Network


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

Hotspot


Dive into the research topics where Catherine M. Fuller is active.

Publication


Featured researches published by Catherine M. Fuller.


Journal of Biological Chemistry | 1996

Regulation of Epithelial Sodium Channels by Short Actin Filaments

Bakhram K. Berdiev; Adriana G. Prat; Horacio F. Cantiello; Dennis A. Ausiello; Catherine M. Fuller; Biljana Jovov; Dale J. Benos; Iskander I. Ismailov

Cytoskeletal elements play an important role in the regulation of ion transport in epithelia. We have studied the effects of actin filaments of different length on the α, β, γ-rENaC (rat epithelial Na+ channel) in planar lipid bilayers. We found the following. 1) Short actin filaments caused a 2-fold decrease in unitary conductance and a 2-fold increase in open probability (Po) of α,β,γ-rENaC. 2) α,β,γ-rENaC could be transiently activated by protein kinase A (PKA) plus ATP in the presence, but not in the absence, of actin. 3) ATP in the presence of actin was also able to induce a transitory activation of α,β,γ-rENaC, although with a shortened time course and with a lower magnitude of change in Po. 4) DNase I, an agent known to prohibit elongation of actin filaments, prevented activation of α,β,γ-rENaC by ATP or PKA plus ATP. 5) Cytochalasin D, added after rundown of α,β,γ-rENaC activity following ATP or PKA plus ATP treatment, produced a second transient activation of α,β,γ-rENaC. 6) Gelsolin, a protein that stabilizes polymerization of actin filaments at certain lengths, evoked a sustained activation of α,β,γ-rENaC at actin/gelsolin ratios of <32:1, with a maximal effect at an actin/gelsolin ratio of 2:1. These results suggest that short actin filaments activate α,β,γ-rENaC. PKA-mediated phosphorylation augments activation of this channel by decreasing the rate of elongation of actin filaments. These results are consistent with the hypothesis that cloned α,β,γ-rENaCs form a core conduction unit of epithelial Na+ channels and that interaction of these channels with other associated proteins, such as short actin filaments, confers regulation to channel activity.


American Journal of Physiology-cell Physiology | 1999

Molecular cloning and transmembrane structure of hCLCA2 from human lung, trachea, and mammary gland

Achim D. Gruber; Kevin D. Schreur; Hong Long Ji; Catherine M. Fuller; Bendicht U. Pauli

The CLCA family of Ca2+-activated Cl- channels has recently been discovered, with an increasing number of closely related members isolated from different species. Here we report the cloning of the second human homolog, hCLCA2, from a human lung cDNA library. Northern blot and RT-PCR analyses revealed additional expression in trachea and mammary gland. A primary translation product of 120 kDa was cleaved into two cell surface-associated glycoproteins of 86 and 34 kDa in transfected HEK-293 cells. hCLCA2 is the first CLCA homolog for which the transmembrane structure has been systematically studied. Glycosylation site scanning and protease protection assays revealed five transmembrane domains with a large, cysteine-rich, amino-terminal extracellular domain. Whole cell patch-clamp recordings of hCLCA2-transfected HEK-293 cells detected a slightly outwardly rectifying anion conductance that was increased in the presence of the Ca2+ ionophore ionomycin and inhibited by DIDS, dithiothreitol, niflumic acid, and tamoxifen. Expression in human trachea and lung suggests that hCLCA2 may play a role in the complex pathogenesis of cystic fibrosis.


Pflügers Archiv: European Journal of Physiology | 2000

Ca2+-Activated Cl– Channels: A Newly Emerging Anion Transport Family

Catherine M. Fuller; Hong Long Ji; Albert Tousson; Randolph C. Elble; Bendicht U. Pauli; Dale J. Benos

Abstract. A new family of chloride transport proteins has recently emerged. These proteins have extensive homology to a protein previously isolated from bovine tracheal epithelium that acts as a Ca2+-sensitive Cl– channel (CaCC) when heterologously expressed or when reconstituted into planar lipid bilayers. Several new members of this family have been identified in human, murine, and bovine epithelia, in addition to some other tissues, and are associated with Ca2+-sensitive conductive chloride transport when heterologously expressed in Xenopus oocytes or HEK 293 cells. The expressed current is also sensitive to inhibitors such as DIDS and niflumic acid. In addition, at least one family member acts as an endothelial cell adhesion molecule. This emerging family may underlie the Ca2+-mediated Cl– conductance responsible for rescue of the cystic fibrosis (CF) knockout mouse from significant airway disease.


Journal of Biological Chemistry | 1996

TRIPLE-BARREL ORGANIZATION OF ENAC, A CLONED EPITHELIAL NA+ CHANNEL

Iskander I. Ismailov; Mouhamed S. Awayda; Bakhram K. Berdiev; James K. Bubien; Joseph E. Lucas; Catherine M. Fuller; Dale J. Benos

A cloned rat epithelial Na+ channel (rENaC) was studied in planar lipid bilayers. Two forms of the channel were examined: channels produced by the α subunit alone and those formed by α, β, and γ subunits. The protein was derived from two sources: either from in vitro translation reaction followed by Sephadex column purification or from heterologous expression in Xenopus oocytes and isolation of plasma membranes. We found that either α-rENaC alone or α- in combination with β- and γ-rENaC, produced highly Na+-selective (PNa/PK = 10), amiloride-sensitive (Kiamil = 170 nM), and mechanosensitive cation channels in planar bilayers. α-rENaC displayed a complicated gating mechanism: there was a nearly constitutively open 13-picosiemens (pS) state and a second 40-pS level that was achieved from the 13-pS level by a 26-pS transition. α-, β-, γ-rENaC showed primarily the 13-pS level. α-rENaC and α,β,γ-rENaC channels studied by patch clamp displayed the same gating pattern, albeit with >2-fold lowered conductance levels, i.e. 6 and 18 pS, respectively. Upon treatment of either channel with the sulfhydryl reducing agent dithiothreitol, both channels fluctuated among three independent 13-pS sublevels. Bathing each channel with a high salt solution (1.5 M NaCl) produced stochastic openings of 19 and 38 pS in magnitude between all three conductance levels. Different combinations of α-, β-, and γ-rENaC in the reconstitution mixture did not produce channels of intermediate conductance levels. These findings suggest that functional ENaC is composed of three identical conducting elements and that their gating is concerted.


Journal of Biological Chemistry | 2009

Knockdown of ASIC1 and Epithelial Sodium Channel Subunits Inhibits Glioblastoma Whole Cell Current and Cell Migration

Niren Kapoor; Rafal Bartoszewski; Yawar J. Qadri; Zsuzsanna Bebok; James K. Bubien; Catherine M. Fuller; Dale J. Benos

High grade gliomas such as glioblastoma multiforme express multiple members of the epithelial sodium channel (ENaC)/Degenerin family, characteristically displaying a basally active amiloride-sensitive cation current not seen in normal human astrocytes or lower grade gliomas. Using quantitative real time PCR, we have shown higher expression of ASIC1, αENaC, and γENaC in D54-MG human glioblastoma multiforme cells compared with primary human astrocytes. We hypothesize that this glioma current is mediated by a hybrid channel composed of a mixture of ENaC and acid-sensing ion channel (ASIC) subunits. To test this hypothesis we made dominant negative cDNAs for ASIC1, αENaC, γENaC, and δENaC. D54-MG cells transfected with the dominant negative constructs for ASIC1, αENaC, or γENaC showed reduced protein expression and a significant reduction in the amiloride-sensitive whole cell current as compared with untransfected D54-MG cells. Knocking down αENaC or γENaC also abolished the high PK+/PNa+ of D54-MG cells. Knocking down δENaC in D54-MG cells reduced δENaC protein expression but had no effect on either the whole cell current or K+ permeability. Using co-immunoprecipitation we show interactions between ASIC1, αENaC, and γENaC, consistent with these subunits interacting with each other to form an ion channel in glioma cells. We also found a significant inhibition of D54-MG cell migration after ASIC1, αENaC, or γENaC knockdown, consistent with the hypothesis that ENaC/Degenerin subunits play an important role in glioma cell biology.


Journal of Biological Chemistry | 2011

The Unfolded Protein Response (UPR)-activated Transcription Factor X-box-binding Protein 1 (XBP1) Induces MicroRNA-346 Expression That Targets the Human Antigen Peptide Transporter 1 (TAP1) mRNA and Governs Immune Regulatory Genes

Rafal Bartoszewski; Joseph W. Brewer; Andras Rab; David K. Crossman; Sylwia Bartoszewska; Niren Kapoor; Catherine M. Fuller; James F. Collawn; Zsuzsa Bebok

Background: The adaptive unfolded protein response (UPR) promotes endoplasmic reticulum (ER) expansion and reduces ER load. Results: UPR-activated XBP1 induces miR-346 expression that targets TAP1. Conclusion: We identify a novel function for XBP1 and an miRNA-mediated pathway for ER load reduction through TAP1. Significance: Novel interventions for protein folding disorders will require an understanding of how microRNAs regulate gene expression during ER stress. To identify endoplasmic reticulum (ER) stress-induced microRNAs (miRNA) that govern ER protein influx during the adaptive phase of unfolded protein response, we performed miRNA microarray profiling and analysis in human airway epithelial cells following ER stress induction using proteasome inhibition or tunicamycin treatment. We identified miR-346 as the most significantly induced miRNA by both classic stressors. miR-346 is encoded within an intron of the glutamate receptor ionotropic delta-1 gene (GRID1), but its ER stress-associated expression is independent of GRID1. We demonstrated that the spliced X-box-binding protein-1 (sXBP1) is necessary and sufficient for ER stress-associated miR-346 induction, revealing a novel role for this unfolded protein response-activated transcription factor. In mRNA profiling arrays, we identified 21 mRNAs that were reduced by both ER stress and miR-346. The target genes of miR-346 regulate immune responses and include the major histocompatibility complex (MHC) class I gene products, interferon-induced genes, and the ER antigen peptide transporter 1 (TAP1). Although most of the repressed mRNAs appear to be indirect targets because they lack specific seeding sites for miR-346, we demonstrate that the human TAP1 mRNA is a direct target of miR-346. The human TAP1 mRNA 3′-UTR contains a 6-mer canonical seeding site for miR-346. Importantly, the ER stress-associated reduction in human TAP1 mRNA and protein levels could be reversed with an miR-346 antagomir. Because TAP function is necessary for proper MHC class I-associated antigen presentation, our results provide a novel mechanistic explanation for reduced MHC class I-associated antigen presentation that was observed during ER stress.


Journal of Biological Chemistry | 1997

Identification of an Amiloride Binding Domain within the α-Subunit of the Epithelial Na+ Channel

Iskander I. Ismailov; Thomas Kieber-Emmons; Chaomei Lin; Bakhram K. Berdiev; Vadim Shlyonsky; Holly K. Patton; Catherine M. Fuller; Roger T. Worrell; Jonathan B. Zuckerman; Weijing Sun; Douglas C. Eaton; Dale J. Benos; Thomas R. Kleyman

Limited information is available regarding domains within the epithelial Na+ channel (ENaC) which participate in amiloride binding. We previously utilized the anti-amiloride antibody (BA7.1) as a surrogate amiloride receptor to delineate amino acid residues that contact amiloride, and identified a putative amiloride binding domain WYRFHY (residues 278–283) within the extracellular domain of αrENaC. Mutations were generated to examine the role of this sequence in amiloride binding. Functional analyses of wild type (wt) and mutant αrENaCs were performed by cRNA expression in Xenopus oocytes and by reconstitution into planar lipid bilayers. Wild type αrENaC was inhibited by amiloride with aK i of 169 nm. Deletion of the entire WYRFHY tract (αrENaC Δ278–283) resulted in a loss of sensitivity of the channel to submicromolar concentrations of amiloride (K i = 26.5 μm). Similar results were obtained when either αrENaC or αrENaC Δ278–283 were co-expressed with wt β- and γrENaC (K i values of 155 nm and 22.8 μm, respectively). Moreover, αrENaC H282D was insensitive to submicromolar concentrations of amiloride (K i = 6.52 μm), whereas αrENaC H282R was inhibited by amiloride with a K i of 29 nm. These mutations do not alter ENaC Na+:K+ selectivity nor single-channel conductance. These data suggest that residues within the tract WYRFHY participate in amiloride binding. Our results, in conjunction with recent studies demonstrating that mutations within the membrane-spanning domains of αrENaC and mutations preceding the second membrane-spanning domains of α-, β-, and γrENaC alters amiloride’s K i , suggest that selected regions of the extracellular loop of αrENaC may be in close proximity to residues within the channel pore.


Journal of Biological Chemistry | 2006

Surface Expression of ASIC2 Inhibits the Amiloride-sensitive Current and Migration of Glioma Cells

Wanda H. Vila-Carriles; Gergely Kovacs; Biljana Jovov; Zhen Hong Zhou; Amit K. Pahwa; Garrett Colby; Ogenna Esimai; G. Yancey Gillespie; Timothy B. Mapstone; James M. Markert; Catherine M. Fuller; James K. Bubien; Dale J. Benos

Gliomas are primary brain tumors with a complex biology characterized by antigenic and genomic heterogeneity and a propensity for invasion into normal brain tissue. High grade glioma cells possess a voltage-independent, amiloride-inhibitable, inward Na+ current. This current does not exist in normal astrocytes or low grade tumor cells. Inhibition of this conductance decreases glioma growth and cell migration making it a potential therapeutic target. Our previous results have shown that the acid-sensing ion channels (ASICs), members of the epithelial Na+ channel (ENaC)/degenerin (DEG) family of ion channels are part of this current pathway. We hypothesized that one member of the ENaC/DEG family, ASIC2, is retained intracellularly and that it is the lack of functional expression of ASIC2 at the cell surface that results in hyperactivity of this conductance in high grade gliomas. In this study we show that the chemical chaperone, glycerol, and the transcriptional regulator, sodium 4-phenylbutyrate, inhibit the constitutively activated inward current and reduce cell growth and migration in glioblastoma multiforme. The results suggest that these compounds induce the movement of ASIC2 to the plasma membrane, and once there, the basally active inward current characteristic of glioma cells is abolished by inherent negative regulatory mechanisms. This in turn compromises the ability of the glioma cell to migrate and proliferate. These results support the hypothesis that the conductance pathway in high grade glioma cells is comprised of ENaC/DEG subunits and that abolishing this channel activity promotes a reversion of a high grade glioma cell to a phenotype resembling that of normal astrocytes.


Biochimica et Biophysica Acta | 1992

Peroxynitrite inhibits sodium uptake in rat colonic membrane vesicles.

Martin L. Bauer; Joseph S. Beckman; Robert J. Bridges; Catherine M. Fuller; Sadis Matalon

Peroxynitrite (ONOO-) is a potent oxidizing agent that initiates lipid peroxidation and sulfhydryl oxidation and may be responsible for a portion of the cytotoxicity attributed to superoxide anion (.O2-). We quantified the extent to which ONOO-, xanthine plus xanthine oxidase (XO) and hydrogen peroxide (H2O2), decreased sodium (Na+) uptake into membrane vesicles derived from colonic cells of dexamethasone-treated rats. Carrier-free 22Na+ uptake into vesicles was measured in the presence of an inside-negative membrane potential, produced by the addition of the potassium ionophore valinomycin (10 microM) after removal of all external potassium by cation exchange chromatography. Preincubation of vesicles with either 100 microM or 1 mM ONOO- for 30 s decreased the amiloride-blockable fraction of Na+ uptake by 27 +/- 7% and 65 +/- 2%, respectively (means +/- S.E.; n greater than or equal to 5; P less than 0.05 from control). However, the amiloride-insensitive part of Na+ uptake was not affected, indicating that there was no overt destruction of these vesicles by these ONOO- concentrations. Decomposed ONOO-, hydrogen peroxide (1 microM-10 mM), or xanthine (500 microM) plus XO (10-30 mU/ml), either in the absence or in the presence of 100 microM FeEDTA, did not decrease Na+ uptake. These data suggest that ONOO- is a potent injurious agent that can compromise Na+ uptake across epithelial cells, possibly by damaging Na+ channels.


Journal of Biological Chemistry | 2007

Heteromeric assembly of acid-sensitive ion channel and epithelial sodium channel subunits.

Robert H. Meltzer; Niren Kapoor; Yawar J. Qadri; Susan J. Anderson; Catherine M. Fuller; Dale J. Benos

Amiloride-sensitive ion channels are formed from homo- or heteromeric combinations of subunits from the epithelial Na+ channel (ENaC)/degenerin superfamily, which also includes the acid-sensitive ion channel (ASIC) family. These channel subunits share sequence homology and topology. In this study, we have demonstrated, using confocal fluorescence resonance energy transfer microscopy and co-immunoprecipitation, that ASIC and ENaC subunits are capable of forming cross-clade intermolecular interactions. We have also shown that combinations of ASIC1 with ENaC subunits exhibit novel electrophysiological characteristics compared with ASIC1 alone. The results of this study suggest that heteromeric complexes of ASIC and ENaC subunits may underlie the diversity of amiloride-sensitive cation conductances observed in a wide variety of tissues and cell types where co-expression of ASIC and ENaC subunits has been observed.

Collaboration


Dive into the Catherine M. Fuller's collaboration.

Top Co-Authors

Avatar

Dale J. Benos

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Hong Long Ji

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar

Iskander I. Ismailov

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

James K. Bubien

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Bakhrom K. Berdiev

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bakhram K. Berdiev

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Biljana Jovov

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

G. Yancey Gillespie

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge