Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 165: 115158, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37473685

ABSTRACT

Diabetic nephropathy (DN) is a serious complication of diabetes mellitus (DM), which currently lacks effective treatments. AMP-activated protein kinase (AMPK) stimulation by chalcones, a class of polyphenols abundantly found in plants, is proposed as a promising therapeutic approach for DM. This study aimed to identify novel chalcone derivatives with improved AMPK-stimulating activity in human podocytes and evaluate their mechanisms of action as well as in vivo efficacy in a mouse model of DN. Among 133 chalcone derivatives tested, the sulfonamide chalcone derivative IP-004 was identified as the most potent AMPK activator in human podocytes. Western blot analyses, intracellular calcium measurements and molecular docking simulation indicated that IP-004 activated AMPK by mechanisms involving direct binding at allosteric site of calcium-dependent protein kinase kinase ß (CaMKKß) without affecting intracellular calcium levels. Interestingly, eight weeks of intraperitoneal administration of IP-004 (20 mg/kg/day) significantly decreased fasting blood glucose level, activated AMPK in the livers, muscles and glomeruli, and ameliorated albuminuria in db/db type II diabetic mice. Collectively, this study identifies a novel chalcone derivative capable of activating AMPK in vitro and in vivo and exhibiting efficacy against hyperglycemia and DN in mice. Further development of AMPK activators based on chalcone derivatives may provide an effective treatment of DN.


Subject(s)
Chalcone , Chalcones , Diabetes Mellitus, Experimental , Diabetic Nephropathies , Hyperglycemia , Mice , Humans , Animals , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , AMP-Activated Protein Kinases/metabolism , Chalcone/pharmacology , Chalcone/therapeutic use , Chalcones/pharmacology , Chalcones/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Calcium , Molecular Docking Simulation , Mice, Inbred C57BL , Mice, Inbred Strains , Hyperglycemia/complications , Hyperglycemia/drug therapy
2.
J Neurochem ; 166(2): 201-214, 2023 07.
Article in English | MEDLINE | ID: mdl-37070532

ABSTRACT

A neurodegenerative disorder is a condition that causes a degeneration of neurons in the central nervous system, leading to cognitive impairment and movement disorders. An accumulation of oxidative stress in neurons contributes to the pathogenesis of neurodegenerative disorders. Over the past few years, several studies have suggested that short-chain fatty acids, metabolites of the gut microbiota, might have a beneficial effect in neurodegenerative disorders. A G protein-coupled receptor 43 (GPR43) plays an important role in modulating oxidative stress and inflammatory processes in several tissues. Interestingly, the downstream signaling pathways activated by GPR43 to modulate oxidative stress differ among tissues. Moreover, the cellular mechanisms underlying GPR43 activation in neuronal cells to handle oxidative stress remain unclear. In this present study, we tested the role of GPR43, which is activated by short-chain fatty acids or a specific GPR43 agonist, in an oxidative stress-induced neuronal cell line (SH-SY5Y) injury. Our findings suggest that a combination of short-chain fatty acids with a physiological function could protect neurons from H2 O2 -induced cell damage. The effect of short-chain fatty acids mixture was abolished by pretreatment with a GPR43 antagonist, indicating this protective effect is a GPR43-dependent mechanism. In addition, a specific GPR43 agonist shows a similar result to that found in short-chain fatty acids mixture. Furthermore, our findings indicate that the downstream activation of GPR43 to protect against oxidative stress-induced neuronal injury is a biased Gq activation signaling of GPR43, which results in the prevention of H2 O2 -induced neuronal apoptosis. In conclusion, our results show new insight into the cellular mechanism of GPR43 and its neuroprotective effect. Taken together, this newly discovered finding suggests that activation of the biased Gq signaling pathway of GPR43 might be a potential therapeutic target for aging-related neurodegeneration.


Subject(s)
Neuroblastoma , Neuroprotective Agents , Humans , Neuroprotective Agents/pharmacology , Fatty Acids, Volatile/pharmacology , Signal Transduction , Oxidative Stress , Receptors, G-Protein-Coupled/metabolism
3.
Nat Commun ; 14(1): 34, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36596803

ABSTRACT

The γ-aminobutyric acid type B (GABAB) receptor is a prototypical family C G protein-coupled receptor (GPCR) that plays a key role in the regulation of synaptic transmission. Although growing evidence suggests that GPCR signaling in neurons might be highly organized in time and space, limited information is available about the mechanisms controlling the nanoscale organization of GABAB receptors and other GPCRs on the neuronal plasma membrane. Using a combination of biochemical assays in vitro, single-particle tracking, and super-resolution microscopy, we provide evidence that the spatial organization and diffusion of GABAB receptors on the plasma membrane are governed by dynamic interactions with filamin A, which tethers the receptors to sub-cortical actin filaments. We further show that GABAB receptors are located together with filamin A in small nanodomains in hippocampal neurons. These interactions are mediated by the first intracellular loop of the GABAB1 subunit and modulate the kinetics of Gαi protein activation in response to GABA stimulation.


Subject(s)
Receptors, GABA-B , Receptors, GABA , Receptors, GABA/metabolism , Filamins , Receptors, GABA-B/metabolism , Cell Membrane/metabolism , gamma-Aminobutyric Acid/metabolism
4.
Molecules ; 27(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35408508

ABSTRACT

Diabetic nephropathy (DN) is a leading cause of end-stage renal disease. An elevated fatty acid plasma concentration leads to podocyte injury and DN progression. This study aimed to identify and characterize cellular mechanisms of natural compounds that inhibit palmitic acid (PA)-induced human podocyte injury. By screening 355 natural compounds using a cell viability assay, 3-hydroxyterphenyllin (3-HT) and candidusin A (CDA), isolated from the marine-derived fungus Aspergillus candidus PSU-AMF169, were found to protect against PA-induced podocyte injury, with half-maximal inhibitory concentrations (IC50) of ~16 and ~18 µM, respectively. Flow cytometry revealed that 3-HT and CDA suppressed PA-induced podocyte apoptosis. Importantly, CDA significantly prevented PA-induced podocyte barrier impairment as determined by 70 kDa dextran flux. Reactive oxygen species (ROS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) direct scavenging assays indicated that both compounds exerted an anti-oxidative effect via direct free radical-scavenging activity. Moreover, 3-HT and CDA upregulated the anti-apoptotic Bcl2 protein. In conclusion, 3-HT and CDA represent fungus-derived bioactive compounds that have a novel protective effect on PA-induced human podocyte apoptosis via mechanisms involving free radical scavenging and Bcl2 upregulation.


Subject(s)
Diabetic Nephropathies , Podocytes , Apoptosis , Diabetic Nephropathies/metabolism , Fungi/metabolism , Humans , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Podocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Terphenyl Compounds
5.
Nat Chem Biol ; 16(9): 946-954, 2020 09.
Article in English | MEDLINE | ID: mdl-32541966

ABSTRACT

G-protein-coupled receptors (GPCRs) are key signaling proteins that mostly function as monomers, but for several receptors constitutive dimer formation has been described and in some cases is essential for function. Using single-molecule microscopy combined with super-resolution techniques on intact cells, we describe here a dynamic monomer-dimer equilibrium of µ-opioid receptors (µORs), where dimer formation is driven by specific agonists. The agonist DAMGO, but not morphine, induces dimer formation in a process that correlates both temporally and in its agonist- and phosphorylation-dependence with ß-arrestin2 binding to the receptors. This dimerization is independent from, but may precede, µOR internalization. These data suggest a new level of GPCR regulation that links dimer formation to specific agonists and their downstream signals.


Subject(s)
Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/metabolism , Single Molecule Imaging/methods , Animals , CHO Cells , Cricetulus , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/chemistry , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Fluorescence Resonance Energy Transfer , Morphine/chemistry , Morphine/pharmacology , Mutation , Naloxone/chemistry , Naloxone/pharmacology , Naltrexone/analogs & derivatives , Naltrexone/chemistry , Naltrexone/pharmacology , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Phosphorylation , Protein Multimerization , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , beta-Arrestins/metabolism
6.
Phys Rev E ; 99(4-1): 042149, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31108610

ABSTRACT

Taking advantage of recent single-particle tracking data, we compare the popular standard mean-squared displacement method with a statistical testing hypothesis procedure for three testing statistics and for two particle types: membrane receptors and the G proteins coupled to them. Each method results in different classifications. For this reason, more rigorous statistical tests are analyzed here in detail. The main conclusion is that the statistical testing approaches might provide good results even for short trajectories, but none of the proposed methods is "the best" for all considered examples; in other words, one needs to combine different approaches to get a reliable classification.

7.
Endocrinology ; 159(8): 2953-2965, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29931263

ABSTRACT

The cytoskeletal protein filamin A (FLNA) has been suggested to play an important role in the responsiveness of GH-secreting pituitary tumors to somatostatin receptor subtype 2 (SSTR2) agonists by regulating SSTR2 expression and signaling. However, the underlying mechanisms are unknown. In this study, we use fast multicolor single-molecule microscopy to image individual SSTR2 and FLNA molecules at the surface of living cells with unprecedented spatiotemporal resolution. We find that SSTR2 and FLNA undergo transient interactions, which occur preferentially along actin fibers and contribute to restraining SSTR2 diffusion. Agonist stimulation increases the localization of SSTR2 along actin fibers and, subsequently, SSTR2 clustering and recruitment to clathrin-coated pits (CCPs). Interfering with FLNA-SSTR2 binding with a dominant-negative FLNA fragment increases SSTR2 mobility, hampers the formation and alignment of SSTR2 clusters along actin fibers, and impairs both SSTR2 recruitment to CCPs and SSTR2 internalization. These findings indicate that dynamic SSTR2-FLNA interactions critically control the nanoscale localization of SSTR2 at the plasma membrane and are required for coupling SSTR2 clustering to internalization. These mechanisms explain the critical role of FLNA in the control of SSTR2 expression and signaling and suggest the possibility of targeting SSTR2-FLNA interactions for the therapy of pharmacologically resistant GH-secreting pituitary tumors.


Subject(s)
Filamins/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Actin Cytoskeleton/metabolism , Adenoma/drug therapy , Adenoma/genetics , Adenoma/metabolism , Animals , CHO Cells , Coated Pits, Cell-Membrane/metabolism , Cricetulus , Filamins/ultrastructure , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/genetics , Growth Hormone-Secreting Pituitary Adenoma/metabolism , HEK293 Cells , Humans , Protein Binding , Protein Transport , Receptors, Somatostatin/agonists , Receptors, Somatostatin/ultrastructure , Single Molecule Imaging
8.
Trends Pharmacol Sci ; 39(2): 109-122, 2018 02.
Article in English | MEDLINE | ID: mdl-29157577

ABSTRACT

G protein-coupled receptors (GPCRs) constitute the largest family of membrane receptors and are of great interest as pharmacological targets. Although the occurrence of GPCR signaling nanodomains has long been hypothesized based on indirect evidence, this and other fundamental aspects of GPCR signaling have been difficult to prove. The advent of single-molecule microscopy methods, which allow direct visualization of individual membrane proteins with unprecedented spatiotemporal resolution, provides unique opportunities to address several of these open questions. Indeed, recent single-molecule studies have revealed that GPCRs and G proteins transiently interact with each other as well as with structural components of the plasma membrane, leading to the formation of dynamic complexes and 'hot spots' for GPCR signaling. Whereas we are only beginning to understand the implications of this unexpected level of complexity, single-molecule approaches are likely to play a crucial role to further dissect the protein-protein interactions that are at the heart of GPCR signaling.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Single Molecule Imaging/methods , Animals , Humans , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
9.
Nature ; 550(7677): 543-547, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29045395

ABSTRACT

G-protein-coupled receptors mediate the biological effects of many hormones and neurotransmitters and are important pharmacological targets. They transmit their signals to the cell interior by interacting with G proteins. However, it is unclear how receptors and G proteins meet, interact and couple. Here we analyse the concerted motion of G-protein-coupled receptors and G proteins on the plasma membrane and provide a quantitative model that reveals the key factors that underlie the high spatiotemporal complexity of their interactions. Using two-colour, single-molecule imaging we visualize interactions between individual receptors and G proteins at the surface of living cells. Under basal conditions, receptors and G proteins form activity-dependent complexes that last for around one second. Agonists specifically regulate the kinetics of receptor-G protein interactions, mainly by increasing their association rate. We find hot spots on the plasma membrane, at least partially defined by the cytoskeleton and clathrin-coated pits, in which receptors and G proteins are confined and preferentially couple. Imaging with the nanobody Nb37 suggests that signalling by G-protein-coupled receptors occurs preferentially at these hot spots. These findings shed new light on the dynamic interactions that control G-protein-coupled receptor signalling.


Subject(s)
Cell Membrane/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Adrenergic/metabolism , Single Molecule Imaging , Animals , Cell Membrane/chemistry , Cell Survival , Clathrin/metabolism , Coated Pits, Cell-Membrane/chemistry , Coated Pits, Cell-Membrane/metabolism , Color , Cytoskeleton/metabolism , Diffusion , Human Umbilical Vein Endothelial Cells , Humans , Kinetics , Mice , Movement , Signal Transduction
10.
Endocrinology ; 157(4): 1613-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26828746

ABSTRACT

A crucial event in female reproduction occurs at midcycle, when a LH peak induces the final maturation of ovarian follicles. LH signals via a G protein-coupled receptor selectively expressed in the outermost follicular cell layers. However, how LH signals are relayed inside these cells and finally to the oocyte is incompletely understood. Here, we monitored LH signaling in intact ovarian follicles of transgenic mice expressing a fluorescent cAMP sensor. We found that LH stimulation induces 2 phases of cAMP signaling in all cell layers surrounding the oocyte. Interfering with LH receptor internalization abolished the second, persistent cAMP phase and partially inhibited oocyte meiosis resumption. These data suggest that persistent cAMP signals from internalized LH receptors contribute to transmitting LH effects inside follicle cells and ultimately to the oocyte. Thus, this study indicates that the recently proposed paradigm of cAMP signaling by internalized G protein-coupled receptors is implicated in receptor function and is physiologically relevant.


Subject(s)
Cyclic AMP/metabolism , Ovarian Follicle/metabolism , Receptors, LH/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Endocytosis/physiology , Female , Fluorescence Resonance Energy Transfer , Luteinizing Hormone/pharmacology , Meiosis/drug effects , Mice, Transgenic , Microscopy, Confocal , Oocytes/drug effects , Oocytes/metabolism , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Signal Transduction/drug effects
11.
Methods Mol Biol ; 1335: 53-66, 2015.
Article in English | MEDLINE | ID: mdl-26260594

ABSTRACT

Assessing the dynamics of individual membrane proteins in living cells is a powerful approach to investigate their assembly, mobility, and function. Here, we describe how to image single G protein-coupled receptors (GPCRs), both in the active and inactive state. This is achieved by combining labeling of GPCRs with bright organic fluorophores and fluorescent imaging by total internal reflection fluorescence microscopy. Using this method, individual tracks of single molecules can be analyzed in parallel with high spatial precision and with frame rates up to 50/s.


Subject(s)
Microscopy, Fluorescence/methods , Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , Calibration , Cricetinae , Cricetulus , Fluorescent Dyes/chemistry , Image Processing, Computer-Assisted , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Staining and Labeling , Transfection
12.
J Vis Exp ; (89): e51784, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-25145374

ABSTRACT

Single-molecule microscopy is emerging as a powerful approach to analyze the behavior of signaling molecules, in particular concerning those aspect (e.g., kinetics, coexistence of different states and populations, transient interactions), which are typically hidden in ensemble measurements, such as those obtained with standard biochemical or microscopy methods. Thus, dynamic events, such as receptor-receptor interactions, can be followed in real time in a living cell with high spatiotemporal resolution. This protocol describes a method based on labeling with small and bright organic fluorophores and total internal reflection fluorescence (TIRF) microscopy to directly visualize single receptors on the surface of living cells. This approach allows one to precisely localize receptors, measure the size of receptor complexes, and capture dynamic events such as transient receptor-receptor interactions. The protocol provides a detailed description of how to perform a single-molecule experiment, including sample preparation, image acquisition and image analysis. As an example, the application of this method to analyze two G-protein-coupled receptors, i.e., ß2-adrenergic and γ-aminobutyric acid type B (GABAB) receptor, is reported. The protocol can be adapted to other membrane proteins and different cell models, transfection methods and labeling strategies.


Subject(s)
Microscopy, Fluorescence/methods , Receptors, Adrenergic, beta-2/chemistry , Receptors, GABA-B/chemistry , Image Processing, Computer-Assisted
13.
Eur J Med Chem ; 63: 629-34, 2013 May.
Article in English | MEDLINE | ID: mdl-23567951

ABSTRACT

Angiotensin II receptor type I (AT1R) is a G-protein coupled receptor involved in regulation of body water-electrolyte balance and blood pressure. Oxidative stress promotes AT1R oligomerization and hyper-responsiveness to its cognate ligand Ang II. In this study, bivalent Ang II, synthesized by linking with aminocaproic acid (Acp) at the N-terminus, was used to induce AT1R dimerization and hyper-responsiveness in AT1R-expressed human embryonic kidney (AT1R-HEK) cells, determined using image correlation spectroscopy (ICS) and by measuring AT1R-mediated change in intracellular Ca(2+) concentration, respectively. In addition, ICS was employed to determine distribution pattern of cell-surface AT1R and its degree of aggregation when stimulated by monomeric (monovalent) and bivalent Ang II under oxidative stress (100 µM H2O2) condition in comparison with normal (unoxidized) AT1R-HEK cells. Bivalent Ang II induced cell-surface AT1R aggregation/clustering but maintained AT1R normal signaling response under oxidative stress condition, whereas stimulation by monomeric Ang II or a mixture of monomeric and Acp-modified Ang II (used in the synthesis of bivalent form) resulted in AT1R hyper-responsiveness. These results suggest that bivalent ligand (viz. Ang II) provides another strategy in the development of novel drugs specifically designed for attenuating aberrant responsiveness of cognate receptor (AT1R) under pathological (oxidative stress) conditions.


Subject(s)
Angiotensin II/pharmacology , Oxidative Stress/drug effects , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Angiotensin II/chemical synthesis , Angiotensin II/chemistry , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Hydrogen Peroxide/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Models, Chemical , Molecular Structure , Oxidants/pharmacology , Protein Multimerization/drug effects , Receptor, Angiotensin, Type 1/chemistry , Receptor, Angiotensin, Type 1/genetics , Spectrometry, Fluorescence
14.
Proc Natl Acad Sci U S A ; 110(2): 743-8, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23267088

ABSTRACT

G-protein-coupled receptors (GPCRs) constitute the largest family of receptors and major pharmacological targets. Whereas many GPCRs have been shown to form di-/oligomers, the size and stability of such complexes under physiological conditions are largely unknown. Here, we used direct receptor labeling with SNAP-tags and total internal reflection fluorescence microscopy to dynamically monitor single receptors on intact cells and thus compare the spatial arrangement, mobility, and supramolecular organization of three prototypical GPCRs: the ß(1)-adrenergic receptor (ß(1)AR), the ß(2)-adrenergic receptor (ß(2)AR), and the γ-aminobutyric acid (GABA(B)) receptor. These GPCRs showed very different degrees of di-/oligomerization, lowest for ß(1)ARs (monomers/dimers) and highest for GABA(B) receptors (prevalently dimers/tetramers of heterodimers). The size of receptor complexes increased with receptor density as a result of transient receptor-receptor interactions. Whereas ß(1)-/ß(2)ARs were apparently freely diffusing on the cell surface, GABA(B) receptors were prevalently organized into ordered arrays, via interaction with the actin cytoskeleton. Agonist stimulation did not alter receptor di-/oligomerization, but increased the mobility of GABA(B) receptor complexes. These data provide a spatiotemporal characterization of ß(1)-/ß(2)ARs and GABA(B) receptors at single-molecule resolution. The results suggest that GPCRs are present on the cell surface in a dynamic equilibrium, with constant formation and dissociation of new receptor complexes that can be targeted, in a ligand-regulated manner, to different cell-surface microdomains.


Subject(s)
Algorithms , Models, Chemical , Multiprotein Complexes/chemistry , Receptors, Adrenergic, beta-1/chemistry , Receptors, Adrenergic, beta-2/chemistry , Receptors, GABA/chemistry , Animals , Bridged Bicyclo Compounds, Heterocyclic , CHO Cells , Carbocyanines , Cricetinae , Cricetulus , Cyclic AMP/metabolism , HEK293 Cells , Humans , Lipids , Microscopy, Fluorescence , Molecular Dynamics Simulation , Plasmids/genetics , Radioligand Assay , Thiazolidines
15.
Biochim Biophys Acta ; 1808(10): 2496-500, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21791198

ABSTRACT

Oxidative stress and hyper-functioning of angiotensin II receptor type I (AT(1)R) are commonly observed in hypertensive patients but the relationship between oxidative stress and AT(1)R function is still unclear. We investigated the effects of H(2)O(2) treatment on AT(1)R-mediated intracellular calcium [Ca(2+)](i) signaling and its cell surface distribution pattern in HEK cells stably expressing EGFP-tagged rat AT(1)R using image correlation spectroscopy (ICS). Following H(2)O(2) treatment (50-800µM), [Ca(2+)](i) was significantly increased upon angiotensin II stimulation. Similarly ICS revealed a significant increase in degree of AT(1)R aggregation in H(2)O(2) treated group during Ang II activation but no difference in cluster density compared with untreated control cells or those with N-acetyl cysteine pretreatment. Thus, oxidative stress-induced AT(1)R hyper-responsiveness can be attributed by an increase in cell surface receptor aggregation state, possibly stemming in part from oxidant-related increase receptor-receptor interactions.


Subject(s)
Oxidative Stress , Receptor, Angiotensin, Type 1/physiology , Spectrum Analysis/methods , Cell Line , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...