Network


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

Hotspot


Dive into the research topics where Aguan Wei is active.

Publication


Featured researches published by Aguan Wei.


Neuron | 1994

Calcium sensitivity of BK-type KCa channels determined by a separable domain

Aguan Wei; Cr Solaro; Christopher J. Lingle; Lawrence Salkoff

High conductance, Ca(2+)-activated (BK-type) K+ channels from mouse (mSlo) and Drosophila (dSlo) differ in their functional properties but share a conserved core resembling voltage-gated K+ channels and a tail appended to the core by a nonconserved linker. We have found that the channel subunit is physically divisible into these two conserved domains and that the core determines such properties as channel open time, conductance, and, probably, voltage dependence, whereas the tail determines apparent Ca2+ sensitivity. Both domains are required for function. We demonstrated the different roles of the core and tail by taking advantage of the functional differences between mSlo and dSlo. Heterologous pairing of cores and tails from mSlo and dSlo showed that single-channel properties were always characteristic of the core species, but that apparent Ca2+ sensitivity was adjusted up or down depending on the species of the tail. Thus, the tail is implicated in the Ca(2+)-sensing role of BK channels.


Neuron | 1991

Shaker, Shal, Shab, and Shaw express independent K+ current systems

Manuel Covarrubias; Aguan Wei; Lawrence Salkoff

Although many K+ channel genes encoding homologous subunits have been cloned, a central question remains: how do these subunits associate to produce the diversity of K+ currents observed in living cells? Previous work has shown that different subunits encoded by the Shaker gene subfamily are able to form heteromultimers, which add to the diversity of currents. However, the unrestrained mixing of subunits from all genes to form hybrid channels would be undesirable for some cells that clearly require functionally discrete K+ currents. We show that Drosophila Shaker, Shal, Shab, and Shaw subunits form functional homomultimers, but that a molecular barrier to heteropolymerization is present. Coexpression of all four K+ channel systems does not alter their individual properties in any way. These experiments also demonstrate that multiple, independent A-current systems together with multiple, independent delayed rectifier systems can coexist in single cells.


Neuron | 2003

The Sodium-Activated Potassium Channel Is Encoded by a Member of the Slo Gene Family

Alex Yuan; Celia M. Santi; Aguan Wei; Zhao-Wen Wang; Kelly Pollak; Michael L. Nonet; Leonard K. Kaczmarek; C. Michael Crowder; Lawrence Salkoff

Na(+)-activated potassium channels (K(Na)) have been identified in cardiomyocytes and neurons where they may provide protection against ischemia. We now report that K(Na) is encoded by the rSlo2 gene (also called Slack), the mammalian ortholog of slo-2 in C. elegans. rSlo2, heterologously expressed, shares many properties of native K(Na) including activation by intracellular Na(+), high conductance, and prominent subconductance states. In addition to activation by Na(+), we report that rSLO-2 channels are cooperatively activated by intracellular Cl(-), similar to C. elegans SLO-2 channels. Since intracellular Na(+) and Cl(-) both rise in oxygen-deprived cells, coactivation may more effectively trigger the activity of rSLO-2 channels in ischemia. In C. elegans, mutational and physiological analysis revealed that the SLO-2 current is a major component of the delayed rectifier. We demonstrate in C. elegans that slo-2 mutants are hypersensitive to hypoxia, suggesting a conserved role for the slo-2 gene subfamily.


Neuron | 1994

Elimination of rapid potassium channel inactivation by phosphorylation of the inactivation gate

Manuel Covarrubias; Aguan Wei; Lawrence Salkoff; Tapan B. Vyas

The effect of protein kinase C (PKC) on rapid N-type inactivation of K+ channels has not been reported previously. We found that PKC specifically eliminates rapid inactivation of a cloned human A-type K+ channel (hKv3.4), converting this channel from a rapidly inactivating A type to a noninactivating delayed rectifier type. Biochemical analysis showed that the N-terminal domain of hKv3.4 is phosphorylated in vitro by PKC, and mutagenesis experiments revealed that two serines within the inactivation gate at the N-terminus are sites of direct PKC action. Moreover, mutating one of these serines to aspartic acid mimics the action of PKC. Serine phosphorylation may thus prevent rapid inactivation by shielding basic residues known to be critical to the function of the inactivation gate. The regulatory mechanism reported here may have substantial effects on signal coding in the nervous system.


Nature Neuroscience | 2000

SLO-2, a K+ channel with an unusual Cl- dependence.

Alex Yuan; Michelle Dourado; Alice Butler; Nina Walton; Aguan Wei; Lawrence Salkoff

The gating of different potassium channels depends on many diverse factors. We now report a unique example of a K+ channel with a Cl− dependence. The slo-2 gene was cloned from Caenorhabditis elegans and is widely expressed in both neurons and muscles; it was highly abundant, as suggested by its high representation in the C. elegans EST database. SLO-2, like its paralogue, SLO-1, was also dependent on Ca2+. We show by site-directed mutagenesis that its requirements for both Cl− and Ca2+ are synergistic and associated with the same functional domain. SLO-2s dependence on Cl− implies that intracellular Cl− homeostasis may be important in regulating cellular excitability through this unusual K+ channel.


The Journal of Neuroscience | 2006

Opposite Regulation of Slick and Slack K+ Channels by Neuromodulators

Celia M. Santi; Gonzalo Ferreira; Bo Yang; Valeswara Rao Gazula; Alice Butler; Aguan Wei; Leonard K. Kaczmarek; Lawrence Salkoff

Slick (Slo2.1) and Slack (Slo2.2) are two novel members of the mammalian Slo potassium channel gene family that may contribute to the resting potentials of cells and control their basal level of excitability. Slo2 channels have sensors that couple channel activity to the intracellular concentrations of Na+ and Cl− ions (Yuan et al., 2003). We now report that activity of both Slo2 channels is controlled by neuromodulators through Gαq-protein coupled receptors (GqPCRs) (the M1 muscarinic receptor and the mGluR1 metabotropic glutamate receptor). Experiments coexpressing channels and receptors in Xenopus oocytes show that Slo2.1 and Slo2.2 channels are modulated in opposite ways: Slo2.1 is strongly inhibited, whereas Slo2.2 currents are strongly activated through GqPCR stimulation. Differential regulation involves protein kinase C (PKC); application of the PKC activator PMA, to cells expressing channels but not receptors, inhibits Slo2.1 whole-cell currents and increases Slo2.2 currents. Synthesis of a chimera showed that the distal carboxyl region of Slo2.1 controls the sensitivity of Slo2.1 to PMA. Slo2 channels have widespread expression in brain (Bhattacharjee et al., 2002, 2005). Using immunocytochemical techniques, we show coexpression of Slo2 channels with the GqPCRs in cortical and hippocampal brain sections and in cultured hippocampal neurons. The differential control of these novel channels by neurotransmitters may elicit long-lasting increases or decreases in neuronal excitability and, because of their widespread distribution, may provide a mechanism to activate or repress electrical activity in many systems of the brain.


Nature Neuroscience | 2009

Na + -activated K + channels express a large delayed outward current in neurons during normal physiology

Gonzalo Budelli; Travis Hage; Aguan Wei; Patricio Rojas; Yuh Jiin Ivy Jong; Karen L. O'Malley; Lawrence Salkoff

One of the largest components of the delayed outward current that is active under physiological conditions in many mammalian neurons, such as medium spiny neurons of the striatum and tufted-mitral cells of the olfactory bulb, has gone unnoticed and is the result of a Na+-activated K+ current. Previous studies of K+ currents in mammalian neurons may have overlooked this large outward component because the sodium channel blocker tetrodotoxin (TTX) is typically used in such studies. We found that TTX also eliminated this delayed outward component in rat neurons as a secondary consequence. Unexpectedly, we found that the activity of a persistent inward sodium current (persistent INa) is highly effective at activating this large Na+-dependent (TTX sensitive) delayed outward current. Using siRNA techniques, we identified SLO2.2 channels as being carriers of this delayed outward current. These findings have far reaching implications for many aspects of cellular and systems neuroscience, as well as clinical neurology and pharmacology.


Neuron | 1997

Behavioral Defects in C. elegans egl-36 Mutants Result from Potassium Channels Shifted in Voltage-Dependence of Activation

Duncan B. Johnstone; Aguan Wei; Alice Butler; Lawrence Salkoff; James H. Thomas

Mutations in the C. elegans egl-36 gene result in defective excitation of egg-laying and enteric muscles. Dominant gain-of-function alleles inhibit enteric and egg-laying muscle contraction, whereas a putative null mutation has no observed phenotype. egl-36 encodes a Shaw-type (Kv3) voltage-dependent potassium channel subunit. In Xenopus oocytes, wild-type egl-36 expresses noninactivating channels with slow activation kinetics. One gain-of-function mutation causes a single amino acid substitution in S6, and the other causes a substitution in the cytoplasmic amino terminal domain. Both mutant alleles produce channels dramatically shifted in their midpoints of activation toward hyperpolarized voltages. An egl-36::gfp fusion is expressed in egg-laying muscles and in a pair of enteric muscle motor neurons. The mutant egl-36 phenotypes can thus be explained by expression in these cells of potassium channels that are inappropriately opened at hyperpolarized potentials, causing decreased excitability due to increased potassium conductance.


Science | 1986

Occult drosophila calcium channels and twinning of calcium and voltage-activated potassium channels

Aguan Wei; Lawrence Salkoff

In the membrane of the flight muscle cells of developing Drosophila a large calcium-sensitive potassium current, IKc, was found. It was present before the development of voltage-activated potassium channels and seems to be the first potassium current to develop in the membrane. Also present in these early cells were large numbers of occult (hidden) calcium channels, which remained inactive until the end of pupal development. These inactive calcium channels could be made to function by injecting adenosine triphosphate or ethyleneglycol tetraacetic acid into the early cells. IKc has kinetic properties resembling the later developing voltage-sensitive current IKv, and is distinct from the fast, transient calcium-dependent outward current IAc, which appears much later in development. IAc closely resembles the voltage-sensitive current IAv, also present in these cells. Thus, both of the voltage-sensitive potassium channel types, IAv and IKv, have similar calcium-sensitive counterparts, IAc and IKc, that are present in the same cells.


Annals of the New York Academy of Sciences | 1999

Genomic Organization of Nematode 4TM K+ Channelsa

Zhao-Wen Wang; Maya T. Kunkel; Aguan Wei; Alice Butler; Lawrence Salkoff

ABSTRACT: As many as 50 genes in the C. elegans genome may encode K+ channels belonging to the novel structural class of two‐pore (4TM) channels. Many 4TM channels can be grouped into channel subfamilies. We analyzed 4TM channels in C. elegans using methods made possible by having complete genomic sequence. Two genes were chosen for comprehensive analysis, n2P16 and n2P17. By comparing the pattern of conservation in genomic DNA sequences between C. elegans and a closely related species, C. briggsae, we were able to identify all coding regions and predict the gene structure for these two genes. Given the extent of the 4TM channel family, we were surprised to discover that n2P17 produced at least six alternative transcripts encoding a constant central region and variable amino‐ and carboxyl‐termini. Blocks of highly conserved DNA sequences in noncoding regions were also apparent and most likely confer important regulatory functions. The interspecies comparison of the deduced channel proteins revealed that the extracellular loop between M1 and P1 is an apparent hot spot for evolutionary change in both channels. This contrasts with the membrane‐spanning domains that are highly conserved. Analysis of intron positions for 36 channels revealed that introns are frequently present at an identical position within the pore region, but very few are located in membrane‐spanning domains.

Collaboration


Dive into the Aguan Wei's collaboration.

Top Co-Authors

Avatar

Lawrence Salkoff

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alice Butler

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alex Yuan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Celia M. Santi

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Manuel Covarrubias

Thomas Jefferson University

View shared research outputs
Top Co-Authors

Avatar

Zhao-Wen Wang

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Gloria L. Fawcett

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael L. Nonet

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gonzalo Ferreira

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge