Network


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

Hotspot


Dive into the research topics where Aylin C. Hanyaloglu is active.

Publication


Featured researches published by Aylin C. Hanyaloglu.


Annual Review of Pharmacology and Toxicology | 2008

Regulation of GPCRs by Endocytic Membrane Trafficking and Its Potential Implications

Aylin C. Hanyaloglu; Mark von Zastrow

The endocytic pathway tightly controls the activity of G protein-coupled receptors (GPCRs). Ligand-induced endocytosis can drive receptors into divergent lysosomal and recycling pathways, producing essentially opposite effects on the strength and duration of cellular signaling via heterotrimeric G proteins, and may also promote distinct signaling events from intracellular membranes. This chapter reviews recent developments toward understanding the molecular machinery and functional implications of GPCR sorting in the endocytic pathway, focusing on mammalian GPCRs whose ligand-induced endocytosis is mediated primarily by clathrin-coated pits. Lysosomal sorting of a number of GPCRs occurs via a highly conserved mechanism requiring covalent tagging of receptors with ubiquitin. There is increasing evidence that additional, noncovalent mechanisms control the sorting of endocytosed GPCRs to lysosomes in mammalian cells. Recycling of several GPCRs to the plasma membrane is also specifically sorted, via a mechanism requiring both receptor-specific and shared sorting proteins. The current data reveal an unprecedented degree of specificity and plasticity in the cellular regulation of mammalian GPCRs by endocytic membrane trafficking. These developments have fundamental implications for GPCR pharmacology, and suggest new mechanisms that could be exploited in GPCR-directed pharmacotherapy.


International Journal of Obesity | 2015

The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents.

Arianna Psichas; Michelle L. Sleeth; Kevin G. Murphy; Lucy Brooks; Gavin A. Bewick; Aylin C. Hanyaloglu; M. A. Ghatei; S.R. Bloom; Gary Frost

Background and Objectives:The gut hormones peptide YY (PYY) and glucagon-like peptide 1 (GLP-1) acutely suppress appetite. The short chain fatty acid (SCFA) receptor, free fatty acid receptor 2 (FFA2) is present on colonic enteroendocrine L cells, and a role has been suggested for SCFAs in appetite regulation. Here, we characterise the in vitro and in vivo effects of colonic propionate on PYY and GLP-1 release in rodents, and investigate the role of FFA2 in mediating these effects using FFA2 knockout mice.Methods:We used Wistar rats, C57BL6 mice and free fatty acid receptor 2 knockout (FFA−/−) mice on a C57BL6 background to explore the impact of the SCFA propionate on PYY and GLP-1 release. Isolated colonic crypt cultures were used to assess the effects of propionate on gut hormone release in vitro. We subsequently developed an in vivo technique to assess gut hormone release into the portal vein following colonic infusion of propionate.Results:Propionate stimulated the secretion of both PYY and GLP-1 from wild-type primary murine colonic crypt cultures. This effect was significantly attenuated in cultures from FFA2−/− mice. Intra-colonic infusion of propionate elevated PYY and GLP-1 levels in jugular vein plasma in rats and in portal vein plasma in both rats and mice. However, propionate did not significantly stimulate gut hormone release in FFA2−/− mice.Conclusions:Intra-colonic administration of propionate stimulates the concurrent release of both GLP-1 and PYY in rats and mice. These data demonstrate that FFA2 deficiency impairs SCFA-induced gut hormone secretion both in vitro and in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Rescue of defective G protein–coupled receptor function in vivo by intermolecular cooperation

Adolfo Rivero-Müller; Yen-Yin Chou; Inhae Ji; Svetlana Lajic; Aylin C. Hanyaloglu; Kim Jonas; Nafis A. Rahman; Tae H. Ji; Ilpo Huhtaniemi

G protein–coupled receptors (GPCRs) are ubiquitous mediators of signaling of hormones, neurotransmitters, and sensing. The old dogma is that a one ligand/one receptor complex constitutes the functional unit of GPCR signaling. However, there is mounting evidence that some GPCRs form dimers or oligomers during their biosynthesis, activation, inactivation, and/or internalization. This evidence has been obtained exclusively from cell culture experiments, and proof for the physiological significance of GPCR di/oligomerization in vivo is still missing. Using the mouse luteinizing hormone receptor (LHR) as a model GPCR, we demonstrate that transgenic mice coexpressing binding-deficient and signaling-deficient forms of LHR can reestablish normal LH actions through intermolecular functional complementation of the mutant receptors in the absence of functional wild-type receptors. These results provide compelling in vivo evidence for the physiological relevance of intermolecular cooperation in GPCR signaling.


The EMBO Journal | 2005

Essential role of Hrs in a recycling mechanism mediating functional resensitization of cell signaling

Aylin C. Hanyaloglu; Emma McCullagh; Mark von Zastrow

Hepatocyte growth factor‐regulated tyrosine kinase substrate (Hrs) is well known to terminate cell signaling by sorting activated receptors to the MVB/lysosomal pathway. Here we identify a distinct role of Hrs in promoting rapid recycling of endocytosed signaling receptors to the plasma membrane. This function of Hrs is specific for receptors that recycle in a sequence‐directed manner, in contrast to default recycling by bulk membrane flow, and is distinguishable in several ways from previously identified membrane‐trafficking functions of Hrs/Vps27p. In particular, Hrs function in sequence‐directed recycling does not require other mammalian Class E gene products involved in MVB/lysosomal sorting, nor is receptor ubiquitination required. Mutational studies suggest that the VHS domain of Hrs plays an important role in sequence‐directed recycling. Disrupting Hrs‐dependent recycling prevented functional resensitization of the β2‐adrenergic receptor, converting the temporal profile of cell signaling by this prototypic G protein‐coupled receptor from sustained to transient. These studies identify a novel function of Hrs in a cargo‐specific recycling mechanism, which is critical to controlling functional activity of the largest known family of signaling receptors.


Trends in Endocrinology and Metabolism | 2002

Applications of novel resonance energy transfer techniques to study dynamic hormone receptor interactions in living cells.

Karin A. Eidne; Karen M. Kroeger; Aylin C. Hanyaloglu

Many aspects of hormone receptor function that are crucial for controlling signal transduction of endocrine pathways can be monitored more accurately with the use of non-invasive, live cell resonance energy transfer (RET) techniques. Fluorescent RET (FRET), and its variation, bioluminescent RET (BRET), can be used to assess the real-time responses to specific hormonal stimuli, whilst preserving the cellular protein network, compartmentalization and spatial arrangement. Both FRET and BRET can be readily adapted to the study of membrane proteins. Here, we focus on their applications to the analysis of interactions involving the superfamily of hormone G-protein-coupled receptors. RET is also emerging as a significant tool for the determination of protein function in general. Such techniques will undoubtedly be of value in determining the functional identities of the vast array of proteins that are encoded by the human genome.


Molecular and Cellular Endocrinology | 2011

Regulation of GPCR signal networks via membrane trafficking.

Frederic Jean-Alphonse; Aylin C. Hanyaloglu

G-protein-coupled receptors (GPCRs) are a superfamily of cell surface signaling proteins that act as central molecular activators and integrators in all endocrine systems. Membrane trafficking of GPCRs is a fundamental process in shaping extensive signaling networks activated by these receptors. Mounting evidence has identified an increasingly complex network of pathways and protein interactions that a GPCR can traverse and associate with, indicating a multi-level system of regulation. This review will discuss the recent developments in how GPCRs are trafficked to the cell surface as newly synthesized receptors, their recruitment to the clathrin-mediated pathway for endocytosis, and their sorting to subsequent divergent post-endocytic fates, focusing primarily on hormone-activated GPCRs. Current models depicting the classic roles membrane trafficking plays in GPCR signaling have evolved to a highly regulated and complex system than previously appreciated. These developments impart key mechanistic information on how spatial and temporal aspects of GPCR signaling may be integrated and could provide pathway-specific targets to be exploited for therapeutic intervention.


Journal of Biological Chemistry | 2001

Casein kinase II sites in the intracellular C-terminal domain of the thyrotropin-releasing hormone receptor and chimeric gonadotropin-releasing hormone receptors contribute to β-arrestin-dependent internalization

Aylin C. Hanyaloglu; Milka Vrecl; Karen M. Kroeger; Lauren E. C. Miles; Hongwei Qian; Walter G. Thomas; Karin A. Eidne

We have previously shown that the mammalian gonadotropin-releasing hormone receptor (GnRHR), a unique G-protein-coupled receptor (GPCR) lacking an intracellular carboxyl tail (C-tail), does not follow a β-arrestin-dependent internalization pathway. However, internalization of a chimeric GnRHR with the thyrotropin-releasing hormone receptor (TRHR) C-tail does utilize β-arrestin. Here, we have investigated the sites within the intracellular C-tail domain that are important for conferring β-arrestin-dependent internalization. In contrast to the chimeric GnRHR with a TRHR C-tail, a chimeric GnRHR with the catfish GnRHR C-tail is not β-arrestin-dependent. Sequence comparisons between these chimeric receptors show three consensus phosphorylation sites for casein kinase II (CKII) in the TRHR C-tail but none in the catfish GnRHR C-tail. We thus investigated a role for CKII sites in determining GPCR internalization via β-arrestin. Sequential introduction of three CKII sites into the chimera with the catfish C-tail (H354D,A366E,G371D) resulted in a change in the pattern of receptor phosphorylation and β-arrestin-dependence, which only occurred when all three sites were introduced. Conversely, mutation of the putative CKII sites (T365A,T371A,S383A) in the C-tail of a β-arrestin-sensitive GPCR, the TRHR, resulted in decreased receptor phosphorylation and a loss of β-arrestin-dependence. Mutation of all three CKII sites was necessary before a loss of β-arrestin-dependence was observed. Visualization of β-arrestin/GFP redistribution confirmed a loss or gain of β-arrestin sensitivity for receptor mutants. Internalization of receptors without C-tail CKII sites was promoted by a phosphorylation-independent β-arrestin mutant (R169E), suggesting that these receptors do not contain the necessary phosphorylation sites required for β-arrestin-dependent internalization. Apigenin, a specific CKII inhibitor, blocked the increase in receptor internalization by β-arrestin, thus providing further support for the involvement of CKII. This study presents evidence of a novel role for C-tail CKII consensus sites in targeting these GPCRs to the β-arrestin-dependent pathway.


Journal of Biological Chemistry | 2015

Single Molecule Analysis of Functionally Asymmetric G Protein-coupled Receptor (GPCR) Oligomers Reveals Diverse Spatial and Structural Assemblies

Kim Jonas; Francesca Fanelli; Ilpo Huhtaniemi; Aylin C. Hanyaloglu

Background: GPCRs form complex oligomers whose role in signaling is poorly understood. Results: Super-resolution imaging of functionally asymmetric oligomers reveals diverse functional and structural organizations and the ability to alter signal responses. Conclusion: GPCR oligomers may fine-tune receptor signaling by altering the functional role of individual protomers. Significance: Distinct oligomers could be exploited pharmacologically to improve efficacy, selectivity, and/or specificity. Formation of G protein-coupled receptors (GPCRs) into dimers and higher order oligomers represents a key mechanism in pleiotropic signaling, yet how individual protomers function within oligomers remains poorly understood. We present a super-resolution imaging approach, resolving single GPCR molecules to ∼8 nm resolution in functional asymmetric dimers and oligomers using dual-color photoactivatable dyes and localization microscopy (PD-PALM). PD-PALM of two functionally defined mutant luteinizing hormone receptors (LHRs), a ligand-binding deficient receptor (LHRB−) and a signaling-deficient (LHRS−) receptor, which only function via intermolecular cooperation, favored oligomeric over dimeric formation. PD-PALM imaging of trimers and tetramers revealed specific spatial organizations of individual protomers in complexes where the ratiometric composition of LHRB− to LHRS− modulated ligand-induced signal sensitivity. Structural modeling of asymmetric LHR oligomers strongly aligned with PD-PALM-imaged spatial arrangements, identifying multiple possible helix interfaces mediating inter-protomer associations. Our findings reveal that diverse spatial and structural assemblies mediating GPCR oligomerization may acutely fine-tune the cellular signaling profile.


Journal of Biological Chemistry | 2007

A Novel Sorting Sequence in the β2-Adrenergic Receptor Switches Recycling from Default to the Hrs-dependent Mechanism

Aylin C. Hanyaloglu; Mark von Zastrow

Plasma membrane recycling of G protein-coupled receptors can occur by at least two distinct mechanisms as follows: a “default” mechanism that occurs nonselectively, and a specifically sorted mechanism that requires the endosome-associated protein Hrs. In this study we have defined a sequence in the β2-adrenergic receptor cytoplasmic tail that confers Hrs dependence on receptor recycling. This sequence resembles acidic dileucine class motifs found in other membrane proteins but is structurally and functionally distinct from previously identified sorting sequences. Mutation of the novel sorting sequence rendered plasma membrane recycling independent of Hrs and independent of a distal PDZ ligand required for Hrs-dependent recycling. We propose that the novel sorting sequence functions to “switch” endocytic trafficking between mechanistically distinct recycling modes, thereby explaining failure of the wild type β2-adrenergic receptor to recycle efficiently by default.


Journal of Biological Chemistry | 2014

Spatially Restricted G Protein-coupled Receptor Activity via Divergent Endocytic Compartments

Frederic Jean-Alphonse; Shanna Bowersox; Stanford Chen; Gemma Beard; Manojkumar A. Puthenveedu; Aylin C. Hanyaloglu

Background: Following ligand-induced internalization, GPCRs are sorted by diverse receptor motifs and protein interactions. Results: Distinct GPCRs are targeted to a pre-early endosome compartment for their sorting and MAPK signaling. Conclusion: GPCR sorting motifs and their interacting proteins provide specificity in endosomal targeting and receptor signaling. Significance: We describe a system to reprogram GPCR signaling at an unprecedented spatial level. Postendocytic sorting of G protein-coupled receptors (GPCRs) is driven by their interactions between highly diverse receptor sequence motifs with their interacting proteins, such as postsynaptic density protein (PSD95), Drosophila disc large tumor suppressor (Dlg1), zonula occludens-1 protein (zo-1) (PDZ) domain proteins. However, whether these diverse interactions provide an underlying functional specificity, in addition to driving sorting, is unknown. Here we identify GPCRs that recycle via distinct PDZ ligand/PDZ protein pairs that exploit their recycling machinery primarily for targeted endosomal localization and signaling specificity. The luteinizing hormone receptor (LHR) and β2-adrenergic receptor (B2AR), two GPCRs sorted to the regulated recycling pathway, underwent divergent trafficking to distinct endosomal compartments. Unlike B2AR, which traffics to early endosomes (EE), LHR internalizes to distinct pre-early endosomes (pre-EEs) for its recycling. Pre-EE localization required interactions of the LHR C-terminal tail with the PDZ protein GAIP-interacting protein C terminus, inhibiting its traffic to EEs. Rerouting the LHR to EEs, or EE-localized GPCRs to pre-EEs, spatially reprograms MAPK signaling. Furthermore, LHR-mediated activation of MAPK signaling requires internalization and is maintained upon loss of the EE compartment. We propose that combinatorial specificity between GPCR sorting sequences and interacting proteins dictates an unprecedented spatiotemporal control in GPCR signal activity.

Collaboration


Dive into the Aylin C. Hanyaloglu's collaboration.

Top Co-Authors

Avatar

Kim Jonas

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karin A. Eidne

University of Western Australia

View shared research outputs
Top Co-Authors

Avatar

Ilpo Huhtaniemi

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ilpo Huhtaniemi

Michigan State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge