Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Proc Natl Acad Sci U S A ; 114(21): E4288-E4295, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28490495

ABSTRACT

Alternative splicing changes the CaV1.2 calcium channel electrophysiological property, but the in vivo significance of such altered channel function is lacking. Structure-function studies of heterologously expressed CaV1.2 channels could not recapitulate channel function in the native milieu of the cardiomyocyte. To address this gap in knowledge, we investigated the role of alternative exon 33 of the CaV1.2 calcium channel in heart function. Exclusion of exon 33 in CaV1.2 channels has been reported to shift the activation potential -10.4 mV to the hyperpolarized direction, and increased expression of CaV1.2Δ33 channels was observed in rat myocardial infarcted hearts. However, how a change in CaV1.2 channel electrophysiological property, due to alternative splicing, might affect cardiac function in vivo is unknown. To address these questions, we generated mCacna1c exon 33-/--null mice. These mice contained CaV1.2Δ33 channels with a gain-of-function that included conduction of larger currents that reflects a shift in voltage dependence and a modest increase in single-channel open probability. This altered channel property underscored the development of ventricular arrhythmia, which is reflected in significantly more deaths of exon 33-/- mice from ß-adrenergic stimulation. In vivo telemetric recordings also confirmed increased frequencies in premature ventricular contractions, tachycardia, and lengthened QT interval. Taken together, the significant decrease or absence of exon 33-containing CaV1.2 channels is potentially proarrhythmic in the heart. Of clinical relevance, human ischemic and dilated cardiomyopathy hearts showed increased inclusion of exon 33. However, the possible role that inclusion of exon 33 in CaV1.2 channels may play in the pathogenesis of human heart failure remains unclear.


Subject(s)
Action Potentials/genetics , Calcium Channels, L-Type/genetics , Long QT Syndrome/genetics , Tachycardia/genetics , Ventricular Premature Complexes/genetics , Action Potentials/physiology , Alternative Splicing/genetics , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Cells, Cultured , Colforsin/pharmacology , Electrophysiological Phenomena/genetics , Heart Failure/genetics , Heart Failure/pathology , Isoproterenol/pharmacology , Long QT Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Nifedipine/pharmacology , Rats , Sequence Deletion/genetics , Tachycardia/pathology , Ventricular Premature Complexes/pathology
2.
J Neurosci ; 38(39): 8364-8377, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30104344

ABSTRACT

Elevated iron deposition has been reported in Parkinson's disease (PD). However, the route of iron uptake leading to high deposition in the substantia nigra is unresolved. Here, we show a mechanism in enhanced Fe2+ uptake via S-nitrosylation of divalent metal transporter 1 (DMT1). While DMT1 could be S-nitrosylated by exogenous nitric oxide donors, in human PD brains, endogenously S-nitrosylated DMT1 was detected in postmortem substantia nigra. Patch-clamp electrophysiological recordings and iron uptake assays confirmed increased Mn2+ or Fe2+ uptake through S-nitrosylated DMT1. We identified two major S-nitrosylation sites, C23 and C540, by mass spectrometry, and DMT1 C23A or C540A substitutions abolished nitric oxide (NO)-mediated DMT1 current increase. To evaluate in vivo significance, lipopolysaccharide (LPS) was stereotaxically injected into the substantia nigra of female and male mice to induce inflammation and production of NO. The intranigral LPS injection resulted in corresponding increase in Fe2+ deposition, JNK activation, dopaminergic neuronal loss and deficit in motoric activity, and these were rescued by the NO synthase inhibitor l-NAME or by the DMT1-selective blocker ebselen. Lentiviral knockdown of DMT1 abolished LPS-induced dopaminergic neuron loss.SIGNIFICANCE STATEMENT Neuroinflammation and high cytoplasmic Fe2+ levels have been implicated in the initiation and progression of neurodegenerative diseases. Here, we report the unexpected enhancement of the functional activity of transmembrane divalent metal transporter 1 (DMT1) by S-nitrosylation. We demonstrated that S-nitrosylation increased DMT1-mediated Fe2+ uptake, and two cysteines were identified by mass spectrometry to be the sites for S-nitrosylation and for enhanced iron uptake. One conceptual advance is that while DMT1 activity could be increased by external acidification because the gating of the DMT1 transporter is proton motive, we discovered that DMT1 activity could also be enhanced by S-nitrosylation. Significantly, lipopolysaccharide-induced nitric oxide (NO)-mediated neuronal death in the substantia nigra could be ameliorated by using l-NAME, a NO synthase inhibitor, or by ebselen, a DMT1-selective blocker.


Subject(s)
Cation Transport Proteins/metabolism , Dopaminergic Neurons/metabolism , Iron/metabolism , Locomotion , Nitric Oxide/chemistry , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Animals , Cation Transport Proteins/chemistry , Female , Humans , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/administration & dosage , Male , Mice, Transgenic
3.
Circulation ; 138(14): 1431-1445, 2018 10 02.
Article in English | MEDLINE | ID: mdl-29650545

ABSTRACT

BACKGROUND: L-type CaV1.2 channels play crucial roles in the regulation of blood pressure. Galectin-1 (Gal-1) has been reported to bind to the I-II loop of CaV1.2 channels to reduce their current density. However, the mechanistic understanding for the downregulation of CaV1.2 channels by Gal-1 and whether Gal-1 plays a direct role in blood pressure regulation remain unclear. METHODS: In vitro experiments involving coimmunoprecipitation, Western blot, patch-clamp recordings, immunohistochemistry, and pressure myography were used to evaluate the molecular mechanisms by which Gal-1 downregulates CaV1.2 channel in transfected, human embryonic kidney 293 cells, smooth muscle cells, arteries from Lgasl1-/- mice, rat, and human patients. In vivo experiments involving the delivery of Tat-e9c peptide and AAV5-Gal-1 into rats were performed to investigate the effect of targeting CaV1.2-Gal-1 interaction on blood pressure monitored by tail-cuff or telemetry methods. RESULTS: Our study reveals that Gal-1 is a key regulator for proteasomal degradation of CaV1.2 channels. Gal-1 competed allosterically with the CaVß subunit for binding to the I-II loop of the CaV1.2 channel. This competitive disruption of CaVß binding led to CaV1.2 degradation by exposing the channels to polyubiquitination. It is notable that we demonstrated that the inverse relationship of reduced Gal-1 and increased CaV1.2 protein levels in arteries was associated with hypertension in hypertensive rats and patients, and Gal-1 deficiency induces higher blood pressure in mice because of the upregulated CaV1.2 protein level in arteries. To directly regulate blood pressure by targeting the CaV1.2-Gal-1 interaction, we administered Tat-e9c, a peptide that competed for binding of Gal-1 by a miniosmotic pump, and this specific disruption of CaV1.2-Gal-1 coupling increased smooth muscle CaV1.2 currents, induced larger arterial contraction, and caused hypertension in rats. In contrasting experiments, overexpression of Gal-1 in smooth muscle by a single bolus of AAV5-Gal-1 significantly reduced blood pressure in spontaneously hypertensive rats. CONCLUSIONS: We have defined molecularly that Gal-1 promotes CaV1.2 degradation by replacing CaVß and thereby exposing specific lysines for polyubiquitination and by masking I-II loop endoplasmic reticulum export signals. This mechanistic understanding provided the basis for targeting CaV1.2-Gal-1 interaction to demonstrate clearly the modulatory role that Gal-1 plays in regulating blood pressure, and offering a potential approach for therapeutic management of hypertension.


Subject(s)
Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Calcium Channels, L-Type/metabolism , Galectin 1/metabolism , Genetic Therapy/methods , Hypertension/therapy , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Peptide Fragments/pharmacology , Animals , Calcium Channels, L-Type/genetics , Case-Control Studies , Dependovirus , Disease Models, Animal , Galectin 1/genetics , Genetic Vectors , HEK293 Cells , Humans , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Male , Membrane Potentials , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Parvovirinae/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Proteolysis , Rats, Inbred SHR , Rats, Inbred WKY
4.
Pflugers Arch ; 471(11-12): 1455-1466, 2019 12.
Article in English | MEDLINE | ID: mdl-31664513

ABSTRACT

Reperfusion therapy is currently the gold standard treatment for acute ischemic stroke. However, reperfusion injuries such as oedema and haemorrhagic transformation largely limit the use of this potent treatment to a narrow time window. Recently, transient receptor potential melastatin 4 (TRPM4) channel has emerged as a potential target for vascular protection in stroke management. Non-specificity and side effects are major concerns for current TRPM4 blockers. The present study was undertaken to develop a novel TRPM4 blocker for stroke management. We report the generation of a TRPM4-specific antibody M4P which binds to a region close to the channel pore. M4P could inhibit TRPM4 current and downregulate TRPM4 surface expression, therefore prevent hypoxia-induced cell swelling. In the rat model of 3-h stroke reperfusion, application of M4P at 2 h after occlusion ameliorated reperfusion injury by improving blood-brain barrier integrity, and enhanced functional recovery. Our results demonstrate that TRPM4 blockade could attenuate reperfusion injury in stroke recanalization. When applied together with reperfusion treatments, TRPM4 blocking antibody has the potential to extend the therapeutic time window for acute ischemic stroke.


Subject(s)
Antibodies, Monoclonal/pharmacology , Reperfusion Injury/drug therapy , Stroke/drug therapy , TRPM Cation Channels/antagonists & inhibitors , Animals , Blood-Brain Barrier/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Disease Models, Animal , Female , Male , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Stroke/metabolism , Up-Regulation/drug effects
5.
Phys Rev Lett ; 122(20): 204301, 2019 May 24.
Article in English | MEDLINE | ID: mdl-31172787

ABSTRACT

Topologically gapless edge states, characterized by topological invariants and Berry's phases of bulk energy bands, provide amazing techniques to robustly control the reflectionless propagation of electrons, photons, and phonons. Recently, a new family of topological phases, dictated by the bulk polarization, has been observed, leading to the discovery of the higher-order topological insulators (HOTIs). So far, the HOTIs have been demonstrated in mechanical and electromagnetic systems and electrical circuits with quantized quadrupole polarization and, more recently, have been experimentally realized in optical and acoustic systems. Here, we realize the higher-order topological states in a two-dimensional (2D) continuous elastic system. We experimentally observe the gapped one-dimensional (1D) edge states, the trivially gapped zero-dimensional (0D) corner states, and the topologically protected 0D corner states. Compared with the trivial corner modes, the topological ones, immunizing against defects, are robustly localized at the obtuse-angled but not the acute-angled corners. The topological shape-dependent corner states open a new route for the design of the topologically protected and reconfigurable 0D localized resonances and provide an excellent platform for the topological transformation of the elastic energy among 2D bulk, 1D edge, and 0D corner modes.

6.
Biophys J ; 114(9): 2095-2106, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29742403

ABSTRACT

The CaV1.2 L-type calcium channel is a key conduit for Ca2+ influx to initiate excitation-contraction coupling for contraction of the heart and vasoconstriction of the arteries and for altering membrane excitability in neurons. Its α1C pore-forming subunit is known to undergo extensive alternative splicing to produce many CaV1.2 isoforms that differ in their electrophysiological and pharmacological properties. Here, we examined the structure-function relationship of human CaV1.2 with respect to the inclusion or exclusion of mutually exclusive exons of the N-terminus exons 1/1a and IS6 segment exons 8/8a. These exons showed tissue selectivity in their expression patterns: heart variant 1a/8a, one smooth-muscle variant 1/8, and a brain isoform 1/8a. Overall, the 1/8a, when coexpressed with CaVß2a, displayed a significant and distinct shift in voltage-dependent activation and inactivation and inactivation kinetics as compared to the other three splice variants. Further analysis showed a clear additive effect of the hyperpolarization shift in V1/2inact of CaV1.2 channels containing exon 1 in combination with 8a. However, this additive effect was less distinct for V1/2act. However, the measured effects were ß-subunit-dependent when comparing CaVß2a with CaVß3 coexpression. Notably, calcium-dependent inactivation mediated by local Ca2+-sensing via the N-lobe of calmodulin was significantly enhanced in exon-1-containing CaV1.2 as compared to exon-1a-containing CaV1.2 channels. At the cellular level, the current densities of the 1/8a or 1/8 variants were significantly larger than the 1a/8a and 1a/8 variants when coexpressed either with CaVß2a or CaVß3 subunit. This finding correlated well with a higher channel surface expression for the exon 1-CaV1.2 isoform that we quantified by protein surface-expression levels or by gating currents. Our data also provided a deeper molecular understanding of the altered biophysical properties of alternatively spliced human CaV1.2 channels by directly comparing unitary single-channel events with macroscopic whole-cell currents.


Subject(s)
Alternative Splicing , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Gene Expression Regulation , Calcium Channels, L-Type/metabolism , Exons/genetics , HEK293 Cells , Humans , Ion Channel Gating/genetics , Kinetics , Protein Domains , Surface Properties
7.
J Acoust Soc Am ; 141(4): 2768, 2017 04.
Article in English | MEDLINE | ID: mdl-28464643

ABSTRACT

This paper presents a homogenization-based interval analysis method for the prediction of coupled structural-acoustic systems involving periodical composites and multi-scale uncertain-but-bounded parameters. In the structural-acoustic system, the macro plate structure is assumed to be composed of a periodically uniform microstructure. The equivalent macro material properties of the microstructure are computed using the homogenization method. By integrating the first-order Taylor expansion interval analysis method with the homogenization-based finite element method, a homogenization-based interval finite element method (HIFEM) is developed to solve a periodical composite structural-acoustic system with multi-scale uncertain-but-bounded parameters. The corresponding formulations of the HIFEM are deduced. A subinterval technique is also introduced into the HIFEM for higher accuracy. Numerical examples of a hexahedral box and an automobile passenger compartment are given to demonstrate the efficiency of the presented method for a periodical composite structural-acoustic system with multi-scale uncertain-but-bounded parameters.

8.
J Biol Chem ; 290(14): 9262-72, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25694430

ABSTRACT

L-type Cav1.2 Ca(2+) channel undergoes extensive alternative splicing, generating functionally different channels. Alternatively spliced Cav1.2 Ca(2+) channels have been found to be expressed in a tissue-specific manner or under pathological conditions. To provide a more comprehensive understanding of alternative splicing in Cav1.2 channel, we systematically investigated the splicing patterns in the neonatal and adult rat hearts. The neonatal heart expresses a novel 104-bp exon 33L at the IVS3-4 linker that is generated by the use of an alternative acceptor site. Inclusion of exon 33L causes frameshift and C-terminal truncation. Whole-cell electrophysiological recordings of Cav1.233L channels expressed in HEK 293 cells did not detect any current. However, when co-expressed with wild type Cav1.2 channels, Cav1.233L channels reduced the current density and altered the electrophysiological properties of the wild type Cav1.2 channels. Interestingly, the truncated 3.5-domain Cav1.233L channels also yielded a dominant negative effect on Cav1.3 channels, but not on Cav3.2 channels, suggesting that Cavß subunits is required for Cav1.233L regulation. A biochemical study provided evidence that Cav1.233L channels enhanced protein degradation of wild type channels via the ubiquitin-proteasome system. Although the physiological significance of the Cav1.233L channels in neonatal heart is still unknown, our report demonstrates the ability of this novel truncated channel to modulate the activity of the functional Cav1.2 channels. Moreover, the human Cav1.2 channel also contains exon 33L that is developmentally regulated in heart. Unexpectedly, human exon 33L has a one-nucleotide insertion that allowed in-frame translation of a full Cav1.2 channel. An electrophysiological study showed that human Cav1.233L channel is a functional channel but conducts Ca(2+) ions at a much lower level.


Subject(s)
Alternative Splicing , Calcium Channels, L-Type/genetics , Myocardium/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Base Sequence , Calcium Channels, L-Type/chemistry , DNA , DNA Primers , Exons , Male , Molecular Sequence Data , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
9.
Pflugers Arch ; 466(3): 563-76, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24043570

ABSTRACT

Transient receptor potential melastatin 4 (TRPM4) is a voltage-dependent, nonselective cation channel. Under pathological conditions, sustained activation of TRPM4 leads to oncotic cell death. Here, we report the upregulation of TRPM4 in vascular endothelium following hypoxia/ischemia in vitro and in vivo. In human umbilical vein endothelial cells, TRPM4 expression was increased at both the mRNA and protein levels following oxygen-glucose deprivation. Blocking TRPM4 with 9-phenanthrol greatly enhanced tube formation on Matrigel. In a rat permanent middle cerebral artery occlusion model, TRPM4 was upregulated in the vascular endothelium within the penumbra region after stroke. TRPM4 expression peaked 1 day post-occlusion and gradually decreased. In vivo siRNA-mediated TRPM4 silencing enhanced angiogenesis and improved capillary integrity. A twofold reduction in infarct volume and a substantial recovery of motor function were observed in animals receiving the siRNA treatment. Interestingly, the protective effect of TRPM4 suppression disappeared 5 days after stroke induction, indicating that TRPM4 upregulation is critical for cerebral damage during the acute phase of stroke. TRPM4 could be a potential therapeutic target for ischemic stroke.


Subject(s)
Infarction, Middle Cerebral Artery/metabolism , Neovascularization, Physiologic , TRPM Cation Channels/metabolism , Animals , Cell Hypoxia , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Genetic Therapy , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/therapy , Locomotion , Male , Phenanthrenes/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Wistar , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , Up-Regulation
10.
Nat Commun ; 15(1): 3978, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38729926

ABSTRACT

A key mechanism employed by plants to adapt to salinity stress involves maintaining ion homeostasis via the actions of ion transporters. While the function of cation transporters in maintaining ion homeostasis in plants has been extensively studied, little is known about the roles of their anion counterparts in this process. Here, we describe a mechanism of salt adaptation in plants. We characterized the chloride channel (CLC) gene AtCLCf, whose expression is regulated by WRKY transcription factor under salt stress in Arabidopsis thaliana. Loss-of-function atclcf seedlings show increased sensitivity to salt, whereas AtCLCf overexpression confers enhanced resistance to salt stress. Salt stress induces the translocation of GFP-AtCLCf fusion protein to the plasma membrane (PM). Blocking AtCLCf translocation using the exocytosis inhibitor brefeldin-A or mutating the small GTPase gene AtRABA1b/BEX5 (RAS GENES FROM RAT BRAINA1b homolog) increases salt sensitivity in plants. Electrophysiology and liposome-based assays confirm the Cl-/H+ antiport function of AtCLCf. Therefore, we have uncovered a mechanism of plant adaptation to salt stress involving the NaCl-induced translocation of AtCLCf to the PM, thus facilitating Cl- removal at the roots, and increasing the plant's salinity tolerance.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Membrane , Chloride Channels , Golgi Apparatus , Salt Stress , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/physiology , Arabidopsis/drug effects , Cell Membrane/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Golgi Apparatus/metabolism , Chloride Channels/metabolism , Chloride Channels/genetics , Gene Expression Regulation, Plant , Protein Transport/drug effects , Salt Tolerance/genetics , Sodium Chloride/pharmacology , Plants, Genetically Modified
11.
Cell Res ; 34(3): 245-257, 2024 03.
Article in English | MEDLINE | ID: mdl-38302740

ABSTRACT

Mutations in the orphan transporter MFSD7c (also known as Flvcr2), are linked to Fowler syndrome. Here, we used Mfsd7c knockout (Mfsd7c-/-) mice and cell-based assays to reveal that MFSD7c is a choline transporter at the blood-brain barrier (BBB). We performed comprehensive metabolomics analysis and detected differential changes of metabolites in the brains and livers of Mfsd7c-/-embryos. Particularly, we found that choline-related metabolites were altered in the brains but not in the livers of Mfsd7c-/- embryos. Thus, we hypothesized that MFSD7c regulates the level of choline in the brain. Indeed, expression of human MFSD7c in cells significantly increased choline uptake. Interestingly, we showed that choline uptake by MFSD7c is greatly increased by choline-metabolizing enzymes, leading us to demonstrate that MFSD7c is a facilitative transporter of choline. Furthermore, single-cell patch clamp analysis showed that the import of choline by MFSD7c is electrogenic. Choline transport function of MFSD7c was shown to be conserved in vertebrates, but not in yeasts. We demonstrated that human MFSD7c is a functional ortholog of HNM1, the yeast choline importer. We also showed that several missense mutations identified in patients exhibiting Fowler syndrome had abolished or reduced choline transport activity. Mice lacking Mfsd7c in endothelial cells of the central nervous system suppressed the import of exogenous choline from blood but unexpectedly had increased choline levels in the brain. Stable-isotope tracing study revealed that MFSD7c was required for exporting choline derived from lysophosphatidylcholine in the brain. Collectively, our work identifies MFSD7c as a choline exporter at the BBB and provides a foundation for future work to reveal the disease mechanisms of Fowler syndrome.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Polycystic Ovary Syndrome , Urination Disorders , Animals , Humans , Mice , Biological Transport , Brain , Choline
13.
Mol Pharmacol ; 84(4): 643-53, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23924992

ABSTRACT

The transcripts of L-type voltage-gated calcium channels (CaV) 1.3 undergo extensive alternative splicing. Alternative splicing, particularly in the C terminus, drastically modifies gating properties of the channel. However, little is known about whether alternative splicing could modulate the pharmacologic properties of CaV1.3 in a manner similar to the paralogous CaV1.2. Here we undertook the screening of different channel splice isoforms harboring splice variations in either the IS6 segment or the C terminus. Unexpectedly, while inclusion of exon 8a or 8, which code for IS6, did not alter dihydropyridine (DHP) sensitivity, distinct pharmacologic properties were observed for the various C-terminal splice isoforms. In the presence of external Ca(2+), fast inactivating splice variants including CaV1.342a and CaV1.343s with intact calmodulin-IQ domain interaction showed consistently low DHP sensitivity. Interestingly, attenuation of calcium-dependent inactivation with overexpression of calmodulin34 did not enhance the sensitivity of CaV1.342a, suggesting that the low DHP sensitivity may not be a result of fast channel inactivation. Alternatively, disruption of calmodulin-IQ domain binding in the CaV1.3Δ41 and full-length CaV1.342 channels was associated with heightened DHP sensitivity. In distinct contrast to the well-known modulation of DHP blockade of CaV1.2 channels, this study has therefore uncovered a novel mechanism for modulation of the pharmacologic properties of CaV1.3 channels through posttranscriptional modification of the C terminus.


Subject(s)
Alternative Splicing/genetics , Calcium Channel Blockers/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Dihydropyridines/metabolism , Protein Isoforms/genetics , Animals , Brain Chemistry/drug effects , Brain Chemistry/genetics , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , HEK293 Cells , Humans , Protein Isoforms/metabolism , Rats
14.
J Biol Chem ; 287(2): 832-47, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22069316

ABSTRACT

The Ca(V)1.4 voltage-gated calcium channel is predominantly expressed in the retina, and mutations to this channel have been associated with human congenital stationary night blindness type-2. The L-type Ca(V)1.4 channel displays distinct properties such as absence of calcium-dependent inactivation (CDI) and slow voltage-dependent inactivation (VDI) due to the presence of an autoinhibitory domain (inhibitor of CDI) in the distal C terminus. We hypothesized that native Ca(V)1.4 is subjected to extensive alternative splicing, much like the other voltage-gated calcium channels, and employed the transcript scanning method to identify alternatively spliced exons within the Ca(V)1.4 transcripts isolated from the human retina. In total, we identified 19 alternative splice variations, of which 16 variations have not been previously reported. Characterization of the C terminus alternatively spliced exons using whole-cell patch clamp electrophysiology revealed a splice variant that exhibits robust CDI. This splice variant arose from the splicing of a novel alternate exon (43*) that can be found in 13.6% of the full-length transcripts screened. Inclusion of exon 43* inserts a stop codon that truncates half the C terminus. The Ca(V)1.4 43* channel exhibited robust CDI, a larger current density, a hyperpolarized shift in activation potential by ∼10 mV, and a slower VDI. Through deletional experiments, we showed that the inhibitor of CDI was responsible for modulating channel activation and VDI, in addition to CDI. Calcium currents in the photoreceptors were observed to exhibit CDI and are more negatively activated as compared with currents elicited from heterologously expressed full-length Ca(V)1.4. Naturally occurring alternative splice variants may in part contribute to the properties of the native Ca(V)1.4 channels.


Subject(s)
Alternative Splicing/physiology , Calcium Channels, L-Type/biosynthesis , Calcium/metabolism , Ion Channel Gating/physiology , Membrane Potentials/physiology , Photoreceptor Cells, Vertebrate/metabolism , Calcium Channels, L-Type/genetics , HEK293 Cells , Humans , Organ Specificity/physiology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
15.
J Biol Chem ; 287(29): 24671-89, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22621926

ABSTRACT

The small hydrophobic (SH) protein is encoded by the human respiratory syncytial virus. Its absence leads to viral attenuation in the context of whole organisms, and it prevents apoptosis in infected cells. Herein, we have examined the structure of SH protein in detergent micelles and in lipid bilayers, by solution NMR and attenuated total reflection-Fourier transform infrared spectroscopy, respectively. We found that SH protein has a single α-helical transmembrane domain and forms homopentamers in several detergents. In detergent micelles, the transmembrane domain is flanked N-terminally by an α-helix that forms a ring around the lumen of the pore and C-terminally by an extended ß-turn. SH protein was found in the plasma membrane of transiently expressing HEK 293 cells, which showed pH-dependent (acid-activated) channel activity. Channel activity was abolished in mutants lacking both native His residues, His(22) and His(51), but not when either His was present. Herein, we propose that the pentameric model of SH protein presented is a physiologically relevant conformation, albeit probably not the only one, in which SH contributes to RSV infection and replication. Viroporins are short (∼100 amino acids) viral membrane proteins that form oligomers of a defined size, act as proton or ion channels, and in general enhance membrane permeability in the host. However, with some exceptions, their precise biological role of their channel activity is not understood. In general, viroporins resemble poorly specialized proteins but are nevertheless critical for viral fitness. In vivo, viruses lacking viroporins usually exhibit an attenuated or weakened phenotype, altered tropism, and diminished pathological effects. We have chosen to study the SH protein, 64 amino acids long, found in the human respiratory syncytial virus because of the effect of RSV on human health and the lack of adequate antivirals. We show that SH protein forms oligomers that behave as ion channels when activated at low pH. This study adds SH protein to a growing group of viroporins that have been structurally characterized. Although the precise biological role of this pentameric channel is still unknown, this report is nevertheless essential to fill some of the many gaps that exist in the understanding of SH protein function.


Subject(s)
Ion Channels/metabolism , Respiratory Syncytial Virus, Human/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Protein Multimerization/physiology , Spectroscopy, Fourier Transform Infrared
16.
Circ Res ; 109(11): 1250-8, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21998324

ABSTRACT

RATIONALE: Ca(V)1.2 channels are essential for excitation-contraction coupling in the cardiovascular system, and alternative splicing optimizes its role. Galectin-1 (Gal-1) has been reported to regulate vascular smooth muscle cell (VSMC) function and play a role in pulmonary hypertension. We have identified Gal-1 multiple times in yeast 2-hybrid assays using the Ca(V)1.2 I-II loop as bait. OBJECTIVE: Our hypothesis is that Gal-1 interacts directly with Ca(V)1.2 channel at the I-II loop to affect arterial constriction. METHODS AND RESULTS: Unexpectedly, Gal-1 was found to selectively bind to the I-II loop only in the absence of alternatively spliced exon 9*. We found that the current densities of Ca(V)1.2(Δ9*) channels were significantly inhibited as a result of decreased functional surface expression due to the binding of Gal-1 at the export signal located on the C-terminus of exon 9. Moreover, the suppression of Gal-1 expression by siRNA in rat A7r5 and isolated VSMCs produced the opposite effect of increased I(Ca,L). The physiological significance of Gal-1 mediated splice variant-specific inhibition of Ca(V)1.2 channels was demonstrated in organ bath culture where rat MAs were reversibly permeabilized with Gal-1 siRNA and the arterial wall exhibited increased K(+)-induced constriction. CONCLUSION: The above data indicated that Gal-1 regulates I(Ca,L) via decreasing the functional surface expression of Ca(V)1.2 channels in a splice variant selective manner and such a mechanism may play a role in modulating vascular constriction.


Subject(s)
Calcium Channels, L-Type/metabolism , Galectin 1/physiology , Muscle, Smooth/metabolism , Vasoconstriction/physiology , Animals , Barium/metabolism , Calcium/metabolism , Calcium Channels, L-Type/genetics , Exons/genetics , Gene Knockdown Techniques , Humans , Ion Channel Gating , Myocytes, Smooth Muscle/metabolism , Protein Binding , Protein Interaction Mapping , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA Splicing , Rats , Two-Hybrid System Techniques , Vasoconstriction/genetics
17.
J Biol Chem ; 286(25): 22186-94, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21525004

ABSTRACT

The prolongation of QT intervals in both mothers and fetuses during the later period of pregnancy implies that higher levels of progesterone may regulate the function of the human ether-a-go-go-related gene (HERG) potassium channel, a key ion channel responsible for controlling the length of QT intervals. Here, we studied the effect of progesterone on the expression, trafficking, and function of HERG channels and the underlying mechanism. Treatment with progesterone for 24 h decreased the abundance of the fully glycosylated form of the HERG channel in rat neonatal cardiac myocytes and HERG-HEK293 cells, a cell line stably expressing HERG channels. Progesterone also concentration-dependently decreased HERG current density, but had no effect on voltage-gated L-type Ca(2+) and K(+) channels. Immunofluorescence microscopy and Western blot analysis show that progesterone preferentially decreased HERG channel protein abundance in the plasma membrane, induced protein accumulation in the dilated endoplasmic reticulum (ER), and increased the protein expression of C/EBP homologous protein, a hallmark of ER stress. Application of 2-hydroxypropyl-ß-cyclodextrin (a sterol-binding agent) or overexpression of Rab9 rescued the progesterone-induced HERG trafficking defect and ER stress. Disruption of intracellular cholesterol homeostasis with simvastatin, imipramine, or exogenous application of cholesterol mimicked the effect of progesterone on HERG channel trafficking. Progesterone may impair HERG channel folding in the ER and/or block its trafficking to the Golgi complex by disrupting intracellular cholesterol homeostasis. Our findings may reveal a novel molecular mechanism to explain the QT prolongation and high risk of developing arrhythmias during late pregnancy.


Subject(s)
Cholesterol/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Homeostasis/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Potassium Channel Blockers/pharmacology , Progesterone/pharmacology , Animals , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetinae , Cricetulus , ERG1 Potassium Channel , Electric Conductivity , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Substrate Specificity , Temperature
18.
J Biol Chem ; 286(49): 42725-42735, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-21998309

ABSTRACT

Ca(V)1.3 channels are unique among the high voltage-activated Ca(2+) channel family because they activate at the most negative potentials and display very rapid calcium-dependent inactivation. Both properties are of crucial importance in neurons of the suprachiasmatic nucleus and substantia nigra, where the influx of Ca(2+) ions at subthreshold membrane voltages supports pacemaking function. Previously, alternative splicing in the Ca(V)1.3 C terminus gives rise to a long (Ca(V)1.3(42)) and a short form (Ca(V)1.3(42A)), resulting in a pronounced activation at more negative voltages and faster inactivation in the latter. It was further shown that the C-terminal modulator in the Ca(V)1.3(42) isoforms modulates calmodulin binding to the IQ domain. Using splice variant-specific antibodies, we determined that protein localization of both splice variants in different brain regions were similar. Using the transcript-scanning method, we further identified alternative splicing at four loci in the C terminus of Ca(V)1.3 channels. Alternative splicing of exon 41 removes the IQ motif, resulting in a truncated Ca(V)1.3 protein with diminished inactivation. Splicing of exon 43 causes a frameshift and exhibits a robust inactivation of similar intensity to Ca(V)1.3(42A). Alternative splicing of exons 44 and 48 are in-frame, altering interaction of the distal modulator with the IQ domain and tapering inactivation slightly. Thus, alternative splicing in the C terminus of Ca(V)1.3 channels modulates its electrophysiological properties, which could in turn alter neuronal firing properties and functions.


Subject(s)
Alternative Splicing , Calcium Channels, L-Type/chemistry , Calcium Channels/chemistry , Amino Acid Sequence , Animals , Brain/metabolism , Electrophysiology/methods , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Molecular Sequence Data , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Spinal Cord/metabolism
19.
Sci Bull (Beijing) ; 67(20): 2069-2075, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36546106

ABSTRACT

Topological phases of matter have been extensively investigated in solid-state materials and classical wave systems with integer dimensions. However, topological states in non-integer dimensions remain almost unexplored. Fractals, being self-similar on different scales, are one of the intriguing complex geometries with non-integer dimensions. Here, we demonstrate fractal higher-order topological states with unprecedented emergent phenomena in a Sierpinski acoustic metamaterial. We uncover abundant topological edge and corner states in the acoustic metamaterial due to the fractal geometry. Interestingly, the numbers of the edge and corner states depend exponentially on the system size, and the leading exponent is the Hausdorff fractal dimension of the Sierpinski carpet. Furthermore, the results reveal the unconventional spectrum and rich wave patterns of the corner states with consistent simulations and experiments. This study thus unveils unconventional topological states in fractal geometry and may inspire future studies of topological phenomena in non-Euclidean geometries.

20.
Hypertension ; 79(12): 2854-2866, 2022 12.
Article in English | MEDLINE | ID: mdl-36263779

ABSTRACT

BACKGROUND: L-type CaV1.2 calcium channel, the primary gateway for Ca2+ influx in smooth muscles, is widely regulated by multiple posttranslational modifications, such as protein kinase-mediated phosphorylation and nitric oxide-induced S-nitrosylation. However, the effect of S-nitrosylation on CaV1.2 channel function and its role in arterial contractility are not well understood. METHODS: Electrophysiological recordings, Ca2+ and confocal imaging, and biochemical assays were used to functionally characterize S-nitrosylated CaV1.2 channels in vitro, while pressure myography and tail-cuff blood pressure measurement were conducted to evaluate the physiological effects of CaV1.2 S-nitrosylation ex vivo and in vivo. RESULTS: S-nitrosylation significantly reduced the CaV1.2 current density by promoting lysosomal degradation that leads to decreased levels of total and surface CaV1.2 channel proteins in a CaVß-independent manner and reducing the open probability of CaV1.2 channel. Mechanistically, the Cys1180 and Cys1280 residues within CaV1.2 channel have been determined as the molecular targets for S-nitrosylation as substitution of either Cys1180 or Cys1280 for serine resulted in substantial reduction of S-nitrosylation levels. Of note, CaV1.2 S-nitrosylation levels were significantly reduced in arteries isolated from both spontaneously hypertensive rats and patients with pulmonary hypertension. Moreover, mouse resistance arteries incubated with S-nitrosocysteine displayed much lower contractility and spontaneously hypertensive rats injected with S-nitrosocysteine also showed significantly reduced blood pressure, suggesting that reduced S-nitrosylation contributes to the upregulation of CaV1.2 channel activity in hypertensive arteries. CONCLUSIONS: This study provides strong evidence that S-nitrosylation-mediated downregulation of CaV1.2 channels is via 2 distinctive mechanisms and the findings offer potential pathways for therapeutic inventions in hypertension.


Subject(s)
Hypertension , Vasoconstriction , Rats , Mice , Animals , Rats, Inbred SHR , Nitric Oxide/metabolism , Muscle, Smooth, Vascular/metabolism , Calcium Channels, L-Type/metabolism , Probability
SELECTION OF CITATIONS
SEARCH DETAIL