Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 386
Filter
1.
Eur J Pharmacol ; 972: 176589, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38631503

ABSTRACT

We explored the vasorelaxant effects of ipragliflozin, a sodium-glucose cotransporter-2 inhibitor, on rabbit femoral arterial rings. Ipragliflozin relaxed phenylephrine-induced pre-contracted rings in a dose-dependent manner. Pre-treatment with the ATP-sensitive K+ channel inhibitor glibenclamide (10 µM), the inwardly rectifying K+ channel inhibitor Ba2+ (50 µM), or the Ca2+-sensitive K+ channel inhibitor paxilline (10 µM) did not influence the vasorelaxant effect. However, the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine (3 mM) reduced the vasorelaxant effect. Specifically, the vasorelaxant response to ipragliflozin was significantly attenuated by pretreatment with the Kv7.X channel inhibitors linopirdine (10 µM) and XE991 (10 µM), the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin (1 µM) and cyclopiazonic acid (10 µM), and the cAMP/protein kinase A (PKA)-associated signaling pathway inhibitors SQ22536 (50 µM) and KT5720 (1 µM). Neither the cGMP/protein kinase G (PKG)-associated signaling pathway nor the endothelium was involved in ipragliflozin-induced vasorelaxation. We conclude that ipragliflozin induced vasorelaxation of rabbit femoral arteries by activating Kv channels (principally the Kv7.X channel), the SERCA pump, and the cAMP/PKA-associated signaling pathway independent of other K+ (ATP-sensitive K+, inwardly rectifying K+, and Ca2+-sensitive K+) channels, cGMP/PKG-associated signaling, and the endothelium.


Subject(s)
Cyclic AMP-Dependent Protein Kinases , Femoral Artery , Glucosides , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Signal Transduction , Thiophenes , Vasodilation , Animals , Rabbits , Femoral Artery/drug effects , Femoral Artery/physiology , Vasodilation/drug effects , Signal Transduction/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Thiophenes/pharmacology , Male , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Vasodilator Agents/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors
2.
J Biol Chem ; 299(5): 104681, 2023 05.
Article in English | MEDLINE | ID: mdl-37030504

ABSTRACT

We report a novel small-molecule screening approach that combines data augmentation and machine learning to identify Food and Drug Administration (FDA)-approved drugs interacting with the calcium pump (Sarcoplasmic reticulum Ca2+-ATPase, SERCA) from skeletal (SERCA1a) and cardiac (SERCA2a) muscle. This approach uses information about small-molecule effectors to map and probe the chemical space of pharmacological targets, thus allowing to screen with high precision large databases of small molecules, including approved and investigational drugs. We chose SERCA because it plays a major role in the excitation-contraction-relaxation cycle in muscle and it represents a major target in both skeletal and cardiac muscle. The machine learning model predicted that SERCA1a and SERCA2a are pharmacological targets for seven statins, a group of FDA-approved 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors used in the clinic as lipid-lowering medications. We validated the machine learning predictions by using in vitro ATPase assays to show that several FDA-approved statins are partial inhibitors of SERCA1a and SERCA2a. Complementary atomistic simulations predict that these drugs bind to two different allosteric sites of the pump. Our findings suggest that SERCA-mediated Ca2+ transport may be targeted by some statins (e.g., atorvastatin), thus providing a molecular pathway to explain statin-associated toxicity reported in the literature. These studies show the applicability of data augmentation and machine learning-based screening as a general platform for the identification of off-target interactions and the applicability of this approach extends to drug discovery.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/metabolism , Myocardium/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Machine Learning
3.
Nat Commun ; 13(1): 4407, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906205

ABSTRACT

The detailed analysis and structural characterization of proteoforms by top-down proteomics (TDP) has gained a lot of interest in biomedical research. Data-dependent acquisition (DDA) of intact proteins is non-trivial due to the diversity and complexity of proteoforms. Dedicated acquisition methods thus have the potential to greatly improve TDP. Here, we present FLASHIda, an intelligent online data acquisition algorithm for TDP that ensures the real-time selection of high-quality precursors of diverse proteoforms. FLASHIda combines fast charge deconvolution algorithms and machine learning-based quality assessment for optimal precursor selection. In an analysis of E. coli lysate, FLASHIda increases the number of unique proteoform level identifications from 800 to 1500 or generates a near-identical number of identifications in one third of the instrument time when compared to standard DDA mode. Furthermore, FLASHIda enables sensitive mapping of post-translational modifications and detection of chemical adducts. As a software extension module to the instrument, FLASHIda can be readily adopted for TDP studies of complex samples to enhance proteoform identification rates.


Subject(s)
Proteome , Proteomics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Escherichia coli/metabolism , Heart , Peptides , Protein Processing, Post-Translational , Proteome/metabolism , Proteomics/methods
4.
Cells ; 11(3)2022 01 27.
Article in English | MEDLINE | ID: mdl-35159243

ABSTRACT

Sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) is a crucial component of the cellular machinery responsible for Ca2+ homeostasis. The selective inhibition of SERCA by thapsigargin (TG) leads to perturbations in Ca2+ signaling, which can trigger endoplasmic reticulum (ER) stress. The unfolded protein response (UPR) pathway is activated in response to ER stress and induces an adaptive response to preserve cell survival or committee cells to programmed death, depending on stress duration and/or level. Early stages of ER stress stimulate mitochondrial metabolism to preserve survival but under chronic ER stress conditions, mitochondrial dysfunction is induced, which, in turn, can enhance inflammation through NLRP3 inflammasome activation. This study was aimed at investigating the role of SERCA inhibition on NLRP3 inflammasome activation in human monocytes, which was evaluated in primary monocytes isolated from healthy individuals and in the THP-1 human monocytic cell line. Findings obtained in both THP-1 and primary monocytes demonstrate that SERCA inhibition triggered by TG does not activate the NLRP3 inflammasome in these innate immune cells since IL-1ß secretion was not affected. Results from THP-1 monocytes showing that SERCA inhibition increases mitochondrial Ca2+ content and fusion, in the absence of changes in ROS levels and membrane potential, support the view that human monocytes counteract ER stress that arises from inhibition of SERCA through modulation of mitochondrial morphology towards mitochondria fusion, thus preventing NLRP3 inflammasome activation. Overall, this work contributes to a better understanding of the molecular mechanisms that modulate the activity of the NLRP3 inflammasome leading to sterile inflammation, which are still poorly understood.


Subject(s)
Inflammasomes , Mitochondrial Dynamics , NLR Family, Pyrin Domain-Containing 3 Protein , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Mitochondria/metabolism , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Thapsigargin/pharmacology
5.
Nat Commun ; 12(1): 6815, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819493

ABSTRACT

Bistable motoneurons of the spinal cord exhibit warmth-activated plateau potential driven by Na+ and triggered by a brief excitation. The thermoregulating molecular mechanisms of bistability and their role in motor functions remain unknown. Here, we identify thermosensitive Na+-permeable Trpm5 channels as the main molecular players for bistability in mouse motoneurons. Pharmacological, genetic or computational inhibition of Trpm5 occlude bistable-related properties (slow afterdepolarization, windup, plateau potentials) and reduce spinal locomotor outputs while central pattern generators for locomotion operate normally. At cellular level, Trpm5 is activated by a ryanodine-mediated Ca2+ release and turned off by Ca2+ reuptake through the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump. Mice in which Trpm5 is genetically silenced in most lumbar motoneurons develop hindlimb paresis and show difficulties in executing high-demanding locomotor tasks. Overall, by encoding bistability in motoneurons, Trpm5 appears indispensable for producing a postural tone in hindlimbs and amplifying the locomotor output.


Subject(s)
Locomotion/physiology , Motor Neurons/metabolism , Paresis/physiopathology , Spinal Cord/physiology , TRPM Cation Channels/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Animals, Newborn , Computer Simulation , Disease Models, Animal , Female , Gene Silencing , HEK293 Cells , Hindlimb/physiology , Humans , Locomotion/drug effects , Male , Mice , Motor Neurons/drug effects , Paresis/genetics , Patch-Clamp Techniques , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ryanodine/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Spinal Cord/cytology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics
6.
Eur J Med Chem ; 224: 113676, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34256125

ABSTRACT

A series of eleven celastrol derivatives was designed, synthesized, and evaluated for their in vitro cytotoxic activities against six human cancer cell lines (A549, HepG2, HepAD38, PC3, DLD-1 Bax-Bak WT and DKO) and three human normal cells (LO2, BEAS-2B, CCD19Lu). To our knowledge, six derivatives were the first example of dipeptide celastrol derivatives. Among them, compound 3 was the most promising derivative, as it exhibited a remarkable anti-proliferative activity and improved selectivity in liver cancer HepAD38 versus human normal hepatocytes, LO2. Compound 6 showed higher selectivity in liver cancer cells against human normal lung fibroblasts, CCD19Lu cell line. The Ca2+ mobilizations of 3 and 6 were also evaluated in the presence and absence of thapsigargin to demonstrate their inhibitory effects on SERCA. Derivatives 3 and 6 were found to induce apoptosis on LO2, HepG2 and HepAD38 cells. The potential docking poses of all synthesized celastrol dipeptides and other known inhibitors were proposed by molecular docking. Finally, 3 inhibited P-gp-mediated drug efflux with greater efficiency than inhibitor verapamil in A549 lung cancer cells. Therefore, celastrol-dipeptide derivatives are potent drug candidates for the treatment of drug-resistant cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Molecular Docking Simulation , Pentacyclic Triterpenes/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , Pentacyclic Triterpenes/metabolism , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/therapeutic use , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Structure-Activity Relationship
7.
Mech Ageing Dev ; 196: 111474, 2021 06.
Article in English | MEDLINE | ID: mdl-33766744

ABSTRACT

We have reported recently that submaximal inhibition of the Sarco Endoplasmic Reticulum Ca2+ ATPase (SERCA) produces an increase in the lifespan of C. elegans worms. We have explored here the mechanism of this increased survival by studying the effect of SERCA inhibition in several mutants of signaling pathways related to longevity. Our data show that the mechanism of the effect is unrelated with the insulin signaling pathway or the sirtuin activity, because SERCA inhibitors increased lifespan similarly in mutants of these pathways. However, the effect required functional mitochondria and both the AMP kinase and TOR pathways, as the SERCA inhibitors were ineffective in the corresponding mutants. The same effects were obtained after reducing SERCA expression with submaximal RNAi treatment. The SERCA inhibitors did not induce ER-stress at the concentrations used, and their effect was not modified by inactivation of the OP50 bacterial food. Altogether, our data suggest that the effect may be due to a reduced ER-mitochondria Ca2+ transfer acting via AMPK activation and mTOR inhibition to promote survival.


Subject(s)
Endoplasmic Reticulum , Insulin/metabolism , Longevity , Mitochondria/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Thapsigargin/pharmacology , AMP-Activated Protein Kinases , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/genetics , Longevity/drug effects , Longevity/physiology , RNAi Therapeutics/methods , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sirtuins/metabolism , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome
8.
Sci Rep ; 11(1): 2846, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531589

ABSTRACT

Skeletal muscle Na+ channels possess Ca2+- and calmodulin-binding sites implicated in Nav1.4 current (INa) downregulation following ryanodine receptor (RyR1) activation produced by exchange protein directly activated by cyclic AMP or caffeine challenge, effects abrogated by the RyR1-antagonist dantrolene which itself increased INa. These findings were attributed to actions of consequently altered cytosolic Ca2+, [Ca2+]i, on Nav1.4. We extend the latter hypothesis employing cyclopiazonic acid (CPA) challenge, which similarly increases [Ca2+]i, but through contrastingly inhibiting sarcoplasmic reticular (SR) Ca2+-ATPase. Loose patch clamping determined Na+ current (INa) families in intact native murine gastrocnemius skeletal myocytes, minimising artefactual [Ca2+]i perturbations. A bespoke flow system permitted continuous INa comparisons through graded depolarizing steps in identical stable membrane patches before and following solution change. In contrast to the previous studies modifying RyR1 activity, and imposing control solution changes, CPA (0.1 and 1 µM) produced persistent increases in INa within 1-4 min of introduction. CPA pre-treatment additionally abrogated previously reported reductions in INa produced by 0.5 mM caffeine. Plots of peak current against voltage excursion demonstrated that 1 µM CPA increased maximum INa by ~ 30%. It only slightly decreased half-maximal activating voltages (V0.5) and steepness factors (k), by 2 mV and 0.7, in contrast to the V0.5 and k shifts reported with direct RyR1 modification. These paradoxical findings complement previously reported downregulatory effects on Nav1.4 of RyR1-agonist mediated increases in bulk cytosolic [Ca2+]. They implicate possible local tubule-sarcoplasmic triadic domains containing reduced [Ca2+]TSR in the observed upregulation of Nav1.4 function following CPA-induced SR Ca2+ depletion.


Subject(s)
Muscle, Skeletal/metabolism , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Caffeine/pharmacology , Calcium Channel Agonists/pharmacology , Calcium Signaling/drug effects , Cells, Cultured , Indoles/pharmacology , Mice , Muscle Fibers, Skeletal , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Patch-Clamp Techniques , Primary Cell Culture , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sodium/metabolism , Up-Regulation/drug effects
9.
J Hematol Oncol ; 14(1): 8, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33407740

ABSTRACT

P-type ATPase inhibitors are among the most successful and widely prescribed therapeutics in modern pharmacology. Clinical transition has been safely achieved for H+/K+ ATPase inhibitors such as omeprazole and Na+/K+-ATPase inhibitors like digoxin. However, this is more challenging for Ca2+-ATPase modulators due to the physiological role of Ca2+ in cardiac dynamics. Over the past two decades, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) modulators have been studied as potential chemotherapy agents because of their Ca2+-mediated pan-cancer lethal effects. Instead, recent evidence suggests that SERCA inhibition suppresses oncogenic Notch1 signaling emerging as an alternative to γ-secretase modulators that showed limited clinical activity due to severe side effects. In this review, we focus on how SERCA inhibitors alter Notch1 signaling and show that Notch on-target-mediated antileukemia properties of these molecules can be achieved without causing overt Ca2+ cellular overload.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Receptors, Notch/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Notch/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/analogs & derivatives , Thapsigargin/pharmacology , Thapsigargin/therapeutic use
10.
J Biol Chem ; 296: 100310, 2021.
Article in English | MEDLINE | ID: mdl-33482198

ABSTRACT

The endoplasmic reticulum (ER) contains various enzymes that metabolize fatty acids (FAs). Given that FAs are the components of membranes, FA metabolic enzymes might be associated with regulation of ER membrane functions. However, it remains unclear whether there is the interplay between FA metabolic enzymes and ER membrane proteins. Trans-2-enoyl-CoA reductase (TER) is an FA reductase present in the ER membrane and catalyzes the last step in the FA elongation cycle and sphingosine degradation pathway. Here we identify sarco(endo)plasmic reticulum Ca2+-ATPase 2b (SERCA2b), an ER Ca2+ pump responsible for Ca2+ accumulation in the ER, as a TER-binding protein by affinity purification from HEK293 cell lysates. We show that TER directly binds to SERCA2b by in vitro assays using recombinant proteins. Thapsigargin, a specific SERCA inhibitor, inhibits this binding. TER binds to SERCA2b through its conserved C-terminal region. TER overexpression suppresses SERCA2b ATPase activity in microsomal membranes of HEK293 cells. Depletion of TER increases Ca2+ storage in the ER and accelerates SERCA2b-dependent Ca2+ uptake to the ER after ligand-induced Ca2+ release. Moreover, depletion of TER reduces the Ca2+-dependent nuclear translocation of nuclear factor of activated T cells 4. These results demonstrate that TER is a negative regulator of SERCA2b, implying the direct linkage of FA metabolism and Ca2+ accumulation in the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Fatty Acids/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Active Transport, Cell Nucleus/genetics , Calcium/metabolism , Calcium Signaling/genetics , Endoplasmic Reticulum/genetics , Enzyme Inhibitors/pharmacology , Fatty Acids/genetics , Gene Expression Regulation, Enzymologic/drug effects , HEK293 Cells , Humans , Ligands , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Protein Binding/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry
11.
Arch Biochem Biophys ; 699: 108764, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33460582

ABSTRACT

The interaction of a dirhamnolipid biosurfactant secreted by Pseudomonas aeruginosa with calcium ATPase from sarcoplasmic reticulum (SR) was studied by means of different approaches, such as enzyme activity, fluorescence spectroscopy, Fourier transform infrared spectroscopy (FTIR), differential scanning calorimetry (DSC), and molecular docking simulations. The ATP hydrolysis activity was fully inhibited by incubation with dirhamnolipid (diRL) up to 0.1 mM concentration, corresponding to a surfactant concentration below membrane solubilization threshold. Surfactant-protein interaction induced conformational changes in the protein observed by an increase in the accessibility of tryptophan residues to the aqueous phase and by changes in the secondary structure of the protein as seen by fluorescence and FTIR spectroscopy. As a consequence, the protein become more unstable and denatured at lower temperatures, as seen by enzyme activity and DSC studies. Finally, these results were explained at molecular level throughout molecular docking simulations. It is concluded that there is a specific dirhamnolipid-protein interaction not related to the surface activity of the surfactant but to the particular physicochemical properties of the biosurfactant molecule.


Subject(s)
Glycolipids/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Surface-Active Agents/metabolism , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glycolipids/chemistry , Molecular Docking Simulation , Protein Binding , Protein Structure, Secondary/drug effects , Pseudomonas aeruginosa/chemistry , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Surface-Active Agents/chemistry
12.
Pflugers Arch ; 473(2): 273-286, 2021 02.
Article in English | MEDLINE | ID: mdl-33108514

ABSTRACT

Pre-blockade of the sarco-endoplasmic reticulum (ER) calcium ATPase (SERCA) with irreversible thapsigargin depresses exocytosis in adrenal bovine chromaffin cells (BCCs). Distinct expression of voltage-dependent Ca2+-channel subtypes and of the Ca2+-induced Ca2+ release (CICR) mechanism in BCCs versus mouse chromaffin cells (MCCs) has been described. We present a parallel study on the effects of the acute SERCA blockade with reversible cyclopizonic acid (CPA), to repeated pulsing with acetylcholine (ACh) at short (15 s) and long intervals (60 s) at 37 °C, allowing the monitoring of the initial size of a ready-release vesicle pool (RRP) and its depletion and recovery in subsequent stimuli. We found (i) strong depression of exocytosis upon ACh pulsing at 15-s intervals and slower depression at 60-s intervals in both cell types; (ii) facilitation of exocytosis upon acute SERCA inhibition, with back to depression upon CPA washout in MCCs; (iii) blockade of exocytosis upon acute SERCA inhibition and pronounced rebound of exocytosis upon CPA washout in BCCs; (iv) basal [Ca2+]c elevation upon stimulation with ACh at short intervals (but not at long intervals) in both cell types; and (v) augmentation of basal [Ca2+]c and inhibition of peak [Ca2+]c amplitude upon CPA treatment in both cell types, with milder effects upon stimulation at 60-s intervals. These results are compatible with the view that while in MCCs the uptake of Ca2+ via SERCA contributes to the mitigation of physiological ACh triggered secretion, in BCCs the uptake of Ca2+ into the ER facilitates such responses likely potentiating a Ca2+-induced Ca2+ release mechanism. These drastic differences in the regulation of ACh-triggered secretion at 37 °C may help to understand different patterns of the regulation of exocytosis by the circulation of Ca2+ at a functional ER Ca2+ store.


Subject(s)
Calcium/metabolism , Catecholamines/metabolism , Chromaffin Cells/drug effects , Endoplasmic Reticulum/drug effects , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Acetylcholine/pharmacology , Animals , Calcium Signaling/drug effects , Cattle , Cells, Cultured , Chromaffin Cells/enzymology , Endoplasmic Reticulum/enzymology , Indoles/pharmacology , Male , Mice, Inbred C57BL , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Species Specificity , Thapsigargin/pharmacology , Time Factors
13.
Biomolecules ; 10(12)2020 12 05.
Article in English | MEDLINE | ID: mdl-33291419

ABSTRACT

In spite of the impressing cytotoxicity of thapsigargin (Tg), this compound cannot be used as a chemotherapeutic drug because of general toxicity, causing unacceptable side effects. Instead, a prodrug targeted towards tumors, mipsagargin, was brought into clinical trials. What substantially reduces the clinical potential is the limited access to Tg and its derivatives and cost-inefficient syntheses with unacceptably low yields. Laser trilobum, which contains a structurally related sesquiterpene lactone, trilobolide (Tb), is successfully cultivated. Here, we report scalable isolation of Tb from L. trilobum and a transformation of Tb to 8-O-(12-aminododecanoyl)-8-O-debutanoylthapsigargin in seven steps. The use of cultivated L. trilobum offers an unlimited source of the active principle in mipsagargin.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Apiaceae/chemistry , Butyrates/chemistry , Chemistry Techniques, Synthetic , Furans/chemistry , Thapsigargin/analogs & derivatives , Antineoplastic Agents, Phytogenic/isolation & purification , Apiaceae/metabolism , Butyrates/isolation & purification , Carbon Dioxide/chemistry , Chromatography, Supercritical Fluid/methods , Fruit/chemistry , Fruit/metabolism , Furans/isolation & purification , Humans , Molecular Structure , Neoplasms/drug therapy , Neoplasms/pathology , Plant Extracts/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Thapsigargin/isolation & purification
14.
Mol Cell ; 80(4): 592-606.e8, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33159855

ABSTRACT

Despite its outstanding clinical success, immune checkpoint blockade remains ineffective in many patients. Accordingly, combination therapy capable of achieving greater antitumor immunity is urgently required. Here, we report that limiting glutamine metabolism in cancer cells bolsters the effectiveness of anti-programmed death ligand-1 (PD-L1) antibody. Inhibition of glutamine utilization increased PD-L1 levels in cancer cells, thereby inactivating co-cultured T cells. Under glutamine-limited conditions, reduced cellular GSH levels caused an upregulation of PD-L1 expression by impairing SERCA activity, which activates the calcium/NF-κB signaling cascade. Consequently, in tumors grown in immunocompetent mice, inhibition of glutamine metabolism decreased the antitumor activity of T cells. In combination with anti-PD-L1, however, glutamine depletion strongly promoted the antitumor efficacy of T cells in vitro and in vivo due to simultaneous increases in Fas/CD95 levels. Our results demonstrate the relevance of cancer glutamine metabolism to antitumor immunity and suggest that co-targeting of glutamine metabolism and PD-L1 represents a promising therapeutic approach.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/metabolism , Glutamine/metabolism , Glutathione/metabolism , Neoplasms/immunology , Neoplasms/prevention & control , T-Lymphocytes/immunology , Aged , Animals , Apoptosis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Proliferation , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
J Biol Chem ; 295(49): 16655-16664, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32972972

ABSTRACT

Viral infection is one environmental factor that may contribute to the initiation of pancreatic ß-cell destruction during the development of autoimmune diabetes. Picornaviruses, such as encephalomyocarditis virus (EMCV), induce a pro-inflammatory response in islets leading to local production of cytokines, such as IL-1, by resident islet leukocytes. Furthermore, IL-1 is known to stimulate ß-cell expression of iNOS and production of the free radical nitric oxide. The purpose of this study was to determine whether nitric oxide contributes to the ß-cell response to viral infection. We show that nitric oxide protects ß-cells against virally mediated lysis by limiting EMCV replication. This protection requires low micromolar, or iNOS-derived, levels of nitric oxide. At these concentrations nitric oxide inhibits the Krebs enzyme aconitase and complex IV of the electron transport chain. Like nitric oxide, pharmacological inhibition of mitochondrial oxidative metabolism attenuates EMCV-mediated ß-cell lysis by inhibiting viral replication. These findings provide novel evidence that cytokine signaling in ß-cells functions to limit viral replication and subsequent ß-cell lysis by attenuating mitochondrial oxidative metabolism in a nitric oxide-dependent manner.


Subject(s)
Encephalomyocarditis virus/physiology , Insulin-Secreting Cells/metabolism , Mitochondria/metabolism , Oxidative Stress , Animals , Apoptosis/drug effects , Cells, Cultured , Female , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/virology , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mice , Mice, Inbred C57BL , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Oxidative Stress/drug effects , Poly I-C/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Unfolded Protein Response/drug effects , Up-Regulation/drug effects , Virus Replication
16.
Viruses ; 12(10)2020 09 27.
Article in English | MEDLINE | ID: mdl-32992478

ABSTRACT

Influenza A virus is a major global pathogen of humans, and there is an unmet need for effective antivirals. Current antivirals against influenza A virus directly target the virus and are vulnerable to mutational resistance. Harnessing an effective host antiviral response is an attractive alternative. We show that brief exposure to low, non-toxic doses of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, promptly elicits an extended antiviral state that dramatically blocks influenza A virus production. Crucially, oral administration of TG protected mice against lethal virus infection and reduced virus titres in the lungs of treated mice. TG-induced ER stress unfolded protein response appears as a key driver responsible for activating a spectrum of host antiviral defences that include an enhanced type I/III interferon response. Our findings suggest that TG is potentially a viable host-centric antiviral for the treatment of influenza A virus infection without the inherent problem of drug resistance.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N8 Subtype/growth & development , Thapsigargin/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , Dogs , Endoplasmic Reticulum Stress/drug effects , Female , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Influenza, Human/drug therapy , Interferon Type I/drug effects , Interferon Type I/immunology , Interferons/drug effects , Interferons/immunology , Mice , Mice, Inbred BALB C , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Swine , Unfolded Protein Response/drug effects , Vero Cells , Interferon Lambda
17.
Arch Biochem Biophys ; 693: 108542, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32818508

ABSTRACT

Spontaneous Ca2+-transient (wave) generation in isolated cardiomyocytes is well established phenomenon which poses a lot of questions about myocardial excitability. Current studies of spontaneous Ca2+-activity in cardiac cells mainly relate to the kinetic characteristics, classification and simulation of Ca2+-events through ryanodine receptor (RyR) activity modeling. Here, for the first time we pay attention to the Ca2+-transients having stationary kinetics for correct estimation of the sarcoplasmic reticulum Ca2+ transport. In cardiomyocytes generating such type of Ca2+-transients, the averaged intracellular calcium ([Ca2+]in) fluorescence practically does not change in time. Stationary Ca2+-transients are observed in different animal models (Wistar, SHR, ground squirrels) revealing a common cardiomyocyte phenomenon. They somewhat depend on external Ca2+ ([Ca2+]ex) because the [Ca2+]ex lowering to 1 µM in the presence of EGTA disrupts Ca2+-wave propagation. At the same time, spontaneous Ca2+-transients do not associated with the forward or reverse mode of Na+/Ca2+ exchanger (NCX), but partially modulated by the L-type Ca2+-channels. Among the sarcoplasmic reticulum targets, RyR and SERCA are crucial for Ca2+-wave generation and sustained self-oscillation activity. Analysis of the spontaneous wave kinetics reveals that both slopes of the rising wave front and the wave front decline are gradually changed during propagation, which well correlates with the RyR and SERCA activity, respectively. On the contrary, in the electrical field-stimulated myocytes, both slope factors are sharply changed corresponding to 'all-or-nothing' rule, which is fundamental principle for action potential in cardiomyocytes. Furthermore, stimulation of single cardiomyocyte using local electrode appears the deterioration in the [Ca2+]in utilization from the cytosol, which limits the time of observation during the protocol. Obtained data suggest that stationary spontaneous Ca2+-transients occurring without actual myocellular excitation represent useful and precise tools for estimation of the sarcoplasmic reticulum Ca2+-transport.


Subject(s)
Calcium/metabolism , Myocytes, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Evoked Potentials , Ion Transport , Rats , Rats, Inbred SHR , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
18.
Sci Rep ; 10(1): 12320, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704072

ABSTRACT

The intrinsic lymphatic contractile activity is necessary for proper lymph transport. Mesenteric lymphatic vessels from high-fructose diet-induced metabolic syndrome (MetSyn) rats exhibited impairments in its intrinsic phasic contractile activity; however, the molecular mechanisms responsible for the weaker lymphatic pumping activity in MetSyn conditions are unknown. Several metabolic disease models have shown that dysregulation of sarcoplasmic reticulum Ca2+ ATPase (SERCA) pump is one of the key determinants of the phenotypes seen in various muscle tissues. Hence, we hypothesized that a decrease in SERCA pump expression and/or activity in lymphatic muscle influences the diminished lymphatic vessel contractions in MetSyn animals. Results demonstrated that SERCA inhibitor, thapsigargin, significantly reduced lymphatic phasic contractile frequency and amplitude in control vessels, whereas, the reduced MetSyn lymphatic contractile activity was not further diminished by thapsigargin. While SERCA2a expression was significantly decreased in MetSyn lymphatic vessels, myosin light chain 20, MLC20 phosphorylation was increased in these vessels. Additionally, insulin resistant lymphatic muscle cells exhibited elevated intracellular calcium and decreased SERCA2a expression and activity. The SERCA activator, CDN 1163 partially restored lymphatic contractile activity in MetSyn lymphatic vessel by increasing phasic contractile frequency. Thus, our data provide the first evidence that SERCA2a modulates the lymphatic pumping activity by regulating phasic contractile amplitude and frequency, but not the lymphatic tone. Diminished lymphatic contractile activity in the vessels from the MetSyn animal is associated with the decreased SERCA2a expression and impaired SERCA2 activity in lymphatic muscle.


Subject(s)
Lymphatic Vessels/pathology , Metabolic Syndrome/enzymology , Muscle Contraction , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Adipose Tissue/drug effects , Aminoquinolines/pharmacology , Animals , Benzamides/pharmacology , Body Composition/drug effects , Cardiomegaly/pathology , Diet , Disease Models, Animal , Feeding Behavior/drug effects , Insulin Resistance , Lymphatic Vessels/drug effects , Male , Models, Biological , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Phosphorylation/drug effects , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Thapsigargin/pharmacology
19.
Int J Mol Sci ; 21(11)2020 Jun 10.
Article in English | MEDLINE | ID: mdl-32532023

ABSTRACT

Sarcoendoplasmic reticulum calcium ATPase (SERCA), a member of the P-type ATPase family of ion and lipid pumps, is responsible for the active transport of Ca2+ from the cytoplasm into the sarcoplasmic reticulum lumen of muscle cells, into the endoplasmic reticulum (ER) of non-muscle cells. X-ray crystallography has proven to be an invaluable tool in understanding the structural changes of SERCA, and more than 70 SERCA crystal structures representing major biochemical states (defined by bound ligand) have been deposited in the Protein Data Bank. Consequently, SERCA is one of the best characterized components of the calcium transport machinery in the cell. Emerging approaches in the field, including spectroscopy and molecular simulation, now help integrate and interpret this rich structural information to understand the conformational transitions of SERCA that occur during activation, inhibition, and regulation. In this review, we provide an overview of the crystal structures of SERCA, focusing on identifying metrics that facilitate structure-based categorization of major steps along the catalytic cycle. We examine the integration of crystallographic data with different biophysical approaches and computational methods to link biochemical and structural states of SERCA that are populated in the cell. Finally, we discuss the challenges and new opportunities in the field, including structural elucidation of functionally important and novel regulatory complexes of SERCA, understanding the structural basis of functional divergence among homologous SERCA regulators, and bridging the gap between basic and translational research directed toward therapeutic modulation of SERCA.


Subject(s)
Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Humans , Phosphorylation , Protein Conformation , Protein Domains , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Thapsigargin/chemistry , Thapsigargin/metabolism
20.
Sci Rep ; 10(1): 8348, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32433555

ABSTRACT

To date current therapies of glioblastoma multiforme (GBM) are largely ineffective. The induction of apoptosis by an unresolvable unfolded protein response (UPR) represents a potential new therapeutic strategy. Here we tested 12ADT, a sarcoendoplasmic reticulum Ca2+ ATPase (SERCA) inhibitor, on a panel of unselected patient-derived neurosphere-forming cells and found that GBM cells can be distinguished into "responder" and "non-responder". By RNASeq analysis we found that the non-responder phenotype is significantly linked with the expression of UPR genes, and in particular ERN1 (IRE1) and ATF4. We also identified two additional genes selectively overexpressed among non-responders, IGFBP3 and IGFBP5. CRISPR-mediated deletion of the ERN1, IGFBP3, IGFBP5 signature genes in the U251 human GBM cell line increased responsiveness to 12ADT. Remarkably, >65% of GBM cases in The Cancer Genome Atlas express the non-responder (ERN1, IGFBP3, IGFBP5) gene signature. Thus, elevated levels of IRE1α and IGFBPs predict a poor response to drugs inducing unresolvable UPR and possibly other forms of chemotherapy helping in a better stratification GBM patients.


Subject(s)
Brain Neoplasms/drug therapy , Endoribonucleases/metabolism , Glioblastoma/drug therapy , Protein Serine-Threonine Kinases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Thapsigargin/pharmacology , Adult , Apoptosis/drug effects , Brain/pathology , Brain/surgery , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/surgery , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/surgery , Humans , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Primary Cell Culture , Progression-Free Survival , Protein Serine-Threonine Kinases/genetics , RNA-Seq , Signal Transduction/genetics , Spheroids, Cellular , Thapsigargin/analogs & derivatives , Thapsigargin/therapeutic use , Tumor Cells, Cultured , Unfolded Protein Response/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...