Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1990, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38443355

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of membrane proteins and are important drug targets. The discovery of drugs targeting these receptors and their G protein signaling properties are based on assays mainly performed with modified receptors expressed in heterologous cells. However, GPCR responses may differ in their native environment. Here, by using highly sensitive Gi/o sensors, we reveal specific properties of Gi/o protein-mediated responses triggered by GABAB, α2 adrenergic and cannabinoid CB1 receptors in primary neurons, different from those in heterologous cells. These include different profiles in the Gi/o protein subtypes-mediated responses, and differences in the potencies of some ligands even at similar receptor expression levels. Altogether, our results show the importance of using biosensors compatible with primary cells for evaluating the activities of endogenous GPCRs in their native environment.


Subject(s)
Neurons , Receptors, G-Protein-Coupled , Signal Transduction , Adrenergic Agents , Biological Assay , GTP-Binding Protein alpha Subunits, Gi-Go
2.
Nat Chem Biol ; 20(7): 857-866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38355723

ABSTRACT

Major depressive disorder, a prevalent and severe psychiatric condition, necessitates development of new and fast-acting antidepressants. Genetic suppression of astrocytic inwardly rectifying potassium channel 4.1 (Kir4.1) in the lateral habenula ameliorates depression-like phenotypes in mice. However, Kir4.1 remains an elusive drug target for depression. Here, we discovered a series of Kir4.1 inhibitors through high-throughput screening. Lys05, the most potent one thus far, effectively suppressed native Kir4.1 channels while displaying high selectivity against established targets for rapid-onset antidepressants. Cryogenic-electron microscopy structures combined with electrophysiological characterizations revealed Lys05 directly binds in the central cavity of Kir4.1. Notably, a single dose of Lys05 reversed the Kir4.1-driven depression-like phenotype and exerted rapid-onset (as early as 1 hour) antidepressant actions in multiple canonical depression rodent models with efficacy comparable to that of (S)-ketamine. Overall, we provided a proof of concept that Kir4.1 is a promising target for rapid-onset antidepressant effects.


Subject(s)
Antidepressive Agents , Potassium Channels, Inwardly Rectifying , Antidepressive Agents/pharmacology , Antidepressive Agents/chemistry , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Mice , Male , Rats , Humans , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Potassium Channel Blockers/pharmacology , Potassium Channel Blockers/chemistry
3.
Am J Physiol Cell Physiol ; 325(1): C79-C89, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37184233

ABSTRACT

G protein-coupled receptors (GPCRs) represent the largest family of membrane proteins and are important drug targets. GPCRs are allosteric machines that transduce an extracellular signal to the cell by activating heterotrimeric G proteins. Herein, we summarize the recent advancements in the molecular activation mechanism of the γ-aminobutyric acid type B (GABAB) and metabotropic glutamate (mGlu) receptors, the most important class C GPCRs that modulate synaptic transmission in the brain. Both are mandatory dimers, this quaternary structure being needed for their function The structures of these receptors in different conformations and in complexes with G proteins have revealed their asymmetric activation. This asymmetry is further highlighted by the recent discovery of mGlu heterodimers, where the eight mGlu subunits can form specific and functional heterodimers. Finally, the development of allosteric modulators has revealed new possibilities for regulating the function of these receptors by targeting the transmembrane dimer interface. This family of receptors never ceases to astonish and serve as models to better understand the diversity and asymmetric functioning of GPCRs.NEW & NOTEWORTHY γ-aminobutyric acid type B (GABAB) and metabotropic glutamate (mGlu) receptors form constitutive dimers, which are required for their function. They serve as models to better understand the diversity and activation of G protein-coupled receptors (GPCRs). The structures of these receptors in different conformations and in complexes with G proteins have revealed their asymmetric activation. This asymmetry is further highlighted by the recent discovery of specific and functional mGlu heterodimers. Allosteric modulators can be developed to target the transmembrane interface and modulate the asymmetry.


Subject(s)
Receptors, Metabotropic Glutamate , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation , Receptors, G-Protein-Coupled , Synaptic Transmission , Glutamic Acid , Receptors, GABA-B/genetics , Receptors, GABA-B/metabolism
4.
Nat Commun ; 13(1): 6365, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36289206

ABSTRACT

G protein-coupled receptors (GPCRs) are important drug targets that mediate various signaling pathways by activating G proteins and engaging ß-arrestin proteins. Despite its importance for the development of therapeutics with fewer side effects, the underlying mechanism that controls the balance between these signaling modes of GPCRs remains largely unclear. Here, we show that assembly into dimers and oligomers can largely influence the signaling mode of the platelet-activating factor receptor (PAFR). Single-particle analysis results show that PAFR can form oligomers at low densities through two possible dimer interfaces. Stabilization of PAFR oligomers through cross-linking increases G protein activity, and decreases ß-arrestin recruitment and agonist-induced internalization significantly. Reciprocally, ß-arrestin prevents PAFR oligomerization. Our results highlight a mechanism involved in the control of receptor signaling, and thereby provide important insights into the relationship between GPCR oligomerization and downstream signaling.


Subject(s)
Platelet Activating Factor , Receptors, G-Protein-Coupled , Platelet Activating Factor/metabolism , Platelet Activating Factor/pharmacology , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Signal Transduction , beta-Arrestin 1/metabolism , GTP-Binding Proteins/metabolism
5.
Nat Chem Biol ; 18(8): 894-903, 2022 08.
Article in English | MEDLINE | ID: mdl-35681029

ABSTRACT

Membrane proteins, including ion channels, receptors and transporters, are often composed of multiple subunits and can form large complexes. Their specific composition in native tissues is difficult to determine and remains largely unknown. In this study, we developed a method for determining the subunit composition of endogenous cell surface protein complexes from isolated native tissues. Our method relies on nanobody-based sensors, which enable proximity detection between subunits in time-resolved Förster resonance energy transfer (FRET) measurements. Additionally, given conformation-specific nanobodies, the activation of these complexes can be recorded in native brain tissue. Applied to the metabotropic glutamate receptors in different brain regions, this approach revealed the clear existence of functional metabotropic glutamate (mGlu)2-mGlu4 heterodimers in addition to mGlu2 and mGlu4 homodimers. Strikingly, the mGlu4 subunits appear to be mainly heterodimers in the brain. Overall, these versatile biosensors can determine the presence and activity of endogenous membrane proteins in native tissues with high fidelity and convenience.


Subject(s)
Glutamic Acid , Receptors, Metabotropic Glutamate , Brain/metabolism , Fluorescence Resonance Energy Transfer/methods , Receptors, Metabotropic Glutamate/metabolism
7.
Sci Signal ; 14(705): eaaz4112, 2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34665640

ABSTRACT

G protein­coupled receptors (GPCRs) activate various mitogen-activated protein kinase (MAPK) pathways to regulate critical cell functions. ß-Arrestins mediate this mechanism for most GPCRs but not the GABAB receptor (GABABR). When coupled to the G protein Gi/o, GABABR phosphorylates the kinases ERK1 and ERK2. Here, we uncovered a distinct ß-arrestin­independent mechanism of MAPK pathway activation by GABABR. We found that GABABR also phosphorylated the kinase JNK downstream of activation of the small guanosine triphosphatases (GTPases) RhoA and Rac1 in primary mouse neurons. However, instead of Gi/o proteins, activation of this RhoA/Rac1-JNK pathway was mediated by G13. This pathway promoted the phosphorylation and accumulation of the postsynaptic scaffolding protein PSD95 and GABABR-mediated neuroprotection in granule neurons. In addition, this pathway synergized with a previously reported GABABR-mediated neuroprotection mediated by a Gi/o-dependent mechanism. GABABR agonists activated G13 with slower kinetics and lower potency than with which they activated Gi/o. Our findings reveal distinct, ß-arrestin­independent, context-specific synergistic mechanisms of MAPK activation by G protein­mediated GPCR signaling.


Subject(s)
Neuroprotection , Receptors, GABA-B , gamma-Aminobutyric Acid
8.
Nature ; 594(7864): 589-593, 2021 06.
Article in English | MEDLINE | ID: mdl-34135509

ABSTRACT

The metabotropic glutamate receptors (mGlus) are involved in the modulation of synaptic transmission and neuronal excitability in the central nervous system1. These receptors probably exist as both homo- and heterodimers that have unique pharmacological and functional properties2-4. Here we report four cryo-electron microscopy structures of the human mGlu subtypes mGlu2 and mGlu7, including inactive mGlu2 and mGlu7 homodimers; mGlu2 homodimer bound to an agonist and a positive allosteric modulator; and inactive mGlu2-mGlu7 heterodimer. We observed a subtype-dependent dimerization mode for these mGlus, as a unique dimer interface that is mediated by helix IV (and that is important for limiting receptor activity) exists only in the inactive mGlu2 structure. The structures provide molecular details of the inter- and intra-subunit conformational changes that are required for receptor activation, which distinguish class C G-protein-coupled receptors from those in classes A and B. Furthermore, our structure and functional studies of the mGlu2-mGlu7 heterodimer suggest that the mGlu7 subunit has a dominant role in controlling dimeric association and G-protein activation in the heterodimer. These insights into mGlu homo- and heterodimers highlight the complex landscape of mGlu dimerization and activation.


Subject(s)
Receptors, Metabotropic Glutamate/chemistry , Cryoelectron Microscopy , Humans , Protein Multimerization , Protein Structure, Tertiary
9.
Front Cell Dev Biol ; 9: 611443, 2021.
Article in English | MEDLINE | ID: mdl-34041234

ABSTRACT

G protein-coupled receptors (GPCRs) represent one of the largest membrane protein families that participate in various physiological and pathological activities. Accumulating structural evidences have revealed how GPCR activation induces conformational changes to accommodate the downstream G protein or ß-arrestin. Multiple GPCR functional assays have been developed based on Förster resonance energy transfer (FRET) and bioluminescence resonance energy transfer (BRET) sensors to monitor the conformational changes in GPCRs, GPCR/G proteins, or GPCR/ß-arrestin, especially over the past two decades. Here, we will summarize how these sensors have been optimized to increase the sensitivity and compatibility for application in different GPCR classes using various labeling strategies, meanwhile provide multiple solutions in functional assays for high-throughput drug screening.

10.
Nature ; 594(7864): 594-598, 2021 06.
Article in English | MEDLINE | ID: mdl-33911284

ABSTRACT

G-protein-coupled receptors (GPCRs) have central roles in intercellular communication1,2. Structural studies have revealed how GPCRs can activate G proteins. However, whether this mechanism is conserved among all classes of GPCR remains unknown. Here we report the structure of the class-C heterodimeric GABAB receptor, which is activated by the inhibitory transmitter GABA, in its active form complexed with Gi1 protein. We found that a single G protein interacts with the GB2 subunit of the GABAB receptor at a site that mainly involves intracellular loop 2 on the side of the transmembrane domain. This is in contrast to the G protein binding in a central cavity, as has been observed with other classes of GPCR. This binding mode results from the active form of the transmembrane domain of this GABAB receptor being different from that of other GPCRs, as it shows no outside movement of transmembrane helix 6. Our work also provides details of the inter- and intra-subunit changes that link agonist binding to G-protein activation in this heterodimeric complex.


Subject(s)
GTP-Binding Proteins/chemistry , Receptors, GABA-B/chemistry , Cryoelectron Microscopy , Humans , Protein Binding , Protein Domains , Protein Multimerization , Protein Structure, Tertiary
11.
Cell Res ; 31(7): 773-790, 2021 07.
Article in English | MEDLINE | ID: mdl-33510386

ABSTRACT

Compelling evidence has revealed that biased activation of G protein-coupled receptor (GPCR) signaling, including angiotensin II (AngII) receptor type 1 (AT1) signaling, plays pivotal roles in vascular homeostasis and injury, but whether a clinically relevant endogenous biased antagonism of AT1 signaling exists under physiological and pathophysiological conditions has not been clearly elucidated. Here, we show that an extracellular matrix protein, cartilage oligomeric matrix protein (COMP), acts as an endogenous allosteric biased modulator of the AT1 receptor and its deficiency is clinically associated with abdominal aortic aneurysm (AAA) development. COMP directly interacts with the extracellular N-terminus of the AT1 via its EGF domain and inhibits AT1-ß-arrestin-2 signaling, but not Gq or Gi signaling, in a selective manner through allosteric regulation of AT1 intracellular conformational states. COMP deficiency results in activation of AT1a-ß-arrestin-2 signaling and subsequent exclusive AAA formation in response to AngII infusion. AAAs in COMP-/- or ApoE-/- mice are rescued by AT1a or ß-arrestin-2 deficiency, or the application of a peptidomimetic mimicking the AT1-binding motif of COMP. Explorations of the endogenous biased antagonism of AT1 receptor or other GPCRs may reveal novel therapeutic strategies for cardiovascular diseases.


Subject(s)
Receptor, Angiotensin, Type 1 , Vascular System Injuries , Animals , Cartilage Oligomeric Matrix Protein , HEK293 Cells , Humans , Mice , Receptor, Angiotensin, Type 1/metabolism , beta-Arrestin 2 , beta-Arrestins/metabolism
12.
Biochem Biophys Res Commun ; 544: 1-7, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33516876

ABSTRACT

BACKGROUND: Our previous study developed ATRQß-001 vaccine, which targets peptide ATR001 from angiotensin Ⅱ (Ang Ⅱ) receptor type 1 (AT1R). The ATRQß-001 vaccine could induce the production of anti-ATR001 monoclonal antibody (McAb-ATR) and inhibit atherosclerosis without feedback activation of the renin-angiotensin system (RAS). This study aims at investigating the underexploited mechanisms of McAb-ATR in ameliorating atherosclerosis. METHODS: AT1R-KO HEK293T cell lines were constructed to identify the specificity of McAb-ATR and key sites of ATRQß-001 vaccine. Beta-arrestin1 knock-out (Arrb1-/-) mice, Beta-arrestin2 knock-out (Arrb2-/-) mice, and low-density lipoprotein receptor knock-out (LDLr-/-) mice were used to detect potential signaling pathways affected by McAb-ATR. The role of McAb-ATR in beta-arrestin and G proteins (Gq or Gi2/i3) signal transduction events was also investigated. RESULTS: McAb-ATR could specifically bind to the Phe182-His183-Tyr184 site of AT1R second extracellular loop (ECL2). The anti-atherosclerotic effect of McAb-ATR disappeared in LDLr-/- mice transplanted with Arrb2-/- mouse bone marrow (BM) and BM-derived macrophages (BMDMs) from Arrb2-/- mice. Furthermore, McAb-ATR inhibited beta-arrestin2-dependent extracellular signal regulated kinase1/2 (ERK1/2) phosphorylation, and promoted beta-arrestin2-mediated nuclear factor kappa B p65 (NFκB p65) inactivity. Compared with conventional AT1R blockers (ARBs), McAb-ATR did not inhibit Ang Ⅱ-induced uncoupling of heterotrimeric G proteins (Gq or Gi2/i3) and Gq-dependent intracellular Ca2+ release, nor cause RAS feedback activation. CONCLUSIONS: Through regulating beta-arrestin2, McAb-ATR ameliorates atherosclerosis without affecting Gq or Gi2/i3 pathways. Due to high selectivity for AT1R and biased interaction with beta-arrestin2, McAb-ATR could serve as a novel strategy for treating atherosclerosis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Atherosclerosis/prevention & control , Receptor, Angiotensin, Type 1/immunology , Receptors, LDL/metabolism , Vaccines, Virus-Like Particle/pharmacology , beta-Arrestin 2/metabolism , Animals , Antibodies, Monoclonal/immunology , Atherosclerosis/immunology , Atherosclerosis/metabolism , Cells, Cultured , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Angiotensin, Type 1/chemistry , Vaccines, Virus-Like Particle/immunology
13.
Circ Res ; 128(2): 262-277, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33208036

ABSTRACT

RATIONALE: The ß2-adrenoceptor (ß2-AR), a prototypical GPCR (G protein-coupled receptor), couples to both Gs and Gi proteins. Stimulation of the ß2-AR is beneficial to humans and animals with heart failure presumably because it activates the downstream Gi-PI3K-Akt cell survival pathway. Cardiac ß2-AR signaling can be regulated by crosstalk or heterodimerization with other GPCRs, but the physiological and pathophysiological significance of this type of regulation has not been sufficiently demonstrated. OBJECTIVE: Here, we aim to investigate the potential cardioprotective effect of ß2-adrenergic stimulation with a subtype-selective agonist, (R,R')-4-methoxy-1-naphthylfenoterol (MNF), and to decipher the underlying mechanism with a particular emphasis on the role of heterodimerization of ß2-ARs with another GPCR, 5-hydroxytryptamine receptors 2B (5-HT2BRs). METHODS AND RESULTS: Using pharmacological, genetic and biophysical protein-protein interaction approaches, we studied the cardioprotective effect of the ß2-agonist, MNF, and explored the underlying mechanism in both in vivo in mice and cultured rodent cardiomyocytes insulted with doxorubicin, hydrogen peroxide (H2O2) or ischemia/reperfusion. In doxorubicin (Dox)-treated mice, MNF reduced mortality and body weight loss, while improving cardiac function and cardiomyocyte viability. MNF also alleviated myocardial ischemia/reperfusion injury. In cultured rodent cardiomyocytes, MNF inhibited DNA damage and cell death caused by Dox, H2O2 or hypoxia/reoxygenation. Mechanistically, we found that MNF or another ß2-agonist zinterol markedly promoted heterodimerization of ß2-ARs with 5-HT2BRs. Upregulation of the heterodimerized 5-HT2BRs and ß2-ARs enhanced ß2-AR-stimulated Gi-Akt signaling and cardioprotection while knockdown or pharmacological inhibition of the 5-HT2BR attenuated ß2-AR-stimulated Gi signaling and cardioprotection. CONCLUSIONS: These data demonstrate that the ß2-AR-stimulated cardioprotective Gi signaling depends on the heterodimerization of ß2-ARs and 5-HT2BRs.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Cardiomyopathies/prevention & control , Fenoterol/analogs & derivatives , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Receptor, Serotonin, 5-HT2B/metabolism , Receptors, Adrenergic, beta-2/metabolism , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiotoxicity , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Doxorubicin , Ethanolamines/pharmacology , Fenoterol/pharmacology , Fibrosis , Hydrogen Peroxide , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Multimerization , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2B/genetics , Receptors, Adrenergic, beta-2/genetics , Signal Transduction
14.
Cell Res ; 30(7): 564-573, 2020 07.
Article in English | MEDLINE | ID: mdl-32494023

ABSTRACT

Metabotropic GABAB G protein-coupled receptor functions as a mandatory heterodimer of GB1 and GB2 subunits and mediates inhibitory neurotransmission in the central nervous system. Each subunit is composed of the extracellular Venus flytrap (VFT) domain and transmembrane (TM) domain. Here we present cryo-EM structures of full-length human heterodimeric GABAB receptor in the antagonist-bound inactive state and in the active state complexed with an agonist and a positive allosteric modulator in the presence of Gi1 protein at a resolution range of 2.8-3.0 Å. Our structures reveal that agonist binding stabilizes the closure of GB1 VFT, which in turn triggers a rearrangement of TM interfaces between the two subunits from TM3-TM5/TM3-TM5 in the inactive state to TM6/TM6 in the active state and finally induces the opening of intracellular loop 3 and synergistic shifting of TM3, 4 and 5 helices in GB2 TM domain to accommodate the α5-helix of Gi1. We also observed that the positive allosteric modulator anchors at the dimeric interface of TM domains. These results provide a structural framework for understanding class C GPCR activation and a rational template for allosteric modulator design targeting the dimeric interface of GABAB receptor.


Subject(s)
Cryoelectron Microscopy , Receptors, GABA-B/ultrastructure , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Models, Molecular , Protein Domains , Protein Multimerization , Receptors, GABA-B/chemistry , Structural Homology, Protein
15.
J Mol Biol ; 432(16): 4596-4611, 2020 07 24.
Article in English | MEDLINE | ID: mdl-32553728

ABSTRACT

Class-A G protein-coupled receptors (GPCRs) are known to homo-dimerize in the membrane. Yet, methods to characterize the structure of GPCR dimer in the native environment are lacking. Accordingly, the molecular basis and functional relevance of the class-A GPCR dimerization remain unclear. Here, we present the dimeric structural model of GPR17 in the cell membrane. The dimer mainly involves transmembrane helix 5 (TM5) at the interface, with F229 in TM5, a critical residue. An F229A mutation makes GPR17 monomeric regardless of the expression level of the receptor. Monomeric mutants of GPR17 display impaired ERK1/2 activation and cannot be properly internalized upon agonist treatment. Conversely, the F229C mutant is cross-linked as a dimer and behaves like wild-type. Importantly, the GPR17 dimer structure has been modeled using sparse inter-protomer FRET distance restraints obtained from fluorescence lifetime imaging microscopy. The same approach can be applied to characterizing the interactions of other important membrane proteins in the cell.


Subject(s)
Cell Membrane/metabolism , Mutation , Nerve Tissue Proteins/chemistry , Receptors, G-Protein-Coupled/chemistry , Animals , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Mice , Microscopy, Fluorescence , Models, Molecular , Nerve Tissue Proteins/genetics , Protein Multimerization , Protein Structure, Secondary , Receptors, G-Protein-Coupled/genetics
16.
FASEB J ; 34(2): 3224-3238, 2020 02.
Article in English | MEDLINE | ID: mdl-31917470

ABSTRACT

Myocardial ischemia-reperfusion injury (MIRI) is common clinical complication, which represents significant challenge in the treatment of acute myocardial infarction (AMI) diseases. Interleukin 35 (IL-35) exhibits anti-inflammatory properties via the engagement of the gp130, IL-12Rß2 and IL-27Rα receptors. However, whether IL-35 plays a beneficial role in the treatment of MIRI and potential underling mechanism are unclear. We showed that IL-35 conferred protection from MIRI as demonstrated by reduced infarct size and cardiac troponin T, improved cardiac function and decreased cardiomyocyte apoptosis in a mouse model. Despite activation of both STAT3 and STAT5 phosphorylation in the heart by IL-35, signal transducers and activators of transcription 3 (STAT3) was essential for mediating the IL-35-mediated protective effect on MIRI using cardiomyocyte-specific STAT3 deficient mice. Furthermore, gp130 was required for the STAT3 activation and cardio-protection induced by IL-35. Interestingly, IL-35 induced gp130 homodimer and gp130/IL-12Rß2 heterodimers in cardiomyocyte. Our results indicate that IL-35 can execute a protective role against MIRI through a novel signaling pathway, IL-35-gp130-STAT3 pathway, in cardiomyocytes, which may be beneficial for the development of novel and effective therapeutic approaches to treat the MIRI.


Subject(s)
Cytokine Receptor gp130/metabolism , Interleukins/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis , Cell Line , Cells, Cultured , Interleukins/pharmacology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Troponin T/metabolism
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-829695

ABSTRACT

Objective@# To investigate the manufacturing procedures of personalized miniscrew-assisted rapid palatal expanders (pMARPE) using digital technologies and to evaluate the effect of the expanders when expanding the midpalatal suture of an adult. @*Methods@# Digital technologies were used to make pMARPE, which was used to treat a 21-year-old woman with maxillary transverse deficiency (MTD). The relevant literature on MARPE was reviewed.@* Results@#PMARPE could be manufactured using intraoral digital scanning, computer-aided design and computer-aided manufacturing(CAD/CAM ), and 3D printing technologies. After expansion, the width of the anterior midpalatal suture, posterior midpalatal suture and maxillary skeletal width increased by 3.9 mm, 3.2 mm and 4.7 mm, respectively. There was no significant change in the inclination of maxillary first molars, and the height of alveolar ridge decreased slightly. It could be seen that using digital technologies to manufacture personalized expanders was possible for MARPE , and the initial stability of miniscrews played an important role in the expansion success rate, the increase of molar inclination is composed of many parts, and the decrease of alveolar ridge height may be overestimated due to the measurement method, as shown by a literature review. @*Conclusion@#The midpalatal suture of an adult patient with MTD could be expanded by pMARPE. However, the effect of this expander on the inclination of the first molar and alveolar bone height needs to be further studied with a larger sample size.

18.
Acta Pharmacol Sin ; 40(4): 460-467, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29946167

ABSTRACT

Metabotropic glutamate receptor 2 (mGlu2) belongs to the group-II metabotropic glutamate (mGlu) receptors and is a neurotransmitter G protein-coupled receptor. The group-II mGlu receptors are promising antipsychotic targets, but the specific role of mGlu2 signaling remains unclear. Receptor tyrosine kinases (RTKs) are also believed to participate in brain pathogenesis. To investigate whether there is any communication between mGlu2 and RTKs, we generated a CHO-mGlu2 cell line that stably expresses mGlu2 and showed that activation of mGlu2 by LY379268, a group II mGlu agonist, was able to transactivate insulin-like growth factor 1 receptor (IGF-1R). We further determined that the Gi/o protein, Gßγ subunits, phospholipase C, and focal adhesion kinase (FAK) were involved in the IGF-1R transactivation signaling axis, which further induced the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2) and cAMP response element-binding protein. In primary mouse cortical neurons, similar signaling pathways were observed when mGlu2 were stimulated by LY487379, an mGlu2 positive allosteric modulator. Transactivation of IGF-1R through FAK in response to mGlu2 should provide a better understanding of the association of mGlu2 with brain disease.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Receptor, IGF Type 1/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acids/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CHO Cells , Cells, Cultured , Cricetulus , Humans , Mice , Phosphorylation , Receptors, Metabotropic Glutamate/agonists
19.
Cell Biol Int ; 42(11): 1511-1522, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30080297

ABSTRACT

Proliferation of the adult hepatocyte population represents a central feature of tissue regeneration after liver injury and resection. This process could be driven by a diverse range of mitogens, such as hepatocyte growth factor (HGF) and fibroblast growth factor (FGF). Among FGF family, FGF2 is closely related to wound repair and cell proliferation. FGF2 does function in the process of angiogenesis in regenerating liver, while fewer reports are concerned with the impact and underlying mechanism of FGF2 on liver cell proliferation. To this end, an immortalized human normal hepatocyte L02 and mouse primary hepatocytes were exposed to FGF2 in this study. We demonstrate that FGF2 significantly enhances liver cell proliferation. Treatment with FGF2 obviously increases the phosphorylation level of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and c-Jun N-terminal kinase (JNK). Activity inhibition or expression down-regulation prove that both ERK1/2 and JNK signaling are required for FGF2-mediated effect on liver cell proliferation. Interestingly, interfering of ERK1/2 signaling results in marked decrease of JNK activation under FGF2 treatment, and JNK signaling is also involved in regulation of FGF2-induced ERK1/2 activation, suggesting that cross-talk between ERK1/2 and JNK signaling is important for FGF2 mitogenic activity. Both ERK1/2 and JNK signal via CREB to function in proliferation impact of FGF2 on liver cells. Taken together, this study reveals that ERK and JNK pathways synergistically regulate FGF2-induced liver cell proliferation via phosphorylating CREB, which will contribute to the understanding of FGF2 impact on liver cell proliferation and liver regeneration.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/pharmacology , Liver/cytology , MAP Kinase Signaling System , Mitogens/pharmacology , Animals , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , MAP Kinase Signaling System/drug effects , Mice , Models, Biological
20.
Nat Struct Mol Biol ; 25(6): 488-495, 2018 06.
Article in English | MEDLINE | ID: mdl-29808000

ABSTRACT

Platelet-activating-factor receptor (PAFR) responds to platelet-activating factor (PAF), a phospholipid mediator of cell-to-cell communication that exhibits diverse physiological effects. PAFR is considered an important drug target for treating asthma, inflammation and cardiovascular diseases. Here we report crystal structures of human PAFR in complex with the antagonist SR 27417 and the inverse agonist ABT-491 at 2.8-Å and 2.9-Å resolution, respectively. The structures, supported by molecular docking of PAF, provide insights into the signal-recognition mechanisms of PAFR. The PAFR-SR 27417 structure reveals an unusual conformation showing that the intracellular tips of helices II and IV shift outward by 13 Å and 4 Å, respectively, and helix VIII adopts an inward conformation. The PAFR structures, combined with single-molecule FRET and cell-based functional assays, suggest that the conformational change in the helical bundle is ligand dependent and plays a critical role in PAFR activation, thus greatly extending knowledge about signaling by G-protein-coupled receptors.


Subject(s)
Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Binding Sites , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Humans , Hydrogen Bonding , Imidazoles/pharmacology , Indoles/pharmacology , Ligands , Molecular Docking Simulation , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/antagonists & inhibitors , Protein Conformation , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...