present investigation was to test the hypothesis that the solid-state physical instability of LEVO precedes the chemical instability leading to product failure. The failure mode was hypothesized to be the dehydration of the crystal hydrate, when exposed to certain humidity and temperature conditions, followed by the oxidation of the API through vacated channels. It was previously reported by the authors that LEVO degradation occurred in the presence of oxygen at a low relative humidity (RH). Furthermore, powder X-ray diffractometry shows changes in the crystal lattice of LEVO initially and through the dehydration stages. Storage of LEVO at RT and 40 °C at 4-6% RH for 12 days shows a decrease in d-spacing of the (00 l) planes. Based on a structure solution from the powder data of the dehydrated material, the basic packing motif persists to varying degrees even when fully dehydrated along with disordering. Therefore, the crystal structure changes of LEVO depend on RH and temperature and are now explicable at the structural level for the first time. This exemplifies the dire need for "new prior knowledge" in generic product development.


Asunto(s)
Desecación/métodos , Tiroxina/química , Tiroxina/farmacocinética , Cristalización , Estabilidad de Medicamentos , Humedad , Polvos , Comprimidos , Temperatura , Difracción de Rayos X/métodos
14.
Bioorg Med Chem Lett ; 28(7): 1223-1227, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29519736

RESUMEN

In the angiogenesis process, integrins, which are members of a family of cell surface transmembrane receptors, play a critical role particularly in blood vessel formation and the local release of vascular growth factors. Thyroid hormones such as l-thyroxine (T4) and 3,5,3'-triiodo-l-thyronine (T3) promote angiogenesis and tumor cell proliferation via integrin αvß3 receptor. At or near an arginine-glycine-aspartate (RGD) recognition site on the binding pocket of integrin αvß3, tetraiodothyroacetic acid (tetrac, a deaminated derivative of T4) is a thyrointegrin receptor antagonist and blocks the actions of T3 and T4 as well as different growth factors-mediated angiogenesis. In this study, we synthesized novel tetrac analogs by modifying the phenolic moiety of tetrac and tested them for their anti-angiogenesis activity using a Matrigel plug model for angiogenesis in mice. Pharmacological activity results showed that tetrac can accommodate numerous modifications and maintain its anti-angiogenesis activity.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Tiroxina/análogos & derivados , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Neoplasias/patología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Relación Estructura-Actividad , Tiroxina/síntesis química , Tiroxina/química , Tiroxina/farmacología
15.
Xenobiotica ; 48(3): 250-257, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28285550

RESUMEN

1. UDP-glucuronosyltransferases (UGTs) are important drug-metabolizing enzymes (DMEs) catalyzing the glucuronidation elimination of various xenobiotics and endogenous substances. Endogenous substances are important regulators for the activity of various UGT isoforms. Triiodothyronine (T3) and thyroxine (T4) are important thyroid hormones essential for normal cellular differentiation and growth. The present study aims to elucidate the inhibition behavior of T3 and T4 on the activity of UGT isoforms. 2. In vitro recombinant UGTs-catalyzed glucuronidation of 4-methylumbelliferone (4-MU) was used to screen the inhibition potential of T3 and T4 on the activity of various UGT isoforms. Initial screening results showed that T4 exerted stronger inhibition potential than T3 on the activity of various UGT isoforms at 100 µM. Inhibition kinetics was determined for the inhibition of T4 on the representative UGT isoforms, including UGT1A1, -1A3, -1A7, -1A8, -1A10 and -2B7. The results showed that T4 competitively inhibited the activity of UGT1A1, -1A3, -1A7, 1A10 and -2B7, and noncompetitively inhibited the activity of UGT1A8. The inhibition kinetic parameters were calculated to be 1.5, 2.4, 11, 9.6, 4.8 and 3.0 µM for UGT1A1, -1A3, -1A7, -1A8, -1A10 and -2B7, respectively. In silico docking method was employed to demonstrate why T4 exerted stronger inhibition than T3 towards UGT1A1. Stronger hydrogen bonds and hydrophobic interaction between T4 and activity cavity of UGT1A1 than T3 contributed to stronger inhibition of T4 towards UGT1A1. 3. In conclusion, more clinical monitoring should be given for the patients with the elevation of T4 level due to stronger inhibition of UGT isoforms-catalyzed metabolism of drugs or endogenous substances by T4.


Asunto(s)
Glucuronosiltransferasa/antagonistas & inhibidores , Tiroxina/farmacología , Triyodotironina/farmacología , Inhibidores Enzimáticos/farmacología , Glucuronosiltransferasa/química , Glucuronosiltransferasa/metabolismo , Humanos , Enlace de Hidrógeno , Himecromona/metabolismo , Simulación del Acoplamiento Molecular , Tiroxina/química , Triyodotironina/química
16.
Nucleic Acids Res ; 44(1): 1-13, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26621913

RESUMEN

Riboswitches are shape-changing regulatory RNAs that bind chemicals and regulate gene expression, directly coupling sensing to cellular actuation. However, it remains unclear how their sequence controls the physics of riboswitch switching and activation, particularly when changing the ligand-binding aptamer domain. We report the development of a statistical thermodynamic model that predicts the sequence-structure-function relationship for translation-regulating riboswitches that activate gene expression, characterized inside cells and within cell-free transcription-translation assays. Using the model, we carried out automated computational design of 62 synthetic riboswitches that used six different RNA aptamers to sense diverse chemicals (theophylline, tetramethylrosamine, fluoride, dopamine, thyroxine, 2,4-dinitrotoluene) and activated gene expression by up to 383-fold. The model explains how aptamer structure, ligand affinity, switching free energy and macromolecular crowding collectively control riboswitch activation. Our model-based approach for engineering riboswitches quantitatively confirms several physical mechanisms governing ligand-induced RNA shape-change and enables the development of cell-free and bacterial sensors for diverse applications.


Asunto(s)
Aptámeros de Nucleótidos/química , Modelos Biológicos , Riboswitch/genética , Técnica SELEX de Producción de Aptámeros , Algoritmos , Aptámeros de Nucleótidos/síntesis química , Técnicas Biosensibles , Dopamina/química , Dopamina/metabolismo , Humanos , Técnicas In Vitro , Mediciones Luminiscentes/métodos , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Pliegue del ARN , Reproducibilidad de los Resultados , Tiroxina/química , Tiroxina/metabolismo , Transcripción Genética
17.
Drug Dev Ind Pharm ; 44(11): 1762-1769, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29973090

RESUMEN

Controversies surround levothyroxine sodium as a drug and product, and are reflected in compendia (USP vs BP) differences in levothyroxine sodium tablets specifications concerning potency limit and dissolution test conditions, and in lack of consensus on several issues such as whether the drug BCS class I or III. We have recently published a clinical study in patients comparing the efficacy of multisource 100 mcg levothyroxine sodium tablets (three sources, two brands, a total of five batches). Clinical efficacy and dissolution rate data varied among the tablet batches studied and indicated that brand/source interchangeability could not be claimed. The efficacy parameters showed good correlation with dissolution data generated under BP 2014, but not under USP 2014 dissolution test conditions. In the present study, we decided to expand the number of tablet batches studied in vitro to a total of 12, to report potency and content uniformity data missing in the clinical study, and to further examine the discrepancy in dissolution results based on the medium used. The wide range of batch age in the studied samples allowed investigating the effect of batch age on in-vitro tablet performance parameters. Generated potency values indicated the prevalence of super-potent tablet batches. The dissolution data reflected the effect of compendia monograph differences in dissolution medium. The results also indicated an inverse relationship between tablet potency and batch age and, between dissolution and batch age. The possible effect of potency results on the generated dissolution data was discussed. Statistical significance of correlations examined was assessed by linear and non-linear regression analysis. Statistical significance was evident for the relation between batch age and BP 2014 dissolution data, compared to USP 2014 dissolution results.


Asunto(s)
Tiroxina/química , Estabilidad de Medicamentos , Inglaterra , Farmacopeas como Asunto , Solubilidad , Comprimidos , Tiroxina/farmacología , Factores de Tiempo , Estados Unidos
18.
Biochim Biophys Acta ; 1860(4): 648-60, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26777880

RESUMEN

BACKGROUND: Natural mutations of R218 in human serum albumin (HSA) result in an increased affinity for L-thyroxine and lead to the autosomal dominant condition of familial dysalbuminemic hyperthyroxinemia. METHODS: Binding was studied by equilibrium dialysis and computer modeling. RESULTS: Ten of 32 other isoforms tested had modified high-affinity hormone binding. L-thyroxine has been reported to bind to four sites (Tr) in HSA; Tr1 and Tr4 are placed in the N-terminal and C-terminal part of the protein, respectively. Site-directed mutagenesis gave new information about all the sites. CONCLUSIONS: It is widely assumed that Tr1 is the primary hormone site, and that this site, on a modified form, is responsible for the above syndrome, but the binding experiments with the genetic variants and displacement studies with marker ligands indicated that the primary site is Tr4. This new assignment of the high-affinity site was strongly supported by results of MM-PBSA analyses and by molecular docking performed on relaxed protein structure. However, dockings also revealed that mutating R218 for a smaller amino acid increases the affinity of Tr1 to such an extent that it can become the high-affinity site. GENERAL SIGNIFICANCE: Placing the high-affinity binding site (Tr4) and the one which can result in familial dysalbuminemic hyperthyroxinemia (Tr1) in two very different parts of HSA is not trivial, because in this way persons with and without the syndrome can have different types of interactions, and thereby complications, when given albumin-bound drugs. The molecular information is also useful when designing drugs based on L-thyroxine analogues.


Asunto(s)
Hipertiroxinemia Disalbuminémica Familiar , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Albúmina Sérica/química , Tiroxina/química , Sitios de Unión , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Tiroxina/metabolismo
19.
Mol Pharmacol ; 90(3): 265-74, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27413119

RESUMEN

Anticonvulsants can increase the risk of developing neurotoxicity in infants; however, the underlying mechanism has not been elucidated to date. Thyroxine [3,5,3',5'-l-tetraiodothyronine (T4)] plays crucial roles in the development of the central nervous system. In this study, we hypothesized that induction of UDP-glucuronosyltransferase 1A1 (UGT1A1)-an enzyme involved in the metabolism of T4-by anticonvulsants would reduce serum T4 levels and cause neurodevelopmental toxicity. Exposure of mice to phenytoin during both the prenatal and postnatal periods significantly induced UGT1A1 and decreased serum T4 levels on postnatal day 14. In the phenytoin-treated mice, the mRNA levels of synaptophysin and synapsin I in the hippocampus were lower than those in the control mice. The thickness of the external granule cell layer was greater in phenytoin-treated mice, indicating that induction of UGT1A1 during the perinatal period caused neurodevelopmental disorders. Exposure to phenytoin during only the postnatal period also caused these neurodevelopmental disorders. A T4 replacement attenuated the increase in thickness of the external granule cell layer, indicating that the reduced T4 was specifically associated with the phenytoin-induced neurodevelopmental disorder. In addition, these neurodevelopmental disorders were also found in the carbamazepine- and pregnenolone-16-α-carbonitrile-treated mice. Our study is the first to indicate that UGT1A1 can control neurodevelopment by regulating serum T4 levels.


Asunto(s)
Glucuronosiltransferasa/biosíntesis , Trastornos del Neurodesarrollo/enzimología , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Encéfalo/patología , Carbamazepina/química , Carbamazepina/farmacología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Inducción Enzimática/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes del Desarrollo , Humanos , Ratones Endogámicos C57BL , Ratones Transgénicos , Leche Humana/metabolismo , Trastornos del Neurodesarrollo/sangre , Trastornos del Neurodesarrollo/genética , Fenitoína/química , Embarazo , Carbonitrilo de Pregnenolona/farmacología , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante , Tiroxina/sangre , Tiroxina/química
20.
Biochem Biophys Res Commun ; 478(3): 1409-15, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27569283

RESUMEN

Angiogenesis is closely associated with osteoblast differentiation. Previously, we demonstrated that bone formation can be accelerated by treatment with COMP-Angiopoietin1, a known angiogenic factor. Angiopoietin1 (Ang1) is a specific growth factor that generates stable and mature vasculature through the Tie2 receptor. In this study, we aimed to identify a novel drug that can activate endogenous Ang1 expression as a pharmacological treatment for bone formation. Therefore, Ang1 expression was examined in U2OS osteoblast-like cells treated with 770 drugs from a library of Food and Drug Administration (FDA)-approved drugs by using ELISA for Ang1. l-thyroxine was selected as a novel drug candidate. l-Thyroxine is a synthetic form of the hormone thyroxine, which is used to treat patients with hypothyroidism. Enzyme-linked immunosorbent assays (ELISAs) were performed to test whether Ang1 is induced in a dose-dependent manner in human osteoblast-like cell lines, U2OS and MG63. The effects of l-thyroxine on osteoblast differentiation and mineralization were evaluated by alkaline phosphatase (ALP) activity and Alizarin red s staining. To determine the molecular mechanism, the expression of proteins related to bone formation and differentiation, such as type I collagen (COL1A1), osteocalcin (OC), bone sialoprotein (BSP), distal-less homeobox 5 (Dlx5), Runt-related transcription factor 2 (Runx2), osterix (OSX), and ALP, was tested by Western blotting analysis. Consequently, l-thyroxine induced Ang1 expression in a dose-dependent manner in both U2OS and M63 cells, which was confirmed by ELISA and Western blotting. Also, l-thyroxine activated ALP activity in U2OS and MG63 cells as well as ALP expression. Furthermore, l-thyroxine enhanced the expression of COL1A1, Runx2, OC, BSP, Dlx5, and OSX mRNA and proteins. Taken together, we demonstrated that l-thyroxine increased Ang1 expression and induces bone formation, differentiation, and mineralization in U2OS and MG63 cell lines, which suggests that l-thyroxine could be a potential bone production agent.


Asunto(s)
Angiopoyetina 1/metabolismo , Diferenciación Celular/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Tiroxina/farmacología , Biomarcadores/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Humanos , Osteoblastos/efectos de los fármacos , Tiroxina/química , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA