Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Thyroid ; 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801167

RESUMEN

BACKGROUND: TRIAC (3,5,3'-triiodothyroacetic acid) is a T3-receptor agonist pharmacologically used in patients to mitigate T3 resistance. It is additionally explored to treat some symptoms of patients with inactivating mutations in the thyroid hormone (TH) transporter MCT8 (SLC16A2). MCT8 is expressed along the blood-brain-barrier, on neurons, astrocytes, and oligodendrocytes. Hence, pathogenic variants in MCT8 limit the access of TH into and their functions within the brain. TRIAC was shown to enter the brain independently of MCT8 and to modulate expression of TH-dependent genes. The aim of the study was to identify transporters that facilitate TRIAC uptake into cells. METHODS: We performed a whole-genome RNAi screen in HepG2 cells stably expressing a T3-receptor-dependent luciferase reporter gene. Validation of hits from the primary and confirmatory secondary screen involved a counter screen with siRNAs and compared the cellular response to TRIAC to the effect of T3, in order to exclude siRNAs targeting the gene expression machinery. MDCK1 cells were stably transfected with cDNA encoding C-terminally myc-tagged versions of the identified TRIAC-preferring transporters. Several individual clones were selected after immunocytochemical characterization for biochemical characterization of their 125I-TRIAC transport activities. RESULTS: We identified SLC22A9 and SLC29A2 as transporters mediating cellular uptake of TRIAC. SLC22A9 encodes the organic anion transporting polypeptide 7 (OAT7), a sodium-independent organic anion transporter expressed in the plasma membrane in brain, pituitary, liver and other organs. Competition with the SLC22A9/OAT7 substrate estrone-3-sulfate reduced 125I-TRIAC uptake. SLC29A2 encodes the equilibrative nucleoside transporter 2 (ENT2), which is ubiquitously expressed including pituitary and brain. Co-incubation with the SLC29A2/ENT2 inhibitor nitrobenzyl-6-thioinosine reduced 125I-TRIAC uptake. Moreover, ABCD1, an ATP-dependent peroxisomal pump, was identified as a 125I-TRIAC exporter in transfected MDCK1 cells. CONCLUSIONS: Knowledge of TRIAC transporter expression patterns, also during brain development, may thus in the future help to interpret observations on TRIAC effects as well as understand why TRIAC may not show a desirable effect on cells or organs not expressing appropriate transporters. The identification of ABCD1 highlights the sensitivity of our established screening assay, but it may not hold significant relevance for patients undergoing TRIAC treatment.

2.
Front Endocrinol (Lausanne) ; 14: 1211473, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37435490

RESUMEN

Introduction: Graves' disease (GD) is an autoimmune disorder caused by autoantibodies against the thyroid stimulating hormone receptor (TSHR) leading to overstimulation of the thyroid gland. Thyroid eye disease (TED) is the most common extra thyroidal manifestation of GD. Therapeutic options to treat TED are very limited and novel treatments need to be developed. In the present study we investigated the effect of linsitinib, a dual small-molecule kinase inhibitor of the insulin-like growth factor 1 receptor (IGF-1R) and the Insulin receptor (IR) on the disease outcome of GD and TED. Methods: Linsitinib was administered orally for four weeks with therapy initiating in either the early ("active") or the late ("chronic") phases of the disease. In the thyroid and the orbit, autoimmune hyperthyroidism and orbitopathy were analyzed serologically (total anti-TSHR binding antibodies, stimulating anti TSHR antibodies, total T4 levels), immunohistochemically (H&E-, CD3-, TNFa- and Sirius red staining) and with immunofluorescence (F4/80 staining). An MRI was performed to quantify in vivo tissue remodeling inside the orbit. Results: Linsitinib prevented autoimmune hyperthyroidism in the early state of the disease, by reducing morphological changes indicative for hyperthyroidism and blocking T-cell infiltration, visualized by CD3 staining. In the late state of the disease linsitinib had its main effect in the orbit. Linsitinib reduced immune infiltration of T-cells (CD3 staining) and macrophages (F4/80 and TNFa staining) in the orbita in experimental GD suggesting an additional, direct effect of linsitinib on the autoimmune response. In addition, treatment with linsitinib normalized the amount of brown adipose tissue in both the early and late group. An in vivo MRI of the late group was performed and revealed a marked decrease of inflammation, visualized by 19F MR imaging, significant reduction of existing muscle edema and formation of brown adipose tissue. Conclusion: Here, we demonstrate that linsitinib effectively prevents development and progression of thyroid eye disease in an experimental murine model for Graves' disease. Linsitinib improved the total disease outcome, indicating the clinical significance of the findings and providing a path to therapeutic intervention of Graves' Disease. Our data support the use of linsitinib as a novel treatment for thyroid eye disease.


Asunto(s)
Enfermedad de Graves , Oftalmopatía de Graves , Inhibidores de Proteínas Quinasas , Receptor IGF Tipo 1 , Animales , Ratones , Enfermedad de Graves/tratamiento farmacológico , Oftalmopatía de Graves/tratamiento farmacológico , Hipertiroidismo , Imidazoles , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor IGF Tipo 1/antagonistas & inhibidores
3.
Magn Reson (Gott) ; 2(1): 355-374, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-37904770

RESUMEN

Dishevelled (Dvl) proteins are important regulators of the Wnt signalling pathway, interacting through their PDZ domains with the Wnt receptor Frizzled. Blocking the Dvl PDZ-Frizzled interaction represents a potential approach for cancer treatment, which stimulated the identification of small-molecule inhibitors, among them the anti-inflammatory drug Sulindac and Ky-02327. Aiming to develop tighter binding compounds without side effects, we investigated structure-activity relationships of sulfonamides. X-ray crystallography showed high complementarity of anthranilic acid derivatives in the GLGF loop cavity and space for ligand growth towards the PDZ surface. Our best binding compound inhibits Wnt signalling in a dose-dependent manner as demonstrated by TOP-GFP assays (IC50∼50 µM) and Western blotting of ß-catenin levels. Real-time PCR showed reduction in the expression of Wnt-specific genes. Our compound interacted with Dvl-1 PDZ (KD=2.4 µM) stronger than Ky-02327 and may be developed into a lead compound interfering with the Wnt pathway.

4.
Neuroendocrinology ; 111(5): 451-464, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32316022

RESUMEN

INTRODUCTION: G protein-coupled receptor (GPCR) mutations are implicated in many diseases. Most inactivating mutations cause receptor misfolding and prevent trafficking to the plasma membrane. Pharmacological chaperones can "rescue" cell surface expression of such mutants, presumably by stabilising correct folding of the nascent protein. OBJECTIVE: Here we examine the scope of intracellularly retained luteinising hormone receptor (LHR) mutants that can be "rescued" by the pharmacological chaperone LHR-Chap, and whether this allosteric agonist can also restore the function of mutant LHRs with deficiencies in hormone binding or hormone-induced signalling. METHODS: Mutant LHRs were expressed in HEK 293-T cells. Cell surface expression/localisation, hormone binding, and hCG/LHR-Chap signalling were determined by ELISA, radioligand binding, and inositol phosphate accumulation assays, respectively. Molecular modelling predicted LHR-Chap interactions. RESULTS: LHR-Chap increased cell surface expression of a subset of retained mutants located in transmembrane helices predicted to be stabilised by LHR-Chap binding. For 3 (T4613.47I, L5024.61P, and S6167.46Y) hCG-responsiveness was increased following treatment. LHRs with mutations in the hormone-binding site (C131ECDR and I152ECDT) or in the hinge region (E354HingeK) had good cell surface expression but poor response to hormone stimulation, yet were responsive to allosteric activation by LHR-Chap. CONCLUSIONS: LHR-Chap, in addition to rescuing cell surface expression of intracellularly retained LHR mutants, can rescue function in mutant receptors with binding and signalling deficiencies that have normal cell surface expression. This demonstration of rescue of multiple elements of LHR dysfunction arising from inactivating mutations offers exceptional potential for treating patients with diseases arising from GPCR mutations in general.


Asunto(s)
Regulación Alostérica , Chaperonas Moleculares , Receptores de HL/agonistas , Células HEK293 , Humanos , Proteínas Mutantes , Mutación , Pliegue de Proteína
5.
Int J Mol Sci ; 22(1)2020 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-33375708

RESUMEN

(1) The human luteinizing hormone (LH)/chorionic gonadotropin (hCG) receptor (LHCGR) discriminates its two hormone ligands and differs from the murine receptor (Lhr) in amino acid residues potentially involved in qualitative discerning of LH and hCG. The latter gonadotropin is absent in rodents. The aim of the study is to identify LHCGR residues involved in hCG/LH discrimination. (2) Eight LHCGR cDNAs were developed, carrying "murinizing" mutations on aminoacidic residues assumed to interact specifically with LH, hCG, or both. HEK293 cells expressing a mutant or the wild type receptor were treated with LH or hCG and the kinetics of cyclic adenosine monophosphate (cAMP) and phosphorylated extracellular signal-regulated kinases 1/2 (pERK1/2) activation was analyzed by bioluminescence resonance energy transfer (BRET). (3) Mutations falling within the receptor leucine reach repeat 9 and 10 (LRR9 and LRR10; K225S +T226I and R247T), of the large extracellular binding domain, are linked to loss of hormone-specific induced cAMP increase, as well as hCG-specific pERK1/2 activation, leading to a Lhr-like modulation of the LHCGR-mediated intracellular signaling pattern. These results support the hypothesis that LHCGR LRR domain is the interaction site of the hormone ß-L2 loop, which differs between LH and hCG, and might be fundamental for inducing gonadotropin-specific signals. (4) Taken together, these data identify LHCGR key residues likely evolved in the human to discriminate LH/hCG specific binding.


Asunto(s)
Aminoácidos/química , Sitios de Unión , Receptores de HL/química , Receptores de HL/metabolismo , Secuencia de Aminoácidos , Gonadotropina Coriónica/metabolismo , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Cinética , Hormona Luteinizante/metabolismo , Proteína Quinasa 1 Activada por Mitógenos , Proteína Quinasa 3 Activada por Mitógenos , Mutación , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de HL/genética , Transducción de Señal
7.
Biochim Biophys Acta Biomembr ; 1862(7): 183279, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32224152

RESUMEN

The functional and structural concept of tight junctions has developed after discovery of claudin and TAMP proteins. Many of these proteins contribute to epi- and endothelial barrier but some, in contrast, form paracellular channels. Claudins form the backbone of tight junction (TJ) strands whereas other proteins regulate TJ dynamics. The current joined double-row model of TJ strands and channels is crucially based on the linear alignment of claudin-15 in the crystal. Molecular dynamics simulations, protein docking, mutagenesis, cellular TJ reconstitution, and electron microscopy studies largely support stability and functionality of the model. Here, we summarize in silico and in vitro data about TJ strand assembly including comparison of claudin crystal structures and alternative models. Sequence comparisons, experimental and structural data substantiate differentiation of classic and non-classic claudins differing in motifs related to strand assembly. Classic claudins seem to share a similar mechanism of strand formation. Interface variations likely contribute to TJ strand flexibility. Combined in vitro/in silico studies are expected to elucidate mechanistic keys determining TJ regulation.


Asunto(s)
Claudinas/química , Conformación Proteica , Uniones Estrechas/química , Uniones Estrechas/genética , Claudinas/genética , Simulación por Computador , Células HEK293 , Humanos , Microscopía Electrónica , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis , Multimerización de Proteína , Uniones Estrechas/ultraestructura
8.
Cell Host Microbe ; 27(2): 238-248.e7, 2020 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-31901518

RESUMEN

Antimicrobial resistance in tuberculosis (TB) is a public health threat of global dimension, worsened by increasing drug resistance. Host-directed therapy (HDT) is an emerging concept currently explored as an adjunct therapeutic strategy for TB. One potential host target is the ligand-activated transcription factor aryl hydrocarbon receptor (AhR), which binds TB virulence factors and controls antibacterial responses. Here, we demonstrate that in the context of therapy, the AhR binds several TB drugs, including front line drugs rifampicin (RIF) and rifabutin (RFB), resulting in altered host defense and drug metabolism. AhR sensing of TB drugs modulates host defense mechanisms, notably impairs phagocytosis, and increases TB drug metabolism. Targeting AhR in vivo with a small-molecule inhibitor increases RFB-treatment efficacy. Thus, the AhR markedly impacts TB outcome by affecting both host defense and drug metabolism. As a corollary, we propose the AhR as a potential target for HDT in TB in adjunct to canonical chemotherapy.


Asunto(s)
Antituberculosos/metabolismo , Mycobacterium tuberculosis , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Tuberculosis/tratamiento farmacológico , Animales , Antituberculosos/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Humanos , Inmunidad Celular/efectos de los fármacos , Mycobacterium marinum/efectos de los fármacos , Mycobacterium marinum/patogenicidad , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Fagocitosis/efectos de los fármacos , Receptores de Hidrocarburo de Aril/metabolismo , Rifabutina/metabolismo , Rifabutina/uso terapéutico , Rifampin/metabolismo , Rifampin/uso terapéutico , Células THP-1 , Resultado del Tratamiento , Tuberculosis/microbiología , Pez Cebra
9.
Eur Thyroid J ; 9(Suppl 1): 66-77, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33511087

RESUMEN

Autoimmune thyroid-stimulating antibodies are activating the thyrotropin receptor (TSHR) in both the thyroid and the eye, but different molecular mechanisms are induced in both organs, leading to Graves' disease (GD) and Graves' orbitopathy (GO), respectively. Therapy with anti-thyroid drugs to reduce hyperthyroidism (GD) by suppressing the biosynthesis of thyroid hormones has only an indirect effect on GO, since it does not causally address pathogenic TSHR activation itself. GO is thus very difficult to treat. The activated TSHR but also the cross-interacting insulin-like growth factor 1 receptor (IGF-1R) contribute to this issue. The TSHR is a heptahelical G-protein-coupled receptor, whereas the IGF-1R is a receptor tyrosine kinase. Despite these fundamental structural differences, both receptors are phosphorylated by G-protein receptor kinases, which enables ß-arrestin binding. Arrestins mediate receptor internalization and also activate the mitogen-activated protein kinase pathway. Moreover, emerging results suggest that arrestin plays a critical role in the cross-interaction of the TSHR and the IGF-1R either in their common signaling pathway and/or during an indirect or potential TSHR/IGF-1R interaction. In this review, novel pharmacological strategies with allosteric small-molecule modulators to treat GO and GD on the level of the TSHR and/or the TSHR/IGF-1R cross-interaction will be discussed. Moreover, monoclonal antibody approaches targeting the TSHR or the IGF-1R and thereby preventing activation of either receptor will be presented. Another chapter addresses the immunomodulation to treat GO using TSHR-derived peptides targeting the human leukocyte antigen DR isotope (HLA-DR), which is a feasible approach to tackle GO, since HLA-DR and TSHR are overexpressed in orbital tissues of GO patients.

10.
Mol Oncol ; 14(2): 261-276, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31825142

RESUMEN

Clostridium perfringens enterotoxin (CPE) can be used to eliminate carcinoma cells that overexpress on their cell surface CPE receptors - a subset of claudins (e.g., Cldn3 and Cldn4). However, CPE cannot target tumors expressing solely CPE-insensitive claudins (such as Cldn1 and Cldn5). To overcome this limitation, structure-guided modifications were used to generate CPE variants that can strongly bind to Cldn1, Cldn2 and/or Cldn5, while maintaining the ability to bind Cldn3 and Cldn4. This enabled (a) targeting of the most frequent endocrine malignancy, namely, Cldn1-overexpressing thyroid cancer, and (b) improved targeting of the most common cancer type worldwide, non-small-cell lung cancer (NSCLC), which is characterized by high expression of several claudins, including Cldn1 and Cldn5. Different CPE variants, including the novel mutant CPE-Mut3 (S231R/S313H), were applied on thyroid cancer (K1 cells) and NSCLC (PC-9 cells) models. In vitro, CPE-Mut3, but not CPEwt, showed Cldn1-dependent binding and cytotoxicity toward K1 cells. For PC-9 cells, CPE-Mut3 improved claudin-dependent cytotoxic targeting, when compared to CPEwt. In vivo, intratumoral injection of CPE-Mut3 in xenograft models bearing K1 or PC-9 tumors induced necrosis and reduced the growth of both tumor types. Thus, directed modification of CPE enables eradication of tumor entities that cannot be targeted by CPEwt, for instance, Cldn1-overexpressing thyroid cancer by using the novel CPE-Mut3.


Asunto(s)
Antineoplásicos/farmacología , Claudinas/metabolismo , Clostridium perfringens/metabolismo , Enterotoxinas/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Claudina-1/química , Claudina-1/genética , Claudina-1/metabolismo , Claudina-3/química , Claudina-3/genética , Claudina-3/metabolismo , Claudina-4/química , Claudina-4/genética , Claudina-4/metabolismo , Claudina-5/química , Claudina-5/genética , Claudina-5/metabolismo , Claudinas/química , Claudinas/genética , Enterotoxinas/química , Enterotoxinas/uso terapéutico , Femenino , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Ratones , Mutagénesis Sitio-Dirigida , Mutación , Necrosis/inducido químicamente , Unión Proteica , Proteínas Recombinantes , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/terapia , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Exp Clin Endocrinol Diabetes ; 128(6-07): 379-382, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31739345

RESUMEN

Thyroid hormones (TH) pass through the plasma membrane into the target cells via transporter proteins. Thyroid hormone transporters that have been identified until now belong to two different solute carrier (SLC) subfamilies i) the major facilitator superfamily (MFS) and ii) the amino acid polyamine-organocation (APC) superfamily. Both are comprised by 12 transmembrane helices, however with different structural topology. The TH transporter MCT8, MCT10 and OATP1C1 are members of the MSF. The l-type amino acid transporters (LATs) are transporting neutral amino acids across the membrane. Two LAT subtypes, LAT1 and LAT2 are members of the APC superfamily, need the escort protein 4F2hc and facilitate uptake but no efflux of TH-subtypes. Homology models of LAT2 that are based on crystal structures of APC transporters guided mutagenesis, revealed molecular structure-function determinants for recognition and transition for import and export of TH-subtypes. The recently solved cryo-EM structure of LAT1 confirmed the structural input. Two other LAT subtypes, LAT3 and LAT4 are members of the MFS. From previous observed negative effect of LAT3 and LAT4 on 3,3'-T2 uptake by LAT1 and LAT2 it was indirectly concluded that LAT3 might export 3,3'-T2. There are still open questions that need to be addressed in order to fully understand the molecular recognition pattern and traversing mechanism of import and export of particular TH-subtypes by LAT1 and LAT2. Moreover, clarification is needed whether LAT3 and LAT4 are exporting TH. Recent new data could not verify the initial hypothesis of TH export by LAT3. Therefore, further investigations are necessary to explain the negative effect of LAT3 on the TH import by LAT2.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Enfermedades del Sistema Endocrino/metabolismo , Transducción de Señal/fisiología , Hormonas Tiroideas/metabolismo , Animales , Humanos
12.
Science ; 366(6472)2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31857448

RESUMEN

Pseudomonas aeruginosa rapidly adapts to altered conditions by quorum sensing (QS), a communication system that it uses to collectively modify its behavior through the production, release, and detection of signaling molecules. QS molecules can also be sensed by hosts, although the respective receptors and signaling pathways are poorly understood. We describe a pattern of regulation in the host by the aryl hydrocarbon receptor (AhR) that is critically dependent on qualitative and quantitative sensing of P. aeruginosa quorum. QS molecules bind to AhR and distinctly modulate its activity. This is mirrored upon infection with P. aeruginosa collected from diverse growth stages and with QS mutants. We propose that by spying on bacterial quorum, AhR acts as a major sensor of infection dynamics, capable of orchestrating host defense according to the status quo of infection.


Asunto(s)
Interacciones Huésped-Patógeno , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Células A549 , Animales , Humanos , Larva , Macrófagos/microbiología , Ratones , Ratones Noqueados , Pseudomonas aeruginosa/genética , Percepción de Quorum/genética , Receptores de Hidrocarburo de Aril/genética , Pez Cebra
13.
Int J Mol Sci ; 20(19)2019 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-31561440

RESUMEN

Claudins regulate paracellular permeability in different tissues. The claudin-binding domain of Clostridium perfringens enterotoxin (cCPE) is a known modulator of a claudin subset. However, it does not efficiently bind to claudin-1 (Cldn1). Cldn1 is a pharmacological target since it is (i) an essential co-receptor for hepatitis C virus (HCV) infections and (ii) a key element of the epidermal barrier limiting drug delivery. In this study, we investigated the potential of a Cldn1-binding cCPE mutant (i) to inhibit HCV entry into hepatocytes and (ii) to open the epidermal barrier. Inhibition of HCV infection by blocking of Cldn1 with cCPE variants was analyzed in the Huh7.5 hepatoma cell line. A model of reconstructed human epidermis was used to investigate modulation of the epidermal barrier by cCPE variants. In contrast to cCPEwt, the Cldn1-binding cCPE-S305P/S307R/S313H inhibited infection of Huh7.5 cells with HCV in a dose-dependent manner. In addition, TJ modulation by cCPE variant-mediated targeting of Cldn1 and Cldn4 opened the epidermal barrier in reconstructed human epidermis. cCPE variants are potent claudin modulators. They can be applied for mechanistic in vitro studies and might also be used as biologics for therapeutic claudin targeting including HCV treatment (host-targeting antivirals) and improvement of drug delivery.


Asunto(s)
Claudinas/metabolismo , Enterotoxinas/metabolismo , Hepatocitos/metabolismo , Piel/metabolismo , Sustitución de Aminoácidos , Línea Celular Tumoral , Claudinas/química , Enterotoxinas/química , Enterotoxinas/farmacología , Epidermis/metabolismo , Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Hepatitis C/metabolismo , Hepatitis C/virología , Humanos , Modelos Moleculares , Conformación Molecular , Unión Proteica , Piel/citología , Internalización del Virus/efectos de los fármacos , Replicación Viral
14.
Mol Pharmacol ; 96(4): 452-462, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31399504

RESUMEN

The large TSH-bound ectodomain of the thyrotropin receptor (TSHR) activates the transmembrane domain (TMD) indirectly via an internal agonist (IA). The ectodomain/TMD interface consists of a converging helix, a Cys-Cys-bridge-linked IA, and extracellular loops (ECL). To investigate the intramolecular course of molecular activation, especially details of the indirect activation, we narrowed down allosteric inhibition sites of negative allosteric modulator (NAM) by mutagenesis, homology modeling, and competition studies with positive allosteric modulator (PAM). From the inhibitory effects of NAM S37a on: 1) chimeras with swapped ectodomain, 2) stepwise N-terminal truncations, 3) distinct constitutively active mutations distributed across the hinge region and ECL, but not across the TMD, we conclude that S37a binds at the ectodomain/TMD interface, between the converging helix, ECL1, and the IA. This is also supported by the noncompetitive inhibition of PAM-C2-activation by S37a in the TSHR-TMD construct lacking the ectodomain. Mutagenesis studies on the IA and ECL were guided by our refined model of the ectodomain/TMD interface and indicate an interaction with the TSHR-specific residues E404 (preceding IA) and H478 (ECL1). At this new allosteric interaction site, NAM S37a blocks both TSH- and PAM-induced activation of the TSHR. Our refined models, mutations, and new allosteric binding pocket helped us to gain more detailed insights into the intramolecular course of TSHR activation at the ectodomain/TMD interface, including the delocalization of the converging helix and rearrangement of the conformation of IA. These changes are embedded between the ECL and cooperatively trigger active conformations of TMD. SIGNIFICANCE STATEMENT: The intramolecular activation mechanisms of the TSHR appear to be distinct from those of other G protein-coupled receptors, as the TSHR has a uniquely large N-terminal ectodomain that includes the hormone binding site and an internal agonist sequence. We present new molecular and structural insights into the interface between ectodomain and transmembrane domain in the TSHR, as well as the transfer of activation to the transmembrane domain. This knowledge is critical for understanding activation or inhibition of the receptor by allosteric ligands. We have identified a new allosteric antagonist binding pocket that is located exactly at this interface and possesses specific features that may allow the generation of potent highly TSHR-selective drugs, of potential value for the treatment of Graves' orbitopathy.


Asunto(s)
Receptores de Tirotropina/química , Receptores de Tirotropina/metabolismo , Tirotropina/metabolismo , Regulación Alostérica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Dominios Proteicos , Receptores de Tirotropina/genética , Homología de Secuencia de Aminoácido , Transducción de Señal
15.
Sci Rep ; 9(1): 10878, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31350436

RESUMEN

As a first host barrier, the skin is constantly exposed to environmental insults that perturb its integrity. Tight regulation of skin homeostasis is largely controlled by the aryl hydrocarbon receptor (AhR). Here, we demonstrate that Henna and its major pigment, the naphthoquinone Lawsone activate AhR, both in vitro and in vivo. In human keratinocytes and epidermis equivalents, Lawsone exposure enhances the production of late epidermal proteins, impacts keratinocyte differentiation and proliferation, and regulates skin inflammation. To determine the potential use of Lawsone for therapeutic application, we harnessed human, murine and zebrafish models. In skin regeneration models, Lawsone interferes with physiological tissue regeneration and inhibits wound healing. Conversely, in a human acute dermatitis model, topical application of a Lawsone-containing cream ameliorates skin irritation. Altogether, our study reveals how a widely used natural plant pigment is sensed by the host receptor AhR, and how the physiopathological context determines beneficial and detrimental outcomes.


Asunto(s)
Dermatitis/tratamiento farmacológico , Queratinocitos/metabolismo , Naftoquinonas/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Piel/metabolismo , Animales , Células Cultivadas , Regeneración Tisular Dirigida , Homeostasis , Humanos , Lawsonia (Planta) , Ratones , Modelos Animales , Naftoquinonas/uso terapéutico , Piel/efectos de los fármacos , Piel/patología , Cicatrización de Heridas , Pez Cebra
16.
Thyroid ; 29(1): 111-123, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30351237

RESUMEN

BACKGROUND: The thyrotropin receptor (TSHR) is the target for autoimmune thyroid stimulating antibodies (TSAb) triggering hyperthyroidism. Whereas elevated thyroid hormone synthesis by the thyroid in Graves' disease can be treated by antithyroid agents, for the pathogenic activation of TSHR in retro-orbital fibroblasts of the eye, leading to Graves' orbitopathy (GO), no causal TSHR directed therapy is available. METHODS: Due to the therapeutic gap for severe GO, TSHR inhibitors were identified by high-throughput screening in Chinese hamster ovary cells expressing the TSHR. Stereo-selective synthesis of the screening hits led to the molecule S37, which contains seven chiral centers. Enantiomeric separation of the molecule S37 resulted in the enantiopure molecule S37a-a micro-molar antagonist of thyrotropin-induced cyclic adenosine monophosphate accumulation in HEK 293 cells expressing the TSHR. RESULTS: The unique rigid bent shape of molecule S37a may mediate the observed high TSHR selectivity. Most importantly, the closely related follitropin and lutropin receptors were not affected by this compound. S37a not only inhibits the TSHR activation by thyrotropin itself but also activation by monoclonal TSAb M22 (human), KSAb1 (murine), and the allosteric small-molecule agonist C2. Disease-related ex vivo studies in HEK 293 cells expressing the TSHR showed that S37a also inhibits cyclic adenosine monophosphate formation by oligoclonal TSAb, which are highly enriched in GO patients' sera. Initial in vivo pharmacokinetic studies revealed no toxicity of S37a and a remarkable 53% oral bioavailability in mice. CONCLUSION: In summary, a novel highly selective inhibitor for the TSHR is presented, which has promising potential for further development for the treatment of GO.


Asunto(s)
Oftalmopatía de Graves/tratamiento farmacológico , Antagonistas de Hormonas/farmacología , Receptores de Tirotropina/antagonistas & inhibidores , Animales , Células CHO , Cricetinae , Cricetulus , Fibroblastos/efectos de los fármacos , Células HEK293 , Antagonistas de Hormonas/uso terapéutico , Humanos , Transducción de Señal/efectos de los fármacos
18.
Horm Metab Res ; 50(12): 894-907, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30366347

RESUMEN

The thyrotropin receptor (TSHR), the lutropin- (LHR), and the follicotropin receptor (FSHR) belong to glycoprotein hormone receptors (GPHR), a subgroup of the class A G-protein coupled receptors. In this review, the unique features of GPHR have been taken into account for their pharmacological interventions: i) The respective hormone and stimulating or blocking antibodies are binding on the large ectodomain that is ii) via a hinge region, containing iii) an internal tethered agonist linked to the transmembrane domain. iv) Multimerization and mechanisms for negative or positive cooperativity of GPHR upon ligand binding and v) dimer- and oligomeric arrangements enabling trans-activation on GPHR signaling are considered. Available knowledge concerning the modulation of the GPHR (mal)-function and associated structural aspects by diverse entities such as antibodies, chaperones, peptides, small molecule agonists, inverse agonists, and antagonists is summarized. The TSHR is important with respect to autoimmune [Graves' disease (GD), Graves' orbitopathy (GO)] or non-autoimmune thyroid dysfunctions and cancer-development. To date there is neither an agonist nor antagonist modulator of pathogenic such as TSHR signaling in the clinics. However, several different ligands monoclonal stimulating and inhibiting antibodies and small molecule drug-like ligands have been reported in the last decade. In special focus are the most recent findings regarding the development and use of small molecule TSHR ligands. Finally, limitations of current knowledge and lack of information are discussed highlighting the need for intensified efforts towards understanding the interplay of TSHR multimers, especially their interaction with drug-like ligands. Important in this context is the biased ligand development.


Asunto(s)
Receptores de Tirotropina/metabolismo , Regulación Alostérica , Animales , Humanos , Ligandos , Mutación/genética , Receptores de Tirotropina/química , Receptores de Tirotropina/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
19.
Biomaterials ; 161: 129-143, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29421550

RESUMEN

The blood-brain barrier (BBB) prevents entry of neurotoxic substances but also that of drugs into the brain. Here, the paracellular barrier is formed by tight junctions (TJs) with claudin-5 (Cldn5) being the main sealing constituent. Transient BBB opening by targeting Cldn5 could improve paracellular drug delivery. The non-toxic C-terminal domain of Clostridium perfringens enterotoxin (cCPE) binds to a subset of claudins, e.g., Cldn3, -4. Structure-based mutagenesis was used to generate Cldn5-binding variants (cCPE-Y306W/S313H and cCPE-N218Q/Y306W/S313H). These cCPE-variants were tested for transient TJ opening using multiple in vitro BBB models: Primary porcine brain endothelial cells, coculture of primary rat brain endothelial cells with astrocytes and mouse cerebEND cells. cCPE-Y306W/S313H and cCPE-N218Q/Y306W/S313H but neither cCPE-wt nor cCPE-Y306A/L315A (not binding to claudins) decreased transendothelial electrical resistance in a concentration-dependent and reversible manner. Furthermore, permeability of carboxyfluorescein (with size of CNS drugs) was increased. cCPE-Y306W/S313H but neither cCPE-wt nor cCPE-Y306A/L315A bound to Cldn5-expressing brain endothelial cells. However, freeze-fracture EM showed that cCPE-Y306W/S313H did not cause drastic TJ breakdown. In sum, Cldn5-binding cCPE-variants enabled mild and transient opening of brain endothelial TJs. Using reliable in vitro BBB models, the results demonstrate that cCPE-based biologics designed to bind Cldn5 improve paracellular drug delivery across the BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Claudina-5/metabolismo , Clostridium perfringens/metabolismo , Enterotoxinas/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/ultraestructura , Encéfalo/metabolismo , Encéfalo/ultraestructura , Células Cultivadas , Células Endoteliales/metabolismo , Enterotoxinas/química , Técnica de Fractura por Congelación , Células HEK293 , Humanos , Microscopía Electrónica , Unión Proteica , Porcinos , Uniones Estrechas/metabolismo , Uniones Estrechas/ultraestructura
20.
J Infect Dis ; 217(1): 147-157, 2017 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-28968861

RESUMEN

Clostridium perfringens enterotoxin (CPE) causes food poisoning and antibiotic-associated diarrhea. It uses some claudin tight junction proteins (eg, claudin-4) as receptors to form Ca2+-permeable pores in the membrane, damaging epithelial cells in small intestine and colon. We demonstrate that only a subpopulation of colonic enterocytes which are characterized by apical dislocation of claudins are CPE-susceptible. CPE-mediated damage was enhanced if paracellular barrier was impaired by Ca2+ depletion, proinflammatory cytokine tumor necrosis factor α, or dedifferentiation. Microscopy, Ca2+ monitoring, and electrophysiological data showed that CPE-mediated cytotoxicity and barrier disruption was limited by extent of CPE-binding. The latter was restricted by accessibility of non-junctional claudin molecules such as claudin-4 at apical membranes. Focal-leaks detected in HT-29/B6 colonic monolayers were verified for native tissue using colon biopsies. These mechanistic findings indicate how CPE-mediated effects may turn from self-limiting diarrhea into severe clinical manifestation such as colonic necrosis-if intestinal barrier dysfunction, eg, during inflammation facilitates claudin accessibility.


Asunto(s)
Claudinas/antagonistas & inhibidores , Infecciones por Clostridium/patología , Clostridium perfringens/patogenicidad , Colon/patología , Enterotoxinas/toxicidad , Enfermedades Transmitidas por los Alimentos/patología , Uniones Estrechas/patología , Línea Celular , Enterocitos/patología , Humanos , Mucosa Intestinal/patología , Permeabilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...