Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Signal ; 118: 111149, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38522808

RESUMEN

G protein-coupled receptors (GPCR) and glycosaminoglycans (GAGs) are two essential components of the cell surface that regulate physiological processes in the body. GPCRs are the most extensive family of transmembrane receptors that control cellular responses to extracellular stimuli, while GAGs are polysaccharides that contribute to the function of the extracellular matrix (ECM). Due to their proximity to the plasma membrane, GAGs participate in signal transduction by interacting with various extracellular molecules and cell surface receptors. GAGs can directly interact with certain GPCRs or their ligands (chemokines, peptide hormones and neuropeptides, structural proteins, and enzymes) from the glutamate receptor family, the rhodopsin receptor family, the adhesion receptor family, and the secretin receptor family. These interactions have recently become an emerging topic, providing a new avenue for understanding how GPCR signaling is regulated. This review discusses our current state of knowledge about the role of GAGs in GPCR signaling and function.


Asunto(s)
Glicosaminoglicanos , Receptores Acoplados a Proteínas G , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , Membrana Celular/metabolismo , Rodopsina/metabolismo
2.
bioRxiv ; 2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37961094

RESUMEN

Since it was proposed as a potential host-directed antiviral agent for SARS-CoV-2, the antiparasitic drug ivermectin has been investigated thoroughly in clinical trials, which have provided insufficient support for its clinical efficacy. To examine the potential for ivermectin to be repurposed as an antiviral agent, we therefore undertook a series of preclinical studies. Consistent with early reports, ivermectin decreased SARS-CoV-2 viral burden in in vitro models at low micromolar concentrations, five- to ten-fold higher than the reported toxic clinical concentration. At similar concentrations, ivermectin also decreased cell viability and increased biomarkers of cytotoxicity and apoptosis. Further mechanistic and profiling studies revealed that ivermectin nonspecifically perturbs membrane bilayers at the same concentrations where it decreases the SARS-CoV-2 viral burden, resulting in nonspecific modulation of membrane-based targets such as G-protein coupled receptors and ion channels. These results suggest that a primary molecular mechanism for the in vitro antiviral activity of ivermectin may be nonspecific membrane perturbation, indicating that ivermectin is unlikely to be translatable into a safe and effective antiviral agent. These results and experimental workflow provide a useful paradigm for performing preclinical studies on (pandemic-related) drug repurposing candidates.

3.
Endocr Rev ; 44(3): 474-491, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36503956

RESUMEN

The classical paradigm of G protein-coupled receptor (GPCR) signaling via G proteins is grounded in a view that downstream responses are relatively transient and confined to the cell surface, but this notion has been revised in recent years following the identification of several receptors that engage in sustained signaling responses from subcellular compartments following internalization of the ligand-receptor complex. This phenomenon was initially discovered for the parathyroid hormone (PTH) type 1 receptor (PTH1R), a vital GPCR for maintaining normal calcium and phosphate levels in the body with the paradoxical ability to build or break down bone in response to PTH binding. The diverse biological processes regulated by this receptor are thought to depend on its capacity to mediate diverse modes of cyclic adenosine monophosphate (cAMP) signaling. These include transient signaling at the plasma membrane and sustained signaling from internalized PTH1R within early endosomes mediated by PTH. Here we discuss recent structural, cell signaling, and in vivo studies that unveil potential pharmacological outputs of the spatial versus temporal dimension of PTH1R signaling via cAMP. Notably, the combination of molecular dynamics simulations and elastic network model-based methods revealed how precise modulation of PTH signaling responses is achieved through structure-encoded allosteric coupling within the receptor and between the peptide hormone binding site and the G protein coupling interface. The implications of recent findings are now being explored for addressing key questions on how location bias in receptor signaling contributes to pharmacological functions, and how to drug a difficult target such as the PTH1R toward discovering nonpeptidic small molecule candidates for the treatment of metabolic bone and mineral diseases.


Asunto(s)
Proteína Relacionada con la Hormona Paratiroidea , Hormona Paratiroidea , Humanos , Hormona Paratiroidea/farmacología , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Transducción de Señal/fisiología , Receptores Acoplados a Proteínas G , AMP Cíclico/metabolismo
4.
J Biol Chem ; 298(9): 102332, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35933010

RESUMEN

The parathyroid hormone (PTH)-related protein (PTHrP) is indispensable for the development of mammary glands, placental calcium ion transport, tooth eruption, bone formation and bone remodeling, and causes hypercalcemia in patients with malignancy. Although mature forms of PTHrP in the body consist of splice variants of 139, 141, and 173 amino acids, our current understanding on how endogenous PTHrP transduces signals through its cognate G-protein coupled receptor (GPCR), the PTH type 1 receptor (PTHR), is largely derived from studies done with its N-terminal fragment, PTHrP1-36. Here, we demonstrate using various fluorescence imaging approaches at the single cell level to measure kinetics of (i) receptor activation, (ii) receptor signaling via Gs and Gq, and (iii) receptor internalization and recycling that the native PTHrP1-141 displays biased agonist signaling properties that are not mimicked by PTHrP1-36. Although PTHrP1-36 induces transient cAMP production, acute intracellular Ca2+ (iCa2+) release and ß-arrestin recruitment mediated by ligand-PTHR interactions at the plasma membrane, PTHrP1-141 triggers sustained cAMP signaling from the plasma membrane and fails to stimulate iCa2+ release and recruit ß-arrestin. Furthermore, we show that the molecular basis for biased signaling differences between PTHrP1-36 and properties of native PTHrP1-141 are caused by the stabilization of a singular PTHR conformation and PTHrP1-141 sensitivity to heparin, a sulfated glycosaminoglycan. Taken together, our results contribute to a better understanding of the biased signaling process of a native protein hormone acting in conjunction with a GPCR.


Asunto(s)
Receptor de Hormona Paratiroídea Tipo 1 , AMP Cíclico/metabolismo , Heparina/metabolismo , Humanos , Ligandos , Conformación Proteica , Receptor de Hormona Paratiroídea Tipo 1/química , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo
5.
Nat Chem Biol ; 18(3): 272-280, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34949836

RESUMEN

Class B G protein-coupled receptors (GPCRs) are notoriously difficult to target by small molecules because their large orthosteric peptide-binding pocket embedded deep within the transmembrane domain limits the identification and development of nonpeptide small molecule ligands. Using the parathyroid hormone type 1 receptor (PTHR) as a prototypic class B GPCR target, and a combination of molecular dynamics simulations and elastic network model-based methods, we demonstrate that PTHR druggability can be effectively addressed. Here we found a key mechanical site that modulates the collective dynamics of the receptor and used this ensemble of PTHR conformers to identify selective small molecules with strong negative allosteric and biased properties for PTHR signaling in cell and PTH actions in vivo. This study provides a computational pipeline to detect precise druggable sites and identify allosteric modulators of PTHR signaling that could be extended to GPCRs to expedite discoveries of small molecules as novel therapeutic candidates.


Asunto(s)
Receptor de Hormona Paratiroídea Tipo 1 , Receptores Acoplados a Proteínas G , Ligandos , Simulación de Dinámica Molecular , Transducción de Señal
6.
Sci Signal ; 14(703): eabc5944, 2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34609896

RESUMEN

The parathyroid hormone (PTH) type 1 receptor (PTHR) is a class B G protein­coupled receptor (GPCR) that regulates mineral ion, vitamin D, and bone homeostasis. Activation of the PTHR by PTH induces both transient cell surface and sustained endosomal cAMP production. To address whether the spatial (location) or temporal (duration) dimension of PTHR-induced cAMP encodes distinct biological outcomes, we engineered a biased PTHR ligand (PTH7d) that elicits cAMP production at the plasma membrane but not at endosomes. PTH7d stabilized a unique active PTHR conformation that mediated sustained cAMP signaling at the plasma membrane due to impaired ß-arrestin coupling to the receptor. Experiments in cells and mice revealed that sustained cAMP production by cell surface PTHR failed to mimic the pharmacological effects of sustained endosomal cAMP production on the abundance of the rate-limiting hydroxylase catalyzing the formation of active vitamin D, as well as increases in circulating active vitamin D and Ca2+ and in bone formation in mice. Thus, similar amounts of cAMP generated by PTHR for similar lengths of time in different cellular locations, plasma membrane and endosomes, mediate distinct physiological responses. These results unveil subcellular signaling location as a means to achieve specificity in PTHR-mediated biological outcomes and raise the prospect of rational drug design based upon spatiotemporal manipulation of GPCR signaling.


Asunto(s)
Hormona Paratiroidea , Receptores de Hormona Paratiroidea , AMP Cíclico
7.
Nat Metab ; 2(3): 243-255, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32694772

RESUMEN

Molecular mechanisms mediating tonic secretion of parathyroid hormone (PTH) in response to hypocalcaemia and hyperparathyroidism (HPT) are unclear. Here we demonstrate increased heterocomplex formation between the calcium-sensing receptor (CaSR) and metabotropic γ-aminobutyric acid (GABA) B1 receptor (GABAB1R) in hyperplastic parathyroid glands (PTGs) of patients with primary and secondary HPT. Targeted ablation of GABAB1R or glutamic acid decarboxylase 1 and 2 in PTGs produces hypocalcaemia and hypoparathyroidism, and prevents PTH hypersecretion in PTGs cultured from mouse models of hereditary HPT and dietary calcium-deficiency. Cobinding of the CaSR/GABAB1R complex by baclofen and high extracellular calcium blocks the coupling of heterotrimeric G-proteins to homomeric CaSRs in cultured cells and promotes PTH secretion in cultured mouse PTGs. These results combined with the ability of PTG to synthesize GABA support a critical autocrine action of GABA/GABAB1R in mediating tonic PTH secretion of PTGs and ascribe aberrant activities of CaSR/GABAB1R heteromer to HPT.


Asunto(s)
Hiperparatiroidismo Secundario/metabolismo , Hormona Paratiroidea/metabolismo , Receptores Sensibles al Calcio/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Calcio/metabolismo , Humanos , Hiperparatiroidismo Secundario/complicaciones , Hipocalcemia/complicaciones , Hipocalcemia/metabolismo , Ratones , Receptores de GABA-B/metabolismo
8.
Nat Chem Biol ; 16(10): 1096-1104, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32632293

RESUMEN

Peptide ligands of class B G-protein-coupled receptors act via a two-step binding process, but the essential mechanisms that link their extracellular binding to intracellular receptor-arrestin interactions are not fully understood. Using NMR, crosslinking coupled to mass spectrometry, signaling experiments and computational approaches on the parathyroid hormone (PTH) type 1 receptor (PTHR), we show that initial binding of the PTH C-terminal part constrains the conformation of the flexible PTH N-terminal signaling epitope before a second binding event occurs. A 'hot-spot' PTH residue, His9, that inserts into the PTHR transmembrane domain at this second step allosterically engages receptor-arrestin coupling. A conformational change in PTHR intracellular loop 3 permits favorable interactions with ß-arrestin's finger loop. These results unveil structural determinants for PTHR-arrestin complex formation and reveal that the two-step binding mechanism proceeds via cooperative fluctuations between ligand and receptor, which extend to other class B G-protein-coupled receptors.


Asunto(s)
Arrestina/metabolismo , Hormona Paratiroidea/metabolismo , Arrestina/química , Fosfatos de Calcio , Microscopía por Crioelectrón , AMP Cíclico , Escherichia coli , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Hormona Paratiroidea/química , Receptores Acoplados a Proteínas G
9.
Proc Natl Acad Sci U S A ; 117(13): 7455-7460, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32184323

RESUMEN

cAMP production upon activation of Gs by G protein-coupled receptors has classically been considered to be plasma membrane-delimited, but a shift in this paradigm has occurred in recent years with the identification of several receptors that continue to signal from early endosomes after internalization. The molecular mechanisms regulating this aspect of signaling remain incompletely understood. Here, we investigated the role of Gq/11 activation by the parathyroid hormone (PTH) type 1 receptor (PTHR) in mediating endosomal cAMP responses. Inhibition of Gq/11 signaling by FR900359 markedly reduced the duration of PTH-induced cAMP production, and this effect was mimicked in cells lacking endogenous Gαq/11 We determined that modulation of cAMP generation by Gq/11 occurs at the level of the heterotrimeric G protein via liberation of cell surface Gßγ subunits, which, in turn, act in a phosphoinositide-3 kinase-dependent manner to promote the assembly of PTHR-ßarrestin-Gßγ signaling complexes that mediate endosomal cAMP responses. These results unveil insights into the spatiotemporal regulation of Gs-dependent cAMP signaling.


Asunto(s)
AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Animales , Arrestinas/metabolismo , Membrana Celular/metabolismo , Depsipéptidos/farmacología , Endosomas/metabolismo , Células HEK293 , Humanos , Ratones , Osteoblastos/metabolismo , Hormona Paratiroidea/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Cultivo Primario de Células , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología , beta-Arrestinas/metabolismo
10.
Trends Endocrinol Metab ; 30(11): 860-874, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31699241

RESUMEN

The parathyroid hormone (PTH) type 1 receptor (PTHR) is the canonical G protein-coupled receptor (GPCR) for PTH and PTH-related protein (PTHrP) and the key regulator of calcium homeostasis and bone turnover. PTHR function is critical for human health to maintain homeostatic control of ionized serum Ca2+ levels and has several unusual signaling features, such as endosomal cAMP signaling, that are well-studied but not structurally understood. In this review, we discuss how recently solved high resolution near-atomic structures of hormone-bound PTHR in its inactive and active signaling states and discovery of extracellular Ca2+ allosterism shed light on the structural basis for PTHR signaling and function.


Asunto(s)
Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Animales , Endosomas/metabolismo , Humanos , Receptor de Hormona Paratiroídea Tipo 1/química , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/fisiología
11.
J Am Chem Soc ; 141(37): 14486-14490, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31496241

RESUMEN

The type-1 parathyroid hormone receptor (PTHR1), which regulates calcium homeostasis and tissue development, has two native agonists, parathyroid hormone (PTH) and PTH-related protein (PTHrP). PTH forms a complex with the PTHR1 that is rapidly internalized and induces prolonged cAMP production from endosomes. In contrast, PTHrP induces only transient cAMP production, which primarily arises from receptors on the cell surface. We show that backbone modification of PTH(1-34)-NH2 and abaloparatide (a PTHrP derivative) with a single homologous ß-amino acid residue can generate biased agonists that induce prolonged cAMP production from receptors at the cell surface. This unique spatiotemporal profile could be useful for distinguishing effects associated with the duration of cAMP production from effects associated with the site of cAMP production.


Asunto(s)
Receptor de Hormona Paratiroídea Tipo 1/química , Secuencia de Aminoácidos , AMP Cíclico/biosíntesis , Células HEK293 , Humanos , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Homología de Secuencia de Aminoácido , Transducción de Señal
12.
Science ; 364(6436): 148-153, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30975883

RESUMEN

The parathyroid hormone receptor-1 (PTH1R) is a class B G protein-coupled receptor central to calcium homeostasis and a therapeutic target for osteoporosis and hypoparathyroidism. Here we report the cryo-electron microscopy structure of human PTH1R bound to a long-acting PTH analog and the stimulatory G protein. The bound peptide adopts an extended helix with its amino terminus inserted deeply into the receptor transmembrane domain (TMD), which leads to partial unwinding of the carboxyl terminus of transmembrane helix 6 and induces a sharp kink at the middle of this helix to allow the receptor to couple with G protein. In contrast to a single TMD structure state, the extracellular domain adopts multiple conformations. These results provide insights into the structural basis and dynamics of PTH binding and receptor activation.


Asunto(s)
Hormona Paratiroidea/química , Receptor de Hormona Paratiroídea Tipo 1/agonistas , Receptor de Hormona Paratiroídea Tipo 1/química , Secuencias de Aminoácidos , Microscopía por Crioelectrón , Humanos , Hormona Paratiroidea/farmacología , Hormona Paratiroidea/fisiología , Unión Proteica , Dominios Proteicos , Receptor de Hormona Paratiroídea Tipo 1/ultraestructura
13.
Proc Natl Acad Sci U S A ; 116(8): 3294-3299, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30718391

RESUMEN

The parathyroid hormone (PTH) and its related peptide (PTHrP) activate PTH receptor (PTHR) signaling, but only the PTH sustains GS-mediated adenosine 3',5'-cyclic monophosphate (cAMP) production after PTHR internalization into early endosomes. The mechanism of this unexpected behavior for a G-protein-coupled receptor is not fully understood. Here, we show that extracellular Ca2+ acts as a positive allosteric modulator of PTHR signaling that regulates sustained cAMP production. Equilibrium and kinetic studies of ligand-binding and receptor activation reveal that Ca2+ prolongs the residence time of ligands on the receptor, thus, increasing both the duration of the receptor activation and the cAMP signaling. We further find that Ca2+ allostery in the PTHR is strongly affected by the point mutation recently identified in the PTH (PTHR25C) as a new cause of hypocalcemia in humans. Using high-resolution and mass accuracy mass spectrometry approaches, we identified acidic clusters in the receptor's first extracellular loop as key determinants for Ca2+ allosterism and endosomal cAMP signaling. These findings coupled to defective Ca2+ allostery and cAMP signaling in the PTHR by hypocalcemia-causing PTHR25C suggest that Ca2+ allostery in PTHR signaling may be involved in primary signaling processes regulating calcium homeostasis.


Asunto(s)
AMP Cíclico/genética , Hipocalcemia/genética , Hormona Paratiroidea/genética , Receptor de Hormona Paratiroídea Tipo 1/genética , Regulación Alostérica/genética , Animales , Células COS , Señalización del Calcio/genética , Chlorocebus aethiops , AMP Cíclico/metabolismo , Humanos , Hipocalcemia/metabolismo , Hipocalcemia/patología , Cinética , Ligandos , Hormona Paratiroidea/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/genética , Mutación Puntual/genética , Unión Proteica/genética , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
14.
J Biol Chem ; 294(4): 1095-1103, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30559293

RESUMEN

cAMP is a ubiquitous second messenger that regulates cellular proliferation, differentiation, attachment, migration, and several other processes. It has become increasingly evident that tight regulation of cAMP accumulation and localization confers divergent yet specific signaling to downstream pathways. Currently, few tools are available that have sufficient spatial and temporal resolution to study location-biased cAMP signaling. Here, we introduce a new fusion protein consisting of a light-activated adenylyl cyclase (bPAC) and luciferase (nLuc). This construct allows dual activation of cAMP production through temporally precise photostimulation or chronic chemical stimulation that can be fine-tuned to mimic physiological levels and duration of cAMP synthesis to trigger downstream events. By targeting this construct to different compartments, we show that cAMP produced in the cytosol and nucleus stimulates proliferation in thyroid cells. The bPAC-nLuc fusion construct adds a new reagent to the available toolkit to study cAMP-regulated processes in living cells.


Asunto(s)
Adenilil Ciclasas/metabolismo , AMP Cíclico/biosíntesis , Activación Enzimática/efectos de la radiación , Luminiscencia , Animales , Proliferación Celular , Células Cultivadas , Células HEK293 , Humanos , Luz , Luciferasas/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA