51
|
Kavanagh JJ, Taylor JL. Voluntary activation of muscle in humans: does serotonergic neuromodulation matter? J Physiol 2022; 600:3657-3670. [PMID: 35864781 PMCID: PMC9541597 DOI: 10.1113/jp282565] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022] Open
Abstract
Ionotropic inputs to motoneurones have the capacity to depolarise and hyperpolarise the motoneurone, whereas neuromodulatory inputs control the state of excitability of the motoneurone. Intracellular recordings of motoneurones from in vitro and in situ animal preparations have provided extraordinary insight into the mechanisms that underpin how neuromodulators regulate neuronal excitability. However, far fewer studies have attempted to translate the findings from cellular and molecular studies into a human model. In this review, we focus on the role that serotonin (5-HT) plays in muscle activation in humans. 5-HT is a potent regulator of neuronal firing rates, which can influence the force that can be generated by muscles during voluntary contractions. We firstly outline structural and functional characteristics of the serotonergic system, and then describe how motoneurone discharge can be facilitated and suppressed depending on the 5-HT receptor subtype that is activated. We then provide a narrative on how 5-HT effects can influence voluntary activation during muscle contractions in humans, and detail how 5-HT may be a mediator of exercise-induced fatigue that arises from the central nervous system.
Collapse
Affiliation(s)
- Justin J. Kavanagh
- Neural Control of Movement laboratoryMenzies Health Institute QueenslandGriffith UniversityGold CoastAustralia
| | - Janet L. Taylor
- Centre for Human Performance, School of Medical and Health SciencesEdith Cowan UniversityPerthAustralia
- Neuroscience Research AustraliaSydneyAustralia
| |
Collapse
|
52
|
Schmoll M, Hinterdobler W. Tools for adapting to a complex habitat: G-protein coupled receptors in Trichoderma. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:65-97. [PMID: 36357080 DOI: 10.1016/bs.pmbts.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Sensing the environment and interpretation of the received signals are crucial competences of living organisms in order to properly adapt to their habitat, succeed in competition and to reproduce. G-protein coupled receptors (GPCRs) are members of a large family of sensors for extracellular signals and represent the starting point of complex signaling cascades regulating a plethora of intracellular physiological processes and output pathways in fungi. In Trichoderma spp. current research involves a wide range of topics from enzyme production, light response and secondary metabolism to sexual and asexual development as well as biocontrol, all of which require delicate balancing of resources in response to the environmental challenges or biotechnological needs at hand, which are crucially impacted by the surroundings of the fungi and their intercellular signaling cascades triggering a precisely tailored response. In this review we summarize recent findings on sensing by GPCRs in Trichoderma, including the function of pheromone receptors, glucose sensing by CSG1 and CSG2, regulation of secondary metabolism by GPR8 and impacts on mycoparasitism by GPR1. Additionally, we provide an overview on structural determinants, posttranslational modifications and interactions for regulation, activation and signal termination of GPCRs in order to inspire future in depth analyses of their function and to understand previous regulatory outcomes of natural and biotechnological processes modulated or enabled by GPCRs.
Collapse
Affiliation(s)
- Monika Schmoll
- Department of Microbiology and Ecosystem Science, Division of Terrestrial Ecosystem Research, University of Vienna, Vienna, Austria.
| | | |
Collapse
|
53
|
Kim GT, Kim EY, Shin SH, Lee H, Lee SH, Sohn KY, Kim JW. Suppression of tumor progression by thioredoxin-interacting protein-dependent adenosine 2B receptor degradation in a PLAG-treated Lewis lung carcinoma-1 model of non-small cell lung cancer. Neoplasia 2022; 31:100815. [PMID: 35728512 PMCID: PMC9209866 DOI: 10.1016/j.neo.2022.100815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 05/23/2022] [Accepted: 06/08/2022] [Indexed: 11/03/2022]
Abstract
PLAG effectively inhibited excessive growth of LLC1 cells in an NSCLC model. PLAG inhibited tumor growth by inducing adenosine 2B receptor (A2BR) degradation. Unlike antagonists, PLAG terminates rather than suppresses signaling pathways. A2BR degradation by PLAG occurs through expression and re-localization of TXNIP.
Extracellular adenosine in the tumor microenvironment plays a vital role in cancer development. Specifically, activation of adenosine receptors affects tumor cell growth and adenosine release. We examined the anti-tumor efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in animal models, revealing the role of PLAG in inhibiting tumor progression by promoting the degradation of adenosine 2B receptors (A2BRs) in tumors. PLAG induced the expression of thioredoxin-interacting protein (TXNIP), a type of α-arrestin that accelerates A2BR internalization by interacting with A2BR complexes containing β-arrestin. Engulfed receptors bound to TXNIP were rapidly degraded after E3 ligase recruitment and ubiquitination, resulting in early termination of intracellular signals that promote tumor overgrowth. However, in control cancer cells, A2BRs bound to protein phosphatase 2A and were returned to the cell membrane instead of being degraded, resulting in continuous receptor-mediated signaling by pathways including the Raf-Erk axis, which promotes tumor proliferation. A TXNIP-silenced cell-implanted mouse model and TXNIP knockout (KO) mice were used to verify that PLAG-mediated suppression of tumor progression is dependent on TXNIP expression. Increased tumor growth was observed in TXNIP-silenced cell-implanted mice, and the anti-tumor effects of PLAG, including delayed tumor overgrowth, were greatly reduced. However, the anti-tumor effects of PLAG were observed in cancer cell-implanted TXNIP-KO mice, which indicates that PLAG produces anti-tumor effects by enhancing TXNIP expression in tumor cells. These essential functions of PLAG, including delaying tumor growth via A2BR degradation, suggest innovative directions for anticancer drug development.
Collapse
Affiliation(s)
- Guen Tae Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 10F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea.
| |
Collapse
|
54
|
Laeremans T, Sands ZA, Claes P, De Blieck A, De Cesco S, Triest S, Busch A, Felix D, Kumar A, Jaakola VP, Menet C. Accelerating GPCR Drug Discovery With Conformation-Stabilizing VHHs. Front Mol Biosci 2022; 9:863099. [PMID: 35677880 PMCID: PMC9170359 DOI: 10.3389/fmolb.2022.863099] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/22/2022] [Indexed: 01/19/2023] Open
Abstract
The human genome encodes 850 G protein-coupled receptors (GPCRs), half of which are considered potential drug targets. GPCRs transduce extracellular stimuli into a plethora of vital physiological processes. Consequently, GPCRs are an attractive drug target class. This is underlined by the fact that approximately 40% of marketed drugs modulate GPCRs. Intriguingly 60% of non-olfactory GPCRs have no drugs or candidates in clinical development, highlighting the continued potential of GPCRs as drug targets. The discovery of small molecules targeting these GPCRs by conventional high throughput screening (HTS) campaigns is challenging. Although the definition of success varies per company, the success rate of HTS for GPCRs is low compared to other target families (Fujioka and Omori, 2012; Dragovich et al., 2022). Beyond this, GPCR structure determination can be difficult, which often precludes the application of structure-based drug design approaches to arising HTS hits. GPCR structural studies entail the resource-demanding purification of native receptors, which can be challenging as they are inherently unstable when extracted from the lipid matrix. Moreover, GPCRs are flexible molecules that adopt distinct conformations, some of which need to be stabilized if they are to be structurally resolved. The complexity of targeting distinct therapeutically relevant GPCR conformations during the early discovery stages contributes to the high attrition rates for GPCR drug discovery programs. Multiple strategies have been explored in an attempt to stabilize GPCRs in distinct conformations to better understand their pharmacology. This review will focus on the use of camelid-derived immunoglobulin single variable domains (VHHs) that stabilize disease-relevant pharmacological states (termed ConfoBodies by the authors) of GPCRs, as well as GPCR:signal transducer complexes, to accelerate drug discovery. These VHHs are powerful tools for supporting in vitro screening, deconvolution of complex GPCR pharmacology, and structural biology purposes. In order to demonstrate the potential impact of ConfoBodies on translational research, examples are presented of their role in active state screening campaigns and structure-informed rational design to identify de novo chemical space and, subsequently, how such matter can be elaborated into more potent and selective drug candidates with intended pharmacology.
Collapse
|
55
|
Engineered Allosteric Regulation of Protein Function. J Mol Biol 2022; 434:167620. [PMID: 35513109 DOI: 10.1016/j.jmb.2022.167620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/27/2022] [Accepted: 04/26/2022] [Indexed: 11/20/2022]
Abstract
Allosteric regulation of proteins has been utilized to study various aspects of cell signaling, from unicellular events to organism-wide phenotypes. However, traditional methods of allosteric regulation, such as constitutively active mutants and inhibitors, lack tight spatiotemporal control. This often leads to unintended signaling consequences that interfere with data interpretation. To overcome these obstacles, researchers employed protein engineering approaches that enable tight control of protein function through allosteric mechanisms. These methods provide high specificity as well as spatial and temporal precision in regulation of protein activity in vitro and in vivo. In this review, we focus on the recent advancements in engineered allosteric regulation and discuss the various bioengineered allosteric techniques available now, from chimeric GPCRs to chemogenetic and optogenetic switches. We highlight the benefits and pitfalls of each of these techniques as well as areas in which future improvements can be made. Additionally, we provide a brief discussion on implementation of engineered allosteric regulation approaches, demonstrating that these tools can shed light on elusive biological events and have the potential to be utilized in precision medicine.
Collapse
|
56
|
Melick CH, Lama-Sherpa TD, Curukovic A, Jewell JL. G-Protein Coupled Receptor Signaling and Mammalian Target of Rapamycin Complex 1 Regulation. Mol Pharmacol 2022; 101:181-190. [PMID: 34965982 PMCID: PMC9092479 DOI: 10.1124/molpharm.121.000302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) senses upstream stimuli to regulate numerous cellular functions such as metabolism, growth, and autophagy. Increased activation of mTOR complex 1 (mTORC1) is typically observed in human disease and continues to be an important therapeutic target. Understanding the upstream regulators of mTORC1 will provide a crucial link in targeting hyperactivated mTORC1 in human disease. In this mini-review, we will discuss the regulation of mTORC1 by upstream stimuli, with a specific focus on G-protein coupled receptor signaling to mTORC1. SIGNIFICANCE STATEMENT: mTORC1 is a master regulator of many cellular processes and is often hyperactivated in human disease. Therefore, understanding the molecular underpinnings of G-protein coupled receptor signaling to mTORC1 will undoubtedly be beneficial for human disease.
Collapse
Affiliation(s)
- Chase H Melick
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Tshering D Lama-Sherpa
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Adna Curukovic
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jenna L Jewell
- Department of Molecular Biology, Harold C. Simmons Comprehensive Cancer, and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
57
|
Hong W, Yang B, He Q, Wang J, Weng Q. New Insights of CCR7 Signaling in Dendritic Cell Migration and Inflammatory Diseases. Front Pharmacol 2022; 13:841687. [PMID: 35281921 PMCID: PMC8914285 DOI: 10.3389/fphar.2022.841687] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
CCR7, collaborated with its ligands CCL19 and CCL21, controls extensive migratory events in the immune system. CCR7-bearing dendritic cells can swarm into T-cell zones in lymph nodes, initiating the antigen presentation and T-cell response. Abnormal expression of CCR7 in dendritic cells will cause a series of inflammatory diseases due to the chaotic dendritic cell trafficking. In this review, we take an in-depth look at the structural–functional domains of CCR7 and CCR7-bearing dendritic cell trajectory to lymph nodes. Then, we summarize the regulatory network of CCR7, including transcriptional regulation, translational and posttranslational regulation, internalization, desensitization, and recycling. Furthermore, the potential strategies of targeting the CCR7 network to regulate dendritic cell migration and to deal with inflammatory diseases are integrated, which not only emphasizes the possibility of CCR7 to be a potential target of immunotherapy but also has an implication on the homing of dendritic cells to benefit inflammatory diseases.
Collapse
Affiliation(s)
- Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- *Correspondence: Qinjie Weng, ; Jiajia Wang,
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Qinjie Weng, ; Jiajia Wang,
| |
Collapse
|
58
|
Activity-based, bioorthogonal imaging of phospholipase D reveals spatiotemporal dynamics of GPCR-Gq signaling. Cell Chem Biol 2022; 29:67-73.e3. [PMID: 34161786 PMCID: PMC8655016 DOI: 10.1016/j.chembiol.2021.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 01/22/2023]
Abstract
Canonically, G-protein-coupled receptor (GPCR) signaling is transient and confined to the plasma membrane (PM). Deviating from this paradigm, the parathyroid hormone receptor (PTHR1) stimulates sustained Gs signaling at endosomes. In addition to Gs, PTHR1 activates Gq signaling; yet, in contrast to the PTHR1-Gs pathway, the spatiotemporal dynamics of the Gq branch of PTHR1 signaling and its relationship to Gs signaling remain largely ill defined. Recognizing that a downstream consequence of Gq signaling is the activation of phospholipase D (PLD) enzymes, we leverage activity-based, bioorthogonal imaging tools for PLD signaling to visualize and quantify the Gq branch of PTHR1 signaling. We establish that PTHR1-Gq signaling is short lived, exclusively at the PM, and antagonized by PTHR1 endocytosis. Our data support a model wherein Gq and Gs compete for ligand-bound receptors at the PM and more broadly highlight the utility of bioorthogonal tools for imaging PLDs as probes to visualize GPCR-Gq signaling.
Collapse
|
59
|
Gupta MK, Beg M, Mohammad S. Editorial: Cell Biology, Physiology and Molecular Pharmacology of G Protein Coupled Receptors. Front Cell Dev Biol 2021; 9:815291. [PMID: 34957128 PMCID: PMC8695997 DOI: 10.3389/fcell.2021.815291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Manveen Kaur Gupta
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic & Case Western Reserve University, Cleveland, OHIO, United States
| | - Muheeb Beg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sameer Mohammad
- Experimental Medicine, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| |
Collapse
|
60
|
Abrol R, Serrano E, Santiago LJ. Development of enhanced conformational sampling methods to probe the activation landscape of GPCRs. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:325-359. [PMID: 35034722 PMCID: PMC11476118 DOI: 10.1016/bs.apcsb.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
G protein-coupled receptors (GPCRs) make up the largest superfamily of integral membrane proteins and play critical signal transduction roles in many physiological processes. Developments in molecular biology, biophysical, biochemical, pharmacological, and computational techniques aimed at these important therapeutic targets are beginning to provide unprecedented details on the structural as well as functional basis of their pleiotropic signaling mediated by G proteins, β arrestins, and other transducers. This pleiotropy presents a pharmacological challenge as the same ligand-receptor interaction can cause a therapeutic effect as well as an undesirable on-target side-effect through different downstream pathways. GPCRs don't function as simple binary on-off switches but as finely tuned shape-shifting machines described by conformational ensembles, where unique subsets of conformations may be responsible for specific signaling cascades. X-ray crystallography and more recently cryo-electron microscopy are providing snapshots of some of these functionally-important receptor conformations bound to ligands and/or transducers, which are being utilized by computational methods to describe the dynamic conformational energy landscape of GPCRs. In this chapter, we review the progress in computational conformational sampling methods based on molecular dynamics and discrete sampling approaches that have been successful in complementing biophysical and biochemical studies on these receptors in terms of their activation mechanisms, allosteric effects, actions of biased ligands, and effects of pathological mutations. Some of the sampled simulation time scales are beginning to approach receptor activation time scales. The list of conformational sampling methods and example uses discussed is not exhaustive but includes representative examples that have pushed the limits of classical molecular dynamics and discrete sampling methods to describe the activation energy landscape of GPCRs.
Collapse
Affiliation(s)
- Ravinder Abrol
- Department of Chemistry and Biochemistry, California State University, Northridge, CA, United States.
| | - Erik Serrano
- Department of Chemistry and Biochemistry, California State University, Northridge, CA, United States
| | - Luis Jaimes Santiago
- Department of Chemistry and Biochemistry, California State University, Northridge, CA, United States
| |
Collapse
|
61
|
McGlone ER, Manchanda Y, Jones B, Pickford P, Inoue A, Carling D, Bloom SR, Tan T, Tomas A. Receptor Activity-Modifying Protein 2 (RAMP2) alters glucagon receptor trafficking in hepatocytes with functional effects on receptor signalling. Mol Metab 2021; 53:101296. [PMID: 34271220 PMCID: PMC8363841 DOI: 10.1016/j.molmet.2021.101296] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/01/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES Receptor Activity-Modifying Protein 2 (RAMP2) is a chaperone protein which allosterically binds to and interacts with the glucagon receptor (GCGR). The aims of this study were to investigate the effects of RAMP2 on GCGR trafficking and signalling in the liver, where glucagon (GCG) is important for carbohydrate and lipid metabolism. METHODS Subcellular localisation of GCGR in the presence and absence of RAMP2 was investigated using confocal microscopy, trafficking and radioligand binding assays in human embryonic kidney (HEK293T) and human hepatoma (Huh7) cells. Mouse embryonic fibroblasts (MEFs) lacking the Wiskott-Aldrich Syndrome protein and scar homologue (WASH) complex and the trafficking inhibitor monensin were used to investigate the effect of halted recycling of internalised proteins on GCGR subcellular localisation and signalling in the absence of RAMP2. NanoBiT complementation and cyclic AMP assays were used to study the functional effect of RAMP2 on the recruitment and activation of GCGR signalling mediators. Response to hepatic RAMP2 upregulation in lean and obese adult mice using a bespoke adeno-associated viral vector was also studied. RESULTS GCGR is predominantly localised at the plasma membrane in the absence of RAMP2 and exhibits remarkably slow internalisation in response to agonist stimulation. Rapid intracellular accumulation of GCG-stimulated GCGR in cells lacking the WASH complex or in the presence of monensin indicates that activated GCGR undergoes continuous cycles of internalisation and recycling, despite apparent GCGR plasma membrane localisation up to 40 min post-stimulation. Co-expression of RAMP2 induces GCGR internalisation both basally and in response to agonist stimulation. The intracellular retention of GCGR in the presence of RAMP2 confers a bias away from β-arrestin-2 recruitment coupled with increased activation of Gαs proteins at endosomes. This is associated with increased short-term efficacy for glucagon-stimulated cAMP production, although long-term signalling is dampened by increased receptor lysosomal targeting for degradation. Despite these signalling effects, only a minor disturbance of carbohydrate metabolism was observed in mice with upregulated hepatic RAMP2. CONCLUSIONS By retaining GCGR intracellularly, RAMP2 alters the spatiotemporal pattern of GCGR signalling. Further exploration of the effects of RAMP2 on GCGR in vivo is warranted.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Yusman Manchanda
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Ben Jones
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Phil Pickford
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - David Carling
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Stephen R Bloom
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Tricia Tan
- Section of Endocrinology and Investigative Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
62
|
Catar RA, Wischnewski O, Chen L, Heidecke H, Rutz C, Schülein R, Dragun D, Philippe A, Kusch A. Non-HLA antibodies targeting angiotensin II type 1 receptors and endothelin-1 type A receptors impair endothelial repair via a β2-arrestin link to the mTOR pathway. Kidney Int 2021; 101:498-509. [PMID: 34757123 DOI: 10.1016/j.kint.2021.09.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
Functional non-HLA antibodies (antibodies to non-human leukocyte antigens) targeting the G protein-coupled receptors angiotensin II type 1 receptor (AT1R) and endothelin-1 type A receptor (ETAR) are implicated in the pathogenesis of transplant vasculopathy. While ERK signaling (a regulator of cell growth) may represent a general cellular response to agonist stimulation, the molecular link between receptor stimulation and development of vascular obliteration has not been fully established. Here we hypothesize involvement of the versatile adaptor proteins, β-arrestins, and the major regulator of cell growth, PI3K/mTOR signaling, in impaired endothelial repair. To test this, human microvascular endothelial cells were treated with AT1R/ETAR antibodies isolated from patients with kidney transplant vasculopathy. These antibodies activated both mTOR complexes via AT1R and ETAR in a PI3K-dependent and ERK-independent manner. The mTOR inhibitor, rapamycin, completely abolished activation of mTORC1 and mTORC2 after long-term treatment with receptor antibodies. Imaging studies revealed that β2- but not β1-arrestin was recruited to ETAR in response to ET1 and patient antibodies but not with antibodies isolated from healthy individuals. Silencing of β2-arrestin by siRNA transfection significantly reduced ERK1/2 and mTORC2 activation. Non-HLA antibodies impaired endothelial repair by AT1R- and ETAR-induced mTORC2 signaling. Thus, we provide evidence that functional AT1R/ETAR antibodies induce ERK1/2 and mTOR signaling involving β2-arrestin in human microvascular endothelium. Hence, our data may provide a translational rational for mTOR inhibitors in combination with receptor blockers in patients with non-HLA receptor recognizing antibodies.
Collapse
Affiliation(s)
- Rusan Ali Catar
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany.
| | - Oskar Wischnewski
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany
| | - Lei Chen
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Department of Nephrology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai City, Guandong Province, People's Republic of China
| | | | - Claudia Rutz
- Leibniz Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V., Berlin, Germany
| | - Ralf Schülein
- Leibniz Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin e.V., Berlin, Germany
| | - Duska Dragun
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, Berlin, Germany
| | - Aurélie Philippe
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, Berlin, Germany
| | - Angelika Kusch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Medizinische Klinik m. S. Nephrologie und internistische Intensivmedizin, Berlin, Germany; Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Center for Cardiovascular Research, Charitéplatz 1, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, Charitéplatz 1, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.
| |
Collapse
|
63
|
Constitutive signal bias mediated by the human GHRHR splice variant 1. Proc Natl Acad Sci U S A 2021; 118:2106606118. [PMID: 34599099 PMCID: PMC8501799 DOI: 10.1073/pnas.2106606118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2021] [Indexed: 11/18/2022] Open
Abstract
The mechanism of functional changes induced by alternative splicing of GHRHR is largely unknown. Here, we demonstrate that GHRH-elicited signal bias toward β-arrestin recruitment is constitutively mediated by SV1. The cryogenic electron microscopy structures of SV1 and molecular dynamics simulations reveal the different functionalities between GHRHR and SV1 at the near-atomic level (i.e., the N termini of GHRHR and SV1 differentiate the downstream signaling pathways, Gs versus β-arrestins). Our findings provide valuable insights into the functional diversity of class B1 GPCRs that may aid in the design of better therapeutic agents against certain cancers. Alternative splicing of G protein–coupled receptors has been observed, but their functions are largely unknown. Here, we report that a splice variant (SV1) of the human growth hormone–releasing hormone receptor (GHRHR) is capable of transducing biased signal. Differing only at the receptor N terminus, GHRHR predominantly activates Gs while SV1 selectively couples to β-arrestins. Based on the cryogenic electron microscopy structures of SV1 in the apo state or GHRH-bound state in complex with the Gs protein, molecular dynamics simulations reveal that the N termini of GHRHR and SV1 differentiate the downstream signaling pathways, Gs versus β-arrestins. As suggested by mutagenesis and functional studies, it appears that GHRH-elicited signal bias toward β-arrestin recruitment is constitutively mediated by SV1. The level of SV1 expression in prostate cancer cells is also positively correlated with ERK1/2 phosphorylation but negatively correlated with cAMP response. Our findings imply that constitutive signal bias may be a mechanism that ensures cancer cell proliferation.
Collapse
|
64
|
Simpson RJ, Boßlau TK, Weyh C, Niemiro GM, Batatinha H, Smith KA, Krüger K. Exercise and adrenergic regulation of immunity. Brain Behav Immun 2021; 97:303-318. [PMID: 34302965 DOI: 10.1016/j.bbi.2021.07.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/07/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022] Open
Abstract
Exercise training has a profound impact on immunity, exerting a multitude of positive effects in indications such as immunosenescence, cancer, viral infections and inflammatory diseases. The immune, endocrine and central nervous systems work in a highly synergistic manner and it has become apparent that catecholamine signaling through leukocyte β-adrenergic receptors (β-ARs) is a key mechanism by which exercise mediates improvements in immune function to help mitigate numerous disease conditions. Central to this is the preferential mobilization and redistribution of effector lymphocytes with potent anti-viral and anti-tumor activity, their interaction with muscle-derived cytokines, and the effects of catecholamine signaling on mitochondrial biogenesis, immunometabolism and the resulting inflammatory response. Here, we review the impact of acute and chronic exercise on adrenergic regulation of immunity in the context of aging, cancer, viral infections and inflammatory disease. We also put forth our contention that exercise interventions designed to improve immunity, prevent disease and reduce inflammation should consider the catecholamine-AR signaling axis as a therapeutic target and ask whether or not the adrenergic signaling machinery can be 'trained' to improve immune responses to stress, disease or during the normal physiological process of aging. Finally, we discuss potential strategies to augment leukocyte catecholamine signaling to boost the effects of exercise on immunity in individuals with desensitized β-ARs or limited exercise tolerance.
Collapse
Affiliation(s)
- Richard J Simpson
- University of Arizona, Department of Nutritional Sciences, Tucson, AZ, USA; University of Arizona, Department of Pediatrics, Tucson, AZ, USA; University of Arizona, Department of Immunobiology, Tucson, AZ, USA; University of Arizona Cancer Center, Tucson, AZ, USA.
| | - Tim K Boßlau
- University of Gießen, Department of Exercise Physiology and Sports Therapy, Gießen, Germany
| | - Christopher Weyh
- University of Gießen, Department of Exercise Physiology and Sports Therapy, Gießen, Germany
| | - Grace M Niemiro
- University of Arizona, Department of Pediatrics, Tucson, AZ, USA; University of Arizona Cancer Center, Tucson, AZ, USA
| | - Helena Batatinha
- University of Arizona, Department of Pediatrics, Tucson, AZ, USA
| | - Kyle A Smith
- University of Arizona, Department of Nutritional Sciences, Tucson, AZ, USA; University of Arizona, Department of Pediatrics, Tucson, AZ, USA
| | - Karsten Krüger
- University of Gießen, Department of Exercise Physiology and Sports Therapy, Gießen, Germany.
| |
Collapse
|
65
|
Rich TC, Leavesley SJ, Brandon AP, Evans CA, Raju SV, Wagener BM. Phosphodiesterase 4 mediates interleukin-8-induced heterologous desensitization of the β 2 -adrenergic receptor. FASEB J 2021; 35:e21946. [PMID: 34555226 DOI: 10.1096/fj.202002712rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/11/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening illness characterized by decreased alveolar-capillary barrier function, pulmonary edema consisting of proteinaceous fluid, and inhibition of net alveolar fluid transport responsible for resolution of pulmonary edema. There is currently no pharmacotherapy that has proven useful to prevent or treat ARDS, and two trials using beta-agonist therapy to treat ARDS demonstrated no effect. Prior studies indicated that IL-8-induced heterologous desensitization of the beta2-adrenergic receptor (β2 -AR) led to decreased beta-agonist-induced mobilization of cyclic adenosine monophosphate (cAMP). Interestingly, phosphodiesterase (PDE) 4 inhibitors have been used in human airway diseases characterized by low intracellular cAMP levels and increases in specific cAMP hydrolyzing activity. Therefore, we hypothesized that PDE4 would mediate IL-8-induced heterologous internalization of the β2 -AR and that PDE4 inhibition would restore beta-agonist-induced functions. We determined that CINC-1 (a functional IL-8 analog in rats) induces internalization of β2 -AR from the cell surface, and arrestin-2, PDE4, and β2 -AR form a complex during this process. Furthermore, we determined that cAMP associated with the plasma membrane was adversely affected by β2 -AR heterologous desensitization. Additionally, we determined that rolipram, a PDE4 inhibitor, reversed CINC-1-induced derangements of cAMP and also caused β2 -AR to successfully recycle back to the cell surface. Finally, we demonstrated that rolipram could reverse CINC-1-mediated inhibition of beta-agonist-induced alveolar fluid clearance in a murine model of trauma-shock. These results indicate that PDE4 plays a role in CINC-1-induced heterologous internalization of the β2 -AR; PDE4 inhibition reverses these effects and may be a useful adjunct in particular ARDS patients.
Collapse
Affiliation(s)
- Thomas C Rich
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, USA.,Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Silas J Leavesley
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, USA.,Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.,Department of Chemical and Biomolecular Engineering, University of South Alabama, Mobile, Alabama, USA
| | - Angela P Brandon
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cilina A Evans
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - S Vamsee Raju
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,UAB Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Brant M Wagener
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
66
|
Zhao J, Stephens T, Zhao Y. Molecular Regulation of Lysophosphatidic Acid Receptor 1 Maturation and Desensitization. Cell Biochem Biophys 2021; 79:477-483. [PMID: 34032994 PMCID: PMC8887818 DOI: 10.1007/s12013-021-00999-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2021] [Indexed: 10/21/2022]
Abstract
Lysophosphatidic acid receptor 1 (LPA1) belongs to the G protein-coupled receptor family. The ligand for LPA1 is LPA, the simplest lysophospholipid. LPA is considered a growth factor and induces cell proliferation, anti-apoptosis, and cell migration. The pro-inflammatory and pro-fibrotic roles of LPA have also been well-demonstrated. Most of the biological functions of LPA are mostly executed through LPA1. The mature form of LPA1 is glycosylated and localized on the plasma membrane. LPA1 is bound to heterotrimetric G proteins and transduces intracellular signaling in response to ligation to LPA. Desensitization of LPA1 negatively regulates LPA1-mediated signaling and the resulting biological functions. Phosphorylation and ubiquitination are well-demonstrated posttranslational modifications of GPCR. In this review, we will discuss our knowledge of LPA1 glycosylation, maturation, and trafficking from the endoplasmic reticulum (ER)/Golgi to the plasma membrane. Moreover, in light of recent findings, we will also discuss molecular regulation of LPA1 internalization and stability.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Physiology and Cell Biology, the Ohio State University, Columbus, OH, USA
| | - Thomas Stephens
- Department of Physiology and Cell Biology, the Ohio State University, Columbus, OH, USA
| | - Yutong Zhao
- Department of Physiology and Cell Biology, the Ohio State University, Columbus, OH, USA.
| |
Collapse
|
67
|
Fréchette L, Degrandmaison J, Binda C, Boisvert M, Côté L, Michaud T, Lalumière MP, Gendron L, Parent JL. Identification of the interactome of the DP1 receptor for Prostaglandin D 2: Regulation of DP1 receptor signaling and trafficking by IQGAP1. Biochim Biophys Acta Gen Subj 2021; 1865:129969. [PMID: 34352343 DOI: 10.1016/j.bbagen.2021.129969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/19/2021] [Accepted: 07/25/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND Mechanisms governing localization, trafficking and signaling of G protein-coupled receptors (GPCRs) are critical in cell function. Protein-protein interactions are determinant in these processes. However, there are very little interacting proteins known to date for the DP1 receptor for prostaglandin D2. METHODS We performed LC-MS/MS analyses of the DP1 receptor interactome in HEK293 cells. To functionally validate our LC-MS/MS data, we studied the implications of the interaction with the IQGAP1 scaffold protein in the trafficking and signaling of DP1. RESULTS In addition to expected interacting proteins such as heterotrimeric G protein subunits, we identified proteins involved in signaling, trafficking, and folding localized in various cell compartments. Endogenous DP1-IQGAP1 co-immunoprecipitation was observed in colon cancer HT-29 cells. The interaction was augmented by DP1 agonist activation in HEK293 cells and GST-pulldown assays showed that IQGAP1 binds to intracellular loops 2 and 3 of DP1. Co-localization of the two proteins was observed by confocal microscopy at the cell periphery and in intracellular vesicles in the basal state. PGD2 treatment resulted in the redistribution of the DP1-IQGAP1 co-localization in the perinuclear vicinity. DP1 receptor internalization was promoted by overexpression of IQGAP1, while it was diminished by IQGAP1 knockdown with DsiRNAs. DP1-mediated ERK1/2 activation was augmented and sustained overtime by overexpression of IQGAP1 when compared to DP1 expressed alone. IQGAP1 knockdown decreased ERK1/2 activation by DP1 stimulation. Interestingly, ERK1/2 signaling by DP1 was increased when IQGAP2 was silenced, while it was impaired by IQGAP3 knockdown. CONCLUSIONS Our findings define the putative DP1 interactome, a patho-physiologically important receptor, and validated the interaction with IQGAP1 in DP1 function. Our data also reveal that IQGAP proteins may differentially regulate GPCR signaling. GENERAL SIGNIFICANCE The identified putative DP1-interacting proteins open multiple lines of research in DP1 and GPCR biology in various cell compartments.
Collapse
Affiliation(s)
- Louis Fréchette
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jade Degrandmaison
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Chantal Binda
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Marilou Boisvert
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Laurie Côté
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Thomas Michaud
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Marie-Pier Lalumière
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Louis Gendron
- Département de Pharmacologie-Physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Département d'Anesthésiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jean-Luc Parent
- Département de Médecine, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Institut de Pharmacologie de Sherbrooke, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Canada; Centre de recherche du Centre Hospitalier de l'Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
68
|
Kalinkovich A, Livshits G. Biased and allosteric modulation of bone cell-expressing G protein-coupled receptors as a novel approach to osteoporosis therapy. Pharmacol Res 2021; 171:105794. [PMID: 34329703 DOI: 10.1016/j.phrs.2021.105794] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 12/16/2022]
Abstract
On the cellular level, osteoporosis (OP) is a result of imbalanced bone remodeling, in which osteoclastic bone resorption outcompetes osteoblastic bone formation. Currently available OP medications include both antiresorptive and bone-forming drugs. However, their long-term use in OP patients, mainly in postmenopausal women, is accompanied by severe side effects. Notably, the fundamental coupling between bone resorption and formation processes underlies the existence of an undesirable secondary outcome that bone anabolic or anti-resorptive drugs also reduce bone formation. This drawback requires the development of anti-OP drugs capable of selectively stimulating osteoblastogenesis and concomitantly reducing osteoclastogenesis. We propose that the application of small synthetic biased and allosteric modulators of bone cell receptors, which belong to the G-protein coupled receptors (GPCR) family, could be the key to resolving the undesired anti-OP drug selectivity. This approach is based on the capacity of these GPCR modulators, unlike the natural ligands, to trigger signaling pathways that promote beneficial effects on bone remodeling while blocking potentially deleterious effects. Under the settings of OP, an optimal anti-OP drug should provide fine-tuned regulation of downstream effects, for example, intermittent cyclic AMP (cAMP) elevation, preservation of Ca2+ balance, stimulation of osteoprotegerin (OPG) and estrogen production, suppression of sclerostin secretion, and/or preserved/enhanced canonical β-catenin/Wnt signaling pathway. As such, selective modulation of GPCRs involved in bone remodeling presents a promising approach in OP treatment. This review focuses on the evidence for the validity of our hypothesis.
Collapse
Affiliation(s)
- Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| | - Gregory Livshits
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel; Adelson School of Medicine, Ariel University, Ariel 4077625, Israel.
| |
Collapse
|
69
|
Jean-Alphonse FG, Sposini S. Confocal and TIRF microscopy based approaches to visualize arrestin trafficking in living cells. Methods Cell Biol 2021; 166:179-203. [PMID: 34752332 DOI: 10.1016/bs.mcb.2021.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Arrestins are key proteins that serve as versatile scaffolds to control and mediate G protein coupled receptors (GPCR) activity. Arrestin control of GPCR functions involves their recruitment from the cytosol to plasma membrane-localized GPCRs and to endosomal compartments, where they mediate internalization, sorting and signaling of GPCRs. Several methods can be used to monitor trafficking of arrestins; however, live fluorescence imaging remains the method of choice to both assess arrestin recruitment to ligand-activated receptors and to monitor its dynamic subcellular localization. Here, we present two approaches based on Total Internal Fluorescence (TIRF) microscopy and confocal microscopy to visualize arrestin trafficking in live cells in real time and to assess their co-localization with the GPCR of interest and their localization at specific subcellular locations.
Collapse
Affiliation(s)
- Frédéric Gaëtan Jean-Alphonse
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France; Université Paris-Saclay, Inria, Inria Saclay-Île-de-France, Palaiseau, France
| | - Silvia Sposini
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, United Kingdom; University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, Bordeaux, France.
| |
Collapse
|
70
|
Olaniru OE, Cheng J, Ast J, Arvaniti A, Atanes P, Huang GC, King AJF, Jones PM, Broichhagen J, Hodson DJ, Persaud SJ. SNAP-tag-enabled super-resolution imaging reveals constitutive and agonist-dependent trafficking of GPR56 in pancreatic β-cells. Mol Metab 2021; 53:101285. [PMID: 34224919 PMCID: PMC8326393 DOI: 10.1016/j.molmet.2021.101285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
Objective Members of the adhesion G protein-coupled receptor (aGPCR) subfamily are important actors in metabolic processes, with GPR56 (ADGRG1) emerging as a possible target for type 2 diabetes therapy. GPR56 can be activated by collagen III, its endogenous ligand, and by a synthetic seven amino-acid peptide (TYFAVLM; P7) contained within the GPR56 Stachel sequence. However, the mechanisms regulating GPR56 trafficking dynamics and agonist activities are not yet clear. Methods Here, we introduced SNAPf-tag into the N-terminal segment of GPR56 to monitor GPR56 cellular activity in situ. Confocal and super-resolution microscopy were used to investigate the trafficking pattern of GPR56 in native MIN6 β-cells and in MIN6 β-cells where GPR56 had been deleted by CRISPR-Cas9 gene editing. Insulin secretion, changes in intracellular calcium, and β-cell apoptosis were determined by radioimmunoassay, single-cell calcium microfluorimetry, and measuring caspase 3/7 activities, respectively, in MIN6 β-cells and human islets. Results SNAP-tag labelling indicated that GPR56 predominantly underwent constitutive internalisation in the absence of an exogenous agonist, unlike GLP-1R. Collagen III further stimulated GPR56 internalisation, whereas P7 was without significant effect. The overexpression of GPR56 in MIN6 β-cells did not affect insulin secretion. However, it was associated with reduced β-cell apoptosis, while the deletion of GPR56 made MIN6 β-cells more susceptible to cytokine-induced apoptosis. P7 induced a rapid increase in the intracellular calcium in MIN6 β-cells (in a GPR56-dependent manner) and human islets, and it also caused a sustained and reversible increase in insulin secretion from human islets. Collagen III protected human islets from cytokine-induced apoptosis, while P7 was without significant effect. Conclusions These data indicate that GPR56 exhibits both agonist-dependent and -independent trafficking in β-cells and suggest that while GPR56 undergoes constitutive signalling, it can also respond to its ligands when required. We have also identified that constitutive and agonist-dependent GPR56 activation is coupled to protect β-cells against apoptosis, offering a potential therapeutic target to maintain β-cell mass in type 2 diabetes. GPR56 predominantly underwent constitutive internalisation in β-cells in the absence of exogenous agonist. The GPR56 agonists, collagen III and P7, showed differential effects on GPR56 trafficking and islet functions. Constitutive and agonist-dependent GPR56 activation is coupled to protection of β-cells against apoptosis.
Collapse
Affiliation(s)
- Oladapo E Olaniru
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK.
| | - Jordan Cheng
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering & Imaging Sciences, 4th floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK
| | - Julia Ast
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Anastasia Arvaniti
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Patricio Atanes
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Guo C Huang
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Aileen J F King
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Peter M Jones
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
71
|
Parathyroid hormone and its related peptides in bone metabolism. Biochem Pharmacol 2021; 192:114669. [PMID: 34224692 DOI: 10.1016/j.bcp.2021.114669] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022]
Abstract
Parathyroid hormone (PTH) is an 84-amino-acid peptide hormone that is secreted by the parathyroid gland. It has different administration modes in bone tissue through which it promotes bone formation (intermittent administration) and bone resorption (continuous administration) and has great potential for application in sbone defect repair. PTH regulates bone metabolism by binding to PTH1R. PTH plays an osteogenic role by acting directly on mesenchymal stem cells, cells with an osteoblastic lineage, osteocytes, and T cells. It also participates as an osteoclast by indirectly acting on osteoclast precursor cells and osteoclasts and directly acting on T cells. In these cells, PTH activates the Wnt signaling, cAMP/PKA, cAMP/PKC, and RANKL/RANK/OPG pathways and other signaling pathways. Although PTH(1-34), also known as teriparatide, has been used clinically, it still has some disadvantages. Developing improved PTH-related peptides is a potential solution to teriparatide's shortcomings. The action mechanism of these PTH-related peptides is not exactly the same as that of PTH. Thus, the mechanisms of PTH and PTH-related peptides in bone metabolism were reviewed in this paper.
Collapse
|
72
|
Tenner B, Zhang JZ, Kwon Y, Pessino V, Feng S, Huang B, Mehta S, Zhang J. FluoSTEPs: Fluorescent biosensors for monitoring compartmentalized signaling within endogenous microdomains. SCIENCE ADVANCES 2021; 7:7/21/eabe4091. [PMID: 34020947 PMCID: PMC8139597 DOI: 10.1126/sciadv.abe4091] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/01/2021] [Indexed: 05/10/2023]
Abstract
Growing evidence suggests that many essential intracellular signaling events are compartmentalized within kinetically distinct microdomains in cells. Genetically encoded fluorescent biosensors are powerful tools to dissect compartmentalized signaling, but current approaches to probe these microdomains typically rely on biosensor fusion and overexpression of critical regulatory elements. Here, we present a novel class of biosensors named FluoSTEPs (fluorescent sensors targeted to endogenous proteins) that combine self-complementing split green fluorescent protein, CRISPR-mediated knock-in, and fluorescence resonance energy transfer biosensor technology to probe compartmentalized signaling dynamics in situ. We designed FluoSTEPs for simultaneously highlighting endogenous microdomains and reporting domain-specific, real-time signaling events including kinase activities, guanosine triphosphatase activation, and second messenger dynamics in live cells. A FluoSTEP for 3',5'-cyclic adenosine monophosphate (cAMP) revealed distinct cAMP dynamics within clathrin microdomains in response to stimulation of G protein-coupled receptors, showcasing the utility of FluoSTEPs in probing spatiotemporal regulation within endogenous signaling architectures.
Collapse
Affiliation(s)
- Brian Tenner
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jason Z Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yonghoon Kwon
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Veronica Pessino
- Graduate Program of Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Siyu Feng
- The UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Bo Huang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Sohum Mehta
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Jin Zhang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
73
|
Peng GE, Pessino V, Huang B, von Zastrow M. Spatial decoding of endosomal cAMP signals by a metastable cytoplasmic PKA network. Nat Chem Biol 2021; 17:558-566. [PMID: 33649598 PMCID: PMC8084946 DOI: 10.1038/s41589-021-00747-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/22/2021] [Indexed: 01/31/2023]
Abstract
G-protein-coupled receptor-regulated cAMP production from endosomes can specify signaling to the nucleus by moving the source of cAMP without changing its overall amount. How this is possible remains unknown because cAMP gradients dissipate over the nanoscale, whereas endosomes typically localize micrometers from the nucleus. We show that the key location-dependent step for endosome-encoded transcriptional control is nuclear entry of cAMP-dependent protein kinase (PKA) catalytic subunits. These are sourced from punctate accumulations of PKA holoenzyme that are densely distributed in the cytoplasm and titrated by global cAMP into a discrete metastable state, in which catalytic subunits are bound but dynamically exchange. Mobile endosomes containing activated receptors collide with the metastable PKA puncta and pause in close contact. We propose that these properties enable cytoplasmic PKA to act collectively like a semiconductor, converting nanoscale cAMP gradients generated from endosomes into microscale elevations of free catalytic subunits to direct downstream signaling.
Collapse
Affiliation(s)
- Grace E Peng
- Program in Cell Biology, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Veronica Pessino
- Graduate Program of Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Swab56 Corp. and Neurophotometrics Ltd, San Diego, CA, USA
| | - Bo Huang
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Mark von Zastrow
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Quantitative Biology Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
74
|
Burgos-Aguilar C, Ferris MJ, Sexton LL, Sun H, Xiao R, Chen R, Childers SR, Howlett AC. Metabotropic glutamate 2,3 receptor stimulation desensitizes agonist activation of G-protein signaling and alters transcription regulators in mesocorticolimbic brain regions. Synapse 2021; 75:e22190. [PMID: 33025628 PMCID: PMC8552243 DOI: 10.1002/syn.22190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/26/2020] [Accepted: 09/06/2020] [Indexed: 01/07/2023]
Abstract
Metabotropic glutamate (mGlu) receptors are regulators of glutamate release and targets for development of therapies for hyperactive glutamatergic signaling. However, the effects of long-term stimulation of mGlu receptors on cellular signaling in the brain have not been described. This study investigated the effects of 2-day and 14-day osmotic mini-pump administration of the mGlu2,3 agonist LY379268 (3.0 mg kg-1 day-1 ) to rats on receptor-mediated G-protein activation and signaling in mesocorticolimbic regions in rat brain sections. A significant reduction in LY379268-stimulated [35 S]GTPγS binding was observed in the 14-day group in some cortical regions, prefrontal cortex, nucleus accumbens, and ventral pallidum. The 14-day LY379268 treatment group exhibited mGlu2 mRNA levels significantly lower in hippocampus, nucleus accumbens, caudate, and ventral pallidum. In both 2-day and 14-day treatment groups immunodetectable phosphorylated cAMP Response Element-Binding protein (CREB) was significantly reduced across all brain regions. In the 2-day group, we observed significantly lower immunodetectable CREB protein across all brain regions, which was subsequently increased in the 14-day group but failed to achieve control values. Neither immunodetectable extracellular signal-regulated kinase (ERK) protein nor phosphorylated ERK from 2-day or 14-day treatment groups differed significantly from control across all brain regions. However, the ratio of phosphorylated ERK to total ERK protein was significantly greater in the 14-day treatment group compared with the control. These results identify compensatory changes to mGlu2,3 signal transduction in rat brains after chronic systemic administration of agonist, which could be predictive of the mechanism of action in human pharmacotherapies.
Collapse
Affiliation(s)
- Carolina Burgos-Aguilar
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Mark J. Ferris
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Lacey L. Sexton
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Haiguo Sun
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Ruoyu Xiao
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Rong Chen
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Steven R. Childers
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| | - Allyn C. Howlett
- Department of Physiology and Pharmacology and Center for the Neurobiology of Addiction Treatment, One Medical Center Blvd., Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| |
Collapse
|
75
|
Moo EV, van Senten JR, Bräuner-Osborne H, Møller TC. Arrestin-Dependent and -Independent Internalization of G Protein-Coupled Receptors: Methods, Mechanisms, and Implications on Cell Signaling. Mol Pharmacol 2021; 99:242-255. [PMID: 33472843 DOI: 10.1124/molpharm.120.000192] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/07/2021] [Indexed: 01/05/2023] Open
Abstract
Agonist-induced endocytosis is a key regulatory mechanism for controlling the responsiveness of the cell by changing the density of cell surface receptors. In addition to the role of endocytosis in signal termination, endocytosed G protein-coupled receptors (GPCRs) have been found to signal from intracellular compartments of the cell. Arrestins are generally believed to be the master regulators of GPCR endocytosis by binding to both phosphorylated receptors and adaptor protein 2 (AP-2) or clathrin, thus recruiting receptors to clathrin-coated pits to facilitate the internalization process. However, many other functions have been described for arrestins that do not relate to their role in terminating signaling. Additionally, there are now more than 30 examples of GPCRs that internalize independently of arrestins. Here we review the methods, pharmacological tools, and cellular backgrounds used to determine the role of arrestins in receptor internalization, highlighting their advantages and caveats. We also summarize key examples of arrestin-independent GPCR endocytosis in the literature and their suggested alternative endocytosis pathway (e.g., the caveolae-dependent and fast endophilin-mediated endocytosis pathways). Finally, we consider the possible function of arrestins recruited to GPCRs that are endocytosed independently of arrestins, including the catalytic arrestin activation paradigm. Technological improvements in recent years have advanced the field further, and, combined with the important implications of endocytosis on drug responses, this makes endocytosis an obvious parameter to include in molecular pharmacological characterization of ligand-GPCR interactions. SIGNIFICANCE STATEMENT: G protein-coupled receptor (GPCR) endocytosis is an important means to terminate receptor signaling, and arrestins play a central role in the widely accepted classical paradigm of GPCR endocytosis. In contrast to the canonical arrestin-mediated internalization, an increasing number of GPCRs are found to be endocytosed via alternate pathways, and the process appears more diverse than the previously defined "one pathway fits all."
Collapse
Affiliation(s)
- Ee Von Moo
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jeffrey R van Senten
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Thor C Møller
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
76
|
Llavero F, Arrazola Sastre A, Luque Montoro M, Martín MA, Arenas J, Lucia A, Zugaza JL. Small GTPases of the Ras superfamily and glycogen phosphorylase regulation in T cells. Small GTPases 2021; 12:106-113. [PMID: 31512989 PMCID: PMC7849735 DOI: 10.1080/21541248.2019.1665968] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/02/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Small GTPases, together with their regulatory and effector molecules, are key intermediaries in the complex signalling pathways that control almost all cellular processes, working as molecular switches to transduce extracellular cues into cellular responses that drive vital functions, such as intracellular transport, biomolecule synthesis, gene activation and cell survival. How all of these networks are linked to metabolic pathways is a subject of intensive study. Because any response to cellular action requires some form of energy input, elucidating how cells coordinate the signals that lead to a tangible response involving metabolism is central to understand cellular activities. In this review, we summarize recent advances in our understanding of the molecular basis of the crosstalk between small GTPases of the Ras superfamily, specifically Rac1 and Ras/Rap1, and glycogen phosphorylase in T lymphocytes. Abbreviations: ADCY: adenylyl cyclase; ADCY6: adenylyl cyclase 6; BCR: B cell receptor; cAMP: 3',5'-cyclic adenosine monophosphate; CRIB: Cdc42/Rac binding domain; DLPFC: dysfunction of the dorsolateral prefrontal cortex; EGFR: epidermal growth factor receptor; Epac2: exchange protein directly activated by cAMP; GDP: guanodine-5'-diphosphate; GPCRs: G protein-coupled receptors; GTP: guanodin-5'-triphosphate; IL2: interleukin 2; IL2-R: interleukin 2 receptor; JAK: janus kinases; MAPK: mitogen-activated protein kinase; O-GlcNAc: O-glycosylation; PAK1: p21 activated kinase 1; PI3K: phosphatidylinositol 3-kinase; PK: phosphorylase kinase; PKA: cAMP-dependent protein kinase A; PKCθ: protein kinase Cθ; PLCγ: phospholipase Cγ; Src: proto-oncogene tyrosine-protein kinase c; STAT: signal transducer and activator of transcription proteins.
Collapse
Affiliation(s)
- Francisco Llavero
- Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Spain
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Alazne Arrazola Sastre
- Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Spain
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, UPV/EHU, Leioa, Spain
| | - Miriam Luque Montoro
- Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Spain
| | - Miguel A. Martín
- Enfermedades Raras, Mitocondriales y Neuromusculares., Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquín Arenas
- Enfermedades Raras, Mitocondriales y Neuromusculares., Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid, Madrid, Spain
- Enfermedades Raras, Mitocondriales y Neuromusculares., Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Center for Biomedical Network Research on Frailty and Healthy Aging (CIBER FES), Instituto de Salud Carlos III, Madrid, Spain
| | - José L. Zugaza
- Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Spain
- Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, UPV/EHU, Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
77
|
Kurtz R, Anderman MF, Shepard BD. GPCRs get fatty: the role of G protein-coupled receptor signaling in the development and progression of nonalcoholic fatty liver disease. Am J Physiol Gastrointest Liver Physiol 2021; 320:G304-G318. [PMID: 33205999 PMCID: PMC8202238 DOI: 10.1152/ajpgi.00275.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), characterized by the abnormal deposition of lipids within the liver not due to alcohol consumption, is a growing epidemic affecting over 30% of the United States population. Both simple fatty liver and its more severe counterpart, nonalcoholic steatohepatitis, represent one of the most common forms of liver disease. Recently, several G protein-coupled receptors have emerged as targets for therapeutic intervention for these disorders. These include those with known hepatic function as well as those involved in global metabolic regulation. In this review, we highlight these emerging therapeutic targets, focusing on several common themes including their activation by microbial metabolites, stimulatory effect on insulin and incretin secretion, and contribution to glucose tolerance. The overlap in ligands, localization, and downstream effects of activation indicate the interdependent nature of these receptors and highlight the importance of this signaling family in the development and prevention of NAFLD.
Collapse
Affiliation(s)
- Ryan Kurtz
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Meghan F. Anderman
- Department of Human Science, Georgetown University, Washington, District of Columbia
| | - Blythe D. Shepard
- Department of Human Science, Georgetown University, Washington, District of Columbia
| |
Collapse
|
78
|
Melanocortin receptor activation alleviates amyloid pathology and glial reactivity in an Alzheimer's disease transgenic mouse model. Sci Rep 2021; 11:4359. [PMID: 33623128 PMCID: PMC7902646 DOI: 10.1038/s41598-021-83932-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder with no disease-modifying treatment. AD progression is characterized by cognitive decline, neuroinflammation, and accumulation of amyloid-beta (Aβ) and neurofibrillary tangles in the brain, leading to neuronal and glial dysfunctions. Neuropeptides govern diverse pathophysiological processes and represent key players in AD pathogenesis, regulating synaptic plasticity, glial cell functions and amyloid pathology. Activation of the pro-opiomelanocortin (POMC)-derived neuropeptide and its receptor from the melanocortin receptor (MCR) family have previously been shown to rescue the impairment in hippocampus-dependent synaptic plasticity in the APP/PS1 mouse model of AD. However, the functional roles of MCR signaling in AD conditions, particularly in glial functions, are largely unknown. In this study, we investigated the potential benefits of MCR activation in AD. In APP/PS1 transgenic mice, we demonstrate that MCR activation mediated by the central administration of its agonist D-Tyr MTII substantially reduces Aβ accumulation, while alleviating global inflammation and astrocytic activation, particularly in the hippocampus. MCR activation prominently reduces the A1 subtype of reactive astrocytes, which is considered a key source of astrocytic neurotoxicity in AD. Concordantly, MCR activation suppresses microglial activation, while enhancing their association with amyloid plaques. The blunted activation of microglia may contribute to the reduction in the neurotoxic phenotypes of astrocytes. Importantly, transcriptome analysis reveals that MCR activation restores the impaired homeostatic processes and microglial reactivity in the hippocampus in APP/PS1 mice. Collectively, our findings demonstrate the potential of MCR signaling as therapeutic target for AD.
Collapse
|
79
|
Zhou S, Wu Q, Lin X, Ling X, Miao J, Liu X, Hu C, Zhang Y, Jia N, Hou FF, Liu Y, Zhou L. Cannabinoid receptor type 2 promotes kidney fibrosis through orchestrating β-catenin signaling. Kidney Int 2021; 99:364-381. [PMID: 33152447 DOI: 10.1016/j.kint.2020.09.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022]
Abstract
The endocannabinoid system has multiple effects. Through interacting with cannabinoid receptor type 1 and type 2, this system can greatly affect disease progression. Previously, we showed that activated cannabinoid receptor type 2 (CB2) mediated kidney fibrosis. However, the underlying mechanisms remain underdetermined. Here, we report that CB2 was upregulated predominantly in kidney tubular epithelial cells in unilateral urinary obstruction and ischemia-reperfusion injury models in mice, and in patients with a variety of kidney diseases. CB2 expression was closely correlated with the progression of kidney fibrosis and accompanied by the activation of β-catenin. Furthermore, CB2 induced the formation of a β-arrestin 1/Src/β-catenin complex, which further triggered the nuclear translocation of β-catenin and caused fibrotic injury. Incubation with XL-001, an inverse agonist to CB2, or knockdown of β-arrestin 1 inhibited CB2-triggered activation of β-catenin and fibrotic injury. Notably, CB2 potentiated Wnt1-induced β-arrestin 1/β-catenin activation and augmented the pathogenesis of kidney fibrosis in mice with unilateral ischemia-reperfusion injury or folic acid-induced nephropathy. Knockdown of β-arrestin 1 inhibited the CB2 agonist AM1241-induced β-catenin activation and kidney fibrosis. By promoter sequence analysis, putative transcription factor binding sites for T-cell factor/lymphoid enhancer factor were found in the promoter regions of the CB2 gene regardless of the species. Overexpression of β-catenin induced the binding of T-cell factor/lymphoid enhancer factor-1 to these sites, promoted the expression of CB2, β-arrestin 1, and the proto-oncogene Src, and triggered their accumulation. Thus, the CB2/β-catenin pathway appears to create a reciprocal activation feedback loop that plays a central role in the pathogenesis of kidney fibrosis.
Collapse
Affiliation(s)
- Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Lin
- Department of Nephrology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chengxiao Hu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yunfang Zhang
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Nan Jia
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Bioland Laboratory (Guangzhou Regenerative Medicine and Health, Guangdong Laboratory), Guangzhou, China.
| |
Collapse
|
80
|
McDonough RC, Gilbert RM, Gleghorn JP, Price C. Targeted Gq-GPCR activation drives ER-dependent calcium oscillations in chondrocytes. Cell Calcium 2021; 94:102363. [PMID: 33550208 DOI: 10.1016/j.ceca.2021.102363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 11/28/2022]
Abstract
The temporal dynamics of calcium signaling are critical regulators of chondrocyte homeostasis and chondrogenesis. Calcium oscillations regulate differentiation and anabolic processes in chondrocytes and their precursors. Attempts to control chondrocyte calcium signaling have been achieved through mechanical perturbations and synthetic ion channel modulators. However, such stimuli can lack both local and global specificity and precision when evoking calcium signals. Synthetic signaling platforms can more precisely and selectively activate calcium signaling, enabling improved dissection of the roles of intracellular calcium ([Ca2+]i) in chondrocyte behavior. One such platform is hM3Dq, a chemogenetic DREADD (Designer Receptors Exclusively Activated by Designer Drugs) that activates calcium signaling via the Gαq-PLCβ-IP3-ER pathway upon administration of clozapine N-oxide (CNO). We previously described the first-use of hM3Dq to precisely mediate targeted, synthetic calcium signals in chondrocyte-like ATDC5 cells. Here, we generated stably expressing hM3Dq-ATDC5 cells to investigate the dynamics of Gαq-GPCR calcium signaling in depth. CNO drove robust calcium responses in a temperature- and concentration-dependent (1 pM-100 μM) manner and elicited elevated levels of oscillatory calcium signaling above 10 nM. hM3Dq-mediated calcium oscillations in ATDC5 cells were reliant on ER calcium stores for both initiation and sustenance, and the downregulation and recovery dynamics of hM3Dq after CNO stimulation align with traditionally reported GPCR recycling kinetics. This study successfully generated a stable hM3Dq cell line to precisely drive Gαq-GPCR-mediated and ER-dependent oscillatory calcium signaling in ATDC5 cells and established a novel tool to elucidate the role that GPCR-mediated calcium signaling plays in chondrocyte biology, cartilage pathology, and cartilage tissue engineering.
Collapse
Affiliation(s)
- Ryan C McDonough
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Rachel M Gilbert
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, United States.
| | - Christopher Price
- Department of Biomedical Engineering, University of Delaware, United States.
| |
Collapse
|
81
|
Kumar GA, Chattopadhyay A. Membrane cholesterol regulates endocytosis and trafficking of the serotonin 1A receptor: Insights from acute cholesterol depletion. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158882. [PMID: 33429076 DOI: 10.1016/j.bbalip.2021.158882] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/24/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis and intracellular trafficking constitute important regulatory features associated with G protein-coupled receptor (GPCR) function. GPCR endocytosis involves several remodeling events at the plasma membrane orchestrated by a concerted interplay of a large number of proteins and membrane lipids. Although considerable literature exists on the protein framework underlying GPCR endocytosis, the role of membrane lipids in this process remains largely unexplored. In order to explore the role of membrane cholesterol (an essential and important lipid in higher eukaryotes) in GPCR endocytosis, we monitored the effect of acute cholesterol depletion using methyl-β-cyclodextrin (MβCD) on endocytosis and intracellular trafficking of the serotonin1A receptor, an important neurotransmitter GPCR. Our results show that the serotonin1A receptor exhibits agonist-induced clathrin-mediated endocytosis with a concentration-dependent inhibition in internalization with increasing concentrations of MβCD, which was restored upon cholesterol replenishment. Interestingly, subsequent to internalization under these conditions, serotonin1A receptors were re-routed toward lysosomal degradation, instead of endosomal recycling observed under normal conditions, thereby implicating membrane cholesterol in modulation of intracellular trafficking of the receptor. This raises the possibility of a novel cholesterol-dependent role of intracellular sorting proteins in GPCR trafficking. These results differ from our previous observations on the endocytosis of the serotonin1A receptor upon statin-induced chronic cholesterol depletion, in terms of endocytic pathway. We conclude that analysis of complex cellular trafficking events such as GPCR endocytosis under acute and chronic cholesterol depletion conditions should be carried out with caution due to fundamental differences underlying these processes.
Collapse
Affiliation(s)
- G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | | |
Collapse
|
82
|
Kim H, Lee HN, Choi J, Seong J. Spatiotemporal Characterization of GPCR Activity and Function during Endosomal Trafficking Pathway. Anal Chem 2021; 93:2010-2017. [PMID: 33400862 DOI: 10.1021/acs.analchem.0c03323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
G protein-coupled receptor (GPCR) is activated by extracellular signals. After their function at plasma membrane, GPCRs are internalized to be desensitized, while emerging evidence suggests that some GPCRs maintain their activity even after internalization. The endosomal trafficking pathway of a prototypic GPCR, β adrenergic receptor 2 (B2AR), is in the range of several hours, however, spatiotemporal B2AR activity during this long-term endosomal trafficking pathway has not been characterized yet. Here, we analyze an agonist-induced real-time B2AR activity and its downstream function at the level of individual vesicles, utilizing a fluorescence resonance energy transfer (FRET)-based B2AR biosensor and cAMP reporters tethered at different trafficking stages of endosomes. Our results report that the internalized B2ARs sustain the activity and maintain the production of cAMP for several hours during the endosomal trafficking pathway. Temporal kinetics of B2AR activity is mathematically well explained by our active-vesicle population model modified from the Ricker model. Therefore, our GPCR monitoring system and a new kinetics model can be applied to understand the spatiotemporal GPCR activity and its downstream function during the endosomal trafficking pathway.
Collapse
Affiliation(s)
- Hyunbin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Hae Nim Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaesik Choi
- Graduate School of Artificial Intelligence, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.,Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
83
|
Abstract
The field of cAMP signaling is witnessing exciting developments with the recognition that cAMP is compartmentalized and that spatial regulation of cAMP is critical for faithful signal coding. This realization has changed our understanding of cAMP signaling from a model in which cAMP connects a receptor at the plasma membrane to an intracellular effector in a linear pathway to a model in which cAMP signals propagate within a complex network of alternative branches and the specific functional outcome strictly depends on local regulation of cAMP levels and on selective activation of a limited number of branches within the network. In this review, we cover some of the early studies and summarize more recent evidence supporting the model of compartmentalized cAMP signaling, and we discuss how this knowledge is starting to provide original mechanistic insight into cell physiology and a novel framework for the identification of disease mechanisms that potentially opens new avenues for therapeutic interventions. SIGNIFICANCE STATEMENT: cAMP mediates the intracellular response to multiple hormones and neurotransmitters. Signal fidelity and accurate coordination of a plethora of different cellular functions is achieved via organization of multiprotein signalosomes and cAMP compartmentalization in subcellular nanodomains. Defining the organization and regulation of subcellular cAMP nanocompartments is necessary if we want to understand the complex functional ramifications of pharmacological treatments that target G protein-coupled receptors and for generating a blueprint that can be used to develop precision medicine interventions.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Miguel J Lobo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
84
|
Ju ZH, Liang X, Ren YY, Shu LW, Yan YH, Cui X. Neurons derived from human-induced pluripotent stem cells express mu and kappa opioid receptors. Neural Regen Res 2021; 16:653-658. [PMID: 33063716 PMCID: PMC8067944 DOI: 10.4103/1673-5374.295341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Neuroprotection studies have shown that induced pluripotent stem (iPS) cells have the possibility to transform neuroprotection research. In the present study, iPS cells were generated from human renal epithelial cells and were then differentiated into neurons. Cells in the iPS-cell group were maintained in stem cell medium. In contrast, cells in the iPS-neuron group were first maintained in neural induction medium and expansion medium containing ROCK inhibitors, and then cultivated in neuronal differentiation medium and neuronal maturation medium to induce the neural stem cells to differentiate into neurons. The expression of relevant markers was compared at different stages of differentiation. Immunofluorescence staining revealed that cells in the iPS-neuron group expressed the neural stem cell markers SOX1 and nestin on day 11 of induction, and neuronal markers TUBB3 and NeuN on day 21 of induction. Polymerase chain reaction results demonstrated that, compared with the iPS-cell group, TUBB3 gene expression in the iPS-neuron group was increased 15.6-fold. Further research revealed that, compared with the iPS-cell group, the gene expression and immunoreactivity of mu opioid receptor in the iPS-neuron group were significantly increased (38.3-fold and 5.7-fold, respectively), but those of kappa opioid receptor had only a slight change (1.33-fold and 1.57-fold increases, respectively). Together, these data indicate that human iPS cells can be induced into mu opioid receptor- and kappa opioid receptor-expressing neurons, and that they may be useful to simulate human opioid receptor function in vitro and explore the underlying mechanisms of human conditions.
Collapse
Affiliation(s)
- Zhi-Hai Ju
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xuan Liang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yao-Yao Ren
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Luo-Wa Shu
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yan-Hong Yan
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xu Cui
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
85
|
Bao Z, Zhou S, Zhou H. Sorting Nexin 27 as a potential target in G protein‑coupled receptor recycling for cancer therapy (Review). Oncol Rep 2020; 44:1779-1786. [PMID: 33000258 PMCID: PMC7551096 DOI: 10.3892/or.2020.7766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
G protein‑coupled receptors (GPCRs) are the largest family of membrane receptors and activate several downstream signaling pathways involved in numerous physiological cellular processes. GPCRs are usually internalized and desensitized by intracellular signals. Numerous studies have shown that several GPCRs interact with sorting nexin 27 (SNX27), a cargo selector of the retromer complex, and are recycled from endosomes to the plasma membrane. Recycled GPCRs usually contain specific C‑terminal postsynaptic density protein 95/Discs large protein/Zonula occludens 1 (PDZ) binding motifs, which are specifically recognized by SNX27, and return to the cell surface as functionally naïve receptors. Aberrant endosome‑to‑membrane recycling of GPCRs mediated by SNX27 may serve a critical role in cancer growth and development. Therefore, SNX27 may be a novel target for cancer therapies.
Collapse
Affiliation(s)
- Zixu Bao
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Clinical Medicine (5+3 Programme), Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shijun Zhou
- Department of Infectious Disease, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Haisheng Zhou
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Center for Scientific Research, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
86
|
Shackley M, Ma Y, Tate EW, Brown AJH, Frost G, Hanyaloglu AC. Short Chain Fatty Acids Enhance Expression and Activity of the Umami Taste Receptor in Enteroendocrine Cells via a Gα i/o Pathway. Front Nutr 2020; 7:568991. [PMID: 33195366 PMCID: PMC7658341 DOI: 10.3389/fnut.2020.568991] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/29/2020] [Indexed: 12/20/2022] Open
Abstract
The short chain fatty acids (SCFAs) acetate, butyrate and propionate, are produced by fermentation of non-digestible carbohydrates by the gut microbiota and regulate appetite, adiposity, metabolism, glycemic control, and immunity. SCFAs act at two distinct G protein coupled receptors (GPCRs), FFAR2 and FFAR3 and are expressed in intestinal enteroendocrine cells (EECs), where they mediate anorectic gut hormone release. EECs also express other GPCRs that act as nutrient sensors, thus SCFAs may elicit some of their health-promoting effects by altering GPCR expression in EECs and enhance gut sensitivity to dietary molecules. Here, we identify that exposure of the murine EEC STC-1 cell line or intestinal organoids to physiological concentrations of SCFAs enhances mRNA levels of the umami taste receptors TASR1 and TASR3, without altering levels of the SCFA GPCRs, FFAR2 and FFAR3. Treatment of EECs with propionate or butyrate, but not acetate, increased levels of umami receptor transcripts, while propionate also reduced CCK expression. This was reversed by inhibiting Gαi/o signaling with pertussis toxin, suggesting that SCFAs act through FFAR2/3 to alter gene expression. Surprisingly, neither a FFAR3 nor a FFAR2 selective ligand could increase TASR1/TASR3 mRNA levels. We assessed the functional impact of increased TASR1/TASR3 expression using unique pharmacological properties of the umami taste receptor; namely, the potentiation of signaling by inosine monophosphate. Activation of umami taste receptor induced inositol-1-phosphate and calcium signaling, and butyrate pretreatment significantly enhanced such signaling. Our study reveals that SCFAs may contribute to EEC adaptation and alter EEC sensitivity to bioactive nutrients.
Collapse
Affiliation(s)
- Matilda Shackley
- Section of Nutrition Research, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom.,Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Yue Ma
- Section of Nutrition Research, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, United Kingdom
| | | | - Gary Frost
- Section of Nutrition Research, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| | - Aylin C Hanyaloglu
- Institute of Reproductive and Developmental Biology, Department of Metabolism, Digestion, and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
87
|
Toneatti R, Shin JM, Shah UH, Mayer CR, Saunders JM, Fribourg M, Arsenovic PT, Janssen WG, Sealfon SC, López-Giménez JF, Benson DL, Conway DE, González-Maeso J. Interclass GPCR heteromerization affects localization and trafficking. Sci Signal 2020; 13:eaaw3122. [PMID: 33082287 PMCID: PMC7717648 DOI: 10.1126/scisignal.aaw3122] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Membrane trafficking processes regulate G protein-coupled receptor (GPCR) activity. Although class A GPCRs are capable of activating G proteins in a monomeric form, they can also potentially assemble into functional GPCR heteromers. Here, we showed that the class A serotonin 5-HT2A receptors (5-HT2ARs) affected the localization and trafficking of class C metabotropic glutamate receptor 2 (mGluR2) through a mechanism that required their assembly as heteromers in mammalian cells. In the absence of agonists, 5-HT2AR was primarily localized within intracellular compartments, and coexpression of 5-HT2AR with mGluR2 increased the intracellular distribution of the otherwise plasma membrane-localized mGluR2. Agonists for either 5-HT2AR or mGluR2 differentially affected trafficking through Rab5-positive endosomes in cells expressing each component of the 5-HT2AR-mGluR2 heterocomplex alone, or together. In addition, overnight pharmacological 5-HT2AR blockade with clozapine, but not with M100907, decreased mGluR2 density through a mechanism that involved heteromerization between 5-HT2AR and mGluR2. Using TAT-tagged peptides and chimeric constructs that are unable to form the interclass 5-HT2AR-mGluR2 complex, we demonstrated that heteromerization was necessary for the 5-HT2AR-dependent effects on mGluR2 subcellular distribution. The expression of 5-HT2AR also augmented intracellular localization of mGluR2 in mouse frontal cortex pyramidal neurons. Together, our data suggest that GPCR heteromerization may itself represent a mechanism of receptor trafficking and sorting.
Collapse
MESH Headings
- Amino Acids/pharmacology
- Animals
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Membrane/metabolism
- Clozapine/pharmacology
- Endosomes/metabolism
- HEK293 Cells
- Humans
- Mice, 129 Strain
- Mice, Knockout
- Microscopy, Confocal
- Multiprotein Complexes/chemistry
- Multiprotein Complexes/metabolism
- Protein Multimerization
- Protein Transport/drug effects
- Receptor, Serotonin, 5-HT2A/chemistry
- Receptor, Serotonin, 5-HT2A/genetics
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptors, Metabotropic Glutamate/chemistry
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Serotonin Antagonists/pharmacology
- Signal Transduction
Collapse
Affiliation(s)
- Rudy Toneatti
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jong M Shin
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Urjita H Shah
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Carl R Mayer
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Justin M Saunders
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Miguel Fribourg
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Translational Transplant Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paul T Arsenovic
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - William G Janssen
- Department Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Stuart C Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan F López-Giménez
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA
- Instituto de Parasitología y Biomedicina "López-Neyra", CSIC, E-18016 Granada, Spain
| | - Deanna L Benson
- Department Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel E Conway
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23220, USA
| | - Javier González-Maeso
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
88
|
Melick CH, Jewell JL. Regulation of mTORC1 by Upstream Stimuli. Genes (Basel) 2020; 11:genes11090989. [PMID: 32854217 PMCID: PMC7565831 DOI: 10.3390/genes11090989] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/22/2020] [Accepted: 08/23/2020] [Indexed: 01/08/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is an evolutionary conserved Ser/Thr protein kinase that senses multiple upstream stimuli to control cell growth, metabolism, and autophagy. mTOR is the catalytic subunit of mTOR complex 1 (mTORC1). A significant amount of research has uncovered the signaling pathways regulated by mTORC1, and the involvement of these signaling cascades in human diseases like cancer, diabetes, and ageing. Here, we review advances in mTORC1 regulation by upstream stimuli. We specifically focus on how growth factors, amino acids, G-protein coupled receptors (GPCRs), phosphorylation, and small GTPases regulate mTORC1 activity and signaling.
Collapse
Affiliation(s)
- Chase H. Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenna L. Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Correspondence:
| |
Collapse
|
89
|
Regulated expression and function of the GABA B receptor in human pancreatic beta cell line and islets. Sci Rep 2020; 10:13469. [PMID: 32778664 PMCID: PMC7417582 DOI: 10.1038/s41598-020-69758-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors are seven transmembrane signaling molecules that are involved in a wide variety of physiological processes. They constitute a large protein family of receptors with almost 300 members detected in human pancreatic islet preparations. However, the functional role of these receptors in pancreatic islets is unknown in most cases. We generated a new stable human beta cell line from neonatal pancreas. This cell line, named ECN90 expresses both subunits (GABBR1 and GABBR2) of the metabotropic GABAB receptor compared to human islet. In ECN90 cells, baclofen, a specific GABAB receptor agonist, inhibits cAMP signaling causing decreased expression of beta cell-specific genes such as MAFA and PCSK1, and reduced insulin secretion. We next demonstrated that in primary human islets, GABBR2 mRNA expression is strongly induced under cAMP signaling, while GABBR1 mRNA is constitutively expressed. We also found that induction and activation of the GABAB receptor in human islets modulates insulin secretion.
Collapse
|
90
|
GGA3 interacts with L-type prostaglandin D synthase and regulates the recycling and signaling of the DP1 receptor for prostaglandin D2 in a Rab4-dependent mechanism. Cell Signal 2020; 72:109641. [DOI: 10.1016/j.cellsig.2020.109641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 12/21/2022]
|
91
|
Deyts C, Clutter M, Pierce N, Chakrabarty P, Ladd TB, Goddi A, Rosario AM, Cruz P, Vetrivel K, Wagner SL, Thinakaran G, Golde TE, Parent AT. APP-Mediated Signaling Prevents Memory Decline in Alzheimer's Disease Mouse Model. Cell Rep 2020; 27:1345-1355.e6. [PMID: 31042463 DOI: 10.1016/j.celrep.2019.03.087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 01/04/2023] Open
Abstract
Amyloid precursor protein (APP) and its metabolites play key roles in Alzheimer's disease (AD) pathophysiology. Whereas short amyloid-β (Aβ) peptides derived from APP are pathogenic, the APP holoprotein serves multiple purposes in the nervous system through its cell adhesion and receptor-like properties. Our studies focused on the signaling mediated by the APP cytoplasmic tail. We investigated whether sustained APP signaling during brain development might favor neuronal plasticity and memory process through a direct interaction with the heterotrimeric G-protein subunit GαS (stimulatory G-protein alpha subunit). Our results reveal that APP possesses autonomous regulatory capacity within its intracellular domain that promotes APP cell surface residence, precludes Aβ production, facilitates axodendritic development, and preserves cellular substrates of memory. Altogether, these events contribute to strengthening cognitive functions and are sufficient to modify the course of AD pathology.
Collapse
Affiliation(s)
- Carole Deyts
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Mary Clutter
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Nicholas Pierce
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Thomas B Ladd
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Anna Goddi
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Pedro Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kulandaivelu Vetrivel
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Steven L Wagner
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Angèle T Parent
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
92
|
Chen J, Chen X, Li S, Jiang Y, Mao H, Zhang R, Ji B, Yan M, Cai X, Wang C. Individual phosphorylation sites at the C-terminus of the apelin receptor play different roles in signal transduction. Redox Biol 2020; 36:101629. [PMID: 32863206 PMCID: PMC7338617 DOI: 10.1016/j.redox.2020.101629] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/16/2022] Open
Abstract
The apelin and Elabela proteins constitute a spatiotemporal double-ligand system that controls apelin receptor (APJ) signal transduction. Phosphorylation of multiple sites within the C-terminus of APJ is essential for the recruitment of β-arrestins. We sought to determine the precise mechanisms by which apelin and Elabela promote APJ phosphorylation, and to elucidate the influence of β-arrestin phosphorylation on G-protein-coupled receptor (GPCR)/β-arrestin-dependent signaling. We used techniques including mass spectrometry (MS), mutation analysis, and bioluminescence resonance energy transfer (BRET) to evaluate the role of phosphorylation sites in APJ-mediated G-protein-dependent and β-dependent signaling. Phosphorylation of APJ occurred at five serine residues in the C-terminal region (Ser335, Ser339, Ser345, Ser348 and Ser369). We also identified two phosphorylation sites in β-arrestin1 and three in β-arrestin2, including three previously identified residues (Ser412, Ser361, and Thr383) and two new sites, Tyr47 in β-arrestin1 and Tyr48 in β-arrestin2. APJ mutations did not affect the phosphorylation of β-arrestins, but it affects the β-arrestin signaling pathway, specifically Ser335 and Ser339. Mutation of Ser335 decreased the ability of the receptor to interact with β-arrestin1/2 and AP2, indicating that APJ affects the β-arrestin signaling pathway by stimulating Elabela. Mutation of Ser339 abolished the capability of the receptor to interact with GRK2 and β-arrestin1/2 upon stimulation with apelin-36, and disrupted receptor internalization and β-arrestin-dependent ERK1/2 activation. Five peptides act on distinct phosphorylation sites at the APJ C-terminus, differentially regulating APJ signal transduction and causing different biological effects. These findings may facilitate screening for drugs to treat cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| | - Xiaoyu Chen
- Department of Physiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, Shandong, China
| | - Sheng Li
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Yunlu Jiang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Huiling Mao
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Rumin Zhang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Bingyuan Ji
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| | - Maocai Yan
- School of Pharmacy, Jining Medical University, Shandong, China
| | - Xin Cai
- Department of Physiology, Weifang Medical University, Weifang, Shandong, China
| | - Chunmei Wang
- Neurobiology Institute, Jining Medical University, Jining, Shandong, China
| |
Collapse
|
93
|
Cheng Y, Chen T, Song J, Teng Z, Wang C, Wang S, Lu G, Feng T, Qi Q, Xi Q, Liu S, Hao L, Zhang Y. Pituitary miRNAs target GHRHR splice variants to regulate GH synthesis by mediating different intracellular signalling pathways. RNA Biol 2020; 17:1754-1766. [PMID: 32508238 DOI: 10.1080/15476286.2020.1778295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Growth hormone (GH), whose synthesis and release are mainly regulated by intracellular signals mediated by growth hormone-releasing hormone receptor (GHRHR), is one of the major pituitary hormones and critical regulators of organism growth, metabolism, and immunoregulation. Pig GHRHR splice variants (SVs) may activate different signalling pathways via the variable C-terminal by alternative splicing, and SVs have the potential to change microRNA (miRNA) binding sites. In this study, we first confirmed the existence of pig GHRHR SVs (i.e., GHRHR, GHRHR SV1 and SV2) and demonstrated the inhibitory effects of critical pituitary miRNAs (i.e., let-7e and miR-328-5p) on GH synthesis and cell proliferation of primary pituitary cells. The SVs of GHRHR targeted by let-7e and miR-328-5p were predicted via bioinformatics analysis and verified by performing dual-luciferase reporter assays and detecting the expression of target transcripts. The differential responses of let-7e, and miR-328-5p to GH-releasing hormone and the changes in signalling pathways mediated by GHRHR suggested that let-7e and miR-328-5p were involved in GH synthesis mediated by GHRHR SVs, indicating that the two miRNAs played different roles by different ways. Finally, results showed that the protein coded by the GHRHR transcript regulated GH through the NO/NOS signalling pathway, whereas that coded by SV1 and SV2 regulated GH through the PKA/CREB signalling pathway, which was confirmed by the changes in signalling pathways after transfecting the expression vectors of GHRHR SVs to GH3 cells. To the best of our knowledge, this paper is the first to report pituitary miRNAs regulate GH synthesis by targeting the different SVs of GHRHR.
Collapse
Affiliation(s)
- Yunyun Cheng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China.,Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| | - Jie Song
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Zhaohui Teng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China.,Research and Development Centre, Dalian Mogue Biotech Co., Ltd , Dalian, China
| | - Chunli Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Siyao Wang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Guanhong Lu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Tianqi Feng
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University , Foshan China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| | - Songcai Liu
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Linlin Hao
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University , Changchun, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University , Guangzhou, China
| |
Collapse
|
94
|
Toms D, Al-Ani A, Sunba S, Tong QYV, Workentine M, Ungrin M. Automated Hypothesis Generation to Identify Signals Relevant in the Development of Mammalian Cell and Tissue Bioprocesses, With Validation in a Retinal Culture System. Front Bioeng Biotechnol 2020; 8:534. [PMID: 32582664 PMCID: PMC7287043 DOI: 10.3389/fbioe.2020.00534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
We have developed an accessible software tool (receptoR) to predict potentially active signaling pathways in one or more cell type(s) of interest from publicly available transcriptome data. As proof-of-concept, we applied it to mouse photoreceptors, yielding the previously untested hypothesis that activin signaling pathways are active in these cells. Expression of the type 2 activin receptor (Acvr2a) was experimentally confirmed by both RT-qPCR and immunochemistry, and activation of this signaling pathway with recombinant activin A significantly enhanced the survival of magnetically sorted photoreceptors in culture. Taken together, we demonstrate that our approach can be easily used to mine publicly available transcriptome data and generate hypotheses around receptor expression that can be used to identify novel signaling pathways in specific cell types of interest. We anticipate that receptoR (available at https://www.ucalgary.ca/ungrinlab/receptoR) will enable more efficient use of limited research resources.
Collapse
Affiliation(s)
- Derek Toms
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Abdullah Al-Ani
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada.,Leaders in Medicine Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Saud Sunba
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Qing Yun Victor Tong
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Matthew Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Mark Ungrin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, AB, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
95
|
Bannert K, Berlin P, Reiner J, Lemcke H, David R, Engelmann R, Lamprecht G. SNX27 regulates DRA activity and mediates its direct recycling by PDZ-interaction in early endosomes at the apical pole of Caco2 cells. Am J Physiol Gastrointest Liver Physiol 2020; 318:G854-G869. [PMID: 32116023 DOI: 10.1152/ajpgi.00374.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
DRA (downregulated in adenoma, SLC26A3) and NHE3 (Na+/H+ exchanger 3, SLC9A3) together mediate intestinal electroneutral NaCl absorption. Both transporters contain PDZ (postsynaptic density 95, disc large, zonula occludens 1) binding motifs and interact with PDZ adaptor proteins regulating their activity and recycling. SNX27 (sorting nexin 27) contains a PDZ domain and is involved in the recycling of cargo proteins including NHE3. The interaction of SNX27 with DRA and its potential role for the activity and recycling of DRA have been evaluated in this study. SNX27 specifically interacts with DRA via its PDZ domain. The knockdown (KD) of SNX27 reduced DRA activity by 50% but was not accompanied by a decrease of DRA surface expression. This indicates that DRA is trafficked to specific functional domains in the plasma membrane in which DRA is particularly active. Consistently, the disruption of lipid raft integrity by methyl-β-cyclodextrin has an inhibitory effect on DRA activity that was strongly reduced after SNX27 KD. In differentiated intestinal Caco2 cells, superresolution microscopy and a novel quantitative axial approach revealed that DRA and SNX27 colocalize in rab5-positive early endosomes at the apical pole. SNX27 regulates the activity of DRA in the apical plasma membrane through binding with its PDZ domain. This interaction occurs in rab5-positive early endosomes at the apical pole of differentiated intestinal Caco2 cells. SNX27 is involved in the direct recycling of DRA to the plasma membrane where it is inserted into lipid rafts facilitating increased activity.NEW & NOTEWORTHY SNX27 has a PDZ domain and is involved in the regulation and recycling of transmembrane proteins. The role of SNX27 on the activity and recycling of the intestinal Cl-/HCO3- exchanger DRA has not yet been studied. This study shows that SNX27 directly interacts with DRA in early endosomes at the apical pole of intestinal Caco2 cells and mediates its direct recycling to facilitate high activity in lipid rafts in the apical plasma membrane.
Collapse
Affiliation(s)
- Karen Bannert
- Department of Gastroenterology and Endocrinology, Center for Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Peggy Berlin
- Department of Gastroenterology and Endocrinology, Center for Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Johannes Reiner
- Department of Gastroenterology and Endocrinology, Center for Internal Medicine, Rostock University Medical Center, Rostock, Germany
| | - Heiko Lemcke
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany.,Department Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany.,Department Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Robby Engelmann
- Institute of Immunology, Rostock University Medical Center, Rostock, Germany
| | - Georg Lamprecht
- Department of Gastroenterology and Endocrinology, Center for Internal Medicine, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
96
|
Shin SH, Jeong J, Kim JH, Sohn KY, Yoon SY, Kim JW. 1-Palmitoyl-2-Linoleoyl-3-Acetyl-rac-Glycerol (PLAG) Mitigates Monosodium Urate (MSU)-Induced Acute Gouty Inflammation in BALB/c Mice. Front Immunol 2020; 11:710. [PMID: 32395118 PMCID: PMC7196669 DOI: 10.3389/fimmu.2020.00710] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/30/2020] [Indexed: 12/25/2022] Open
Abstract
Acute gouty arthritis is an auto-inflammatory disease caused by the deposition of monosodium urate (MSU) crystals in joints or tissues. Excessive neutrophil recruitment into gouty lesions is a general clinical sign and induces a pain phenotype. Attenuation of successive periods of neutrophil infiltration might be a beneficial approach to achieve therapeutic efficacy. In this study, the activity of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in attenuation of excess neutrophil infiltration was assessed in gout-induced lesions of BALB/c mice. Neutrophil infiltration in MSU-induced gouty lesions was analyzed using immunohistochemical staining. ELISA and RT-PCR were used to measure attenuation of expression of the major neutrophil chemoattractant, CXC motif chemokine ligand 8 (CXCL8), in a PLAG-treated animal model and in cells in vitro. The animal model revealed massive increased neutrophil infiltration in the MSU-induced gouty lesions, but the PLAG-treated mice had significantly reduced neutrophil numbers in these lesions. The results also indicated that the MSU crystals stimulated a damage-associated molecular pattern that was recognized by the P2Y6 purinergic receptor. This MSU-stimulated P2Y6 receptor was destined to intracellular trafficking. During intracellular endosomal trafficking of the receptor, endosome-dependent signaling provided expression of CXCL8 chemokines for neutrophil recruitment. PLAG accelerated initiation of the intracellular trafficking of the P2Y6 receptor and returning the receptor to the membrane. This process shortened the intracellular retention time of the receptor anchoring endosome and subsequently attenuated endosome-dependent signaling for CXCL8 expression. These study results suggested that PLAG could be used for resolution of acute inflammation induced in gout lesions.
Collapse
Affiliation(s)
- Su-Hyun Shin
- Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| | - Jinseon Jeong
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Seoul, South Korea
| | - Joo Heon Kim
- Department of Pathology, EulJi University School of Medicine, Daejeon, South Korea
| | - Ki-Young Sohn
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Seoul, South Korea
| | - Sun Young Yoon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Seoul, South Korea
| | - Jae Wha Kim
- Division of Systems Biology and Bioengineering, Cell Factory Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
97
|
Gröper J, König GM, Kostenis E, Gerke V, Raabe CA, Rescher U. Exploring Biased Agonism at FPR1 as a Means to Encode Danger Sensing. Cells 2020; 9:cells9041054. [PMID: 32340221 PMCID: PMC7226602 DOI: 10.3390/cells9041054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/28/2022] Open
Abstract
Ligand-based selectivity in signal transduction (biased signaling) is an emerging field of G protein-coupled receptor (GPCR) research and might allow the development of drugs with targeted activation profiles. Human formyl peptide receptor 1 (FPR1) is a GPCR that detects potentially hazardous states characterized by the appearance of N-formylated peptides that originate from either bacteria or mitochondria during tissue destruction; however, the receptor also responds to several non-formylated agonists from various sources. We hypothesized that an additional layer of FPR signaling is encoded by biased agonism, thus allowing the discrimination of the source of threat. We resorted to the comparative analysis of FPR1 agonist-evoked responses across three prototypical GPCR signaling pathways, i.e., the inhibition of cAMP formation, receptor internalization, and ERK activation, and analyzed cellular responses elicited by several bacteria- and mitochondria-derived ligands. We also included the anti-inflammatory annexinA1 peptide Ac2-26 and two synthetic ligands, the W-peptide and the small molecule FPRA14. Compared to the endogenous agonists, the bacterial agonists displayed significantly higher potencies and efficacies. Selective pathway activation was not observed, as both groups were similarly biased towards the inhibition of cAMP formation. The general agonist bias in FPR1 signaling suggests a source-independent pathway selectivity for transmission of pro-inflammatory danger signaling.
Collapse
Affiliation(s)
- Jieny Gröper
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.G.); (V.G.)
- Cells in Motion” Interfaculty Centre, University of Muenster, 48149 Muenster, Germany
| | - Gabriele M. König
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany; (G.M.K.); (E.K.)
| | - Evi Kostenis
- Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115 Bonn, Germany; (G.M.K.); (E.K.)
| | - Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.G.); (V.G.)
- Cells in Motion” Interfaculty Centre, University of Muenster, 48149 Muenster, Germany
| | - Carsten A. Raabe
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.G.); (V.G.)
- Institute of Experimental Pathology, Center for Molecular Biology of Inflammation, University of Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
- Correspondence: (C.A.R.); (U.R.); Tel.: +49-(0)251-835-2132 (C.A.R.); +49-(0)251-835-2121(U.R.)
| | - Ursula Rescher
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany; (J.G.); (V.G.)
- Cells in Motion” Interfaculty Centre, University of Muenster, 48149 Muenster, Germany
- Correspondence: (C.A.R.); (U.R.); Tel.: +49-(0)251-835-2132 (C.A.R.); +49-(0)251-835-2121(U.R.)
| |
Collapse
|
98
|
Xu Z, Xie Z, Sun J, Huang S, Chen Y, Li C, Sun X, Xia B, Tian L, Guo C, Li F, Pi G. Gut Microbiome Reveals Specific Dysbiosis in Primary Osteoporosis. Front Cell Infect Microbiol 2020; 10:160. [PMID: 32373553 PMCID: PMC7186314 DOI: 10.3389/fcimb.2020.00160] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/25/2020] [Indexed: 12/14/2022] Open
Abstract
Object: Primary osteoporosis (PO) is the most common bone disease, which is characterized by decreased bone mass, damage of bone tissue microstructure, increased bone fragility, and is prone to fracture. Gut microbiome may be involved in bone metabolism of PO through gut-brain axis regulation of immune system and endocrine system, however, the specific mechanism is still unclear. The purpose of this study was to characterize the gut microbiome of patients with PO and its possible role in the occurrence and development of the disease. Methods: Fecal samples were collected from 48 PO patients and 48 healthy controls (HC). The composition of gut microbiome community was analyzed by 16s rDNA amplification sequencing, and the difference of gut microbiome composition between PO patients and HC individuals was compared. PICRUSt was also used to predict the biological function of gut microbiome in patients with PO, and to explore its possible role in the occurrence and development of this disease. The classification model is constructed by random forest algorithm so as to screen the key biomarkers. Result: The diversity of gut microorganisms in PO patients was significantly higher than that in HC group (p < 0.05) and there was significant difference in microbial composition in PO group. The abundance of Dialister (0.036 vs. 0.004, p < 0.001) and Faecalibacterium (0.331 vs. 0.132, p < 0.001) were significantly enriched which were the key flora related to PO. Although no significant correlation between bone mineral density and the richness of microbial communities are found, PICRUST results show that there are a wide range of potential pathways between gut microbiome and PO patients, including genetic information processing, metabolism, environmental information processing, cellular processes, human diseases, and organic systems. Notably, the discriminant model based on dominant microflora can effectively distinguish PO from HC (AUC = 93.56). Conclusions: The findings show that PO is related to the change of gut microbiome, especially the enriched Dialister and Faecalibacterium genera, which give new clues to understand the disease and provide markers for the diagnosis and new strategies for intervention treatment of the disease.
Collapse
Affiliation(s)
- Zhiming Xu
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zheng Xie
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jianguang Sun
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shilei Huang
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yangyang Chen
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Chengxiang Li
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xing Sun
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Bicheng Xia
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lingyang Tian
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Caijuan Guo
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Feng Li
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guofu Pi
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
99
|
Melick CH, Meng D, Jewell JL. A-kinase anchoring protein 8L interacts with mTORC1 and promotes cell growth. J Biol Chem 2020; 295:8096-8105. [PMID: 32312749 DOI: 10.1074/jbc.ac120.012595] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/14/2020] [Indexed: 01/28/2023] Open
Abstract
mTOR complex 1 (mTORC1) senses nutrients to mediate anabolic processes within the cell. Exactly how mTORC1 promotes cell growth remains unclear. Here, we identified a novel mTORC1-interacting protein called protein kinase A anchoring protein 8L (AKAP8L). Using biochemical assays, we found that the N-terminal region of AKAP8L binds to mTORC1 in the cytoplasm. Importantly, loss of AKAP8L decreased mTORC1-mediated processes such as translation, cell growth, and cell proliferation. AKAPs anchor protein kinase A (PKA) through PKA regulatory subunits, and we show that AKAP8L can anchor PKA through regulatory subunit Iα. Reintroducing full-length AKAP8L into cells restored mTORC1-regulated processes, whereas reintroduction of AKAP8L missing the N-terminal region that confers the interaction with mTORC1 did not. Our results suggest a multifaceted role for AKAPs in the cell. We conclude that mTORC1 appears to regulate cell growth, perhaps in part through AKAP8L.
Collapse
Affiliation(s)
- Chase H Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jenna L Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390 .,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
100
|
Hutchings CJ. A review of antibody-based therapeutics targeting G protein-coupled receptors: an update. Expert Opin Biol Ther 2020; 20:925-935. [PMID: 32264722 DOI: 10.1080/14712598.2020.1745770] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION G protein-coupled receptors (GPCRs) play key roles in many biological functions and are linked to many diseases across all therapeutic areas. As such, GPCRs represent a significant opportunity for antibody-based therapeutics. AREAS COVERED The structure of the major GPCR families is summarized in the context of choice of antigen source employed in the drug discovery process and receptor biology considerations which may impact on targeting strategies. An overview of the therapeutic GPCR-antibody target landscape and the diversity of current therapeutic programs is provided along with summary case studies for marketed antibody drugs or those in advanced clinical studies. Antibodies in early clinical studies and the emergence of next-generation modalities are also highlighted. EXPERT OPINION The GPCR-antibody pipeline has progressed significantly with a number of technical developments enabling the successful resolution of some of the challenges previously encountered and this has contributed to the growing interest in antibody-based therapeutics addressing this target class.
Collapse
|