Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Phys Rev Lett ; 126(3): 036802, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33543950

ABSTRACT

Topological superconductivity holds promise for fault-tolerant quantum computing. While planar Josephson junctions are attractive candidates to realize this exotic state, direct phase measurements as the fingerprint of the topological transition are missing. By embedding two gate-tunable Al/InAs Josephson junctions in a loop geometry, we measure a π jump in the junction phase with an increasing in-plane magnetic field B_{∥}. This jump is accompanied by a minimum of the critical current, indicating a closing and reopening of the superconducting gap, strongly anisotropic in B_{∥}. Our theory confirms that these signatures of a topological transition are compatible with the emergence of Majorana bound states.

2.
Nano Lett ; 21(5): 1915-1920, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33617256

ABSTRACT

Epitaxial Al-InAs heterostructures appear as a promising materials platform for exploring mesoscopic and topological superconductivity. A unique property of Josephson junction field effect transistors (JJ-FETs) fabricated on these heterostructures is the ability to tune the supercurrent using a metallic gate. Here, we report the fabrication and measurement of gate-tunable Al-InAs JJ-FETs in which the gate dielectric in contact with the InAs is produced by mechanically exfoliated hexagonal boron nitride (h-BN) followed by dry transfer. We discuss a versatile fabrication process that enables compatibility between layered material transfer and Al-InAs heterostructures that allows us to achieve full gate-tunability of supercurrent by using only 5 nm thick h-BN flakes. Our study shows that pristine properties of epitaxial Josephson junctions, such as product of normal resistance and critical current, IcRn, are preserved. Furthermore, complementary measurements confirm that using h-BN dielectric changes the channel density less when compared to atomic layer deposition of Al2O3.

3.
Nat Commun ; 12(1): 78, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33397966

ABSTRACT

Josephson junctions hosting Majorana fermions have been predicted to exhibit a 4π periodic current phase relation. One experimental consequence of this periodicity is the disappearance of odd steps in Shapiro steps experiments. Experimentally, missing odd Shapiro steps have been observed in a number of materials systems with strong spin-orbit coupling and have been interpreted in the context of topological superconductivity. Here we report on missing odd steps in topologically trivial Josephson junctions fabricated on InAs quantum wells. We ascribe our observations to the high transparency of our junctions allowing Landau-Zener transitions. The probability of these processes is shown to be independent of the drive frequency. We analyze our results using a bi-modal transparency distribution which demonstrates that only few modes carrying 4π periodic current are sufficient to describe the disappearance of odd steps. Our findings highlight the elaborate circumstances that have to be considered in the investigation of the 4π Josephson junctions in relationship to topological superconductivity.

4.
Nat Commun ; 11(1): 212, 2020 Jan 10.
Article in English | MEDLINE | ID: mdl-31924783

ABSTRACT

In a standard Josephson junction the current is zero when the phase difference between superconducting leads is zero. This condition is protected by parity and time-reversal symmetries. However, the combined presence of spin-orbit coupling and magnetic field breaks these symmetries and can lead to a finite supercurrent even when the phase difference is zero. This is the so called anomalous Josephson effect-the hallmark effect of superconducting spintronics-which can be characterized by the corresponding anomalous phase shift. Here we report the observation of a tunable anomalous Josephson effect in InAs/Al Josephson junctions measured via a superconducting quantum interference device. By gate controlling the density of InAs, we are able to tune the spin-orbit coupling in the Josephson junction. This gives us the ability to tune the anomalous phase, and opens new opportunities for superconducting spintronics, and new possibilities for realizing and characterizing topological superconductivity.

5.
Sci Rep ; 9(1): 8820, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217439

ABSTRACT

Neuropeptide release in the brain has traditionally been difficult to observe. Existing methods lack temporal and spatial resolution that is consistent with the function and size of neurons. We use cultured "sniffer cells" to improve the temporal and spatial resolution of observing neuropeptide release. Sniffer cells were created by stably transfecting Chinese Hamster Ovary (CHO) cells with plasmids encoding the rat angiotensin type 1a receptor and a genetically encoded Ca2+ sensor. Isolated, cultured sniffer cells showed dose-dependent increases in fluorescence in response to exogenously applied angiotensin II and III, but not other common neurotransmitters. Sniffer cells placed on the median preoptic nucleus (a presumptive site of angiotensin release) displayed spontaneous activity and evoked responses to either electrical or optogenetic stimulation of the subfornical organ. Stable sniffer cell lines could be a viable method for detecting neuropeptide release in vitro, while still being able to distinguish differences in neuropeptide concentration.


Subject(s)
Angiotensin II/metabolism , Neurons/metabolism , Animals , Brain/metabolism , CHO Cells , Cricetinae , Cricetulus , Fluorescence , Male , Optogenetics , Rats, Sprague-Dawley
6.
J Neuroendocrinol ; 31(8): e12752, 2019 08.
Article in English | MEDLINE | ID: mdl-31136029

ABSTRACT

Salt-loading (SL) impairs GABAA inhibition of arginine vasopressin (AVP) neurones in the supraoptic nucleus (SON) of the hypothalamus. Based on previous studies, we hypothesised that SL activates tyrosine receptor kinase B (TrkB), down-regulating the activity of K+ /Cl- co-transporter2 (KCC2) and up-regulating Na+ /K+ /Cl- co-transporter1 (NKCC1). These changes in chloride transport would result in increased [Cl- ]i in SON AVP neurones. The study combined virally-mediated chloride imaging with ClopHensorN with a single-cell western blot analysis. An adeno-associated virus with ClopHensorN and a vasopressin promoter (AAV2-0VP1-ClopHensorN) was bilaterally injected in the SON of adult male Sprague-Dawley rats that were either euhydrated (Eu) or salt-loaded (SL) for 7 days. Acutely dissociated SON neurones expressing ClopHensorN were tested for decreases or increases in [Cl- ]i in response to focal application of the GABAA agonist muscimol (100 µmol L-1 ). SON AVP neurones from Eu rats showed muscimol-induced chloride influx (P < 0.05;23/35). SON AVP neurones from SL rats either significantly increased chloride efflux (P < 0.05;27/39) or did not change chloride flux (12/39). The SON AVP neurones that responded to muscimol appeared to be viable and expressed KCC2 and ß-actin. Neurones that did not respond during chloride imaging did not show KCC2 and ß-actin protein expression. The KCC2 antagonist (VU0240551,10 µmol L-1 ) significantly blocked the chloride influx in cells from Eu rats but did not affect cells from SL rats. A NKCC1 antagonist (bumetanide,10 µmol L-1 ) significantly blocked the chloride efflux in cells from SL rats but had no effect on cells from Eu rats. Blocking NKCC1 using bumetanide had less of an effect on the muscimol-induced Cl- influx in Eu rat neurones compared to the KCC2 antagonist. The TrkB antagonist (AnA-12) (50 µmol L-1 ) and protein kinase inhibitor (K252a) (100 nmol L-1 ) each significantly blocked chloride efflux in SON AVP neurones from SL rats. Salt-loading increases [Cl- ]i in SON AVP neurones via a TrKB-KCC2-NKCC1-dependent mechanism in rats.


Subject(s)
Arginine Vasopressin/metabolism , Neurons/drug effects , Sodium Chloride/pharmacology , Supraoptic Nucleus/drug effects , Animals , Arginine Vasopressin/genetics , Biosensing Techniques , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Male , Neurons/cytology , Neurons/metabolism , Optical Imaging/methods , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Supraoptic Nucleus/diagnostic imaging , Supraoptic Nucleus/metabolism
7.
Skeletal Radiol ; 47(5): 661-669, 2018 May.
Article in English | MEDLINE | ID: mdl-29218391

ABSTRACT

OBJECTIVE: To determine the diagnostic yield of CT-guided percutaneous biopsy of densely sclerotic bone lesions. MATERIALS AND METHODS: We retrospectively analyzed CT-guided percutaneous bone biopsies performed at our institution from September 2008 through August 2011 (329 cases) and from September 2012 through August 2015 (324 cases) after adoption of a battery-powered drill system (OnControl). Bone lesions were included in the analysis if they were >70% sclerotic by visual inspection, had a density > 2 times that of adjacent trabecular bone, and had an attenuation of ≥250 HU. Pathological fractures, diskitis-osteomyelitis, and osteoid osteomas were excluded. Eligible cases were characterized by lesion location, maximum lesion diameter, mean density, biopsy needle type and gauge, reported complications, and histological diagnosis. Clinical and imaging follow-up was used to confirm histological diagnosis. Cases in which a benign histological diagnosis could not be confirmed by imaging over a minimum period of 1 year were excluded. RESULTS: A total of 37 biopsies of sclerotic bone lesions met the inclusion criteria, 17 of which were performed with a power drill needle and 20 of which were performed with a manually driven needle. The mean lesion density was 604.1 HU. The overall diagnostic yield was 78.4%; overall diagnostic accuracy was 94.6%, and the false-negative rate was 5.4%. Diagnostic yield and accuracy were 82.4% and 100% respectively, with a power drill and 75% and 90% respectively, with a manual device. Diagnostic yield for lesions ≥700 HU was 90% (9 out of 10). CONCLUSION: Densely sclerotic bone lesions are amenable to percutaneous needle biopsy.


Subject(s)
Bone Diseases/pathology , Image-Guided Biopsy/methods , Radiography, Interventional/methods , Tomography, X-Ray Computed/methods , Adult , Aged , Aged, 80 and over , Bone Diseases/diagnostic imaging , Bone Diseases/surgery , Female , Humans , Male , Middle Aged , Retrospective Studies
8.
Skeletal Radiol ; 47(1): 107-116, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28952012

ABSTRACT

PURPOSE: To compare diagnostic performance of a 5-min knee MRI protocol to that of a standard knee MRI. MATERIALS AND METHODS: One hundred 3 T (100 patients, mean 38.8 years) and 50 1.5 T (46 patients, mean 46.4 years) MRIs, consisting of 5 fast, 2D multi-planar fast-spin-echo (FSE) sequences and five standard multiplanar FSE sequences, from two academic centers (1/2015-1/2016), were retrospectively reviewed by four musculoskeletal radiologists. Agreement between fast and standard (interprotocol agreement) and between standard (intraprotocol agreement) readings for meniscal, ligamentous, chondral, and bone pathology was compared for interchangeability. Frequency of major findings, sensitivity, and specificity was also tested for each protocol. RESULTS: Interprotocol agreement using fast MRI was similar to intraprotocol agreement with standard MRI (83.0-99.5%), with no excess disagreement (≤ 1.2; 95% CI, -4.2 to 3.8%), across all structures. Frequency of major findings (1.1-22.4% across structures) on fast and standard MRI was not significantly different (p ≥ 0.215), except more ACL tears on fast MRI (p = 0.021) and more cartilage defects on standard MRI (p < 0.001). Sensitivities (59-100%) and specificities (73-99%) of fast and standard MRI were not significantly different for meniscal and ligament tears (95% CI for difference, -0.08-0.08). For cartilage defects, fast MRI was slightly less sensitive (95% CI for difference, -0.125 to -0.01) but slightly more specific (95% CI for difference, 0.01-0.5) than standard MRI. CONCLUSION: A fast 5-min MRI protocol is interchangeable with and has similar accuracy to a standard knee MRI for evaluating internal derangement of the knee.


Subject(s)
Knee Injuries/diagnostic imaging , Magnetic Resonance Imaging/methods , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Retrospective Studies , Sensitivity and Specificity
9.
Sci Rep ; 7(1): 5075, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698564

ABSTRACT

The molecular components of store-operated Ca2+ influx channels (SOCs) in proliferative and migratory vascular smooth muscle cells (VSMCs) are quite intricate with many channels contributing to SOCs. They include the Ca2+-selective Orai1 and members of the transient receptor potential canonical (TRPC) channels, which are activated by the endoplasmic reticulum Ca2+ sensor STIM1. The scaffolding protein Homer assembles SOC complexes, but its role in VSMCs is not well understood. Here, we asked whether these SOC components and Homer1 are present in the same complex in VSMCs and how Homer1 contributes to VSMC SOCs, proliferation, and migration leading to neointima formation. Homer1 expression levels are upregulated in balloon-injured vs. uninjured VSMCs. Coimmunoprecipitation assays revealed the presence and interaction of all SOC components in the injured VSMCs, where Homer1 interacts with Orai1 and various TRPC channels. Accordingly, knockdown of Homer1 in cultured VSMCs partially inhibited SOCs, VSMC migration, and VSMC proliferation. Neointimal area was reduced after treatment with an adeno-associated viral vector expressing a short hairpin RNA against Homer1 mRNA (AAV-shHomer1). These findings stress the role of multiple Ca2+ influx channels in VSMCs and are the first to show the role of Homer proteins in VSMCs and its importance in neointima formation.


Subject(s)
Cell Movement , Homer Scaffolding Proteins/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima/pathology , ORAI1 Protein/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Calcium Signaling , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Proliferation , Cells, Cultured , Gene Knockdown Techniques , Male , Protein Binding , Rats, Sprague-Dawley
10.
Radiol Clin North Am ; 54(5): 801-15, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27545421

ABSTRACT

Repetitive, high-velocity overhead throwing can lead to several adaptive changes in the throwing shoulder, which over time lead to structural microtrauma and eventually overt injury. MR imaging is a useful imaging modality to evaluate these changes and to characterize their acuity and severity. Understanding the throwing motion and the effects of this motion on the structures of the shoulder can help radiologists to recognize these findings and provide useful information to referring physicians, which may affect the treatment of these athletes. This article reviews shoulder pathomechanics and MR imaging findings in overhead throwing athletes.


Subject(s)
Athletic Injuries/diagnostic imaging , Baseball/injuries , Cumulative Trauma Disorders/diagnostic imaging , Magnetic Resonance Imaging/methods , Shoulder Injuries/diagnostic imaging , Soft Tissue Injuries/diagnostic imaging , Diagnosis, Differential , Humans , Shoulder Joint/diagnostic imaging
13.
Am J Physiol Renal Physiol ; 306(9): F1069-80, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24623143

ABSTRACT

The present study was conducted to determine whether and how store-operated Ca(2+) entry (SOCE) in glomerular mesangial cells (MCs) was altered by high glucose (HG) and diabetes. Human MCs were treated with either normal glucose or HG for different time periods. Cyclopiazonic acid-induced SOCE was significantly greater in the MCs with 7-day HG treatment and the response was completely abolished by GSK-7975A, a selective inhibitor of store-operated Ca(2+) channels. Similarly, the inositol 1,4,5-trisphosphate-induced store-operated Ca(2+) currents were significantly enhanced in the MCs treated with HG for 7 days, and the enhanced response was abolished by both GSK-7975A and La(3+). In contrast, receptor-operated Ca(2+) entry in MCs was significantly reduced by HG treatment. Western blotting showed that HG increased the expression levels of STIM1 and Orai1 in cultured MCs. A significant HG effect occurred at a concentration as low as 10 mM, but required a minimum of 7 days. The HG effect in cultured MCs was recapitulated in renal glomeruli/cortex of both type I and II diabetic rats. Furthermore, quantitative real-time RT-PCR revealed that a 6-day HG treatment significantly increased the mRNA expression level of STIM1. However, the expressions of STIM2 and Orai1 transcripts were not affected by HG. Taken together, these results suggest that HG/diabetes enhanced SOCE in MCs by increasing STIM1/Orai1 protein expressions. HG upregulates STIM1 by promoting its transcription but increases Orai1 protein through a posttranscriptional mechanism.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Diabetic Nephropathies/metabolism , Glucose/metabolism , Ion Channel Gating , Mesangial Cells/metabolism , Animals , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Signaling/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/genetics , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Ion Channel Gating/drug effects , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesangial Cells/drug effects , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , ORAI1 Protein , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Stromal Interaction Molecule 1 , Time Factors , Transcriptional Activation , Up-Regulation
14.
J Biol Chem ; 289(10): 6372-6382, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24464579

ABSTRACT

Transient receptor potential canonical (TRPC) channels mediate a critical part of the receptor-evoked Ca(2+) influx. TRPCs are gated open by the endoplasmic reticulum Ca(2+) sensor STIM1. Here we asked which stromal interaction molecule 1 (STIM1) and TRPC domains mediate the interaction between them and how this interaction is used to open the channels. We report that the STIM1 Orai1-activating region domain of STIM1 interacts with the TRPC channel coiled coil domains (CCDs) and that this interaction is essential for opening the channels by STIM1. Thus, disruption of the N-terminal (NT) CCDs by triple mutations eliminated TRPC surface localization and reduced binding of STIM1 to TRPC1 and TRPC5 while increasing binding to TRPC3 and TRPC6. Single mutations in TRPC1 NT or C-terminal (CT) CCDs reduced interaction and activation of TRPC1 by STIM1. Remarkably, single mutations in the TRPC3 NT CCD enhanced interaction and regulation by STIM1. Disruption in the TRPC3 CT CCD eliminated regulation by STIM1 and the enhanced interaction caused by NT CCD mutations. The NT CCD mutations converted TRPC3 from a TRPC1-dependent to a TRPC1-independent, STIM1-regulated channel. TRPC1 reduced the FRET between BFP-TRPC3 and TRPC3-YFP and between CFP-TRPC3-YFP upon stimulation. Accordingly, knockdown of TRPC1 made TRPC3 STIM1-independent. STIM1 dependence of TRPC3 was reconstituted by the TRPC1 CT CCD alone. Knockout of Trpc1 and Trpc3 similarly inhibited Ca(2+) influx, and inhibition of Trpc3 had no further effect on Ca(2+) influx in Trpc1(-/-) cells. Cell stimulation enhanced the formation of Trpc1-Stim1-Trpc3 complexes. These findings support a model in which the TRPC3 NT and CT CCDs interact to shield the CT CCD from interaction with STIM1. The TRPC1 CT CCD dissociates this interaction to allow the STIM1 Orai1-activating region within STIM1 access to the TRPC3 CT CCD and regulation of TRPC3 by STIM1. These studies provide evidence that the TRPC channel CCDs participate in channel gating.


Subject(s)
Ion Channel Gating , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Interaction Domains and Motifs , TRPC Cation Channels/metabolism , Animals , Calcium Channels/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , ORAI1 Protein , RNA Interference , Stromal Interaction Molecule 1 , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics
15.
J Cell Biol ; 202(1): 71-9, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23816623

ABSTRACT

Ca(2+) influx by store-operated Ca(2+) channels (SOCs) mediates all Ca(2+)-dependent cell functions, but excess Ca(2+) influx is highly toxic. The molecular components of SOC are the pore-forming Orai1 channel and the endoplasmic reticulum Ca(2+) sensor STIM1. Slow Ca(2+)-dependent inactivation (SCDI) of Orai1 guards against cell damage, but its molecular mechanism is unknown. Here, we used homology modeling to identify a conserved STIM1(448-530) C-terminal inhibitory domain (CTID), whose deletion resulted in spontaneous clustering of STIM1 and full activation of Orai1 in the absence of store depletion. CTID regulated SCDI by determining access to and interaction of the STIM1 inhibitor SARAF with STIM1 Orai1 activation region (SOAR), the STIM1 domain that activates Orai1. CTID had two lobes, STIM1(448-490) and STIM1(490-530), with distinct roles in mediating access of SARAF to SOAR. The STIM1(448-490) lobe restricted, whereas the STIM1(490-530) lobe directed, SARAF to SOAR. The two lobes cooperated to determine the features of SCDI. These findings highlight the central role of STIM1 in SCDI and provide a molecular mechanism for SCDI of Orai1.


Subject(s)
Calcium Channels/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Amino Acid Sequence , Calcium/metabolism , Calcium Channels/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Conserved Sequence , HEK293 Cells , Humans , Intracellular Calcium-Sensing Proteins , Membrane Proteins/genetics , Models, Molecular , Neoplasm Proteins/genetics , ORAI1 Protein , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Sequence Deletion , Stromal Interaction Molecule 1 , Structural Homology, Protein , Structure-Activity Relationship
16.
J Biol Chem ; 288(18): 12852-65, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23525112

ABSTRACT

This study was carried out to explore the molecular mechanism for down-regulation of TRPC6 expression in the reactive oxygen species (ROS)/PKC signaling in kidney cells. In cultured human mesangial cells, H2O2 and TNF-α inhibited TRPC6 mRNA expression in a time-dependent manner. Inhibition of NF-κB reversed both H2O2- and phorbol 12-myristate 13-acetate (PMA)-induced decrease in TRPC6 protein expression. Activation of NF-κB by knocking down IκBα using siRNA could mimic the suppressive effect of ROS/PKC on TRPC6. a Ca(2+) imaging study showed that activation and inhibition of NF-κB significantly decreased and increased the TRPC6-mediated Ca(2+) entry, respectively. Further experiments showed that PMA, but not its inactive analog 4α-phorbol 12, 13-didecanoate (4α-PDD), caused phosphorylation of IκBα and stimulated the nuclear translocation of NF-κB p50 and p65 subunits. The PMA-dependent IκBα phosphorylation was significantly inhibited by Gö6976. Electrophoretic mobility shift assay revealed that PMA stimulated DNA binding activity of NF-κB. Furthermore, specific knockdown of p65, but not p50, prevented an H2O2 inhibitory effect on TRPC6 protein expression, suggesting p65 as a predominant NF-κB subunit repressing TRPC6. In agreement with a major role of p65, chromatin immunoprecipitation assays showed that PMA treatment induced p65 binding to the TRPC6 promoter. Moreover, PMA treatment increased the association of p65 with histone deacetylase (HDAC) and decreased histone acetylation at the TRPC6 promoter. Consistently, knockdown of HDAC2 by siRNA or inhibition of HDAC with trichostatin A prevented a H2O2-induced decrease in TRPC6 mRNA and protein expressions, respectively. Taken together, our findings imply an important role of NF-κB in a negative regulation of TRPC6 expression at the gene transcription level in kidney cells.


Subject(s)
Gene Expression Regulation/drug effects , Hydrogen Peroxide/pharmacology , Kidney/metabolism , NF-kappa B p50 Subunit/metabolism , Oxidants/pharmacology , Protein Kinase C/metabolism , TRPC Cation Channels/biosynthesis , Transcription Factor RelA/metabolism , Carbazoles/pharmacology , Carcinogens/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Kidney/cytology , NF-kappa B p50 Subunit/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Response Elements/physiology , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor RelA/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
18.
Exp Biol Med (Maywood) ; 237(2): 111-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22282397

ABSTRACT

Canonical transient receptor potential (TRPC) channel proteins have been identified as downstream molecules in a G protein-coupled receptor signaling pathway and are involved in a variety of cell functions due to their ability to regulate intracellular calcium signaling. TRPC channel physiology has been an increasingly interesting and relevant topic over the last decade, and the outcomes from various studies have advanced our understanding of TRPC function in the normal state. Recently, attention has turned to whether or not TRPC proteins are implicated in diseases. Emerging evidence suggests a significant contribution of several isoforms of TRPC proteins to cardiovascular as well as renal diseases. This review focuses on the implication of TRPC proteins as they pertain to diabetes. We summarize the recent findings by other investigators as well as ourselves and additionally discuss the important role of TRPC proteins in the development of various diabetic complications, such as diabetic nephropathy and diabetic vasculopathy. The underlying mechanisms which contribute to these complications are also outlined. Lastly, we elaborate on the role of TRPC proteins as a potential therapeutic target for treating diabetes-associated diseases.


Subject(s)
Diabetes Mellitus/metabolism , Receptors, G-Protein-Coupled/metabolism , TRPC Cation Channels/metabolism , Animals , Atherosclerosis/metabolism , Calcium/chemistry , Calcium/metabolism , Calcium Signaling , Diabetes Complications/metabolism , Diabetes Complications/pathology , Glucose/metabolism , Humans , Models, Biological , Protein Isoforms/chemistry , Reactive Oxygen Species , Signal Transduction , TRPC Cation Channels/physiology
19.
Mol Pharmacol ; 81(4): 510-26, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22210847

ABSTRACT

We addressed the requirement for stromal interaction molecule 1 (STIM1), the endoplasmic reticulum (ER) Ca(2+)-sensor, and Orai1, a Ca(2+) selective channel, in regulating Ca(2+) entry through the store-operated channels mouse transient receptor potential canonical (TRPC) 4 or human TRPC1. Studies were made using murine and human lung endothelial cells (ECs) challenged with thrombin known to induce Ca(2+) entry via TRPC1/4. Deletion or knockdown of TRPC4 abolished Ca(2+) entry secondary to depletion of ER Ca(2+) stores, preventing the disruption of the endothelial barrier. Knockdown of STIM1 (but not of Orai1or Orai3) or expression of the dominant-negative STIM1(K684E-K685E) mutant in ECs also suppressed Ca(2+) entry secondary to store depletion. Ectopic expression of WT-STIM1 or WT-Orai1 in TRPC4(-/-)-ECs failed to rescue Ca(2+) entry; however, WT-TRPC4 expression in TRPC4(-/-)-ECs restored Ca(2+) entry indicating the requirement for TRPC4 in mediating store-operated Ca(2+) entry. Moreover, expression of the dominant-negative Orai1(R91W) mutant or Orai3(E81W) mutant in WT-ECs failed to prevent thrombin-induced Ca(2+) entry. In contrast, expression of the dominant-negative TRPC4(EE647-648KK) mutant in WT-ECs markedly reduced thrombin-induced Ca(2+) entry. In ECs expressing YFP-STIM1, ER-store Ca(2+) depletion induced formation of fluorescent membrane puncta in WT but not in TRPC4(-/-) cells, indicating that mobilization of STIM1 and engagement of its Ca(2+) sensing function required TRPC4 expression. Coimmunoprecipitation studies showed coupling of TRPC1 and TRPC4 with STIM1 on depletion of ER Ca(2+) stores. Thus, TRPC1 and TRPC4 can interact with STIM1 to form functional store-operated Ca(2+)-entry channels, which are essential for mediating Ca(2+) entry-dependent disruption of the endothelial barrier.


Subject(s)
Calcium/metabolism , Endothelium, Vascular/metabolism , Membrane Glycoproteins/physiology , TRPC Cation Channels/physiology , Animals , Blotting, Western , Calcium Channels , Cells, Cultured , Endothelium, Vascular/cytology , Mice , Mice, Knockout , RNA, Small Interfering , Stromal Interaction Molecule 1
20.
Cell Signal ; 24(4): 899-906, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22201561

ABSTRACT

The transient receptor potential (TRPC) family of Ca²âº permeable, non-selective cation channels is abundantly expressed in the brain, and can function as store-operated (SOC) and store-independent channels depending on their interaction with the ER Ca²âº sensor STIM1. TRPC1 and TRPC5 have critical roles in neurite outgrowth, however which of their functions regulate neurite outgrowth is unknown. In this study, we investigated the effects of TRPC channels and their STIM1-induced SOC activity on neurite outgrowth of PC12 cells. We report that PC12 cell differentiation down-regulates TRPC5 expression, whereas TRPC1 expression is retained. TRPC1 and TRPC5 interact with STIM1 through the STIM1 ERM domain. Transfection of TRPC1 and TRPC5 increased the receptor-activated Ca²âº influx that was markedly augmented by the co-expression of STIM1. Topical expression of TRPC1 in PC12 cells markedly increased neurite outgrowth while that of TRPC5 suppressed neurite outgrowth. Suppression of neurite outgrowth by TRPC5 requires the channel function of TRPC5. However, strikingly, multiple lines of evidence show that the TRPC1-induced neurite outgrowth was independent of TRPC1-mediated Ca²âº influx. Thus, a) TRPC1 and TRPC5 similarly increased Ca²âº influx but only TRPC1 induced neurite outgrowth, b) the constitutively STIM1(D76A) mutant that activates Ca²âº influx by TRPC and Orai channels did not increase neurite outgrowth, c) co-expression of TRPC5 with TRPC1 suppressed the effect of TRPC1 on neurite outgrowth, d) and most notable, channel-dead pore mutant of TRPC1 increased neurite outgrowth to the same extent as TRPC1(WT). Suppression of TRPC1-induced neurite outgrowth by TRPC5 was due to a marked reduction in the surface expression of TRPC1. We conclude that the regulation of neurite outgrowth by TRPC1 is independent of Ca²âº influx and TRPC1-promoted neurite outgrowth depends on the surface expression of TRPC1. It is likely that TRPC1 acts as a scaffold at the cell surface to assemble a signaling complex to stimulate neurite outgrowth.


Subject(s)
Calcium/metabolism , Neurites/physiology , Signal Transduction/genetics , TRPC Cation Channels/genetics , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Differentiation , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Proliferation , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Ion Channel Gating , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neurogenesis , PC12 Cells , Protein Structure, Tertiary , Rats , Stromal Interaction Molecule 1 , TRPC Cation Channels/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...