Network


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

Hotspot


Dive into the research topics where Vera Y. Moiseenkova-Bell is active.

Publication


Featured researches published by Vera Y. Moiseenkova-Bell.


Nature Communications | 2016

Structure of the full-length TRPV2 channel by cryo-EM

Kevin Huynh; Matthew R. Cohen; Jiansen Jiang; Amrita Samanta; David T. Lodowski; Z. Hong Zhou; Vera Y. Moiseenkova-Bell

Transient receptor potential (TRP) proteins form a superfamily Ca2+-permeable cation channels regulated by a range of chemical and physical stimuli. Structural analysis of a ‘minimal TRP vanilloid subtype 1 (TRPV1) elucidated a mechanism of channel activation by agonists through changes in its outer pore region. Though homologous to TRPV1, other TRPV channels (TRPV2–6) are insensitive to TRPV1 activators including heat and vanilloids. To further understand the structural basis of TRPV channel function, we determined the structure of full-length TRPV2 at ∼5u2009Å resolution by cryo-electron microscopy. Like TRPV1, TRPV2 contains two constrictions, one each in the pore-forming upper and lower gates. The agonist-free full-length TRPV2 has wider upper and lower gates compared with closed and agonist-activated TRPV1. We propose these newly revealed TRPV2 structural features contribute to diversity of TRPV channels.


The Journal of Membrane Biology | 2014

Application of Amphipols for Structure–Functional Analysis of TRP Channels

Kevin Huynh; Matthew R. Cohen; Vera Y. Moiseenkova-Bell

Amphipathic polymers (amphipols), such as A8-35 and SApol, are a new tool for stabilizing integral membrane proteins in detergent-free conditions for structural and functional studies. Transient receptor potential (TRP) ion channels function as tetrameric protein complexes in a diverse range of cellular processes including sensory transduction. Mammalian TRP channels share ~20xa0% sequence similarity and are categorized into six subfamilies: TRPC (canonical), TRPV (vanilloid), TRPA (ankyrin), TRPM (melastatin), TRPP (polycystin), and TRPML (mucolipin). Due to the inherent difficulties in purifying eukaryotic membrane proteins, structural studies of TRP channels have been limited. Recently, A8-35 was essential in resolving the molecular architecture of the nociceptor TRPA1 and led to the determination of a high-resolution structure of the thermosensitive TRPV1 channel by cryo-EM. Newly developed maltose-neopentyl glycol (MNG) detergents have also proven to be useful in stabilizing TRP channels for structural analysis. In this review, we will discuss the impacts of amphipols and MNG detergents on structural studies of TRP channels by cryo-EM. We will compare how A8-35 and MNG detergents interact with the hydrophobic transmembrane domains of TRP channels. In addition, we will discuss what these cryo-EM studies reveal on the importance of screening different types of surfactants toward determining high-resolution structures of TRP channels.


Nature Structural & Molecular Biology | 2018

Structural basis of TRPV5 channel inhibition by econazole revealed by cryo-EM

Taylor E.T. Hughes; David T. Lodowski; Kevin W. Huynh; Aysenur Yazici; John del Rosario; Abhijeet Kapoor; Sandip Basak; Amrita Samanta; Xu Han; Sudha Chakrapani; Z. Hong Zhou; Marta Filizola; Tibor Rohacs; Seungil Han; Vera Y. Moiseenkova-Bell

The transient receptor potential vanilloid 5 (TRPV5) channel is a member of the transient receptor potential (TRP) channel family, which is highly selective for Ca2+, that is present primarily at the apical membrane of distal tubule epithelial cells in the kidney and plays a key role in Ca2+ reabsorption. Here we present the structure of the full-length rabbit TRPV5 channel as determined using cryo-EM in complex with its inhibitor econazole. This structure reveals that econazole resides in a hydrophobic pocket analogous to that occupied by phosphatidylinositides and vanilloids in TRPV1, thus suggesting conserved mechanisms for ligand recognition and lipid binding among TRPV channels. The econazole-bound TRPV5 structure adopts a closed conformation with a distinct lower gate that occludes Ca2+ permeation through the channel. Structural comparisons between TRPV5 and other TRPV channels, complemented with molecular dynamics (MD) simulations of the econazole-bound TRPV5 structure, allowed us to gain mechanistic insight into TRPV5 channel inhibition by small molecules.The cryo-EM structure of the full-length TRPV5 channel in complex with inhibitor econazole reveals a domain-swapped architecture andxa0provides insights into mechanisms of inhibition.


Current Topics in Membranes | 2014

Structure of Thermally Activated TRP Channels

Matthew R. Cohen; Vera Y. Moiseenkova-Bell

Temperature sensation is important for adaptation and survival of organisms. While temperature has the potential to affect all biological macromolecules, organisms have evolved specific thermosensitive molecular detectors that are able to transduce temperature changes into physiologically relevant signals. Among these thermosensors are ion channels from the transient receptor potential (TRP) family. Prime candidates include TRPV1-4, TRPA1, and TRPM8 (the so-called thermoTRP channels), which are expressed in sensory neurons and gated at specific temperatures. Electrophysiological and thermodynamic approaches have been employed to determine the nature by which thermoTRPs detect temperature and couple temperature changes to channel gating. To further understand how thermoTRPs sense temperature, high-resolution structures of full-length thermoTRPs channels will be required. Here, we will discuss current progress in unraveling the structures of thermoTRP channels.


Nature Communications | 2018

Cryo-EM structure of 5-HT3A receptor in its resting conformation

Sandip Basak; Yvonne Gicheru; Amrita Samanta; Sudheer K. Molugu; Wei Huang; Maria la de Fuente; Taylor E.T. Hughes; Derek J. Taylor; Marvin T. Nieman; Vera Y. Moiseenkova-Bell; Sudha Chakrapani

Serotonin receptors (5-HT3AR) directly regulate gut movement, and drugs that inhibit 5-HT3AR function are used to control emetic reflexes associated with gastrointestinal pathologies and cancer therapies. The 5-HT3AR function involves a finely tuned orchestration of three domain movements that include the ligand-binding domain, the pore domain, and the intracellular domain. Here, we present the structure from the full-length 5-HT3AR channel in the apo-state determined by single-particle cryo-electron microscopy at a nominal resolution of 4.3u2009Å. In this conformation, the ligand-binding domain adopts a conformation reminiscent of the unliganded state with the pore domain captured in a closed conformation. In comparison to the 5-HT3AR crystal structure, the full-length channel in the apo-conformation adopts a more expanded conformation of all the three domains with a characteristic twist that is implicated in gating.Serotonin receptor (5-HT3AR), a pentameric ligand-gated ion channel, regulates numerous gastrointestinal functions. Here the authors provide a cryo-electron microscopic structure from the full-length 5-HT3AR in the apo-state which corresponds to a resting conformation of the channel.


The Journal of General Physiology | 2018

Structural insights into the molecular mechanism of mouse TRPA1 activation and inhibition

Amrita Samanta; Janna Kiselar; Ruth A. Pumroy; Seungil Han; Vera Y. Moiseenkova-Bell

Pain, though serving the beneficial function of provoking a response to dangerous situations, is an unpleasant sensory and emotional experience. Transient receptor potential ankyrin 1 (TRPA1) is a member of the transient receptor potential (TRP) cation channel family and is localized in “nociceptors,” where it plays a key role in the transduction of chemical, inflammatory, and neuropathic pain. TRPA1 is a Ca2+-permeable, nonselective cation channel that is activated by a large variety of structurally unrelated electrophilic and nonelectrophilic chemical compounds. Electrophilic ligands are able to activate TRPA1 channels by interacting with critical cysteine residues on the N terminus of the channels via covalent modification and/or disulfide bonds. Activation by electrophilic compounds is dependent on their thiol-reactive moieties, accounting for the structural diversity of the group. On the other hand, nonelectrophilic ligands do not interact with critical cysteines on the channel, so the structural diversity of this group is unexplained. Although near-atomic-resolution structures of TRPA1 were resolved recently by cryo-electron microscopy, in the presence of both agonists and antagonists, detailed mechanisms of channel activation and inhibition by these modulators could not be determined. Here, we investigate the effect of both electrophilic and nonelectrophilic ligands on TRPA1 channel conformational rearrangements with limited proteolysis and mass spectrometry. Collectively, our results reveal that channel modulation results in conformational rearrangements in the N-terminal ankyrin repeats, the pre-S1 helix, the TRP-like domain, and the linker regions of the channel.


Journal of Lipid Research | 2017

Allosteric modulation of the substrate specificity of acyl-CoA wax alcohol acyltransferase 2

Jason M. Arne; Made Airanthi K. Widjaja-Adhi; Taylor E.T. Hughes; Kevin Huynh; Josie A. Silvaroli; Sylwia Chelstowska; Vera Y. Moiseenkova-Bell; Marcin Golczak

The esterification of alcohols with fatty acids is a universal mechanism to form inert storage forms of sterols, di- and triacylglycerols, and retinoids. In ocular tissues, formation of retinyl esters is an essential step in the enzymatic regeneration of the visual chromophore (11-cis-retinal). Acyl-CoA wax alcohol acyltransferase 2 (AWAT2), also known as multifunctional O-acyltransferase (MFAT), is an integral membrane enzyme with a broad substrate specificity that has been shown to preferentially esterify 11-cis-retinol and thus contribute to formation of a readily available pool of cis retinoids in the eye. However, the mechanism by which this promiscuous enzyme can gain substrate specificity is unknown. Here, we provide evidence for an allosteric modulation of the enzymatic activity by 11-cis retinoids. This regulation is independent from cellular retinaldehyde-binding protein (CRALBP), the major cis-retinoid binding protein. This positive-feedback regulation leads to decreased esterification rates for 9-cis, 13-cis, or all-trans retinols and thus enables preferential synthesis of 11-cis-retinyl esters. Finally, electron microscopy analyses of the purified enzyme indicate that this allosteric effect does not result from formation of functional oligomers. Altogether, these data provide the experimental basis for understanding regulation of AWAT2 substrate specificity.


Archive | 2018

Transient receptor potential (TRP) channels

Amrita Samanta; Taylor E.T. Hughes; Vera Y. Moiseenkova-Bell

Transient Receptor Potential (TRP) channels are evolutionarily conserved integral membrane proteins. The mammalian TRP superfamily of ionxa0channels consists of 28 cation permeable channels that are grouped into six subfamilies based on sequence homology (Fig. 6.1). The canonical TRP (TRPC) subfamily is known for containingxa0the founding member of mammalian TRP channels. The vanilloid TRP (TRPV) subfamily has been extensively studied due to the heat sensitivity of its founding member. The melastatin-related TRP (TRPM) subfamily includes some of the few known bi-functional ion channels, which contain functional enzymatic domains. The ankyrin TRP (TRPA) subfamily consists of a single chemo-nociceptor that has been proposed to be a target for analgesics. The mucolipin TRP (TRPML) subfamily channels are found primarily in intracellular compartments and were discovered based on their critical role in type IV mucolipidosis (ML-IV). The polycystic TRP (TRPP) subfamily is a diverse group of proteins implicated in autosomal dominant polycystic kidney disease (ADPKD). Overall, this superfamily of channels is involved in a vast array of physiological and pathophysiological processes making the study of these channels imperative to our understanding of subcellular biochemistry.


Nature Communications | 2018

Structural insights on TRPV5 gating by endogenous modulators.

T.E.T. Hughes; R.A. Pumroy; Aysenur Yazici; Kasimova; E.C. Fluck; K.W. Huynh; A. Samanta; S.K. Molugu; Z.H. Zhou; V. Carnevale; Tibor Rohacs; Vera Y. Moiseenkova-Bell


Nature Communications | 2018

Cryo-EM structure of 5-HT

Sandip Basak; Yvonne Gicheru; Amrita Samanta; Sudheer K. Molugu; Wei Huang; M. Fuente; Taylor E.T. Hughes; Derek J. Taylor; Marvin T. Nieman; Vera Y. Moiseenkova-Bell; Sudha Chakrapani

Collaboration


Dive into the Vera Y. Moiseenkova-Bell's collaboration.

Top Co-Authors

Avatar

Amrita Samanta

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Taylor E.T. Hughes

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Matthew R. Cohen

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Kevin Huynh

Baker IDI Heart and Diabetes Institute

View shared research outputs
Top Co-Authors

Avatar

David T. Lodowski

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Sandip Basak

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Sudha Chakrapani

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Z. Hong Zhou

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Derek J. Taylor

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge