Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Ann Noninvasive Electrocardiol ; 27(6): e13002, 2022 11.
Article in English | MEDLINE | ID: mdl-36087037

ABSTRACT

OBJECTIVE: To explore short-term changes after left bundle branch pacing (LBBP) using echocardiography and computed tomography (CT), especially for postoperative ventricular septal perforation. METHODS: Between January and September 2019, 33 patients with atrioventricular block underwent LBBP at Beijing Anzhen Hospital. All the patients were evaluated using electrocardiography, pacing, parameters and echocardiographic measurements, including for major complications, during the 1, 3, 6, 12 and 24-month follow-up. Interval perforations were examined during a 1-month follow-up echocardiogram and CT. RESULTS: Left bundle branch pacing was successfully performed in 100% (33/33) of patients. The mean seizure threshold was stable and unchanged postoperatively at the 1, 3, 6, 12 and 24-month follow-up. The paced QRS duration of the LBBP was 119.72 ± 2.53 ms and <130 ms in all patients. Unipolar impedance during the procedure was higher than 500 Ω (662.00 ± 181.50 Ω). No ventricular septal perforation occurred at the end of the procedure. At the 1-month follow-up, two patients reported transthoracic echocardiography, with CT revealing septal lead perforation. Through CT, two other patients were found to have septal lead perforation, and echocardiography indicated that the pacing lead had penetrated the interventricular septum and entered the left subendocardium. At the 1, 3, 6, 12 and 24-month follow-up, these four patients exhibited no significant increase in pacing threshold or impedance (p > .05). No ventricular thrombus or stroke was detected. CONCLUSION: Permanent LBBP is safe and feasible in patients with bradycardia. Echocardiography and/or CT can more accurately evaluate changes in cardiac structure and function after LBBP.


Subject(s)
Bundle of His , Cardiac Pacing, Artificial , Humans , Cardiac Pacing, Artificial/adverse effects , Cardiac Pacing, Artificial/methods , Electrocardiography/methods , Echocardiography/methods , Tomography, X-Ray Computed , Tomography , Treatment Outcome
2.
Nature ; 604(7907): 763-770, 2022 04.
Article in English | MEDLINE | ID: mdl-35418678

ABSTRACT

Adhesion G-protein-coupled receptors (aGPCRs) are important for organogenesis, neurodevelopment, reproduction and other processes1-6. Many aGPCRs are activated by a conserved internal (tethered) agonist sequence known as the Stachel sequence7-12. Here, we report the cryogenic electron microscopy (cryo-EM) structures of two aGPCRs in complex with Gs: GPR133 and GPR114. The structures indicate that the Stachel sequences of both receptors assume an α-helical-bulge-ß-sheet structure and insert into a binding site formed by the transmembrane domain (TMD). A hydrophobic interaction motif (HIM) within the Stachel sequence mediates most of the intramolecular interactions with the TMD. Combined with the cryo-EM structures, biochemical characterization of the HIM motif provides insight into the cross-reactivity and selectivity of the Stachel sequences. Two interconnected mechanisms, the sensing of Stachel sequences by the conserved 'toggle switch' W6.53 and the constitution of a hydrogen-bond network formed by Q7.49/Y7.49 and the P6.47/V6.47φφG6.50 motif (φ indicates a hydrophobic residue), are important in Stachel sequence-mediated receptor activation and Gs coupling. Notably, this network stabilizes kink formation in TM helices 6 and 7 (TM6 and TM7, respectively). A common Gs-binding interface is observed between the two aGPCRs, and GPR114 has an extended TM7 that forms unique interactions with Gs. Our structures reveal the detailed mechanisms of aGPCR activation by Stachel sequences and their Gs coupling.


Subject(s)
Peptides , Receptors, G-Protein-Coupled , Binding Sites , Cryoelectron Microscopy , Protein Domains , Protein Structure, Secondary , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
3.
Proc Natl Acad Sci U S A ; 119(15): e2117004119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35394864

ABSTRACT

GPR126 is a member of the adhesion G protein-coupled receptors (aGPCRs) that is essential for the normal development of diverse tissues, and its mutations are implicated in various pathological processes. Here, through screening 34 steroid hormones and their derivatives for cAMP production, we found that progesterone (P4) and 17-hydroxyprogesterone (17OHP) could specifically activate GPR126 and trigger its downstream Gi signaling by binding to the ligand pocket in the seven-transmembrane domain of the C-terminal fragment of GPR126. A detailed mutagenesis screening according to a computational simulated structure model indicated that K1001ECL2 and F1012ECL2 are key residues that specifically recognize 17OHP but not progesterone. Finally, functional analysis revealed that progesterone-triggered GPR126 activation promoted cell growth in vitro and tumorigenesis in vivo, which involved Gi-SRC pathways in a triple-negative breast cancer model. Collectively, our work identified a membrane receptor for progesterone/17OHP and delineated the mechanisms by which GPR126 participated in potential tumor progression in triple-negative breast cancer, which will enrich our understanding of the functions and working mechanisms of both the aGPCR member GPR126 and the steroid hormone progesterone.


Subject(s)
Progesterone , Receptors, G-Protein-Coupled , Receptors, Progesterone , Triple Negative Breast Neoplasms , 17-alpha-Hydroxyprogesterone/metabolism , Cell Line, Tumor , Humans , Progesterone/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
J Biol Chem ; 296: 100174, 2021.
Article in English | MEDLINE | ID: mdl-33303626

ABSTRACT

The adhesion GPCR ADGRG2, also known as GPR64, is a critical regulator of male fertility that maintains ion/pH homeostasis and CFTR coupling. The molecular basis of ADGRG2 function is poorly understood, in part because no endogenous ligands for ADGRG2 have been reported, thus limiting the tools available to interrogate ADGRG2 activity. It has been shown that ADGRG2 can be activated by a peptide, termed p15, derived from its own N-terminal region known as the Stachel sequence. However, the low affinity of p15 limits its utility for ADGRG2 characterization. In the current study, we used alanine scanning mutagenesis to examine the critical residues responsible for p15-induced ADGRG2 activity. We next designed systematic strategies to optimize the peptide agonist of ADGRG2, using natural and unnatural amino acid substitutions. We obtained an optimized ADGRG2 Stachel peptide T1V/F3Phe(4-Me) (VPM-p15) that activated ADGRG2 with significantly improved (>2 orders of magnitude) affinity. We then characterized the residues in ADGRG2 that were important for ADGRG2 activation in response to VPM-p15 engagement, finding that the toggle switch W6.53 and residues of the ECL2 region of ADGRG2 are key determinants for VPM-p15 interactions and VPM-p15-induced Gs or arrestin signaling. Our study not only provides a useful tool to investigate the function of ADGRG2 but also offers new insights to guide further optimization of Stachel peptides to activate adhesion GPCR members.


Subject(s)
Peptides/metabolism , Protein Engineering/methods , Receptors, G-Protein-Coupled/chemistry , Amino Acid Substitution , Animals , Binding Sites , Gene Expression , HEK293 Cells , Humans , Kinetics , Ligands , Mice , Models, Molecular , Mutagenesis, Site-Directed , Peptides/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Transgenes
5.
Neurol Res ; 42(8): 670-675, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32573395

ABSTRACT

OBJECTIVE: Underlying Coronary Artery Disease (CAD) complicated by Mental Stress-Induced Myocardial Ischemia (MSIMI) has been linked with an increased risk for adverse cardiovascular events and even sudden death. However, the underlying mechanisms of MSIMI remain unknown. In this study, we investigated cytokine levels at baseline inflammation status and during acute inflammatory responses to mental stress in patients with known CAD who presented with MSIMI. METHOD: 77 patients with known CAD were recruited and all underwent echocardiography before and during arithmetic stress task. MSIMI was diagnosed by new or worsening wall motion abnormalities greater than or equal to a 5% reduction of left ventricle ejection fraction. Inflammatory markers were measured both before and immediately after the Mental Stress (MS) by ELISA kits. Repeated measures models were used to report the responses and mixed linear regression models were used to report the differences between MSIMI negative and positive patients. RESULT: MS induced a significant increase in Stromal Cell-Derived Factor-1α (SDF-1α) and Monocyte Chemoattractant Protein-1 (MCP-1) in all subjects; 20.78% of the patients with known CAD developed MSIMI during the arithmetic task. MSIMI positive patients had significantly lower baseline levels of Interleukin-1ß (IL-1ß) and Tumor Necrosis Factor-α (TNF-α), but a higher response in levels of SDF-1α than MSIMI negative patients. CONCLUSION: MS can induce acute inflammatory responses. MSIMI is associated with lower levels of IL-1ß and TNF-α at baseline and higher levels of SDF-1α in response to MS.


Subject(s)
Cytokines/blood , Inflammation/blood , Myocardial Ischemia/blood , Stress, Psychological/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Female , Humans , Inflammation/complications , Inflammation Mediators/blood , Male , Middle Aged , Myocardial Ischemia/complications , Stress, Psychological/complications
6.
BMC Cardiovasc Disord ; 20(1): 141, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188408

ABSTRACT

BACKGROUND: Depression has been recognized as an independent risk factor of coronary heart disease (CHD). Moreover, there is interrelationship of both depression and CHD. However, the potential pathophysiological mechanisms remain unknown. It might be influenced by genetic and environmental factors. According to recent researches, there is potential association between serotonin transporter gene-linked polymorphic region (5-HTTLPR) polymorphism and risk of depression in CHD patients, but the results are still inconclusive. Therefore, we performed this meta-analysis based on unadjusted and adjusted data to ascertain a more precise conclusion. METHODS: We searched relevant articles through PubMed, Embase, Web of Science, Chinese BioMedical Literature (CBM) and Chinese National Knowledge Infrastructure (CNKI) databases up to August 26, 2019. Study selection and data extraction were accomplished by two authors independently. The strength of the correlation was assessed via odds ratios (ORs) with their 95% confidence intervals (95%CIs). RESULTS: This meta-analysis enrolled six observational studies. Based on unadjusted data, there was significant relationship between 5-HTTLPR polymorphism and depression risk in CHD patients under all genetic models (S vs. L: OR = 1.31, 95%CI = 1.07-1.60; SS vs. LL: OR = 1.73, 95%CI = 1.12-2.67; LS vs. LL: OR = 1.47, 95%CI = 1.13-1.92; LS + SS vs. LL: OR = 1.62, 95%CI = 1.25-2.09; SS vs. LL + LS: OR = 1.33, 95%CI = 1.02-1.74). The results of adjusted data further strengthened this relationship (SS vs. LL: OR = 1.89, 95%CI = 1.28-2.80; LS vs. LL: OR = 1.69, 95%CI = 1.14-2.51; LS + SS vs. LL: OR = 1.80, 95%CI = 1.25-2.59). Subgroup analyses based on ethnicity and major depressive disorder revealed similar results to that of the overall analysis. No evidence of publication bias was observed. CONCLUSIONS: Our results suggest that 5-HTTLPR polymorphism may have an important effect on the risk of depression among patients with CHD, and carriers of the S allele of 5-HTTLPR are more vulnerable to depression.


Subject(s)
Affect , Coronary Disease/genetics , Depression/genetics , Polymorphism, Genetic , Serotonin Plasma Membrane Transport Proteins/genetics , Coronary Disease/epidemiology , Coronary Disease/psychology , Depression/epidemiology , Depression/psychology , Genetic Predisposition to Disease , Humans , Phenotype , Risk Factors
7.
Chin Med J (Engl) ; 133(2): 134-140, 2020 Jan 20.
Article in English | MEDLINE | ID: mdl-31880742

ABSTRACT

BACKGROUND: The symptomatic bradyarrhythmia is Class I indication for pacing therapy which is not a radical cure. The present study aimed to assess the feasibility and to present the initial results of the restricted ablation of the parasympathetic innervation surrounding sinus and atrioventricular (AV) nodes for treating patients with bradyarrhythmia. METHODS: A total of 13 patients with cardiogenic syncope were included from May 2008 to June 2015. Under the guidance of fluoroscopy and /or three-dimensional geometry by 64-slice spiral computed tomography, atrial activation sequence in sinus rhythm was mapped. Chamber geometry was reconstructed manually or automatically using the Niobe II magnetic navigation system integrated with the CARTO-remote magnetic technology (RMT) system. Cardioneuroablation was targeted at the high-amplitude fractionated electrograms surrounding the regions of His bundle and the site with the earliest activation in sinus rhythm. Areas surrounding the sinus node, AV node, and the phrenic nerve were avoided. RESULTS: Thirteen patients completed the studies. Ablation was successfully performed in 12 patients and failed in one. The high-frequency potential was recorded in atrial electrograms surrounding the sinus or AV nodes in all the patients and disappeared in 15 s after radiofrequency applications. The vagal reaction was observed before the improvement of the sinus and AV node function. No complications occurred during the procedures. Patients were followed up for a mean of 13.0 ±â€Š5.9 months. During the follow up ten patients remained free of symptoms, and two patients had a permanent cardiac pacemaker implanted due to spontaneous recurrence of syncope. The heart rate of post-ablation was higher than pre-ablation (69.0 ±â€Š11.0 vs. 49.0 ±â€Š10.0 beats/min, t = 4.56, P = 0.008). The sinus node recovery time, Wenckebach block point, and atrium-His bundle interval were significantly shorter after ablation (1386.0 ±â€Š165.0 vs. 921.0 ±â€Š64.0 ms, t = 7.45, P = 0.002; 590.0 ±â€Š96.0 vs. 464.0 ±â€Š39.0 ms, t = 2.38, P = 0.023; 106.0 ±â€Š5.0 vs. 90.0 ±â€Š12.0 ms, t = 9.80, P = 0.013 before and after ablation procedure, respectively). CONCLUSIONS: Ablation of sinoatrial and AV nodal peripheral fibrillar myocardium electrical activity might provide a new treatment to ameliorate paroxysmal sinus node dysfunction, high degree AV block, and vagal-mediated syncope.


Subject(s)
Atrioventricular Block/physiopathology , Atrioventricular Block/therapy , Atrioventricular Node/innervation , Bradycardia/physiopathology , Bradycardia/therapy , Catheter Ablation/methods , Sick Sinus Syndrome/therapy , Adult , Cardiac Pacing, Artificial , Electrocardiography , Female , Heart Atria/physiopathology , Humans , Male , Middle Aged , Retrospective Studies , Sick Sinus Syndrome/physiopathology , Tomography, Spiral Computed
8.
Chin Med J (Engl) ; 132(12): 1390-1399, 2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31205095

ABSTRACT

BACKGROUND: Mental stress-induced myocardial ischemia (MSIMI) is closely associated with adverse cardiac events in patients with coronary artery disease (CAD) and we aimed to determine whether biomarkers and blood pressure could be potential predictors of MSIMI. METHODS: This study enrolled 82 patients with documented CAD between June 1, 2017 and November 9, 2017. Patient blood samples were obtained at resting period and at the end of mental arithmetic. Then, patients were assigned to MSIMI positive group and MSIMI negative group. The main statistical methods included linear regression, receiver operating characteristic (ROC) curves, and logistic regression. RESULTS: Patients with CAD with MSIMI had significantly greater median resting N-terminal pro-brain natriuretic peptide (NT-proBNP, 141.02 [45.85-202.76] pg/mL vs. 57.95 [27.06-117.64] pg/mL; Z = -2.23, P = 0.03) and mean systolic blood pressure (SBP) (145.56 ±â€Š16.87 mmHg vs. 134.92 ±â€Š18.16 mmHg, Z = -2.13, P = 0.04) when compared with those without MSIMI. After 5-min mental stress task, those who developed MSIMI presented higher elevation of median post-stressor high sensitivity cardiac troponin I (hs-cTnI, 0.020 [0.009-0.100] ng/mL vs. 0.009 [0.009-0.010] ng/mL; Z = -2.45, P = 0.01), post-stressor NT-proBNP (138.96 [39.93-201.56] pg/mL vs. 61.55 [25.66-86.50] pg/mL; Z = -2.15, P = 0.03) compared with those without MSIMI. Using the ROC curves, and after the adjustment for basic characteristics, the multiple logistic regression analysis showed that patients presenting a post-stressor hs-cTnI ≥ 0.015 ng/mL had seven-fold increase in the risk of developing MSIMI (odds ratio [OR]: 7.09; 95% confidence interval [CI]: 1.65-30.48; P = 0.009), a rest NT-proBNP ≥ 80.51 pg/mL had nearly eight-fold increase (OR: 7.85; 95% CI: 1.51-40.82; P = 0.014), a post-stressor NT-proBNP ≥ 98.80 pg/mL had 35-fold increase (OR: 34.96; 95% CI: 3.72-328.50; P = 0.002), a rest SBP ≥ 129.50 mmHg had 11-fold increase (OR: 11.42; 95% CI: 1.21-108.17; P = 0.034). CONCLUSIONS: The present study shows that CAD patients with higher hs-cTnI level, and/or greater NT-proBNP and/or SBP are at higher risk of suffering from MSIMI when compared with those without MSIMI, indicating that hs-cTnI, NT-proBNP, SBP might be potential predictors of MSIMI.


Subject(s)
Coronary Artery Disease/complications , Myocardial Ischemia/etiology , Stress, Psychological/complications , Aged , Anxiety/blood , Anxiety/complications , Biomarkers/blood , Blood Pressure/physiology , C-Reactive Protein/metabolism , Coronary Artery Disease/blood , Depression/blood , Depression/complications , Electrocardiography , Female , Humans , Male , Middle Aged , Myocardial Ischemia/blood , Natriuretic Peptide, Brain/blood , Odds Ratio , Peptide Fragments/blood , Predictive Value of Tests , Prospective Studies , ROC Curve , Stress, Psychological/blood , Tomography, Emission-Computed, Single-Photon , Troponin I/blood , Troponin T/blood
9.
J Cell Mol Med ; 23(7): 4653-4665, 2019 07.
Article in English | MEDLINE | ID: mdl-31044530

ABSTRACT

Severe reduction in the ß-cell number (collectively known as the ß-cell mass) contributes to the development of both type 1 and type 2 diabetes. Recent pharmacological studies have suggested that increased pancreatic ß-cell proliferation could be due to specific inhibition of adenosine kinase (ADK). However, genetic evidence for the function of pancreatic ß-cell ADK under physiological conditions or in a pathological context is still lacking. In this study, we crossed mice carrying LoxP-flanked Adk gene with Ins2-Cre mice to acquire pancreatic ß -cell ADK deficiency (Ins2-Cre± Adkfl/fl ) mice. Our results revealed that Ins2-Cre+/- Adkfl/fl mice showed improved glucose metabolism and ß-cell mass in younger mice, but showed normal activity in adult mice. Moreover, Ins2-Cre± Adkfl/fl mice were more resistant to streptozotocin (STZ) induced hyperglycaemia and pancreatic ß-cell damage in adult mice. In conclusion, we found that ADK negatively regulates ß-cell replication in young mice as well as under pathological conditions, such as STZ induced pancreatic ß-cell damage. Our study provided genetic evidence that specific inhibition of pancreatic ß-cell ADK has potential for anti-diabetic therapy.


Subject(s)
Adenosine Kinase/genetics , Gene Deletion , Glucose/metabolism , Homeostasis , Hyperglycemia/chemically induced , Hyperglycemia/enzymology , Insulin-Secreting Cells/enzymology , Aging/pathology , Animals , Cell Count , Cell Proliferation , Mice, Knockout , Streptozocin , Time Factors
11.
Nat Chem Biol ; 14(9): 876-886, 2018 09.
Article in English | MEDLINE | ID: mdl-30120361

ABSTRACT

Signals from 800 G-protein-coupled receptors (GPCRs) to many SH3 domain-containing proteins (SH3-CPs) regulate important physiological functions. These GPCRs may share a common pathway by signaling to SH3-CPs via agonist-dependent arrestin recruitment rather than through direct interactions. In the present study, 19F-NMR and cellular studies revealed that downstream of GPCR activation engagement of the receptor-phospho-tail with arrestin allosterically regulates the specific conformational states and functional outcomes of remote ß-arrestin 1 proline regions (PRs). The observed NMR chemical shifts of arrestin PRs were consistent with the intrinsic efficacy and specificity of SH3 domain recruitment, which was controlled by defined propagation pathways. Moreover, in vitro reconstitution experiments and biophysical results showed that the receptor-arrestin complex promoted SRC kinase activity through an allosteric mechanism. Thus, allosteric regulation of the conformational states of ß-arrestin 1 PRs by GPCRs and the allosteric activation of downstream effectors by arrestin are two important mechanisms underlying GPCR-to-SH3-CP signaling.


Subject(s)
Allosteric Regulation , Arrestin/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , src Homology Domains , Cells, Cultured , HEK293 Cells , Humans
12.
J Biol Chem ; 293(42): 16226-16241, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30154244

ABSTRACT

Slingshots are phosphatases that modulate cytoskeleton dynamics, and their activities are tightly regulated in different physiological contexts. Recently, abnormally elevated Slingshot activity has been implicated in many human diseases, such as cancer, Alzheimer's disease, and vascular diseases. Therefore, Slingshot-specific inhibitors have therapeutic potential. However, an enzymological understanding of the catalytic mechanism of Slingshots and of their activation by actin is lacking. Here, we report that the N-terminal region of human Slingshot2 auto-inhibits its phosphatase activity in a noncompetitive manner. pH-dependent phosphatase assays and leaving-group dependence studies suggested that the N-terminal domain of Slingshot2 regulates the stability of the leaving group of the product during catalysis by modulating the general acid Asp361 in the catalytic VYD loop. F-actin binding relieved this auto-inhibition and restored the function of the general acid. Limited tryptic digestion and biophysical studies identified large conformational changes in Slingshot2 after the F-actin binding. The dissociation of N-terminal structural elements, including Leu63, and the exposure of the loop between α-helix-2 and ß-sheet-3 of the phosphatase domain served as the structural basis for Slingshot activation via F-actin binding in vitro and via neuregulin stimulation in cells. Moreover, we designed a FlAsH-BRET-based Slingshot2 biosensor whose readout was highly correlated with the in vivo phosphatase activities of Slingshot2. Our results reveal the auto-inhibitory mechanism and allosteric activation mechanisms of a human Slingshot phosphatase. They also contribute to the design of new strategies to study Slingshot regulation in various cellular contexts and to screen for new activators/inhibitors of Slingshot activity.


Subject(s)
Allosteric Regulation , Phosphoprotein Phosphatases/metabolism , Actins/metabolism , Biosensing Techniques/methods , Catalysis , Catalytic Domain , Humans , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/chemistry , Protein Binding , Protein Structure, Secondary
13.
Elife ; 72018 02 02.
Article in English | MEDLINE | ID: mdl-29393851

ABSTRACT

Luminal fluid reabsorption plays a fundamental role in male fertility. We demonstrated that the ubiquitous GPCR signaling proteins Gq and ß-arrestin-1 are essential for fluid reabsorption because they mediate coupling between an orphan receptor ADGRG2 (GPR64) and the ion channel CFTR. A reduction in protein level or deficiency of ADGRG2, Gq or ß-arrestin-1 in a mouse model led to an imbalance in pH homeostasis in the efferent ductules due to decreased constitutive CFTR currents. Efferent ductule dysfunction was rescued by the specific activation of another GPCR, AGTR2. Further mechanistic analysis revealed that ß-arrestin-1 acts as a scaffold for ADGRG2/CFTR complex formation in apical membranes, whereas specific residues of ADGRG2 confer coupling specificity for different G protein subtypes, this specificity is critical for male fertility. Therefore, manipulation of the signaling components of the ADGRG2-Gq/ß-arrestin-1/CFTR complex by small molecules may be an effective therapeutic strategy for male infertility.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Fertility , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 1/metabolism , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , beta-Arrestin 1/genetics
14.
J Clin Invest ; 127(7): 2631-2646, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28604389

ABSTRACT

Somatostatin secreted by pancreatic δ cells mediates important paracrine interactions in Langerhans islets, including maintenance of glucose metabolism through the control of reciprocal insulin and glucagon secretion. Disruption of this circuit contributes to the development of diabetes. However, the precise mechanisms that control somatostatin secretion from islets remain elusive. Here, we found that a super-complex comprising the cullin 4B-RING E3 ligase (CRL4B) and polycomb repressive complex 2 (PRC2) epigenetically regulates somatostatin secretion in islets. Constitutive ablation of CUL4B, the core component of the CRL4B-PRC2 complex, in δ cells impaired glucose tolerance and decreased insulin secretion through enhanced somatostatin release. Moreover, mechanistic studies showed that the CRL4B-PRC2 complex, under the control of the δ cell-specific transcription factor hematopoietically expressed homeobox (HHEX), determines the levels of intracellular calcium and cAMP through histone posttranslational modifications, thereby altering expression of the Cav1.2 calcium channel and adenylyl cyclase 6 (AC6) and modulating somatostatin secretion. In response to high glucose levels or urocortin 3 (UCN3) stimulation, increased expression of cullin 4B (CUL4B) and the PRC2 subunit histone-lysine N-methyltransferase EZH2 and reciprocal decreases in Cav1.2 and AC6 expression were found to regulate somatostatin secretion. Our results reveal an epigenetic regulatory mechanism of δ cell paracrine interactions in which CRL4B-PRC2 complexes, Cav1.2, and AC6 expression fine-tune somatostatin secretion and facilitate glucose homeostasis in pancreatic islets.


Subject(s)
Cullin Proteins/metabolism , Insulin/metabolism , Multienzyme Complexes/metabolism , Paracrine Communication , Somatostatin-Secreting Cells/metabolism , Somatostatin/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cullin Proteins/genetics , Cyclic AMP/metabolism , Epigenesis, Genetic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin Secretion , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Somatostatin/genetics , Somatostatin-Secreting Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Nat Commun ; 8: 14335, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181498

ABSTRACT

Acute hormone secretion triggered by G protein-coupled receptor (GPCR) activation underlies many fundamental physiological processes. GPCR signalling is negatively regulated by ß-arrestins, adaptor molecules that also activate different intracellular signalling pathways. Here we reveal that TRV120027, a ß-arrestin-1-biased agonist of the angiotensin II receptor type 1 (AT1R), stimulates acute catecholamine secretion through coupling with the transient receptor potential cation channel subfamily C 3 (TRPC3). We show that TRV120027 promotes the recruitment of TRPC3 or phosphoinositide-specific phospholipase C (PLCγ) to the AT1R-ß-arrestin-1 signalling complex. Replacing the C-terminal region of ß-arrestin-1 with its counterpart on ß-arrestin-2 or using a specific TAT-P1 peptide to block the interaction between ß-arrestin-1 and PLCγ abolishes TRV120027-induced TRPC3 activation. Taken together, our results show that the GPCR-arrestin complex initiates non-desensitized signalling at the plasma membrane by coupling with ion channels. This fast communication pathway might be a common mechanism of several cellular processes.


Subject(s)
Catecholamines/metabolism , Receptor, Angiotensin, Type 1/agonists , TRPC Cation Channels/metabolism , beta-Arrestin 1/metabolism , beta-Arrestin 2/metabolism , Animals , Calcium/metabolism , Estrenes/pharmacology , HEK293 Cells , Humans , Ligands , Mice, Knockout , Oligopeptides/pharmacology , Phospholipase C gamma/metabolism , Pyrrolidinones/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , beta-Arrestin 1/chemistry
16.
BMC Cardiovasc Disord ; 17(1): 16, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28061808

ABSTRACT

BACKGROUND: Whether diabetes mellitus (DM) is a predictor of long-term adverse clinical outcomes after repeat drug eluting stent (DES) implantation for DES in-stent restenosis (ISR) remains controversial. We sought to evaluate the effect of DM on the long-term clinical outcomes in patients undergoing repeat DES implantation for DES-ISR lesions. METHODS: In the present study, 254 patients with DES-ISR were divided into DM or non-DM groups according to the presence or absence of DM. All patients received repeat 2nd generation DES implantation for DES-ISR. The occurrences of major adverse cardiac events (MACEs) over a 2-year follow-up period were compared between the two groups. MACEs were defined as cardiac death, myocardial infarction (MI), and target lesion revascularization (TLR). MACE free survival was investigated with Kaplan-Meier curve analysis. Cox regression analysis was used to identify factors associated with MACEs. RESULTS: Baseline clinical characteristics were similar between groups, except for the prevalence of early restenosis (lower) in the DM group. Differences in angiographic and procedural characteristics were not significant between groups. The rates of 2-year MACE (30.9 vs. 26.0%; P = 0.453) and TLR (24.7 vs. 19.7%; P = 0.411) were similar between groups. MACE-free survival and TLR-free survival were also similar between groups (P = 0.441 and P = 0.807). Subgroup analysis suggested a significant difference in the MACE (39.0 vs.15.3%, P < 0.001) and TLR occurrence (30.5 vs.8.2%, P < 0.001) and TLR-free survival (lower in early subgroup, P < 0.001) between early and late occurrence of ISR in the non-DM group of patients but not in the DM group. After adjustment for all significant clinical variables, Cox regression analysis indicated that DM was not associated with MACEs (hazard ratio [HR] 1.531, 95% confidence interval [CI] 0.882-2.658, P =0.130). Non-focal type ISR and early ISR were predictors of MACEs (HR 2.671, 95% CI 1.468-4.858,P = 0.001; HR 4.703, 95% CI 2.725-8.117, P < 0.001, respectively). CONCLUSIONS: Patients with DM have similar 2-year clinical outcomes to patients without DM when repeat 2nd generation DES was used for treatment of DES-ISR. DM is not the predictor of long-term prognosis in patients undergoing repeat 2nd generation DES for DES-ISR.


Subject(s)
Diabetes Mellitus , Drug-Eluting Stents/adverse effects , Graft Occlusion, Vascular/epidemiology , Percutaneous Coronary Intervention/adverse effects , Sirolimus/pharmacology , China/epidemiology , Coronary Angiography , Female , Follow-Up Studies , Graft Occlusion, Vascular/diagnosis , Graft Occlusion, Vascular/etiology , Humans , Immunosuppressive Agents/pharmacology , Incidence , Male , Middle Aged , Prevalence , Reoperation , Retrospective Studies , Time Factors
17.
Biol Psychiatry ; 81(8): 654-670, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27916196

ABSTRACT

BACKGROUND: Stress is a conserved physiological response in mammals. Whereas moderate stress strengthens memory to improve reactions to previously experienced difficult situations, too much stress is harmful. METHODS: We used specific ß-adrenergic agonists, as well as ß2-adrenergic receptor (ß2AR) and arrestin knockout models, to study the effects of adaptive ß2AR activation on cognitive function using Morris water maze and object recognition experiments. We used molecular and cell biological approaches to elucidate the signaling subnetworks. RESULTS: We observed that the duration of the adaptive ß2AR activation determines its consequences on learning and memory. Short-term formoterol treatment, for 3 to 5 days, improved cognitive function; however, prolonged ß2AR activation, for more than 6 days, produced harmful effects. We identified the activation of several signaling networks downstream of ß2AR, as well as an essential role for arrestin and lactate metabolism in promoting cognitive ability. Whereas Gs-protein kinase A-cyclic adenosine monophosphate response element binding protein signaling modulated monocarboxylate transporter 1 expression, ß-arrestin-1 controlled expression levels of monocarboxylate transporter 4 and lactate dehydrogenase A through the formation of a ß-arrestin-1/phospho-mitogen-activated protein kinase/hypoxia-inducible factor-1α ternary complex to upregulate lactate metabolism in astrocyte-derived U251 cells. Conversely, long-term treatment with formoterol led to the desensitization of ß2ARs, which was responsible for its decreased beneficial effects. CONCLUSIONS: Our results not only revealed that ß-arrestin-1 regulated lactate metabolism to contribute to ß2AR functions in improved memory formation, but also indicated that the appropriate management of one specific stress pathway, such as through the clinical drug formoterol, may exert beneficial effects on cognitive abilities.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Lactic Acid/metabolism , Learning/physiology , Memory/physiology , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Stress, Psychological/metabolism , beta-Arrestin 1/metabolism , Adrenergic beta-2 Receptor Agonists/administration & dosage , Animals , Astrocytes/metabolism , Cell Line , Formoterol Fumarate/administration & dosage , Hippocampus/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Receptors, Adrenergic, beta-2/genetics , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Transcriptome
18.
Chin Med J (Engl) ; 129(24): 2913-2919, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27958222

ABSTRACT

BACKGROUND: Our previous studies have demonstrated that the levels of 5-hydroxytryptamine (5-HT) and 5-HT 2A receptor (5-HT2AR) in serum and platelet were associated with depression and myocardial infarction (MI), and pretreatment with ginseng fruit saponins (GFS) before MI and depression had an effect on the 5-HT system. In this study, the effects of GFS on the 5-HT system in the Sprague-Dawley (SD) rats with MI, depression, and MI + depression were evaluated. METHODS: A total of eighty SD rats were allocated to four groups: MI, depression, MI + depression, and control groups (n = 20 in each group). Each group included two subgroups (n = 10 in each subgroup): Saline treatment subgroup and GFS treatment subgroup. The levels of 5-HT, 5-HT2AR, and serotonin transporter (SERT) were quantified in serum, platelet lysate, and brain tissue through the enzyme-linked immunosorbent assay method, respectively. RESULTS: Compared with those in the saline treatment subgroups, the levels of 5-HT in serum and platelet lysate statistically significantly increased in the GFS treatment subgroups of MI, depression, and MI + depression groups (serum: all P = 0.000; platelet lysate: P = 0.002, 0.000, 0.000, respectively). However, the 5-HT levels in brain homogenate significantly decreased in the GFS treatment subgroups compared with those in the saline treatment subgroups in MI and depression groups (P = 0.025 and 0.044 respectively), and no significant difference was observed between saline and GFS treatment subgroups in MI + depression group (P = 0.663). Compared with that in GFS treatment subgroup of control group, the 5-HT2AR levels in the platelet lysate significantly decreased in GFS treatment subgroups of MI, depression, and MI + depression groups (all P = 0.000). Compared to those in the saline treatment subgroups, the serum SERT levels significantly decreased in the GFS treatment subgroups in MI, depression, and MI + depression groups (P = 0.009, 0.038, and P = 0.001, respectively), while the SERT levels of platelet lysate significantly decreased in GFS treatment subgroup of MI group (P = 0.000), significantly increased in GFS treatment subgroup of depression group (P = 0.019), and slightly changed in GFS treatment subgroup of MI + depression group (P = 0.219). No significant changes for SERT levels in brain homogenate could be found between the saline and GFS treatment subgroups in MI, depression, and MI + depression groups (P = 0.421, 0.076 and P = 0.642). CONCLUSIONS: This study indicated that GFS might inhibit the reuptake of 5-HT from serum to platelet according to decreased 5-HT2AR in platelet and SERT in serum and platelet. The change of 5-HT in serum after GFS treatment was inconsistent with that in the brain. It seemed that GFS could not pass through the blood-brain barrier to affect the central serotonergic system.


Subject(s)
Depression/drug therapy , Myocardial Infarction/drug therapy , Panax/chemistry , Saponins/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2B/metabolism , Saponins/chemistry , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism
19.
Cell Rep ; 15(6): 1345-58, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27134172

ABSTRACT

PTPN12 is an important tumor suppressor that plays critical roles in various physiological processes. However, the molecular basis underlying the substrate specificity of PTPN12 remains uncertain. Here, enzymological and crystallographic studies have enabled us to identify two distinct structural features that are crucial determinants of PTPN12 substrate specificity: the pY+1 site binding pocket and specific basic charged residues along its surface loops. Key structurally plastic regions and specific residues in PTPN12 enabled recognition of different HER2 phosphorylation sites and regulated specific PTPN12 functions. In addition, the structure of PTPN12 revealed a CDK2 phosphorylation site in a specific PTPN12 loop. Taken together, our results not only provide the working mechanisms of PTPN12 for desphosphorylation of its substrates but will also help in designing specific inhibitors of PTPN12.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 12/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/metabolism , Humans , Kinetics , Models, Molecular , Peptides/chemistry , Phosphorylation , Phosphoserine/metabolism , Protein Structure, Secondary , Substrate Specificity
20.
ChemMedChem ; 10(12): 1980-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26553423

ABSTRACT

Slingshot proteins form a small group of dual-specific phosphatases that modulate cytoskeleton dynamics through dephosphorylation of cofilin and Lim kinases (LIMK). Small chemical compounds with Slingshot-inhibiting activities have therapeutic potential against cancers or infectious diseases. However, only a few Slingshot inhibitors have been investigated and reported, and their cellular activities have not been examined. In this study, we identified two rhodanine-scaffold-based para-substituted benzoic acid derivatives as competitive Slingshot inhibitors. The top compound, (Z)-4-((4-((4-oxo-2-thioxo-3-(o-tolyl)thiazolidin-5-ylidene)methyl)phenoxy)methyl)benzoic acid (D3) had an inhibition constant (Ki) of around 4 µm and displayed selectivity over a panel of other phosphatases. Moreover, compound D3 inhibited cell migration and cofilin dephosphorylation after nerve growth factor (NGF) or angiotensin II stimulation. Therefore, our newly identified Slingshot inhibitors provide a starting point for developing Slingshot-targeted therapies.


Subject(s)
Benzoates/chemistry , Benzoic Acid/chemistry , Enzyme Inhibitors/chemistry , Phosphoprotein Phosphatases/antagonists & inhibitors , Rhodanine/analogs & derivatives , Animals , Benzoates/metabolism , Benzoates/pharmacology , Benzoic Acid/metabolism , Benzoic Acid/pharmacology , Cell Movement/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Kinetics , Lim Kinases/metabolism , Nerve Growth Factor/pharmacology , PC12 Cells , Phosphoprotein Phosphatases/metabolism , Phosphorylation/drug effects , Protein Binding , Rats , Rhodanine/chemistry , Rhodanine/metabolism , Rhodanine/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...