Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.969
1.
Cell Calcium ; 120: 102883, 2024 Jun.
Article En | MEDLINE | ID: mdl-38643716

The basal and glucose-induced insulin secretion from pancreatic beta cells is a tightly regulated process that is triggered in a Ca2+-dependent fashion and further positively modulated by substances that raise intracellular levels of adenosine 3',5'-cyclic monophosphate (cAMP) or by certain antidiabetic drugs. In a previous study, we have temporally resolved the subplasmalemmal [Ca2+]i dynamics in beta cells that are characterized by trains of sharply delimited spikes, reaching peak values up to 5 µM. Applying total internal reflection fluorescence (TIRF) microscopy and synaptopHluorin to visualize fusion events of individual granules, we found that several fusion events can coincide within 50 to 150 ms. To test whether subplasmalemmal [Ca2+]i microdomains around single or clustered Ca2+ channels may cause a synchronized release of insulin-containing vesicles, we applied simultaneous dual-color TIRF microscopy and monitored Ca2+ fluctuations and exocytotic events in INS-1 cells at high frame rates. The results indicate that fusions can be triggered by subplasmalemmal Ca2+ spiking. This, however, does account for a minority of fusion events. About 90 %-95 % of fusion events either happen between Ca2+ spikes or incidentally overlap with subplasmalemmal Ca2+ spikes. We conclude that only a fraction of exocytotic events in glucose-induced and tolbutamide- or forskolin-enhanced insulin release from INS-1 cells is tightly coupled to Ca2+ microdomains around voltage-gated Ca2+ channels.


Calcium , Exocytosis , Insulin-Secreting Cells , Insulin , Microscopy, Fluorescence , Insulin-Secreting Cells/metabolism , Calcium/metabolism , Animals , Rats , Insulin/metabolism , Exocytosis/drug effects , Calcium Signaling , Insulin Secretion/drug effects , Glucose/metabolism , Secretory Vesicles/metabolism
2.
J Biol Chem ; 299(9): 105119, 2023 09.
Article En | MEDLINE | ID: mdl-37527778

Serratia marcescens is an opportunistic human pathogen involved in antibiotic-resistant hospital acquired infections. Upon contact with the host epithelial cell and prior to internalization, Serratia induces an early autophagic response that is entirely dependent on the ShlA toxin. Once Serratia invades the eukaryotic cell and multiples inside an intracellular vacuole, ShlA expression also promotes an exocytic event that allows bacterial egress from the host cell without compromising its integrity. Several toxins, including ShlA, were shown to induce ATP efflux from eukaryotic cells. Here, we demonstrate that ShlA triggered a nonlytic release of ATP from Chinese hamster ovary (CHO) cells. Enzymatic removal of accumulated extracellular ATP (eATP) or pharmacological blockage of the eATP-P2Y2 purinergic receptor inhibited the ShlA-promoted autophagic response in CHO cells. Despite the intrinsic ecto-ATPase activity of CHO cells, the effective concentration and kinetic profile of eATP was consistent with the established affinity of the P2Y2 receptor and the known kinetics of autophagy induction. Moreover, eATP removal or P2Y2 receptor inhibition also suppressed the ShlA-induced exocytic expulsion of the bacteria from the host cell. Blocking α5ß1 integrin highly inhibited ShlA-dependent autophagy, a result consistent with α5ß1 transactivation by the P2Y2 receptor. In sum, eATP operates as the key signaling molecule that allows the eukaryotic cell to detect the challenge imposed by the contact with the ShlA toxin. Stimulation of P2Y2-dependent pathways evokes the activation of a defensive response to counteract cell damage and promotes the nonlytic clearance of the pathogen from the infected cell.


Autophagy , Host-Pathogen Interactions , Integrin alpha5beta1 , Receptors, Purinergic P2Y2 , Serratia , Toxins, Biological , Animals , Cricetinae , Adenosine Triphosphate/metabolism , Autophagy/drug effects , CHO Cells , Cricetulus , Exocytosis/drug effects , Host-Pathogen Interactions/drug effects , Integrin alpha5beta1/antagonists & inhibitors , Integrin alpha5beta1/metabolism , Receptors, Purinergic P2Y2/metabolism , Serratia/chemistry , Serratia/drug effects , Serratia/physiology , Toxins, Biological/pharmacology , Humans
3.
Neuromolecular Med ; 25(1): 125-135, 2023 03.
Article En | MEDLINE | ID: mdl-36436129

Lithium is a mood stabilizer broadly used to prevent and treat symptoms of mania and depression in people with bipolar disorder (BD). Little is known, however, about its mode of action. Here, we analyzed the impact of lithium on synaptic vesicle (SV) cycling at presynaptic terminals releasing glutamate, a neurotransmitter previously implicated in BD and other neuropsychiatric conditions. We used the pHluorin-based synaptic tracer vGpH and a fully automated image processing pipeline to quantify the effect of lithium on both SV exocytosis and endocytosis in hippocampal neurons. We found that lithium selectively reduces SV exocytic rates during electrical stimulation, and markedly slows down SV recycling post-stimulation. Analysis of single-bouton responses revealed the existence of functionally distinct excitatory synapses with varying sensitivity to lithium-some terminals show responses similar to untreated cells, while others are markedly impaired in their ability to recycle SVs. While the cause of this heterogeneity is unclear, these data indicate that lithium interacts with the SV machinery and influences glutamate release in a large fraction of excitatory synapses. Together, our findings show that lithium down modulates SV cycling, an effect consistent with clinical reports indicating hyperactivation of glutamate neurotransmission in BD.


Glutamic Acid , Lithium Compounds , Synapses , Synaptic Vesicles , Lithium Compounds/pharmacology , Glutamic Acid/metabolism , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Action Potentials/drug effects , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Hippocampus/pathology , Exocytosis/drug effects , Endocytosis/drug effects , Animals , Rats , Cells, Cultured
4.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article En | MEDLINE | ID: mdl-35163009

The fusion of membranes is a central part of the physiological processes involving the intracellular transport and maturation of vesicles and the final release of their contents, such as neurotransmitters and hormones, by exocytosis. Traditionally, in this process, proteins, such SNAREs have been considered the essential components of the fusion molecular machinery, while lipids have been seen as merely structural elements. Nevertheless, sphingosine, an intracellular signalling lipid, greatly increases the release of neurotransmitters in neuronal and neuroendocrine cells, affecting the exocytotic fusion mode through the direct interaction with SNAREs. Moreover, recent studies suggest that FTY-720 (Fingolimod), a sphingosine structural analogue used in the treatment of multiple sclerosis, simulates sphingosine in the promotion of exocytosis. Furthermore, this drug also induces the intracellular fusion of organelles such as dense vesicles and mitochondria causing cell death in neuroendocrine cells. Therefore, the effect of sphingosine and synthetic derivatives on the heterologous and homologous fusion of organelles can be considered as a new mechanism of action of sphingolipids influencing important physiological processes, which could underlie therapeutic uses of sphingosine derived lipids in the treatment of neurodegenerative disorders and cancers of neuronal origin such neuroblastoma.


Exocytosis/drug effects , Neuroendocrine Cells/metabolism , Sphingosine/metabolism , Animals , Biological Transport , Humans , Membrane Fusion , SNARE Proteins/metabolism , Sphingosine/pharmacology
5.
Life Sci ; 296: 120433, 2022 May 01.
Article En | MEDLINE | ID: mdl-35219696

AIMS: Neurotransmitter release from the synaptic vesicles can occur through two modes of exocytosis: "full-collapse" or "kiss-and-run". Here we investigated how increasing the nerve activity and pharmacological stimulation of adrenoceptors can influence the mode of exocytosis in the motor nerve terminal. METHODS: Recording of endplate potentials with intracellular microelectrodes was used to estimate acetylcholine release. A fluorescent dye FM1-43 and its quenching with sulforhodamine 101 were utilized to visualize synaptic vesicle recycling. KEY FINDINGS: An increase in the frequency of stimulation led to a decrease in the rate of FM1-43 unloading despite the higher number of quanta released. High frequency activity promoted neurotransmitter release via the kiss-and-run mechanism. This was confirmed by experiments utilizing (I) FM1-43 dye quencher, that is able to pass into the synaptic vesicle via fusion pore, and (II) loading of FM1-43 by compensatory endocytosis. Noradrenaline and specific α2-adrenoreceptors agonist, dexmedetomidine, controlled the mode of synaptic vesicle recycling at high frequency activity. Their applications favored neurotransmitter release via full-collapse exocytosis rather than the kiss-and-run pathway. SIGNIFICANCE: At the diaphragm neuromuscular junctions, neuronal commands are translated into contractions necessary for respiration. During stress, an increase in discharge rate of the phrenic nerve shifts the exocytosis from the full-collapse to the kiss-and-run mode. The stress-related molecule, noradrenaline, restricts neurotransmitter release in response to a high frequency activity, and prevents the shift in the mode of exocytosis through α2-adrenoceptor activation. This may be a component of the mechanism that limits overstimulation of the respiratory system during stress.


Exocytosis/physiology , Neuromuscular Junction/physiology , Receptors, Adrenergic/metabolism , Acetylcholine/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Dexmedetomidine/pharmacology , Evoked Potentials/drug effects , Exocytosis/drug effects , Fluorescent Dyes/pharmacokinetics , Mice, Inbred BALB C , Neuromuscular Junction/drug effects , Neurotransmitter Agents/metabolism , Norepinephrine/metabolism , Norepinephrine/pharmacology , Pyridinium Compounds/pharmacokinetics , Quaternary Ammonium Compounds/pharmacokinetics , Receptors, Adrenergic, alpha-2/metabolism , Synaptic Vesicles/metabolism
6.
Cells ; 11(2)2022 01 12.
Article En | MEDLINE | ID: mdl-35053370

In this study, we identified a novel pyrazole-based derivative (P3C) that displayed potent cytotoxicity against 27 human cancer cell lines derived from different tissue origins with 50% cytotoxic concentrations (CC50) in the low micromolar and nanomolar range, particularly in two triple-negative breast cancer (TNBC) cell lines (from 0.25 to 0.49 µM). In vitro assays revealed that P3C induces reactive oxygen species (ROS) accumulation leading to mitochondrial depolarization and caspase-3/7 and -8 activation, suggesting the participation of both the intrinsic and extrinsic apoptotic pathways. P3C caused microtubule disruption, phosphatidylserine externalization, PARP cleavage, DNA fragmentation, and cell cycle arrest on TNBC cells. In addition, P3C triggered dephosphorylation of CREB, p38, ERK, STAT3, and Fyn, and hyperphosphorylation of JNK and NF-kB in TNBC cells, indicating the inactivation of both p38MAPK/STAT3 and ERK1/2/CREB signaling pathways. In support of our in vitro assays, transcriptome analyses of two distinct TNBC cell lines (MDA-MB-231 and MDA-MB-468 cells) treated with P3C revealed 28 genes similarly affected by the treatment implicated in apoptosis, oxidative stress, protein kinase modulation, and microtubule stability.


Pyrazoles/toxicity , Signal Transduction , Triple Negative Breast Neoplasms/pathology , Caspases/metabolism , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Exocytosis/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Microtubules/drug effects , Microtubules/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Phosphatidylserines/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Pyrazoles/chemistry , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Triple Negative Breast Neoplasms/genetics , Tubulin/metabolism
7.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article En | MEDLINE | ID: mdl-35055082

Nerve growth factor (NGF) is known to intensify pain in various ways, so perturbing pertinent effects without negating its essential influences on neuronal functions could help the search for much-needed analgesics. Towards this goal, cultured neurons from neonatal rat trigeminal ganglia-a locus for craniofacial sensory nerves-were used to examine how NGF affects the Ca2+-dependent release of a pain mediator, calcitonin gene-related peptide (CGRP), that is triggered by activating a key signal transducer, transient receptor potential vanilloid 1 (TRPV1) with capsaicin (CAP). Measurements utilised neurons fed with or deprived of NGF for 2 days. Acute re-introduction of NGF induced Ca2+-dependent CGRP exocytosis that was inhibited by botulinum neurotoxin type A (BoNT/A) or a chimera of/E and/A (/EA), which truncated SNAP-25 (synaptosomal-associated protein with Mr = 25 k) at distinct sites. NGF additionally caused a Ca2+-independent enhancement of the neuropeptide release evoked by low concentrations (<100 nM) of CAP, but only marginally increased the peak response to ≥100 nM. Notably, BoNT/A inhibited CGRP exocytosis evoked by low but not high CAP concentrations, whereas/EA effectively reduced responses up to 1 µM CAP and inhibited to a greater extent its enhancement by NGF. In addition to establishing that sensitisation of sensory neurons to CAP by NGF is dependent on SNARE-mediated membrane fusion, insights were gleaned into the differential ability of two regions in the C-terminus of SNAP-25 (181-197 and 198-206) to support CAP-evoked Ca2+-dependent exocytosis at different intensities of stimulation.


Calcitonin Gene-Related Peptide/biosynthesis , Capsaicin/pharmacology , Nerve Growth Factor/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/metabolism , Animals , Botulinum Toxins, Type A/pharmacology , Calcium/metabolism , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Exocytosis/drug effects , Nerve Growth Factor/pharmacology , Proteolysis , Rats , Synaptosomal-Associated Protein 25/metabolism
8.
Angew Chem Int Ed Engl ; 61(1): e202111853, 2022 01 03.
Article En | MEDLINE | ID: mdl-34734656

Alpha-synuclein (α-Syn) localizes at presynaptic terminal and modulates synaptic functions. Increasing evidence demonstrate that α-Syn oligomers, forming at the early of aggregation, are cytotoxic and is thus related to brain neurodegenerative diseases. Herein, we find that vitamin D (VD) can reduce neurocytotoxicity. The reduced neurocytotoxicity might be attributed to the less amount of large-sized α-Syn oligomers inhibited by VD, measured by electrochemical collision at single particle level, which are not observable with traditionally ensembled method. Single-cell amperometry (SCA) results show that VD can recover the amount of neurotransmitter release during exocytosis induced by α-Syn oligomers, further verifying the neuroprotection of VD. Our study reveals the neuroprotective role of VD through inhibiting α-Syn aggregation, which is envisioned to be of great importance in treatment and prevention of the neurodegenerative diseases.


Electrochemical Techniques , Vitamin D/pharmacology , alpha-Synuclein/antagonists & inhibitors , Cell Line, Tumor , Exocytosis/drug effects , Humans , Models, Molecular , Protein Aggregates/drug effects , Vitamin D/chemistry , alpha-Synuclein/metabolism
9.
J Nutr Biochem ; 99: 108864, 2022 01.
Article En | MEDLINE | ID: mdl-34606907

Vitamin D3 is associated with improvements in insulin resistance and glycemia. In this study, we investigated the short-term effect of 1α,25(OH)2 Vitamin D3 (1,25-D3) and cholecalciferol (vitamin D3) on the glycemia and insulin sensitivity of control and dexamethasone-induced insulin-resistance rats. 45Ca2+ influx responses to 1,25-D3 and its role in insulin secretion were investigated in isolated pancreatic islets from control rats. In vivo, 5 d treatment with 1,25-D3 (i.p.) prevented insulin resistance in dexamethasone-treated rats. Treatment with 1,25-D3 improved the activities of hepatic enzymes, serum lipids and calcium concentrations in insulin-resistant rats. 25-D3 (o.g.) does not affect insulin resistance. In pancreatic islets, 1,25-D3 increased insulin secretion and stimulated rapid response 45Ca2+ influx. The stimulatory effect of 1,25-D3 on 45Ca2+ influx was decreased by diazoxide, apamine, thapsigargin, dantrolene, 2-APB, nifedipine, TEA, PKA, PKC, and cytoskeleton inhibitor, while it was increased by glibenclamide and N-ethylmaleimide. The stimulatory effect of 1,25-D3 on 45Ca2+ influx involves the activation of L-type VDCC, K+-ATP, K+-Ca2+, and Kv channels, which augment cytosolic calcium. These ionic changes mobilize calcium from stores and downstream activation of PKC, PKA tethering vesicle traffic and fusion at the plasma membrane for insulin secretion. This is the first study highlighting the unprecedented role of 1,25-D3 (short-term effect) in the regulation of glucose homeostasis and on prevention of insulin resistance. Furthermore, this study shows the intracellular ß-cell signal transduction of 1,25-D3 through the modulation of pivotal ionic channels and proteins exhibiting a coordinated exocytosis of vesicles for insulin secretion.


Cholecalciferol/analogs & derivatives , Exocytosis/drug effects , Insulin Resistance , Insulin Secretion/drug effects , Insulin/metabolism , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cholecalciferol/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Rats , Rats, Wistar
10.
Biochem Pharmacol ; 194: 114821, 2021 12.
Article En | MEDLINE | ID: mdl-34748819

The pancreatic ß-cells control insulin secretion in the body to regulate glucose homeostasis, and ß-cell stress and dysfunction is characteristic of Type 2 Diabetes. Pharmacological targeting of the ß-cell to increase insulin secretion is typically utilised, however, extended use of common drugs such as sulfonylureas are known to result in secondary failure. Moreover, there is evidence they may induce ß-cell failure in the long term. Within ß-cells, insulin secretory granules (ISG) serve as compartments to store, process and traffic insulin for exocytosis. There is now growing evidence that ISG exist in multiple populations, distinct in their protein composition, motility, age, and capacity for secretion. In this review, we discuss the implications of a heterogenous ISG population in ß-cells and highlight the need for more understanding into how unique ISG populations may be targeted in anti-diabetic therapies.


Diabetes Mellitus, Type 2/drug therapy , Drug Delivery Systems/methods , Exocytosis/drug effects , Hypoglycemic Agents/administration & dosage , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Animals , Diabetes Mellitus, Type 2/metabolism , Exocytosis/physiology , Humans , Hypoglycemic Agents/metabolism , Insulin/metabolism , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism
11.
Cell Rep ; 37(5): 109923, 2021 11 02.
Article En | MEDLINE | ID: mdl-34731611

The dense variant surface glycoprotein (VSG) coat of African trypanosomes represents the primary host-pathogen interface. Antigenic variation prevents clearing of the pathogen by employing a large repertoire of antigenically distinct VSG genes, thus neutralizing the host's antibody response. To explore the epitope space of VSGs, we generate anti-VSG nanobodies and combine high-resolution structural analysis of VSG-nanobody complexes with binding assays on living cells, revealing that these camelid antibodies bind deeply inside the coat. One nanobody causes rapid loss of cellular motility, possibly due to blockage of VSG mobility on the coat, whose rapid endocytosis and exocytosis are mechanistically linked to Trypanosoma brucei propulsion and whose density is required for survival. Electron microscopy studies demonstrate that this loss of motility is accompanied by rapid formation and shedding of nanovesicles and nanotubes, suggesting that increased protein crowding on the dense membrane can be a driving force for membrane fission in living cells.


Cell Membrane/drug effects , Cell Movement/drug effects , Single-Domain Antibodies/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Variant Surface Glycoproteins, Trypanosoma/immunology , Animals , Antibody Specificity , Binding Sites, Antibody , Camelids, New World/immunology , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endocytosis/drug effects , Epitopes , Exocytosis/drug effects , Protein Binding , Single-Domain Antibodies/immunology , Single-Domain Antibodies/metabolism , Trypanocidal Agents/immunology , Trypanocidal Agents/metabolism , Trypanosoma brucei brucei/immunology , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/ultrastructure , Trypanosomiasis, African/immunology , Trypanosomiasis, African/metabolism , Trypanosomiasis, African/parasitology , Variant Surface Glycoproteins, Trypanosoma/metabolism
12.
Angew Chem Int Ed Engl ; 60(44): 23552-23556, 2021 10 25.
Article En | MEDLINE | ID: mdl-34363735

Electrochemical methods were used to explore the exocytotic nature of serotonin (5-HT) release in human carcinoid BON cells, an in vitro human enterochromaffin cell model, to understand the mechanisms operating the release of gut-derived 5-HT in the intestinal mucosal epithelium. We show that the fractional vesicular 5-HT release in BON cells is 80 % compared to previous work in pancreatic beta cells (34 %). The fractional release increased from 80 % in control BON cells to 87 % with 5-HT preincubation and nearly 100 % with the combination of 5-HT and the 5-HT4 autoreceptor agonist, cisapride. Thus, partial release is the primary mechanism of exocytosis in BON cells, resulting in a variable amount of the vesicular content being released. Factors that control secretion of 5-HT from enterochromaffin cells or BON cells are important as partial release provides a mechanism for development of effective therapeutic strategies to treat gastrointestinal diseases.


Electrochemical Techniques , Enterochromaffin Cells/drug effects , Nanotechnology , Serotonin/pharmacology , Drug Liberation , Exocytosis/drug effects , Gastrointestinal Diseases/drug therapy , Humans , Serotonin/chemistry
13.
Br J Anaesth ; 127(4): 587-599, 2021 Oct.
Article En | MEDLINE | ID: mdl-34384592

BACKGROUND: The cellular and molecular mechanisms by which general anaesthesia occurs is poorly understood. Hippocampal interneurone subpopulations, which are critical regulators of cognitive function, have diverse neurophysiological and synaptic properties, but their responses to anaesthetics are unclear. METHODS: We used live-cell imaging of fluorescent biosensors expressed in mouse hippocampal neurones to delineate interneurone subtype-specific effects of isoflurane on synaptic vesicle exocytosis. The role of voltage-gated sodium channel (Nav) subtype expression in determining isoflurane sensitivity was probed by overexpression or knockdown of specific Nav subtypes in identified interneurones. RESULTS: Clinically relevant concentrations of isoflurane differentially inhibited synaptic vesicle exocytosis: to 83.1% (11.7%) of control in parvalbumin-expressing interneurones, and to 58.6% (13.3%) and 64.5% (8.5%) of control in somatostatin-expressing interneurones and glutamatergic neurones, respectively. The relative expression of Nav1.1 (associated with lower sensitivity) and Nav1.6 (associated with higher sensitivity) determined the sensitivity of exocytosis to isoflurane. CONCLUSIONS: Isoflurane inhibits synaptic vesicle exocytosis from hippocampal glutamatergic neurones and GABAergic interneurones in a cell-type-specific manner depending on their expression of voltage-gated sodium channel subtypes.


Anesthetics, Inhalation/pharmacology , Hippocampus/drug effects , Isoflurane/pharmacology , gamma-Aminobutyric Acid/metabolism , Animals , Exocytosis/drug effects , Female , Gene Knockdown Techniques , Hippocampus/metabolism , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Synaptic Transmission/drug effects , Voltage-Gated Sodium Channels/genetics
14.
Bioorg Chem ; 115: 105231, 2021 10.
Article En | MEDLINE | ID: mdl-34388485

The analgesic peptide DD04107 (Pal-EEMQRR-NH2) and its acetylated analogue inhibit α-calcitonin gene-related peptide (α-CGRP) exocytotic release from primary sensory neurons. Examining the crystal structure of the SNARE-Synaptotagmin-1(Syt1) complex, we hypothesized that these peptides could inhibit neuronal exocytosis by binding to Syt1, hampering at least partially its interaction with the SNARE complex. To address this hypothesis, we first interrogate the role of individual side-chains on the inhibition of α-CGRP release, finding that E1, M3, Q4 and R6 residues were crucial for activity. CD and NMR conformational analysis showed that linear peptides have tendency to adopt α-helical conformations, but the results with cyclic analogues indicated that this secondary structure is not needed for activity. Isothermal titration calorimetry (ITC) measurements demonstrate a direct interaction of some of these peptides with Syt1-C2B domain, but not with Syt7-C2B region, indicating selectivity. As expected for a compound able to inhibit α-CGRP release, cyclic peptide derivative Pal-E-cyclo[EMQK]R-NH2 showed potent in vivo analgesic activity, in a model of inflammatory pain. Molecular dynamics simulations provided a model consistent with KD values for the interaction of peptides with Syt1-C2B domain, and with their biological activity. Altogether, these results identify Syt1 as a potential new analgesic target.


Analgesics/pharmacology , Lipopeptides/pharmacology , Pain/drug therapy , Synaptotagmin I/antagonists & inhibitors , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/metabolism , Dose-Response Relationship, Drug , Exocytosis/drug effects , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Male , Mice , Molecular Dynamics Simulation , Molecular Structure , Pain/metabolism , Structure-Activity Relationship , Synaptotagmin I/metabolism
15.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166246, 2021 12 01.
Article En | MEDLINE | ID: mdl-34403739

Alzheimer's disease (AD) pathology progresses gradually via anatomically connected brain regions. Direct transfer of amyloid-ß1-42 oligomers (oAß) between connected neurons has been shown, however, the mechanism is not fully revealed. We observed formation of oAß induced tunneling nanotubes (TNTs)-like nanoscaled f-actin containing membrane conduits, in differentially differentiated SH-SY5Y neuronal models. Time-lapse images showed that oAß propagate from one cell to another via TNT-like structures. Preceding the formation of TNT-like conduits, we detected oAß-induced plasma membrane (PM) damage and calcium-dependent repair through lysosomal-exocytosis, followed by massive endocytosis to re-establish the PM. Massive endocytosis was monitored by an influx of the membrane-staining dye TMA-DPH and PM damage was quantified by propidium iodide influx in the absence of Ca2+. The massive endocytosis eventually caused accumulation of internalized oAß in Lamp1 positive multivesicular bodies/lysosomes via the actin cytoskeleton remodulating p21-activated kinase1 (PAK1) dependent endocytic pathway. Three-dimensional quantitative confocal imaging, structured illumination superresolution microscopy, and flowcytometry quantifications revealed that oAß induces activation of phospho-PAK1, which modulates the formation of long stretched f-actin extensions between cells. Moreover, the formation of TNT-like conduits was inhibited by preventing PAK1-dependent internalization of oAß using the small-molecule inhibitor IPA-3, a highly selective cell-permeable auto-regulatory inhibitor of PAK1. The present study reveals that the TNT-like conduits are probably instigated as a consequence of oAß induced PM damage and repair process, followed by PAK1 dependent endocytosis and actin remodeling, probably to maintain cell surface expansion and/or membrane tension in equilibrium.


Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Disulfides/pharmacology , Naphthols/pharmacology , p21-Activated Kinases/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Brain/drug effects , Brain/pathology , Cell Membrane/drug effects , Cell Membrane/pathology , Endocytosis/drug effects , Exocytosis/drug effects , Humans , Lysosomes/drug effects , Nanotubes/chemistry , p21-Activated Kinases/antagonists & inhibitors
16.
Cell Rep ; 36(8): 109609, 2021 08 24.
Article En | MEDLINE | ID: mdl-34433018

Agonists for glucagon-like-peptide-1 receptor (GLP-1R) are currently used for the treatment of type 2 diabetes and obesity. Their benefits have been centered on pancreas and hypothalamus, but their roles in other organ systems are not well understood. We studied the action of GLP-1R on secretions of adrenal medulla. Exendin-4, a synthetic analog of GLP-1, increases the synthesis and the release of catecholamines (CAs) by increasing cyclic AMP (cAMP) production, without apparent participation of cAMP-regulated guanine nucleotide exchange factor (Epac). Exendin-4, when incubated for 24 h, increases CA synthesis by promoting the activation of tyrosine hydroxylase. Short incubation (20 min) increases the quantum size of exocytotic events by switching exocytosis from partial to full fusion. Our results give a strong support to the role of GLP-1 in the fine control of exocytosis.


Chromaffin Cells/metabolism , Exenatide/pharmacology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/drug effects , Animals , Chromaffin Cells/drug effects , Cyclic AMP/metabolism , Diabetes Mellitus, Type 2/metabolism , Exenatide/metabolism , Exocytosis/drug effects , Exocytosis/physiology , Glucagon-Like Peptide-1 Receptor/metabolism , Guanine Nucleotide Exchange Factors/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Rats , Receptors, Glucagon/drug effects , Receptors, Glucagon/metabolism
17.
Cell Rep ; 36(1): 109318, 2021 07 06.
Article En | MEDLINE | ID: mdl-34233185

The immunological synapse is a complex structure that decodes stimulatory signals into adapted lymphocyte responses. It is a unique window to monitor lymphocyte activity because of development of systematic quantitative approaches. Here we demonstrate the applicability of high-content imaging to human T and natural killer (NK) cells and develop a pipeline for unbiased analysis of high-definition morphological profiles. Our approach reveals how distinct facets of actin cytoskeleton remodeling shape immunological synapse architecture and affect lytic granule positioning. Morphological profiling of CD8+ T cells from immunodeficient individuals allows discrimination of the roles of the ARP2/3 subunit ARPC1B and the ARP2/3 activator Wiskott-Aldrich syndrome protein (WASP) in immunological synapse assembly. Single-cell analysis further identifies uncoupling of lytic granules and F-actin radial distribution in ARPC1B-deficient lymphocytes. Our study provides a foundation for development of morphological profiling as a scalable approach to monitor primary lymphocyte responsiveness and to identify complex aspects of lymphocyte micro-architecture.


Cell Shape , Imaging, Three-Dimensional , Killer Cells, Natural/cytology , T-Lymphocytes/cytology , Actin-Related Protein 2-3 Complex/deficiency , Actin-Related Protein 2-3 Complex/metabolism , Adolescent , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , Cell Line , Cell Shape/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Exocytosis/drug effects , Humans , Immunological Synapses/drug effects , Immunological Synapses/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Male , Organoselenium Compounds/pharmacology , Organosilicon Compounds/pharmacology , Single-Cell Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Thiones/pharmacology , Uracil/analogs & derivatives , Uracil/pharmacology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/metabolism
18.
Neuropharmacology ; 195: 108632, 2021 09 01.
Article En | MEDLINE | ID: mdl-34097947

Cardiovascular side effects of varenicline and a case report of a hypertensive crisis in a varenicline-prescribed patient with pheochromocytoma have been reported. The goal of the present study was to determine whether such side effects might derive, in part, from increased exocytosis of secretory vesicles and subsequent catecholamine release triggered by varenicline in human chromaffin cells of the adrenal gland. In this study, we performed electrophysiological plasma membrane capacitance and carbon fiber amperometry experiments to evaluate the effect of varenicline on exocytosis and catecholamine release, respectively, at concentrations reached during varenicline therapy (100 nM). Experiments were conducted in the absence or presence of nicotine, at plasma concentrations achieved right after smoking (250 nM) or steady-state concentrations (110 nM), in chromaffin cells of the adrenal gland obtained from human organ donors. Cells were stimulated with short pulses (10 ms) of acetylcholine (ACh; 300 µM) applied at 0.2 Hz, in order to closer mimic the physiological situation at the splanchnic nerve-chromaffin cell synapse. In addition, rat chromaffin cells were used to compare the effects obtained in cells from a more readily available species. Varenicline increased the exocytosis of secretory vesicles in human and rat chromaffin cells in the presence of nicotine, effects that were not due to an increase of plasma membrane capacitance or currents triggered by the nicotinic agonists alone. These results should be considered in nicotine addiction therapies when varenicline is used.


Catecholamines/metabolism , Chromaffin Cells/drug effects , Exocytosis/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Varenicline/pharmacology , Acetylcholine/pharmacology , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Animals , Cattle , Chromaffin Cells/metabolism , Humans , Rats
19.
Int J Mol Sci ; 22(9)2021 May 05.
Article En | MEDLINE | ID: mdl-34063076

Platelet function is developmentally regulated. Healthy neonates do not spontaneously bleed, but their platelets are hypo-reactive to several agonists. The mechanisms underlying immature platelet function in neonates are incompletely understood. This critical issue remains challenging for the establishment of age-specific reference ranges. In this study, we evaluated platelet reactivity of five pediatric age categories, ranging from healthy full-term neonates up to adolescents (11-18 years) in comparison to healthy adults (>18 years) by flow cytometry. We confirmed that platelet hypo-reactivity detected by fibrinogen binding, P-selectin, and CD63 surface expression was most pronounced in neonates compared to other pediatric age groups. However, maturation of platelet responsiveness varied with age, agonist, and activation marker. In contrast to TRAP and ADP, collagen-induced platelet activation was nearly absent in neonates. Granule secretion markedly remained impaired at least up to 10 years of age compared to adults. We show for the first time that neonatal platelets are deficient in thrombospondin-1, and exogenous platelet-derived thrombospondin-1 allows platelet responsiveness to collagen. Platelets from all pediatric age groups normally responded to the C-terminal thrombospondin-1 peptide RFYVVMWK. Thus, thrombospondin-1 deficiency of neonatal platelets might contribute to the relatively impaired response to collagen, and platelet-derived thrombospondin-1 may control distinct collagen-induced platelet responses.


Aging/physiology , Blood Platelets/metabolism , Collagen/pharmacology , Thrombospondin 1/pharmacology , Adenosine Diphosphate/pharmacology , Adolescent , Adult , Blood Platelets/drug effects , Child , Crotalid Venoms/pharmacology , Exocytosis/drug effects , Humans , Infant , Infant, Newborn , Lectins, C-Type , Peptides/pharmacology , Platelet Activation/drug effects , Receptors, Proteinase-Activated/metabolism , Thrombospondin 1/chemistry
20.
Adv Biol Regul ; 80: 100809, 2021 05.
Article En | MEDLINE | ID: mdl-33932728

Non-coding RNAs (ncRNAs) play important and diverse roles in mammalian cell biology and pathology. Although the functions of an increasing number of ncRNAs have been identified, the mechanisms underlying ncRNA gene expression remain elusive and are incompletely understood. Here, we investigated ncRNA gene expression in Michigan cancer foundation 7 (MCF7), a malignant breast cancer cell line, on treatment of tetraarsenic oxide (TAO), a potential anti-cancer drug. Our genomic analyses found that TAO up- or down-regulated ncRNA genes genome-wide. A subset of identified ncRNAs with critical biological and clinical functions were validated by real-time quantitative polymerase chain reaction. Intriguingly, these TAO-regulated genes included CDKN2B-AS, HOXA11-AS, SHH, and DUSP5 that are known to interact with or be targeted by polycomb repressive complexes (PRCs). In addition, the PRC subunits were enriched in these TAO-regulated ncRNA genes and TAO treatment deregulated the expression of PRC subunits. Strikingly, TAO decreased the cellular and gene-specific levels of EZH2 expression and H3K27me3. In particular, TAO reduced EZH2 and H3K27me3 and increased transcription at MALAT1 gene. Inhibiting the catalytic activity of EZH2 using GSK343 increased representative TAO-inducible ncRNA genes. Together, our findings suggest that the expression of a subset of ncRNA genes is regulated by PRC2 and that TAO could be a potent epigenetic regulator through PRCs to modulate the ncRNA gene expression in MCF7 cells.


Antineoplastic Agents/pharmacology , Arsenic Trioxide/pharmacology , Histones/genetics , Polycomb-Group Proteins/genetics , RNA, Untranslated/genetics , Transcriptome , Autophagy/drug effects , Autophagy/genetics , Cell Cycle/drug effects , Cell Cycle/genetics , Computational Biology/methods , DNA Repair/drug effects , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Exocytosis/drug effects , Gene Expression Regulation, Neoplastic , Gene Ontology , Genome, Human , HEK293 Cells , Histones/metabolism , Humans , MCF-7 Cells , Molecular Sequence Annotation , Polycomb-Group Proteins/classification , Polycomb-Group Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Untranslated/classification , RNA, Untranslated/metabolism
...