Network


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

Hotspot


Dive into the research topics where Ryan T. Strachan is active.

Publication


Featured researches published by Ryan T. Strachan.


Science Signaling | 2011

Distinct Phosphorylation Sites on the β2-Adrenergic Receptor Establish a Barcode That Encodes Differential Functions of β-Arrestin

Kelly N. Nobles; Kunhong Xiao; Seungkirl Ahn; Arun K. Shukla; Christopher M. Lam; Sudarshan Rajagopal; Ryan T. Strachan; Teng-Yi Huang; Erin A. Bressler; Makoto R. Hara; Sudha K. Shenoy; Steven P. Gygi; Robert J. Lefkowitz

Different patterns of GPCR phosphorylation produce distinct conformations of β-arrestin and specific downstream responses. Cracking a Phosphorylation Code Not only can ligands for G protein–coupled receptors (GPCRs) trigger signaling through two completely different pathways—G protein–mediated and β-arrestin–mediated—Nobles et al. report that phosphorylation of one of these receptors, the β2-adrenergic receptor, by isoform-specific GPCR kinases (GRKs) produces distinct phosphorylation patterns that influence β-arrestin conformation and induce distinct downstream responses. As noted in the Perspective by Liggett, GPCRs are the largest class of signaling proteins in the human genome and are common targets of clinically used therapeutic agents. Drugs that bias signaling down G protein–coupled pathways or the β-arrestin pathways already exist. That the β-arrestin pathways depend on the specific GRK-induced “barcode” triggered by receptor activation has implications for understanding the effects of existing drugs and the development of selective therapies targeting specific β-arrestin–mediated pathways. Phosphorylation of G protein–coupled receptors (GPCRs, which are also known as seven-transmembrane spanning receptors) by GPCR kinases (GRKs) plays essential roles in the regulation of receptor function by promoting interactions of the receptors with β-arrestins. These multifunctional adaptor proteins desensitize GPCRs, by reducing receptor coupling to G proteins and facilitating receptor internalization, and mediate GPCR signaling through β-arrestin–specific pathways. Detailed mapping of the phosphorylation sites on GPCRs targeted by individual GRKs and an understanding of how these sites regulate the specific functional consequences of β-arrestin engagement may aid in the discovery of therapeutic agents targeting individual β-arrestin functions. The β2-adrenergic receptor (β2AR) has many serine and threonine residues in the carboxyl-terminal tail and the intracellular loops, which are potential sites of phosphorylation. We monitored the phosphorylation of the β2AR at specific sites upon stimulation with an agonist that promotes signaling by both G protein–mediated and β-arrestin–mediated pathways or with a biased ligand that promotes signaling only through β-arrestin–mediated events in the presence of the full complement of GRKs or when either GRK2 or GRK6 was depleted. We correlated the specific and distinct patterns of receptor phosphorylation by individual GRKs with the functions of β-arrestins and propose that the distinct phosphorylation patterns established by different GRKs establish a “barcode” that imparts distinct conformations to the recruited β-arrestin, thus regulating its functional activities.


Nature | 2011

A stress response pathway regulates DNA damage through β2-adrenoreceptors and β-arrestin-1.

Makoto R. Hara; Jeffrey J. Kovacs; Erin J. Whalen; Sudarshan Rajagopal; Ryan T. Strachan; Wayne Grant; Aaron J. Towers; Barbara Williams; Christopher M. Lam; Kunhong Xiao; Sudha K. Shenoy; Simon G. Gregory; Seungkirl Ahn; Derek R. Duckett; Robert J. Lefkowitz

The human mind and body respond to stress, a state of perceived threat to homeostasis, by activating the sympathetic nervous system and secreting the catecholamines adrenaline and noradrenaline in the ‘fight-or-flight’ response. The stress response is generally transient because its accompanying effects (for example, immunosuppression, growth inhibition and enhanced catabolism) can be harmful in the long term. When chronic, the stress response can be associated with disease symptoms such as peptic ulcers or cardiovascular disorders, and epidemiological studies strongly indicate that chronic stress leads to DNA damage. This stress-induced DNA damage may promote ageing, tumorigenesis, neuropsychiatric conditions and miscarriages. However, the mechanisms by which these DNA-damage events occur in response to stress are unknown. The stress hormone adrenaline stimulates β2-adrenoreceptors that are expressed throughout the body, including in germline cells and zygotic embryos. Activated β2-adrenoreceptors promote Gs-protein-dependent activation of protein kinase A (PKA), followed by the recruitment of β-arrestins, which desensitize G-protein signalling and function as signal transducers in their own right. Here we elucidate a molecular mechanism by which β-adrenergic catecholamines, acting through both Gs–PKA and β-arrestin-mediated signalling pathways, trigger DNA damage and suppress p53 levels respectively, thus synergistically leading to the accumulation of DNA damage. In mice and in human cell lines, β-arrestin-1 (ARRB1), activated via β2-adrenoreceptors, facilitates AKT-mediated activation of MDM2 and also promotes MDM2 binding to, and degradation of, p53, by acting as a molecular scaffold. Catecholamine-induced DNA damage is abrogated in Arrb1-knockout (Arrb1−/−) mice, which show preserved p53 levels in both the thymus, an organ that responds prominently to acute or chronic stress, and in the testes, in which paternal stress may affect the offspring’s genome. Our results highlight the emerging role of ARRB1 as an E3-ligase adaptor in the nucleus, and reveal how DNA damage may accumulate in response to chronic stress.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Rapid modulation of spine morphology by the 5-HT2A serotonin receptor through kalirin-7 signaling

Kelly A. Jones; Deepak P. Srivastava; John A. Allen; Ryan T. Strachan; Bryan L. Roth; Peter Penzes

The 5-HT2A serotonin receptor is the most abundant serotonin receptor subtype in the cortex and is predominantly expressed in pyramidal neurons. The 5-HT2A receptor is a target of several hallucinogens, antipsychotics, anxiolytics, and antidepressants, and it has been associated with several psychiatric disorders, conditions that are also associated with aberrations in dendritic spine morphogenesis. However, the role of 5-HT2A receptors in regulating dendritic spine morphogenesis in cortical neurons is unknown. Here we show that the 5-HT2A receptor is present in a subset of spines, in addition to dendritic shafts. It colocalizes with PSD-95 and with multiple PDZ protein-1 (MUPP1) in a subset of dendritic spines of rat cortical pyramidal neurons. MUPP1 is enriched in postsynaptic density (PSD) fractions, is targeted to spines in pyramidal neurons, and enhances the localization of 5-HT2A receptors to the cell periphery. 5-HT2A receptor activation by the 5-HT2 receptor agonist DOI induced a transient increase in dendritic spine size, as well as phosphorylation of p21-activated kinase (PAK) in cultured cortical neurons. PAK is a downstream target of the neuronal Rac guanine nucleotide exchange factor (RacGEF) kalirin-7 that is important for spine remodeling. Kalirin-7 regulates dendritic spine morphogenesis in neurons but its role in neuromodulator signaling has not been investigated. We show that peptide interference that prevents the localization of kalirin-7 to the postsynaptic density disrupts DOI-induced PAK phosphorylation and spine morphogenesis. These results suggest a potential role for serotonin signaling in modulating spine morphology and kalirin-7s function at cortical synapses.


Nature | 2016

Allosteric nanobodies reveal the dynamic range and diverse mechanisms of G-protein-coupled receptor activation

Dean P. Staus; Ryan T. Strachan; Aashish Manglik; Biswaranjan Pani; Alem W. Kahsai; Tae Hun Kim; Laura M. Wingler; Seungkirl Ahn; Arnab K. Chatterjee; Ali Masoudi; Andrew C. Kruse; Els Pardon; Jan Steyaert; William I. Weis; R. Scott Prosser; Brian K. Kobilka; Tommaso Costa; Robert J. Lefkowitz

G-protein-coupled receptors (GPCRs) modulate many physiological processes by transducing a variety of extracellular cues into intracellular responses. Ligand binding to an extracellular orthosteric pocket propagates conformational change to the receptor cytosolic region to promote binding and activation of downstream signalling effectors such as G proteins and β-arrestins. It is well known that different agonists can share the same binding pocket but evoke unique receptor conformations leading to a wide range of downstream responses (‘efficacy’). Furthermore, increasing biophysical evidence, primarily using the β2-adrenergic receptor (β2AR) as a model system, supports the existence of multiple active and inactive conformational states. However, how agonists with varying efficacy modulate these receptor states to initiate cellular responses is not well understood. Here we report stabilization of two distinct β2AR conformations using single domain camelid antibodies (nanobodies)—a previously described positive allosteric nanobody (Nb80) and a newly identified negative allosteric nanobody (Nb60). We show that Nb60 stabilizes a previously unappreciated low-affinity receptor state which corresponds to one of two inactive receptor conformations as delineated by X-ray crystallography and NMR spectroscopy. We find that the agonist isoprenaline has a 15,000-fold higher affinity for β2AR in the presence of Nb80 compared to the affinity of isoprenaline for β2AR in the presence of Nb60, highlighting the full allosteric range of a GPCR. Assessing the binding of 17 ligands of varying efficacy to the β2AR in the absence and presence of Nb60 or Nb80 reveals large ligand-specific effects that can only be explained using an allosteric model which assumes equilibrium amongst at least three receptor states. Agonists generally exert efficacy by stabilizing the active Nb80-stabilized receptor state (R80). In contrast, for a number of partial agonists, both stabilization of R80 and destabilization of the inactive, Nb60-bound state (R60) contribute to their ability to modulate receptor activation. These data demonstrate that ligands can initiate a wide range of cellular responses by differentially stabilizing multiple receptor states.


ACS Chemical Biology | 2013

Conformation guides molecular efficacy in docking screens of activated β-2 adrenergic G protein coupled receptor

Dahlia R. Weiss; Seungkirl Ahn; Maria F. Sassano; Andrew Kleist; Xiao Zhu; Ryan T. Strachan; Bryan L. Roth; Robert J. Lefkowitz; Brian K. Shoichet

A prospective, large library virtual screen against an activated β2-adrenergic receptor (β2AR) structure returned potent agonists to the exclusion of inverse-agonists, providing the first complement to the previous virtual screening campaigns against inverse-agonist-bound G protein coupled receptor (GPCR) structures, which predicted only inverse-agonists. In addition, two hits recapitulated the signaling profile of the co-crystal ligand with respect to the G protein and arrestin mediated signaling. This functional fidelity has important implications in drug design, as the ability to predict ligands with predefined signaling properties is highly desirable. However, the agonist-bound state provides an uncertain template for modeling the activated conformation of other GPCRs, as a dopamine D2 receptor (DRD2) activated model templated on the activated β2AR structure returned few hits of only marginal potency.


Molecular Pharmacology | 2014

Regulation of β2-Adrenergic Receptor Function by Conformationally Selective Single-Domain Intrabodies

Dean P. Staus; Laura M. Wingler; Ryan T. Strachan; Søren Rasmussen; Els Pardon; Seungkirl Ahn; Jan Steyaert; Brian K. Kobilka; Robert J. Lefkowitz

The biologic activity induced by ligand binding to orthosteric or allosteric sites on a G protein–coupled receptor (GPCR) is mediated by stabilization of specific receptor conformations. In the case of the β2 adrenergic receptor, these ligands are generally small-molecule agonists or antagonists. However, a monomeric single-domain antibody (nanobody) from the Camelid family was recently found to allosterically bind and stabilize an active conformation of the β2-adrenergic receptor (β2AR). Here, we set out to study the functional interaction of 18 related nanobodies with the β2AR to investigate their roles as novel tools for studying GPCR biology. Our studies revealed several sequence-related nanobody families with preferences for active (agonist-occupied) or inactive (antagonist-occupied) receptors. Flow cytometry analysis indicates that all nanobodies bind to epitopes displayed on the intracellular receptor surface; therefore, we transiently expressed them intracellularly as “intrabodies” to test their effects on β2AR-dependent signaling. Conformational specificity was preserved after intrabody conversion as demonstrated by the ability for the intracellularly expressed nanobodies to selectively bind agonist- or antagonist-occupied receptors. When expressed as intrabodies, they inhibited G protein activation (cyclic AMP accumulation), G protein–coupled receptor kinase (GRK)–mediated receptor phosphorylation, β-arrestin recruitment, and receptor internalization to varying extents. These functional effects were likely due to either steric blockade of downstream effector (Gs, β-arrestin, GRK) interactions or stabilization of specific receptor conformations which do not support effector coupling. Together, these findings strongly implicate nanobody-derived intrabodies as novel tools to study GPCR biology.


Journal of Biological Chemistry | 2014

Divergent Transducer-specific Molecular Efficacies Generate Biased Agonism at a G Protein-coupled Receptor (GPCR) *

Ryan T. Strachan; Jinpeng Sun; David H. Rominger; Jonathan D. Violin; Seungkirl Ahn; Alex R.B. Thomsen; Xiao Zhu; Andrew Kleist; Tommaso Costa; Robert J. Lefkowitz

Background: Biased agonism is an incompletely understood phenomenon describing the unequal activation of different signal transduction pathways by a G protein-coupled receptor (GPCR). Results: A cell-free approach using GPCR-transducer fusion proteins (G-protein or β-arrestin) quantifies signaling in vitro to elucidate the molecular basis of biased agonism. Conclusion: Differences in ligand-receptor-transducer coupling account for biased agonism in cells. Significance: Biased agonism is a bona fide molecular property of GPCR ligands. The concept of “biased agonism” arises from the recognition that the ability of an agonist to induce a receptor-mediated response (i.e. “efficacy”) can differ across the multiple signal transduction pathways (e.g. G protein and β-arrestin (βarr)) emanating from a single GPCR. Despite the therapeutic promise of biased agonism, the molecular mechanism(s) whereby biased agonists selectively engage signaling pathways remain elusive. This is due in large part to the challenges associated with quantifying ligand efficacy in cells. To address this, we developed a cell-free approach to directly quantify the transducer-specific molecular efficacies of balanced and biased ligands for the angiotensin II type 1 receptor (AT1R), a prototypic GPCR. Specifically, we defined efficacy in allosteric terms, equating shifts in ligand affinity (i.e. KLo/KHi) at AT1R-Gq and AT1R-βarr2 fusion proteins with their respective molecular efficacies for activating Gq and βarr2. Consistent with ternary complex model predictions, transducer-specific molecular efficacies were strongly correlated with cellular efficacies for activating Gq and βarr2. Subsequent comparisons across transducers revealed that biased AT1R agonists possess biased molecular efficacies that were in strong agreement with the signaling bias observed in cellular assays. These findings not only represent the first measurements of the thermodynamic driving forces underlying differences in ligand efficacy between transducers but also support a molecular mechanism whereby divergent transducer-specific molecular efficacies generate biased agonism at a GPCR.


Journal of Biological Chemistry | 2014

Allosteric modulation of β-arrestin-biased angiotensin II type 1 receptor signaling by membrane stretch.

Wei Tang; Ryan T. Strachan; Robert J. Lefkowitz; Howard A. Rockman

Background: AT1Rs function as mechanosensors to activate β-arrestin-dependent signaling. Results: Osmotic stretch induces active conformations of the AT1R and β-arrestin2 that are exclusively stabilized by β-arrestin-biased agonists. Conclusion: Membrane stretch functions as an allosteric modulator to selectively enhance β-arrestin-biased signaling. Significance: Membrane stretch induces a biased conformation of the AT1R similar to that of β-arrestin-biased agonists. It has recently been appreciated that the angiotensin II type 1 receptor (AT1R), a prototypic member of the G protein-coupled receptor superfamily, also functions as a mechanosensor. Specifically, mechanical stretch activates the AT1R to promote downstream signaling mediated exclusively by the multifunctional scaffold protein, β-arrestin, in a manner consistent with previously identified β-arrestin-biased ligands. However, the ligand-independent mechanism by which mechanical stretch promotes β-arrestin-biased signaling remains unknown. Implicit in the concept of biased agonism (i.e. the ability of an agonist to activate a subset of receptor-mediated signaling pathways) is the notion that distinct active conformations of the receptor mediate differential activation of signaling pathways. Here we determined whether mechanical stretch stabilizes distinct β-arrestin-activating conformations of the AT1R by using β-arrestin2-biased agonists as conformational probes in pharmacological and biophysical assays. When tested at cells expressing the AT1R fused to β-arrestin (AT1R-β-arrestin2), we found that osmotic stretch increased the binding affinity and potency of the β-arrestin-biased agonist TRV120023, with no effect on the balanced agonist AngII. In addition, the effect of osmotic stretch on ERK activation was markedly augmented in cells expressing the AT1R-β-arrestin2 fusion compared with the wild type AT1R and completely blocked in cells expressing the AT1R-Gq fusion. Biophysical experiments with an intramolecular BRET β-arrestin2 biosensor revealed that osmotic stretch and TRV120023 activate AT1Rs to stabilize β-arrestin2 active conformations that differ from those stabilized by the AT1R activated by angiotensin II. Together, these data support a novel ligand-independent mechanism whereby mechanical stretch allosterically stabilizes specific β-arrestin-biased active conformations of the AT1R and has important implications for understanding pathophysiological AT1R signaling.


Nature Chemical Biology | 2016

Conformationally selective RNA aptamers allosterically modulate the [beta]2-adrenoceptor

Alem W. Kahsai; James W. Wisler; Jungmin Lee; Seungkirl Ahn; Thomas Joseph Cahill; S. Moses Dennison; Dean P. Staus; Alex R.B. Thomsen; Kara Anasti; Biswaranjan Pani; Laura M. Wingler; Hemant Desai; Kristin M. Bompiani; Ryan T. Strachan; Xiaoxia Qin; S. Munir Alam; Bruce A. Sullenger; Robert J. Lefkowitz

G-protein-coupled receptor (GPCR) ligands function by stabilizing multiple, functionally distinct receptor conformations. This property underlies how “biased agonists” activate specific subsets of a given receptor’s signaling profile. However, stabilization of distinct active GPCR conformations to enable structural characterization of mechanisms underlying GPCR activation remains difficult. These challenges have accentuated the need for receptor tools that allosterically stabilize and regulate receptor function via unique, previously unappreciated mechanisms. Here, utilizing a highly diverse RNA library combined with advanced selection strategies involving state-of-the-art next-generation sequencing and bioinformatics analyses, we identify RNA aptamers that bind a prototypical GPCR, β2-adrenoceptor (β2AR). Using biochemical, pharmacological, and biophysical approaches, we demonstrate that these aptamers bind with nanomolar affinity at defined surfaces of the receptor, allosterically stabilizing active, inactive, and ligand-specific receptor conformations. The discovery of RNA aptamers as allosteric GPCR modulators significantly expands the diversity of ligands available to study the structural and functional regulation of GPCRs.


British Journal of Pharmacology | 2012

Chronic treatment in vivo with β‐adrenoceptor agonists induces dysfunction of airway β2‐adrenoceptors and exacerbates lung inflammation in mice

Rui Lin; Simone Degan; Barbara S. Theriot; Bernard M. Fischer; Ryan T. Strachan; Jiurong Liang; Richard A. Pierce; Mary E. Sunday; Paul W. Noble; Monica Kraft; Arnold R. Brody; Julia Kl Walker

BACKGROUND AND PURPOSE Inhalation of a β‐adrenoceptor agonist (β‐agonist) is first‐line asthma therapy, used for both prophylaxis against, and acute relief of, bronchoconstriction. However, repeated clinical use of β‐agonists leads to impaired bronchoprotection and, in some cases, adverse patient outcomes. Mechanisms underlying this β2‐adrenoceptor dysfunction are not well understood, due largely to the lack of a comprehensive animal model and the uncertainty as to whether or not bronchorelaxation in mice is mediated by β2‐adrenoceptors. Thus, we aimed to develop a mouse model that demonstrated functional β‐agonist‐induced β2‐adrenoceptor desensitization in the context of allergic inflammatory airway disease.

Collaboration


Dive into the Ryan T. Strachan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bryan L. Roth

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Els Pardon

Vrije Universiteit Brussel

View shared research outputs
Researchain Logo
Decentralizing Knowledge