Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Joseph R. Hume is active.

Publication


Featured researches published by Joseph R. Hume.


Nature | 1997

Molecular identification of a volume-regulated chloride channel

Dayue Duan; Cathy Winter; Suzanne Cowley; Joseph R. Hume; Burton Horowitz

A volume-regulated chloride current ( I Cl.vol) is ubiquitously present in mammalian cells, and is required for the regulation of electrical activity, cell volume, intracellular pH, immunological responses, cell proliferation and differentiation. However, the molecule responsible for I Cl.vol has yet to be determined. Although three putative chloride channel proteins expressed from cloned genes (P-glycoprotein, p I Cln (ref. 5) and ClC-2 (ref. 6)) have been proposed to be the molecular equivalent of I Cl.vol, neither P-glycoprotein nor p I Cln is thought to be a chloride channel or part thereof,, and the properties of expressed ClC-2 channels differ from native I Cl.vol (refs. 3, 6). Here we report that functional expression in NIH/3T3 cells of a cardiac clone of another member of the ClC family, ClC-3, results in a large basally active chloride conductance, which is strongly modulated by cell volume and exhibits many properties identical to those of I Cl.vol in native cells,. A mutation of asparagine to lysine at position 579 at the end of the transmembrane domains of ClC-3 abolishes the outward rectification and changes the anion selectivity from I− > Cl− to Cl− > I− but leaves swelling activation intact. Because ClC-3 is a channel protein belonging to a large gene family of chloride channels,, these results indicate that ClC-3 encodes I Cl.vol in many native mammalian cells.


Circulation Research | 1995

[Ca2+]i Inhibition of K+ Channels in Canine Pulmonary Artery : Novel Mechanism for Hypoxia-Induced Membrane Depolarization

Joseph M. Post; Craig H. Gelband; Joseph R. Hume

Experiments were performed on smooth muscle cells isolated from canine pulmonary artery to identify the type of K+ channel modulated by hypoxia and examine the possible role of [Ca2+]i in hypoxic K+ channel inhibition. Whole-cell patch-clamp experiments revealed that hypoxia (induced by the O2 scavenger, sodium dithionite) reduced macroscopic K+ currents, an effect that could be prevented by strong intracellular buffering of [Ca2+]i. The inhibitory effects of hypoxia were mimicked by acute exposure of cells to caffeine and could be prevented by caffeine pretreatment, suggesting an important obligatory role of [Ca2+]i in hypoxic inhibition of K+ currents. Exposure of cells to low concentrations of 4-aminopyridine (4-AP, 1 mmol/L) prevented hypoxic inhibition of macroscopic K+ currents, whereas low concentrations of tetraethylammonium were without effect, suggesting that the target K+ channel inhibited by hypoxia is a voltage-dependent delayed rectifier K+ channel, which is inhibited by [Ca2+]i. Hypoxia failed to consistently modify the activity of large-conductance (118 picosiemens [pS] in physiological K+) Ca(2+)-activated K+ channels in inside-out membrane patches but reduced open probability of smaller-conductance (25-pS) delayed rectifier K+ channels in cell-attached membrane patches. In inside-out membrane patches, 1 mumol/L Ca2+ added to the cytoplasmic surface significantly reduced open probability of small-conductance (25-pS) 4-AP-sensitive delayed rectifier K+ channels. Whole-cell current measurements using symmetrical K+ to increase driving force for small currents active near the cells resting membrane potential revealed the presence of a 4-AP-sensitive K+ current that activated near -65 mV and was inhibited by hypoxia.(ABSTRACT TRUNCATED AT 250 WORDS)


British Journal of Pharmacology | 1997

Prominent role of intracellular Ca2+ release in hypoxic vasoconstriction of canine pulmonary artery

Rita Jabr; Helen Toland; Craig H. Gelband; Xiao Xia Wang; Joseph R. Hume

1 The possible role of sarcoplasmic reticulum (SR) Ca2+ stores in hypoxic pulmonary vasoconstriction (HPV) is not well understood. In order to assess the possible role of intracellular Ca2+ release from SR Ca2+ stores in HPV, we examined the effects of: (1) ryanodine (10 μM) depletion of intracellular Ca2+ stores, and (2) thapsigargin (THAPS, 2 μM) or cyclopiazonic acid (CPA, 10 μM) depletion of intracellular Ca2+ stores on HPV in canine pulmonary artery. 2 Isometric tension was measured from arterial ring suspended in Krebs‐Henseliet solution (K‐H) bubbled with 95%O2/5%CO2. Hypoxia was induced by bubbling phenylephrine (PE, 1 μM) precontracted rings with 95%N2/5%CO2. HPV was observed in both intact and endothelial‐denuded arteries and expressed as % of maximal KCl contraction (%Tkmax)=21.3±3.2%; n=13 and 21.7±4%; n=4, respectively. 3 When SR caffeine sensitive Ca2+ stores were depleted by pretreatment with ryanodine and brief caffeine (15 mM) exposure, the hypoxic response was signifcantly reduced to 19.1±9.2% of the control hypoxic contraction (n=7; P<0.001) with little or no effect on PE or KCl contractions. On the other hand, in normoxic rings pretreated with THAPS or CPA, the PE responses were significantly reduced (%Tkmax=18.2±3.1% compared to 39.0±3.9% in control; n=16; P<0.001; %Tkmax=3.4±1.6% compared to 49.9±7.9% in control; n=6; P<0.001; respectively) with no significant effect on caffeine‐induced contractions, suggesting that both THAPS and CPA preferentially deplete InsP3‐sensitive Ca2+ stores, without affecting the caffeine‐sensitive Ca2+ store; consistent with the existence of separate and independent InsP3 and caffeine‐sensitive Ca2+ stores in this preparation. 4 When hypoxia was induced in the presence of THAPS or CPA, developed tension was significantly larger than control (%Tkmax=64.5±6.0%; n=16; P<0.05%; %Tkmax=78.2±15%; n=6; P<0.05; respectively), was partially blocked by nisoldipine (10 μM) and ryanodine (%Tkmax=20.3±3.7%; n=6), and nearly completely blocked by SK&F 96365 (50 μM). However, the actions of SK&F 96365 appeared to be nonselective since this compound also significantly reduced contractions elicited by KCl, PE and caffeine. 5 Finally, evidence was obtained suggesting: (a) that at least some of the Ca2+ released from the caffeine‐ and ryanodine‐sensitive Ca2+ stores by hypoxia may be taken up and buffered by the InsP3‐sensitive Ca2+ stores, and (b) the apparent dependence of HPV on extracellular Ca2+ entry pathways may be partially due to the dependence of the Ca2+ content of intracellular SR Ca2+ stores on sarcolemmal Ca2+ entry pathways. 6 These data suggest that caffeine‐ and ryanodine‐sensitive SR Ca2+ stores contribute significantly to HPV under normal conditions and, in the presence of THAPS or CPA, an additional nisoldipine‐ and ryanodine‐insensitive Ca2+ entry pathway is evoked by hypoxia.


Circulation Research | 1993

Regulation of Ca2+ channels by cAMP and cGMP in vascular smooth muscle cells.

Tomohisa Ishikawa; Joseph R. Hume; Kathleen D. Keef

Whole-cell Ca2+ channel currents in rabbit portal vein cells were recorded using the amphotericin B-perforated patch-clamp technique at 35 degrees C. This technique allowed recording of stable inward currents in the absence of run-down for more than 30 minutes. Depolarizing voltage steps from a holding potential of -70 mV elicited voltage-dependent inward currents. The voltage dependence of inward currents measured in either 2.5 mmol/L Ba(2+)- or 2.5 mmol/L Ca(2+)-containing solution were very similar. However, maximum Ba2+ current (obtained at around +10 mV) was approximately 1.5-fold larger than maximum Ca2+ current. Changing the holding potential from -70 to -40 mV decreased inward currents but did not shift the voltage dependence significantly. Inward currents were also completely blocked by the dihydropyridine Ca2+ channel blocker, nicardipine (10 mumol/L), suggesting the presence of predominantly L-type Ca2+ channels in rabbit portal vein cells. Isoproterenol caused small increases in the amplitude of Ba2+ currents in a concentration-dependent manner (10 nmol/L to 1 mumol/L), which were reversed with propranolol. Forskolin (1 mumol/L) or 8-bromo-cAMP (0.1 mmol/L) also caused small increases in the amplitude of Ba2+ currents, suggesting that the stimulatory actions of isoproterenol are importantly linked to the production of cAMP. Higher concentrations of of isoproterenol (10 mumol/L) or forskolin (10 mumol/L) caused a transient increase in Ba2+ currents followed by f decrease in current amplitude. Higher doses of 8-bromo-cAMP (1 mmol/L) and low doses of 8-bromo-cGMP (0.1 mmol/L) inhibited Ba2+ currents, increased the rate of current inactivation, and produced a negative voltage shift in steady-state availability. These results indicate that low concentrations of intracellular cAMP produce modest increases in Ca2+ channel activity, whereas cGMP and higher concentrations of cAMP result in inhibition of Ca2+ channel activity in vascular smooth muscle cells. The observed similarities of cGMP and high concentrations of cAMP on Ba2+ current amplitude, kinetics, and steady-state inactivation suggest mediation by a common mechanism, possibly involving activation of cGMP-dependent protein kinase.


The Journal of Physiology | 1998

Functional and molecular expression of volume‐regulated chloride channels in canine vascular smooth muscle cells

Jun Yamazaki; Dayue Duan; Robert Janiak; Karri Kuenzli; Burton Horowitz; Joseph R. Hume

1 We examined the possibility of functional and molecular expression of volume‐regulated Cl− channels in vascular smooth muscle using the whole‐cell patch‐clamp technique and quantitative reverse transcriptase‐polymerase chain reaction (RT‐PCR) on cells from canine pulmonary and renal arteries. 2 Decreasing external osmolarity induced cell swelling, which was accompanied by activation of Cl−‐dependent outward‐rectifying membrane currents with an anion permeability sequence of SCN− > I− > Br− > Cl− > aspartate−. These currents were sensitive to block by DIDS, extracellular ATP and the antioestrogen compound tamoxifen. 3 Experiments were performed to determine whether the molecular form of the volume‐regulated chloride channel (ClC‐3) is expressed in pulmonary and renal arteries. Quantitative RT‐PCR confirmed expression of ClC‐3 in both types of smooth muscle. ClC‐3 expression was 76.4 % of β‐actin in renal artery and 48.0 % of β‐actin in pulmonary artery. 4 We conclude that volume‐regulated Cl− channels are expressed in vascular smooth muscle cells and exhibit functional properties similar to those found in other types of cells, presumably contributing to the regulation of cell volume, electrical activity and, possibly, myogenic tone.


Journal of Biological Chemistry | 2002

ClC-3 is a fundamental molecular component of volume-sensitive outwardly rectifying Cl- channels and volume regulation in HeLa cells and Xenopus laevis oocytes.

Marcela A. Hermoso; Christina M. Satterwhite; Yaniré Naty Andrade; Jorge Hidalgo; Sean M. Wilson; Burton Horowitz; Joseph R. Hume

Volume-sensitive osmolyte and anion channels (VSOACs) are activated upon cell swelling in most vertebrate cells. Native VSOACs are believed to be a major pathway for regulatory volume decrease (RVD) through efflux of chloride and organic osmolytes. ClC-3 has been proposed to encode native VSOACs in Xenopus laevis oocytes and in some mammalian cells, including cardiac and vascular smooth muscle cells. The relationship between the ClC-3 chloride channel, the native volume-sensitive osmolyte and anion channel (VSOAC) currents, and cell volume regulation in HeLa cells andX. laevis oocytes was investigated using ClC-3 antisense. In situ hybridization in HeLa cells, semiquantitative and real-time PCR, and immunoblot studies in HeLa cells and X. laevis oocytes demonstrated the presence of ClC-3 mRNA and protein, respectively. Exposing both cell types to hypotonic solutions induced cell swelling and activated native VSOACs. Transient transfection of HeLa cells with ClC-3 antisense oligonucleotide or X. laevis oocytes injected with antisense cRNA abolished the native ClC-3 mRNA transcript and protein and significantly reduced the density of native VSOACs activated by hypotonically induced cell swelling. In addition, antisense against native ClC-3 significantly impaired the ability of HeLa cells and X. laevis oocytes to regulate their volume. These results suggest that ClC-3 is an important molecular component underlying VSOACs and the RVD process in HeLa cells and X. laevis oocytes.


Circulation Research | 1996

Unitary Cl− Channels Activated by Cytoplasmic Ca2+ in Canine Ventricular Myocytes

Mei Lin Collier; Paul C. Levesque; James L. Kenyon; Joseph R. Hume

Recent whole-cell studies have shown that Ca(2+)-activated Cl- currents contribute to the Ca(2+)-dependent 4-aminopyridine-insensitive component of the transient outward current and to the arrhythmogenic transient inward current in rabbit and canine cardiac cells. These Cl(-)-sensitive currents are activated by Ca2+ release from the sarcoplasmic reticulum and are inhibited by anion transport blockers; however, the unitary single channels responsible have yet to be identified. We used inside-out patches from canine ventricular myocytes and conditions under which the only likely permeant ion is Cl- to identify 4-aminopyridine-resistant unitary Ca(2+)-activated Cl- channels, Ca2+ applied to the cytoplasmic surface of membrane patches activated small-conductance (1.0 to 1.3 pS) channels. These channels were Cl- selective, with rectification properties that could be described by the Goldman-Hodgkin-Katz current equation. Channel activity exhibited time independence when cytoplasmic Ca2+ was held constant and was blocked by the anion transport blockers, DIDS and niflumic acid. Ca2+ (ranging from pCa > or = 6 to pCa 3) applied to the cytoplasmic surface of inside-out patches increased, in a dose-dependent manner, NPo, where N is the number of channels opened and Po is open probability. At negative membrane potentials (-60 to -130 mV), an estimate of the dependence of NPo on cytoplasmic Ca2+ yielded an apparent Kd of 150.2 mumol/L. At pCa 3, an average channel density of approximately equal to 3 microns-2 was estimated. Calculations based on these estimates of cytoplasmic Ca2+ sensitivity and channel current amplitude and density suggest that these small-conductance Cl- channels contribute significant whole-cell membrane current in response to changes in intracellular Ca2+ within the physiological range. We suggest that these small-conductance Ca(2+)-activated Cl- channels underlie the transient Ca(2+)-activated 4-aminopyridine-insensitive current, which contributes to phase-1 repolarization, and under conditions of Ca2+ overload, these channels may generate transient inward currents, contributing to the development of triggered cardiac arrhythmias.


Circulation Research | 1997

Inhibitory Effects of Glibenclamide on Cystic Fibrosis Transmembrane Regulator, Swelling-Activated, and Ca2+-Activated Cl− Channels in Mammalian Cardiac Myocytes

Jun Yamazaki; Joseph R. Hume

Recent studies have provided evidence that sulfonylureas, in addition to blocking ATP-sensitive K+ (KATP) channels, also inhibit cystic fibrosis transmembrane regulator (CFTR) Cl- channels in epithelial and cardiac cells. The purpose of this study was to test whether the sulfonylurea glibenclamide might also inhibit other types of cardiac Cl- channels. Whole-cell patch-clamp techniques were used to compare the effects of glibenclamide on CFTR Cl- currents in guinea pig ventricular myocytes, swelling-activated Cl- currents in guinea pig atrial myocytes, and Ca(2+)-activated Cl- currents in canine ventricular myocytes. Glibenclamide markedly inhibited CFTR Cl- currents in a voltage-independent manner at 22 degrees C, with estimated IC50 values of 12.5 and 11.0 mumol/L at +50 and -100 mV, respectively. The outwardly rectifying swelling-activated Cl- current in atrial cells was less sensitive to glibenclamide, and the block exhibited voltage dependence. At 22 degrees C, the estimated IC50 values were 193 and 470 mumol/L at +50 and -100 mV, respectively, and block was enhanced at 35 degrees C. Macroscopic Cl- currents activated by a rise in intracellular Ca2+, induced by either Ca(2+)-induced Ca2+ release or by external application of the Ca2+ ionophore A23187, were also markedly inhibited at 22 degrees C by glibenclamide in a voltage-independent manner. The estimated IC50 values were 61.5 and 69.9 mumol/L at +50 and -100 mV, respectively. These results suggest that glibenclamide, an inhibitor of cardiac CFTR Cl- channels, also inhibits swelling-activated and Ca(2+)-activated Cl- channels at higher concentrations. The results also suggest that studies attributing the beneficial or deleterious effects of sulfonylurea compounds in the heart solely to blockade of KATP channels should use submicromolar concentrations of these agents to minimize possible secondary interactions with cardiac Cl- channels.


The Journal of Physiology | 2004

Altered properties of volume‐sensitive osmolyte and anion channels (VSOACs) and membrane protein expression in cardiac and smooth muscle myocytes from Clcn3‐/‐ mice

Shintaro Yamamoto-Mizuma; Ge-Xin Wang; Luis L. Liu; Kathleen M. Schegg; William J. Hatton; Dayue Duan; Burton Horowitz; Fred S. Lamb; Joseph R. Hume

ClC‐3, a member of the large superfamily of ClC voltage‐dependent Cl– channels, has been proposed as a molecular candidate responsible for volume‐sensitive osmolyte and anion channels (VSOACs) in some cells, including heart and vascular smooth muscle. However, the reported presence of native VSOACs in at least two cell types from transgenic ClC‐3 disrupted (Clcn3−/−) mice casts considerable doubt on this proposed role for ClC‐3. We compared several properties of native VSOACs and examined mRNA transcripts and membrane protein expression profiles in cardiac and pulmonary arterial smooth muscle cells from Clcn3+/+ and Clcn3−/− mice to: (1) test the hypothesis that native VSOACs are unaltered in cells from Clcn3−/− mice, and (2) test the possibility that targeted inactivation of the Clcn3 gene using a conventional murine global knock‐out approach may result in compensatory changes in expression of other membrane proteins. Our experiments demonstrate that VSOAC currents in myocytes from Clcn3+/+ and Clcn3−/− mice are remarkably similar in terms of activation and inactivation kinetics, steady‐state current densities, rectification, anion selectivity (I− > Cl−≫ Asp−) and sensitivity to block by glibenclamide, niflumic acid, DIDS and extracellular ATP. However, additional experiments revealed several significant differences in other fundamental properties of native VSOACs recorded from atrial and smooth muscle cells from Clcn3−/− mice, including: differences in regulation by endogenous protein kinase C, differential sensitivity to block by anti‐ClC‐3 antibodies, and differential sensitivities to [ATP]i and free [Mg2+]i. These results suggest that in response to Clcn3 gene deletion, there may be compensatory changes in expression of other proteins that alter VSOAC channel subunit composition or associated regulatory subunits that give rise to VSOACs with different properties. Consistent with this hypothesis, in atria from Clcn3−/− mice compared to Clcn3+/+ mice, quantitative analysis of ClC mRNA expression levels revealed significant increases in transcripts for ClC‐1, ClC‐2, and ClC‐3, and protein expression profiles obtained using two‐dimensional polyacrylamide gel electrophoresis revealed complex changes in at least 35 different unidentified membrane proteins in cells from Clcn3−/− mice. These findings emphasize that caution needs to be exercised in simple attempts to interpret the phenotypic consequences of conventional global Clcn3 gene inactivation.


The Journal of Physiology | 2009

TRPC1 and STIM1 mediate capacitative Ca2+ entry in mouse pulmonary arterial smooth muscle cells

Lih Chyuan Ng; Mary D. McCormack; Judith A. Airey; Cherie A. Singer; Phillip S. Keller; Xiao-Ming Shen; Joseph R. Hume

Previous studies in pulmonary arterial smooth muscle cells (PASMCs) showed that the TRPC1 channel mediates capacitative Ca2+ entry (CCE), but the molecular signal(s) that activate TRPC1 in PASMCs remains unknown. The aim of the present study was to determine if TRPC1 mediates CCE through activation of STIM1 protein in mouse PASMCs. In primary cultured mouse PASMCs loaded with fura‐2, cyclopiazonic acid (CPA) caused a transient followed by a sustained rise in intracellular Ca2+ concentration ([Ca2+]i). The transient but not the sustained rise in [Ca2+]i was partially inhibited by nifedipine. In addition, CPA increased the rate of Mn2+ quench of fura‐2 fluorescence that was inhibited by SKF 96365, Ni2+, La3+ and Gd3+, exhibiting pharmacological properties characteristic of CCE. The nifedipine‐insensitive sustained rise in [Ca2+]i and the increase in Mn2+ quench of fura‐2 fluorescence caused by CPA were both inhibited in cells pretreated with antibody raised against an extracellular epitope of TRPC1. Moreover, STIM1 siRNA reduced the rise in [Ca2+]i and Mn2+ quench of fura‐2 fluorescence caused by CPA, whereas overexpression of STIM1 resulted in a marked increase in these responses. RT‐PCR revealed TRPC1 and STIM1 mRNAs, and Western blot analysis identified TRPC1 and STIM1 proteins in mouse PASMCs. Furthermore, TRPC1 was found to co‐immunoprecipitate with STIM1, and the precipitation level of TRPC1 was increased in cells subjected to store depletion. Taken together, store depletion causes activation of voltage‐operated Ca2+ entry and CCE. These data provide direct evidence that CCE is mediated by TRPC1 channel through activation of STIM1 in mouse PASMCs.

Collaboration


Dive into the Joseph R. Hume's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge