Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.795
Filter
1.
Nature ; 628(8009): 910-918, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38570680

ABSTRACT

OSCA/TMEM63 channels are the largest known family of mechanosensitive channels1-3, playing critical roles in plant4-7 and mammalian8,9 mechanotransduction. Here we determined 44 cryogenic electron microscopy structures of OSCA/TMEM63 channels in different environments to investigate the molecular basis of OSCA/TMEM63 channel mechanosensitivity. In nanodiscs, we mimicked increased membrane tension and observed a dilated pore with membrane access in one of the OSCA1.2 subunits. In liposomes, we captured the fully open structure of OSCA1.2 in the inside-in orientation, in which the pore shows a large lateral opening to the membrane. Unusually for ion channels, structural, functional and computational evidence supports the existence of a 'proteo-lipidic pore' in which lipids act as a wall of the ion permeation pathway. In the less tension-sensitive homologue OSCA3.1, we identified an 'interlocking' lipid tightly bound in the central cleft, keeping the channel closed. Mutation of the lipid-coordinating residues induced OSCA3.1 activation, revealing a conserved open conformation of OSCA channels. Our structures provide a global picture of the OSCA channel gating cycle, uncover the importance of bound lipids and show that each subunit can open independently. This expands both our understanding of channel-mediated mechanotransduction and channel pore formation, with important mechanistic implications for the TMEM16 and TMC protein families.


Subject(s)
Calcium Channels , Cryoelectron Microscopy , Ion Channel Gating , Mechanotransduction, Cellular , Humans , Anoctamins/chemistry , Anoctamins/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium Channels/ultrastructure , Lipids/chemistry , Liposomes/metabolism , Liposomes/chemistry , Models, Molecular , Nanostructures/chemistry
2.
J Chem Inf Model ; 63(21): 6768-6777, 2023 11 13.
Article in English | MEDLINE | ID: mdl-37871325

ABSTRACT

Transient receptor potential vanilloid 5 (TRPV5) is a calcium-selective TRP channel that plays a crucial role in calcium homeostasis regulation. However, there are still many issues that need to be addressed, such as the specific conformational transition of TRPV5 and the specific functions of each structure in cation gating. Here, we build a model of the calcium ion transport protein from Xenopus oocytes in the presence of the lipid membrane and water molecules. Due to the activation process of ion channels are global and collective, coarse-grained molecular dynamics (CG-MD) simulations of the potential of mean force along the conformational transition pathway are performed. The CG-MD simulations show that the S6 helix plays a vital role in the TRPV5 conformational transition. Most importantly, these simulated trajectories indicate that the activation of ion channels happens before the extension and rotation of S6 helices, revealing that TRPV5 has a unique gating mechanism different from TRPV6. The present work demonstrates how the mechanical force acting on the S6 helix opens the TRPV5 channel gates. These results deepen our understanding of the TRPV5 gating mechanism.


Subject(s)
Calcium , Molecular Dynamics Simulation , Calcium/metabolism , Molecular Conformation , Calcium Channels/chemistry , TRPV Cation Channels
4.
Angew Chem Int Ed Engl ; 62(29): e202302812, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37148162

ABSTRACT

Ziconotide (ω-conotoxin MVIIA) is an approved analgesic for the treatment of chronic pain. However, the need for intrathecal administration and adverse effects have limited its widespread application. Backbone cyclization is one way to improve the pharmaceutical properties of conopeptides, but so far chemical synthesis alone has been unable to produce correctly folded and backbone cyclic analogues of MVIIA. In this study, an asparaginyl endopeptidase (AEP)-mediated cyclization was used to generate backbone cyclic analogues of MVIIA for the first time. Cyclization using six- to nine-residue linkers did not perturb the overall structure of MVIIA, and the cyclic analogues of MVIIA showed inhibition of voltage-gated calcium channels (CaV 2.2) and substantially improved stability in human serum and stimulated intestinal fluid. Our study reveals that AEP transpeptidases are capable of cyclizing structurally complex peptides that chemical synthesis cannot achieve and paves the way for further improving the therapeutic value of conotoxins.


Subject(s)
Conotoxins , omega-Conotoxins , Humans , omega-Conotoxins/pharmacology , omega-Conotoxins/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use , Conotoxins/pharmacology , Calcium Channels/chemistry , Calcium Channel Blockers/pharmacology
5.
Channels (Austin) ; 17(1): 2191773, 2023 12.
Article in English | MEDLINE | ID: mdl-37075164

ABSTRACT

One of nature's exceptions was discovered when a Cav3 T-type channel was observed to switch phenotype from a calcium channel into a sodium channel by neutralizing an aspartate residue in the high field strength (HFS) +1 position within the ion selectivity filter. The HFS+1 site is dubbed a "beacon" for its location at the entryway just above the constricted, minimum radius of the HFS site's electronegative ring. A classification is proposed based on the occupancy of the HFS+1 "beacon" which correlates with the calcium- or sodium-selectivity phenotype. If the beacon is a glycine, or neutral, non-glycine residue, then the cation channel is calcium-selective or sodium-permeable, respectively (Class I). Occupancy of a beacon aspartate are calcium-selective channels (Class II) or possessing a strong calcium block (Class III). A residue lacking in position of the sequence alignment for the beacon are sodium channels (Class IV). The extent to which animal channels are sodium-selective is dictated in the occupancy of the HFS site with a lysine residue (Class III/IV). Governance involving the beacon solves the quandary the HFS site as a basis for ion selectivity, where an electronegative ring of glutamates at the HFS site generates a sodium-selective channel in one-domain channels but generates a calcium-selective channel in four-domain channels. Discovery of a splice variant in an exceptional channel revealed nature's exploits, highlighting the "beacon" as a principal determinant for calcium and sodium selectivity, encompassing known ion channels composed of one and four domains, from bacteria to animals.


Subject(s)
Aspartic Acid , Calcium , Animals , Calcium/metabolism , Amino Acid Sequence , Calcium Channels/chemistry , Sodium Channels/metabolism , Sodium/metabolism
6.
Nat Struct Mol Biol ; 30(6): 735-739, 2023 06.
Article in English | MEDLINE | ID: mdl-36973510

ABSTRACT

Gabapentinoid drugs for pain and anxiety act on the CaVα2δ-1 and CaVα2δ-2 subunits of high-voltage-activated calcium channels (CaV1s and CaV2s). Here we present the cryo-EM structure of the gabapentin-bound brain and cardiac CaV1.2/CaVß3/CaVα2δ-1 channel. The data reveal a binding pocket in the CaVα2δ-1 dCache1 domain that completely encapsulates gabapentin and define CaVα2δ isoform sequence variations that explain the gabapentin binding selectivity of CaVα2δ-1 and CaVα2δ-2.


Subject(s)
Calcium Channels , Gabapentin , Calcium Channels/chemistry
7.
Trends Plant Sci ; 28(6): 673-684, 2023 06.
Article in English | MEDLINE | ID: mdl-36740491

ABSTRACT

Across phyla, voltage-gated ion channels (VGICs) allow excitability. The vacuolar two-pore channel AtTPC1 from the tiny mustard plant Arabidopsis thaliana has emerged as a paradigm for deciphering the role of voltage and calcium signals in membrane excitation. Among the numerous experimentally determined structures of VGICs, AtTPC1 was the first to be revealed in a closed and resting state, fueling speculation about structural rearrangements during channel activation. Two independent reports on the structure of a partially opened AtTPC1 channel protein have led to working models that offer promising insights into the molecular switches associated with the gating process. We review new structure-function models and also discuss the evolutionary impact of two-pore channels (TPCs) on K+ homeostasis and vacuolar excitability.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/chemistry , Calcium Channels/metabolism , Vacuoles/metabolism
8.
Asian J Androl ; 25(4): 528-532, 2023.
Article in English | MEDLINE | ID: mdl-36571327

ABSTRACT

Beta-defensin 20 (DEFB20) is widely expressed in the epididymis with gene features involved in epididymal sperm maturation. However, the action mechanism and function of DEFB20 in sperm maturation are still unclear. One of the important roles of beta-defensin is the ion channel activity. The cation channel sperm-associated protein (CatSper) alpha is an ion channel protein found on the sperm surface. This study aimed to investigate the interaction between DEFB20 and CatSper1-4 protein in relation to the sperm maturation process. Protein sequences were obtained from the National Center for Biotechnology Information (NCBI). Protein modeling and validation were carried out by using the Robetta modeling server and the Ramachandran plot method. Rosetta web server was used for the docking analysis. The results revealed a natural interaction between DEFB20 and CatSper1-4. The interaction occurred at the cation channel (close to the casein kinase II), ion transport protein, and kinase c phosphorylation of the CatSper1-4 active site. The DEFB20 region interacting with CatSper2-4 was the beta-defensin domain, while with CatSper1 was the non-beta-defensin domain. Based on the analysis, DEFB20 may interact with CatSper α subunits, particularly CatsSper1, to affect ion channel activity during sperm maturation.


Subject(s)
Calcium Channels , beta-Defensins , Male , Humans , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels/metabolism , beta-Defensins/metabolism , Sperm Maturation , Semen/metabolism , Spermatozoa/metabolism , Cations/metabolism , Sperm Motility
9.
Cell Calcium ; 109: 102684, 2023 01.
Article in English | MEDLINE | ID: mdl-36495796

ABSTRACT

Recent structural studies have shown that the carboxyl-terminus of many TRP channels, including TRPC3, are folded into a horizontal rib helix that is connected to the vertical pole helix, which play roles in inter-structural interactions and multimerization. In a previous work we identified I807 located in the pole helix with a role in regulation of TRPC3 by STIM1 (Lee et al., 2014, Liu et al., 2022). To further determine the role of the pole helix in TRPC3 function, here we identified key hydrophobic residues in the pole helix that form tight tunnel-like structure and used mutations to probe their role in TRPC3 regulation by Ca2+ and Calmodulin. Our findings suggest that the hydrophobic starch formed by the I807-L818 residues has several roles, it modulates gating of TRPC3 by Ca2+, affects channel selectivity and the channel Ca2+ permeability. Mutations of I807, I811, L814 and L818 all attenuated the Ca2+-dependent inactivation (CDI) of TRPC3, with I807 having the most prominent effect. The extent of modulation of the CDI depended on the degree of hydrophobicity of I807. Moreover, the TRPC3(I807S) mutant showed altered channel monovalent ion selectivity and increased Ca2+ permeability, without affecting the channel permeability to Mg2+ and Ba2+ and without changing the pore diameter. The CDI of TRPC3 was reduced by an inactive calmodulin mutant and by a pharmacological inhibitor of calmodulin, which was eliminated by the I807S mutation. Notably, deletion of STIM1 caused similar alteration of TRPC3 properties. Taken together, these findings reveal a role of the pole helix in CDI, in addition to its potential role in channel multimerization that required gating of TRPC3 by STIM1. Since all TRPC and most TRP channels have pole helix structures, our findings raise the possibility that the pole helix may have similar roles in all the TRP family.


Subject(s)
Calcium Channels , Calcium , Calmodulin , TRPC Cation Channels , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calmodulin/chemistry , Hydrophobic and Hydrophilic Interactions , Mutation , TRPC Cation Channels/genetics , TRPC Cation Channels/chemistry , Humans
10.
Nature ; 610(7932): 532-539, 2022 10.
Article in English | MEDLINE | ID: mdl-36163289

ABSTRACT

Plant intracellular nucleotide-binding leucine-rich repeat receptors (NLRs) detect pathogen effectors to trigger immune responses1. Indirect recognition of a pathogen effector by the dicotyledonous Arabidopsis thaliana coiled-coil domain containing NLR (CNL) ZAR1 induces the formation of a large hetero-oligomeric protein complex, termed the ZAR1 resistosome, which functions as a calcium channel required for ZAR1-mediated immunity2-4. Whether the resistosome and channel activities are conserved among plant CNLs remains unknown. Here we report the cryo-electron microscopy structure of the wheat CNL Sr355 in complex with the effector AvrSr356 of the wheat stem rust pathogen. Direct effector binding to the leucine-rich repeats of Sr35 results in the formation of a pentameric Sr35-AvrSr35 complex, which we term the Sr35 resistosome. Wheat Sr35 and Arabidopsis ZAR1 resistosomes bear striking structural similarities, including an arginine cluster in the leucine-rich repeats domain not previously recognized as conserved, which co-occurs and forms intramolecular interactions with the 'EDVID' motif in the coiled-coil domain. Electrophysiological measurements show that the Sr35 resistosome exhibits non-selective cation channel activity. These structural insights allowed us to generate new variants of closely related wheat and barley orphan NLRs that recognize AvrSr35. Our data support the evolutionary conservation of CNL resistosomes in plants and demonstrate proof of principle for structure-based engineering of NLRs for crop improvement.


Subject(s)
Calcium Channels , Cryoelectron Microscopy , NLR Proteins , Plant Proteins , Receptors, Immunologic , Triticum , Arabidopsis/immunology , Arabidopsis/metabolism , Arginine , Calcium Channels/chemistry , Calcium Channels/immunology , Calcium Channels/metabolism , Cations/metabolism , Leucine , NLR Proteins/chemistry , NLR Proteins/immunology , NLR Proteins/metabolism , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Immunity , Plant Proteins/chemistry , Plant Proteins/immunology , Plant Proteins/metabolism , Receptors, Immunologic/chemistry , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Triticum/immunology , Triticum/metabolism , Amino Acid Motifs , Conserved Sequence , Electrophysiology
11.
J Biol Chem ; 298(5): 101838, 2022 05.
Article in English | MEDLINE | ID: mdl-35339491

ABSTRACT

Calcium homeostasis modulator 1 (CALHM1) is a voltage- and Ca2+-gated ATP channel that plays an important role in neuronal signaling. However, as the previously reported CALHM structures are all in the ATP-conducting state, the gating mechanism of ATP permeation is still elusive. Here, we report cryo-EM reconstructions of two Danio rerio CALHM1 heptamers with ordered or flexible long C-terminal helices at resolutions of 3.2 Å and 2.9 Å, respectively, and one D. rerio CALHM1 octamer with flexible long C-terminal helices at a resolution of 3.5 Å. Structural analysis shows that the heptameric CALHM1s are in an ATP-nonconducting state with a central pore diameter of approximately 6.6 Å. Compared with those inside the octameric CALHM1, the N-helix inside the heptameric CALHM1 is in the "down" position to avoid steric clashing with the adjacent TM1 helix. Molecular dynamics simulations show that as the N-helix moves from the "down" position to the "up" position, the pore size of ATP molecule permeation increases significantly. Our results provide important information for elucidating the mechanism of ATP molecule permeation in the CALHM1 channel.


Subject(s)
Adenosine Triphosphate , Calcium Channels , Zebrafish Proteins , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calcium Channels/chemistry , Cryoelectron Microscopy , Homeostasis , Zebrafish , Zebrafish Proteins/chemistry
12.
Molecules ; 27(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35209100

ABSTRACT

Voltage-gated calcium channels (VGCCs) are widely expressed in the brain, heart and vessels, smooth and skeletal muscle, as well as in endocrine cells. VGCCs mediate gene transcription, synaptic and neuronal structural plasticity, muscle contraction, the release of hormones and neurotransmitters, and membrane excitability. Therefore, it is not surprising that VGCC dysfunction results in severe pathologies, such as cardiovascular conditions, neurological and psychiatric disorders, altered glycemic levels, and abnormal smooth muscle tone. The latest research findings and clinical evidence increasingly show the critical role played by VGCCs in autism spectrum disorders, Parkinson's disease, drug addiction, pain, and epilepsy. These findings outline the importance of developing selective calcium channel inhibitors and modulators to treat such prevailing conditions of the central nervous system. Several small molecules inhibiting calcium channels are currently used in clinical practice to successfully treat pain and cardiovascular conditions. However, the limited palette of molecules available and the emerging extent of VGCC pathophysiology require the development of additional drugs targeting these channels. Here, we provide an overview of the role of calcium channels in neurological disorders and discuss possible strategies to generate novel therapeutics.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Animals , Calcium Channel Agonists/therapeutic use , Calcium Channel Blockers/therapeutic use , Calcium Channels/chemistry , Calcium Channels/classification , Calcium Channels/genetics , Clinical Studies as Topic , Disease Management , Disease Susceptibility , Drug Discovery , Drug Evaluation, Preclinical , Humans , Ligands , Nervous System Diseases/diagnosis , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Treatment Outcome
13.
Molecules ; 27(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35164225

ABSTRACT

Excessive release of glutamate induces excitotoxicity and causes neuronal damage in several neurodegenerative diseases. Natural products have emerged as potential neuroprotective agents for preventing and treating neurological disorders. Dehydrocorydaline (DHC), an active alkaloid compound isolated from Corydalis yanhusuo, possesses neuroprotective capacity. The present study investigated the effect of DHC on glutamate release using a rat brain cortical synaptosome model. Our results indicate that DHC inhibited 4-aminopyridine (4-AP)-evoked glutamate release and elevated intrasynaptosomal calcium levels. The inhibitory effect of DHC on 4-AP-evoked glutamate release was prevented in the presence of the vesicular transporter inhibitor bafilomycin A1 and the N- and P/Q-type Ca2+ channel blocker ω-conotoxin MVIIC but not the intracellular inhibitor of Ca2+ release dantrolene or the mitochondrial Na+/Ca2+ exchanger inhibitor CGP37157. Moreover, the inhibitory effect of DHC on evoked glutamate release was prevented by the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) inhibitor PD98059. Western blotting data in synaptosomes also showed that DHC significantly decreased the level of ERK1/2 phosphorylation and synaptic vesicle-associated protein synapsin I, the main presynaptic target of ERK. Together, these results suggest that DHC inhibits presynaptic glutamate release from cerebrocortical synaptosomes by suppressing presynaptic voltage-dependent Ca2+ entry and the MAPK/ERK/synapsin I signaling pathway.


Subject(s)
Alkaloids/pharmacology , Calcium/metabolism , Cerebral Cortex/drug effects , Corydalis/chemistry , Glutamic Acid/metabolism , Nerve Tissue/drug effects , Neuroprotective Agents/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Cerebral Cortex/metabolism , Male , Nerve Tissue/metabolism , Rats , Rats, Sprague-Dawley
14.
FEBS Open Bio ; 12(4): 811-826, 2022 04.
Article in English | MEDLINE | ID: mdl-35060355

ABSTRACT

Mitochondrial calcium homeostasis plays critical roles in cell survival and aerobic metabolism in eukaryotes. The calcium uniporter is a highly selective calcium ion channel consisting of several subunits. Mitochondrial calcium uniporter (MCU) and essential MCU regulator (EMRE) are core subunits of the calcium uniporter required for calcium uptake activity in the mitochondria. Recent 3D structure analysis of the MCU-EMRE complex reconstituted in nanodiscs revealed that the human MCU exists as a tetramer forming a channel pore, with EMRE bound to each MCU at a 1 : 1 ratio. However, the stoichiometry of MCU and EMRE in the mitochondria has not yet been investigated. We here quantitatively examined the protein levels of MCU and EMRE in the mitochondria from mouse tissues by using characterized antibodies and standard proteins. Unexpectedly, the number of EMRE molecules was lower than that of MCU; moreover, the ratios between MCU and EMRE were significantly different among tissues. Statistical calculations based on our findings suggest that a MCU tetramer binding to 4 EMREs may exist, but at low levels in the mitochondrial inner membrane. In brain mitochondria, the majority of MCU tetramers bind to 2 EMREs; in mitochondria in liver, kidney, and heart, MCU tetramers bind to 1 EMRE; and in kidney and heart, almost half of MCU tetramers bound to no EMRE. We propose here a novel stoichiometric model of the MCU-EMRE complex in mitochondria.


Subject(s)
Calcium Channels , Mitochondria , Animals , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , HeLa Cells , Humans , Mice , Mitochondria/metabolism
15.
Mol Cell ; 81(23): 4784-4798.e7, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34800360

ABSTRACT

Calcium influx through plasma membrane calcium release-activated calcium (CRAC) channels, which are formed of hexamers of Orai1, is a potent trigger for many important biological processes, most notably in T cell-mediated immunity. Through a bioinformatics-led cell biological screen, we have identified Orai1 as a substrate for the rhomboid intramembrane protease RHBDL2. We show that RHBDL2 prevents stochastic calcium signaling in unstimulated cells through conformational surveillance and cleavage of inappropriately activated Orai1. A conserved disease-linked proline residue is responsible for RHBDL2's recognizing the active conformation of Orai1, which is required to sharpen switch-like signaling triggered by store-operated calcium entry. Loss of RHBDL2 control of CRAC channel activity causes severe dysregulation of downstream CRAC channel effectors, including transcription factor activation, inflammatory cytokine expression, and T cell activation. We propose that this surveillance function may represent an ancient activity of rhomboid proteases in degrading unwanted signaling proteins.


Subject(s)
ORAI1 Protein/chemistry , Peptide Hydrolases/chemistry , Serine Endopeptidases/metabolism , Animals , Calcium/metabolism , Calcium Channels/chemistry , Calcium Signaling/physiology , Cell Membrane/metabolism , Computational Biology , Drosophila melanogaster , HEK293 Cells , Humans , Ion Channel Gating , Lymphocyte Activation , Membrane Proteins/metabolism , Mutation , Protein Binding , Protein Conformation , Signal Transduction , Stochastic Processes
16.
Nat Commun ; 12(1): 6284, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725357

ABSTRACT

TRPV6 is a calcium-selective ion channel implicated in epithelial Ca2+ uptake. TRPV6 inhibitors are needed for the treatment of a broad range of diseases associated with disturbed calcium homeostasis, including cancers. Here we combine cryo-EM, calcium imaging, and mutagenesis to explore molecular bases of human TRPV6 inhibition by the antifungal drug econazole and the universal ion channel blocker ruthenium red (RR). Econazole binds to an allosteric site at the channel's periphery, where it replaces a lipid. In contrast, RR inhibits TRPV6 by binding in the middle of the ion channel's selectivity filter and plugging its pore like a bottle cork. Despite different binding site locations, both inhibitors induce similar conformational changes in the channel resulting in closure of the gate formed by S6 helices bundle crossing. The uncovered molecular mechanisms of TRPV6 inhibition can guide the design of a new generation of clinically useful inhibitors.


Subject(s)
Antifungal Agents/chemistry , Calcium Channel Blockers/chemistry , Calcium Channels/chemistry , Econazole/chemistry , Ruthenium Red/chemistry , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/chemistry , Binding Sites , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Humans , Models, Molecular , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
17.
Food Funct ; 12(22): 11387-11398, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34672304

ABSTRACT

Cancer is a major threat to human health worldwide, yet the clinical therapies remain unsatisfactory. In this study, we found that a Tetrastigma hemsleyanum leaves flavone (TLF) intervention could achieve tumor inhibition. Besides, neochlorogenic acid (NA), which had the highest absorbance peak in the HPLC profile of TLF, showed superior anti-proliferation ability over TLF, and could effectively trigger apoptosis, restrain migration, and facilitate cytoskeleton collapse, suggesting its key role in TLF's anticancer property. Molecular docking analysis suggested that NA was capable of binding with mitochondrial Ca2+ uniporter (MCU), and further experiments confirmed that NA upregulated the MCU level to permit excess calcium ion influx, leading to mitochondrial calcium imbalance, dysfunction, structure alteration, and ROS elevation. Moreover, tumor-bearing mice were applied to further confirm the excellent tumor inhibition ability of NA under Ca2+-abundant conditions. Therefore, this study uncovered that NA could effectively trigger robust MCU-mediated calcium overload cancer therapy, which could be utilized in novel strategies for future cancer treatment.


Subject(s)
Antineoplastic Agents , Calcium Channels , Calcium , Chlorogenic Acid/analogs & derivatives , Quinic Acid/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Calcium/chemistry , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Cell Survival/drug effects , Chlorogenic Acid/chemistry , Chlorogenic Acid/metabolism , Flavones/metabolism , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Neoplasms, Experimental , Plant Leaves/chemistry , Quinic Acid/chemistry , Quinic Acid/metabolism , Vitaceae/chemistry
18.
PLoS One ; 16(10): e0258112, 2021.
Article in English | MEDLINE | ID: mdl-34610038

ABSTRACT

Calmodulin, a ubiquitous eukaryotic calcium sensor responsible for the regulation of many fundamental cellular processes, is a highly flexible protein and exhibits an unusually wide range of conformations. Furthermore, CaM is known to interact with more than 300 cellular targets. Molecular dynamics (MD) simulation trajectories suggest that EF-hand loops show different magnitudes of flexibility. Therefore, the four EF-hand motifs have different affinities for Ca2+ ions, which enables CaM to function on wide range of Ca2+ ion concentrations. EF-hand loops are 2-3 times more flexible in apo CaM whereas least flexible in Ca2+/CaM-IQ motif complexes. We report a unique intermediate conformation of Ca2+/CaM while transitioning from extended to compact form. We also report the complex formation process between Ca2+/CaM and IQ CaM-binding motifs. Our results showed how IQ motif recognise its binding site on the CaM and how CaM transforms from extended to compact form upon binding to IQ motif.


Subject(s)
Calcium Channels , Calcium/metabolism , Calmodulin , Molecular Dynamics Simulation , Binding Sites , Calcium Channels/chemistry , Calcium Channels/metabolism , Calmodulin/chemistry , Calmodulin/metabolism , Protein Binding , Protein Conformation
19.
Nat Commun ; 12(1): 5802, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608145

ABSTRACT

Two-pore channels (TPCs) are a ubiquitous family of cation channels that localize to acidic organelles in animals and plants to regulate numerous Ca2+-dependent events. Little is known about TPCs in unicellular organisms despite their ancient origins. Here, we characterize a TPC from Toxoplasma gondii, the causative agent of toxoplasmosis. TgTPC is a member of a novel clad of TPCs in Apicomplexa, distinct from previously identified TPCs and only present in coccidians. We show that TgTPC localizes not to acidic organelles but to the apicoplast, a non-photosynthetic plastid found in most apicomplexan parasites. Conditional silencing of TgTPC resulted in progressive loss of apicoplast integrity, severely affecting growth and the lytic cycle. Isolation of TPC null mutants revealed a selective role for TPCs in replication independent of apicoplast loss that required conserved residues within the pore-lining region. Using a genetically-encoded Ca2+ indicator targeted to the apicoplast, we show that Ca2+ signals deriving from the ER but not from the extracellular space are selectively transmitted to the lumen. Deletion of the TgTPC gene caused reduced apicoplast Ca2+ uptake and membrane contact site formation between the apicoplast and the ER. Fundamental roles for TPCs in maintaining organelle integrity, inter-organelle communication and growth emerge.


Subject(s)
Calcium Channels/metabolism , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Toxoplasma/metabolism , Amino Acid Sequence , Apicoplasts/metabolism , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Signaling , DNA/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mutation , Organelle Biogenesis , Phylogeny , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
20.
Int J Mol Sci ; 22(20)2021 Oct 19.
Article in English | MEDLINE | ID: mdl-34681928

ABSTRACT

Ca2+ entry through Cav1.3 Ca2+ channels plays essential roles in diverse physiological events. We employed yeast-two-hybrid (Y2H) assays to mine novel proteins interacting with Cav1.3 and found Snapin2, a synaptic protein, as a partner interacting with the long carboxyl terminus (CTL) of rat Cav1.3L variant. Co-expression of Snapin with Cav1.3L/Cavß3/α2δ2 subunits increased the peak current density or amplitude by about 2-fold in HEK-293 cells and Xenopus oocytes, without affecting voltage-dependent gating properties and calcium-dependent inactivation. However, the Snapin up-regulation effect was not found for rat Cav1.3S containing a short CT (CTS) in which a Snapin interaction site in the CTL was deficient. Luminometry and electrophysiology studies uncovered that Snapin co-expression did not alter the membrane expression of HA tagged Cav1.3L but increased the slope of tail current amplitudes plotted against ON-gating currents, indicating that Snapin increases the opening probability of Cav1.3L. Taken together, our results strongly suggest that Snapin directly interacts with the CTL of Cav1.3L, leading to up-regulation of Cav1.3L channel activity via facilitating channel opening probability.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/metabolism , Up-Regulation , Vesicular Transport Proteins/metabolism , Animals , Binding Sites , Female , HEK293 Cells , Humans , Protein Domains , Rats , Two-Hybrid System Techniques , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...