1
|
Zhang C, Zhang S, Wang G, Huang X, Xu S, Wang D, Guo C, Wang Y. Genomics and transcriptomics identify quantitative trait loci affecting growth-related traits in silver pomfret (Pampus argenteus). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101414. [PMID: 39813916 DOI: 10.1016/j.cbd.2025.101414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
Pampus argenteus, a species distributed throughout the Indo-West Pacific, plays a significant role in the yield of aquaculture species. However, cultured P. argenteus has always been characterised by unbalanced growth synchronisation among individuals, slow growth rate, and lack of excellent germplasm resources. Therefore, we conducted mass selection for fast-growing strain P. argenteus for several consecutive years. Various genetic improvement programs have modified its genome sequence through selective pressure, leaving nucleotide signals that can be detected at the genomic level. In the present study, we combined bulked segregant analysis and transcriptome sequencing to identify candidate single nucleotide polymorphisms (SNPs) and key genes for growth-related traits in P. argenteus. A total of 7,280,936 SNPs and 2,212,379 insertions/deletions were identified in the extreme phenotypes of the fast-growing and slow-growing groups. Based on the examination of SNP frequency differences and sliding-window analysis, 42 SNPs were identified as candidate markers. Moreover, 14 of the 42 SNPs linked to growth-related traits were confirmed to be credible SNPs, and eight growth-related genes were screened, namely myb-binding protein 1 A, insulin A/B chains, α-1B adrenoceptor, engulfment and cell motility protein 3, myosin light chain kinase family member 4, insulin receptor located, unconventional myosin-9b, and matrilin-1. An optimal three-factor model (SNP4&SNP12&SNP14) was constructed using the generalized multifactor dimensionality reduction method, and its accuracy was verified as 67.72 %. These results may benefit genetic studies and accelerate genetic improvement of fast-growing strains of P. argenteus.
Collapse
Affiliation(s)
- Cheng Zhang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Shun Zhang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Guanlin Wang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Xiang Huang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Shanliang Xu
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Danli Wang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China
| | - Chunyang Guo
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China.
| | - Yajun Wang
- National Engineering Research Laboratory of marine biotechnology and Engineering, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo University, Zhejiang, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Zhejiang, Ningbo 315211, China.
| |
Collapse
|
2
|
Wang L, Jin Y, Zhi Y, Li Z, Wang M, Wang B, Wang X. Effects of melatonin in polycystic ovary syndrome: is there Hippo pathway crosstalk? J Ovarian Res 2025; 18:101. [PMID: 40369589 PMCID: PMC12076993 DOI: 10.1186/s13048-025-01642-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder among reproductive women, characterized by hyperandrogenism, oligo-ovulation and polycystic ovarian morphology. Incorporating complementary medicine alongside traditional lifestyle therapies for PCOS may offer additional benefits for affected women. Melatonin (MT), a hormone secreted by the pineal gland, has emerged as a potential treatment for regulating ovarian function in PCOS. However, the specific effects and underlying mechanisms of MT on PCOS need to be elucidated. METHODS This review consolidates evidence from randomized controlled trials, original research articles, systematic reviews, and meta-analyses regarding MT supplementation in PCOS, with a particular focus on its interaction with the Hippo pathway, to provide a comprehensive overview of current knowledge. RESULTS Current evidence suggests that MT plays a role in modulating PCOS through various mechanisms and is associated with the Hippo pathway. However, several uncertainties and key limitations in the existing literature must be addressed before these treatments can be integrated into standard clinical practice. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Yuanyuan Jin
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Yuanyuan Zhi
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Zhenzhen Li
- Department of Pathology, Shandong Provincial Maternal and Child Health Care Hospital, Qingdao University, Jinan, 250014, China
| | - Meili Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China
| | - Boda Wang
- Emergency Department, Xinji Town Central Health Center, Guanxian County, Liaocheng, 252500, China
| | - Xinbo Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
- Key Laboratory of Maternal & Fetal Medicine of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| |
Collapse
|
3
|
He X, Zou J, Chen Q, Qin X, Liu Y, Zeng L, Su H. Microbial and transcriptional response of Acropora valida and Turbinaria peltata to Vibrio coralliilyticus challenge: insights into corals disease resistance. BMC Microbiol 2024; 24:288. [PMID: 39095694 PMCID: PMC11295391 DOI: 10.1186/s12866-024-03438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Coral diseases are significant drivers of global coral reef degradation, with pathogens dominated by Vibrio coralliilyticus playing a prominent role in the development of coral diseases. Coral phenotype, symbiotic microbial communities, and host transcriptional regulation have been well-established as factors involved in determining coral disease resistance, but the underlying mechanisms remain incompletely understood. METHODS This study employs high-throughput sequencing to analyse the symbiotic microbial and transcriptional response of the hosts in order to evaluate the disease resistance of Acropora valida and Turbinaria peltata exposed to Vibrio coralliilyticus. RESULTS A. valida exhibited pronounced bleaching and tissue loss within 7 h of pathogen infection, whereas T. peltata showed no signs of disease throughout the experiment. Microbial diversity analyses revealed that T. peltata had a more flexible microbial community and a higher relative abundance of potential beneficial bacteria compared to A. valida. Although Vibrio inoculation resulted in a more significant decrease in the Symbiodiniaceae density of A. valida compared to that of T. peltata, it did not lead to recombination of the coral host and Symbiodiniaceae in either coral species. RNA-seq analysis revealed that the interspecific differences in the transcriptional regulation of hosts after Vibrio inoculation. Differentially expressed genes in A. valida were mainly enriched in the pathways associated with energy supply and immune response, such as G protein-coupled receptor signaling, toll-like receptor signaling, regulation of TOR signaling, while these genes in T. peltata were mainly involved in the pathway related to immune homeostasis and ion transport, such as JAK-STAT signaling pathway and regulation of ion transport. CONCLUSIONS Pathogenic challenges elicit different microbial and transcriptional shifts across coral species. This study offers novel insights into molecular mechanisms of coral resistance to disease.
Collapse
Affiliation(s)
- Xucong He
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Jie Zou
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China
- School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Qiqi Chen
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China
- School of Resources, Environment and Materials, Guangxi University, Nanning, 530004, China
| | - Xiao Qin
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Yuan Liu
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Lujia Zeng
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China
| | - Hongfei Su
- Coral Reef Research Center of China, Guangxi Laboratory On the Study of Coral Reefs in the South China Sea, School of Marine Sciences, Guangxi University, Nanning, 530004, China.
| |
Collapse
|
4
|
Tzortzini E, Kolocouris A. Molecular Biophysics of Class A G Protein Coupled Receptors-Lipids Interactome at a Glance-Highlights from the A 2A Adenosine Receptor. Biomolecules 2023; 13:957. [PMID: 37371538 DOI: 10.3390/biom13060957] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/25/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are embedded in phospholipid membrane bilayers with cholesterol representing 34% of the total lipid content in mammalian plasma membranes. Membrane lipids interact with GPCRs structures and modulate their function and drug-stimulated signaling through conformational selection. It has been shown that anionic phospholipids form strong interactions between positively charged residues in the G protein and the TM5-TM6-TM 7 cytoplasmic interface of class A GPCRs stabilizing the signaling GPCR-G complex. Cholesterol with a high content in plasma membranes can be identified in more specific sites in the transmembrane region of GPCRs, such as the Cholesterol Consensus Motif (CCM) and Cholesterol Recognition Amino Acid Consensus (CRAC) motifs and other receptor dependent and receptor state dependent sites. Experimental biophysical methods, atomistic (AA) MD simulations and coarse-grained (CG) molecular dynamics simulations have been applied to investigate these interactions. We emphasized here the impact of phosphatidyl inositol-4,5-bisphosphate (PtdIns(4,5)P2 or PIP2), a minor phospholipid component and of cholesterol on the function-related conformational equilibria of the human A2A adenosine receptor (A2AR), a representative receptor in class A GPCR. Several GPCRs of class A interacted with PIP2 and cholesterol and in many cases the mechanism of the modulation of their function remains unknown. This review provides a helpful comprehensive overview for biophysics that enter the field of GPCRs-lipid systems.
Collapse
Affiliation(s)
- Efpraxia Tzortzini
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Antonios Kolocouris
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece
| |
Collapse
|
5
|
Proteomic Analysis of the Role of the Adenylyl Cyclase-cAMP Pathway in Red Blood Cell Mechanical Responses. Cells 2022; 11:cells11071250. [PMID: 35406814 PMCID: PMC8997765 DOI: 10.3390/cells11071250] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Red blood cell (RBC) deformability is modulated by the phosphorylation status of the cytoskeletal proteins that regulate the interactions of integral transmembrane complexes. Proteomic studies have revealed that receptor-related signaling molecules and regulatory proteins involved in signaling cascades are present in RBCs. In this study, we investigated the roles of the cAMP signaling mechanism in modulating shear-induced RBC deformability and examined changes in the phosphorylation of the RBC proteome. We implemented the inhibitors of adenylyl cyclase (SQ22536), protein kinase A (H89), and phosphodiesterase (PDE) (pentoxifylline) to whole blood samples, applied 5 Pa shear stress (SS) for 300 s with a capillary tubing system, and evaluated RBC deformability using a LORRCA MaxSis. The inhibition of signaling molecules significantly deteriorated shear-induced RBC deformability (p < 0.05). Capillary SS slightly increased the phosphorylation of RBC cytoskeletal proteins. Tyrosine phosphorylation was significantly elevated by the modulation of the cAMP/PKA pathway (p < 0.05), while serine phosphorylation significantly decreased as a result of the inhibition of PDE (p < 0.05). AC is the core element of this signaling pathway, and PDE works as a negative feedback mechanism that could have potential roles in SS-induced RBC deformability. The cAMP/PKA pathway could regulate RBC deformability during capillary transit by triggering significant alterations in the phosphorylation state of RBCs.
Collapse
|
6
|
Abstract
PURPOSE Our understanding of thyroid-associated ophthalmopathy (TAO, A.K.A Graves' orbitopathy, thyroid eye disease) has advanced substantially, since one of us (TJS) wrote the 2010 update on TAO, appearing in this journal. METHODS PubMed was searched for relevant articles. RESULTS Recent insights have resulted from important studies conducted by many different laboratory groups around the World. A clearer understanding of autoimmune diseases in general and TAO specifically emerged from the use of improved research methodologies. Several key concepts have matured over the past decade. Among them, those arising from the refinement of mouse models of TAO, early stage investigation into restoring immune tolerance in Graves' disease, and a hard-won acknowledgement that the insulin-like growth factor-I receptor (IGF-IR) might play a critical role in the development of TAO, stand out as important. The therapeutic inhibition of IGF-IR has blossomed into an effective and safe medical treatment. Teprotumumab, a β-arrestin biased agonist monoclonal antibody inhibitor of IGF-IR has been studied in two multicenter, double-masked, placebo-controlled clinical trials demonstrated both effectiveness and a promising safety profile in moderate-to-severe, active TAO. Those studies led to the approval by the US FDA of teprotumumab, currently marketed as Tepezza for TAO. We have also learned far more about the putative role that CD34+ fibrocytes and their derivatives, CD34+ orbital fibroblasts, play in TAO. CONCLUSION The past decade has been filled with substantial scientific advances that should provide the necessary springboard for continually accelerating discovery over the next 10 years and beyond.
Collapse
Affiliation(s)
- E J Neag
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
- Michigan State University College of Osteopathic Medicine, East Lansing, MI, USA
| | - T J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, Brehm Tower, 1000 Wall Street, Ann Arbor, MI, 48105, USA.
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
7
|
Shen Y, Luchetti A, Fernandes G, Do Heo W, Silva AJ. The emergence of molecular systems neuroscience. Mol Brain 2022; 15:7. [PMID: 34983613 PMCID: PMC8728933 DOI: 10.1186/s13041-021-00885-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
Systems neuroscience is focused on how ensemble properties in the brain, such as the activity of neuronal circuits, gives rise to internal brain states and behavior. Many of the studies in this field have traditionally involved electrophysiological recordings and computational approaches that attempt to decode how the brain transforms inputs into functional outputs. More recently, systems neuroscience has received an infusion of approaches and techniques that allow the manipulation (e.g., optogenetics, chemogenetics) and imaging (e.g., two-photon imaging, head mounted fluorescent microscopes) of neurons, neurocircuits, their inputs and outputs. Here, we will review novel approaches that allow the manipulation and imaging of specific molecular mechanisms in specific cells (not just neurons), cell ensembles and brain regions. These molecular approaches, with the specificity and temporal resolution appropriate for systems studies, promise to infuse the field with novel ideas, emphases and directions, and are motivating the emergence of a molecularly oriented systems neuroscience, a new discipline that studies how the spatial and temporal patterns of molecular systems modulate circuits and brain networks, and consequently shape the properties of brain states and behavior.
Collapse
Affiliation(s)
- Yang Shen
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Alessandro Luchetti
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Giselle Fernandes
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Alcino J Silva
- Departments of Neurobiology, Psychiatry and Biobehavioral Sciences, and Psychology, Integrative Center for Learning and Memory, and Brain Research Institute, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Ng KF, Chen TC, Stacey M, Lin HH. Role of ADGRG1/GPR56 in Tumor Progression. Cells 2021; 10:cells10123352. [PMID: 34943858 PMCID: PMC8699533 DOI: 10.3390/cells10123352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
Cellular communication plays a critical role in diverse aspects of tumorigenesis including tumor cell growth/death, adhesion/detachment, migration/invasion, angiogenesis, and metastasis. G protein-coupled receptors (GPCRs) which constitute the largest group of cell surface receptors are known to play fundamental roles in all these processes. When considering the importance of GPCRs in tumorigenesis, the adhesion GPCRs (aGPCRs) are unique due to their hybrid structural organization of a long extracellular cell-adhesive domain and a seven-transmembrane signaling domain. Indeed, aGPCRs have been increasingly shown to be associated with tumor development by participating in tumor cell interaction and signaling. ADGRG1/GPR56, a representative tumor-associated aGPCR, is recognized as a potential biomarker/prognostic factor of specific cancer types with both tumor-suppressive and tumor-promoting functions. We summarize herein the latest findings of the role of ADGRG1/GPR56 in tumor progression.
Collapse
Affiliation(s)
- Kwai-Fong Ng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (K.-F.N.); (T.-C.C.)
| | - Tse-Ching Chen
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (K.-F.N.); (T.-C.C.)
| | - Martin Stacey
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK;
| | - Hsi-Hsien Lin
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (K.-F.N.); (T.-C.C.)
- Division of Rheumatology, Allergy, and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung 20401, Taiwan
- Center for Medical and Clinical Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence:
| |
Collapse
|
9
|
Güran A, Ji Y, Fang P, Pan KT, Urlaub H, Avkiran M, Lenz C. Quantitative Analysis of the Cardiac Phosphoproteome in Response to Acute β-Adrenergic Receptor Stimulation In Vivo. Int J Mol Sci 2021; 22:12584. [PMID: 34830474 PMCID: PMC8618155 DOI: 10.3390/ijms222212584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
β-adrenergic receptor (β-AR) stimulation represents a major mechanism of modulating cardiac output. In spite of its fundamental importance, its molecular basis on the level of cell signalling has not been characterised in detail yet. We employed mass spectrometry-based proteome and phosphoproteome analysis using SuperSILAC (spike-in stable isotope labelling by amino acids in cell culture) standardization to generate a comprehensive map of acute phosphoproteome changes in mice upon administration of isoprenaline (ISO), a synthetic β-AR agonist that targets both β1-AR and β2-AR subtypes. Our data describe 8597 quantitated phosphopeptides corresponding to 10,164 known and novel phospho-events from 2975 proteins. In total, 197 of these phospho-events showed significantly altered phosphorylation, indicating an intricate signalling network activated in response to β-AR stimulation. In addition, we unexpectedly detected significant cardiac expression and ISO-induced fragmentation of junctophilin-1, a junctophilin isoform hitherto only thought to be expressed in skeletal muscle. Data are available via ProteomeXchange with identifier PXD025569.
Collapse
Affiliation(s)
- Alican Güran
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, St Thomas’ Hospital, Westminster Bridge Road, London SE1 7EH, UK; (A.G.); (M.A.)
| | - Yanlong Ji
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany; (Y.J.); (P.F.); (K.-T.P.); (H.U.)
- Hematology/Oncology, Department of Medicine II, Johann Wolfgang Goethe University, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Johann Wolfgang Goethe University, 60590 Frankfurt am Main, Germany
| | - Pan Fang
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany; (Y.J.); (P.F.); (K.-T.P.); (H.U.)
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou 215123, China
| | - Kuan-Ting Pan
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany; (Y.J.); (P.F.); (K.-T.P.); (H.U.)
- Hematology/Oncology, Department of Medicine II, Johann Wolfgang Goethe University, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Johann Wolfgang Goethe University, 60590 Frankfurt am Main, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany; (Y.J.); (P.F.); (K.-T.P.); (H.U.)
- Department of Clinical Chemistry, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Metin Avkiran
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King’s College London, St Thomas’ Hospital, Westminster Bridge Road, London SE1 7EH, UK; (A.G.); (M.A.)
| | - Christof Lenz
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany; (Y.J.); (P.F.); (K.-T.P.); (H.U.)
- Department of Clinical Chemistry, University Medical Center Goettingen, 37075 Goettingen, Germany
| |
Collapse
|
10
|
Liu S, Trupiano MX, Simon J, Guo J, Anton ES. The essential role of primary cilia in cerebral cortical development and disorders. Curr Top Dev Biol 2021; 142:99-146. [PMID: 33706927 DOI: 10.1016/bs.ctdb.2020.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Primary cilium, first described in the 19th century in different cell types and organisms by Alexander Ecker, Albert Kolliker, Aleksandr Kowalevsky, Paul Langerhans, and Karl Zimmermann (Ecker, 1844; Kolliker, 1854; Kowalevsky, 1867; Langerhans, 1876; Zimmermann, 1898), play an essential modulatory role in diverse aspects of nervous system development and function. The primary cilium, sometimes referred to as the cell's 'antennae', can receive wide ranging inputs from cellular milieu, including morphogens, growth factors, neuromodulators, and neurotransmitters. Its unique structural and functional organization bequeaths it the capacity to hyper-concentrate signaling machinery in a restricted cellular domain approximately one-thousandth the volume of cell soma. Thus enabling it to act as a signaling hub that integrates diverse developmental and homestatic information from cellular milieu to regulate the development and function of neural cells. Dysfunction of primary cilia contributes to the pathophysiology of several brain malformations, intellectual disabilities, epilepsy, and psychiatric disorders. This review focuses on the most essential contributions of primary cilia to cerebral cortical development and function, in the context of neurodevelopmental disorders and malformations. It highlights the recent progress made in identifying the mechanisms underlying primary cilia's role in cortical progenitors, neurons and glia, in health and disease. A future challenge will be to translate these insights and advances into effective clinical treatments for ciliopathies.
Collapse
Affiliation(s)
- Siling Liu
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Mia X Trupiano
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jeremy Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jiami Guo
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States.
| |
Collapse
|
11
|
Simões SC, Balico-Silva AL, Parreiras-E-Silva LT, Bitencourt ALB, Bouvier M, Costa-Neto CM. Signal Transduction Profiling of Angiotensin II Type 1 Receptor With Mutations Associated to Atrial Fibrillation in Humans. Front Pharmacol 2021; 11:600132. [PMID: 33424609 PMCID: PMC7786401 DOI: 10.3389/fphar.2020.600132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 11/18/2020] [Indexed: 11/24/2022] Open
Abstract
The AT1 receptor (AT1R) has a major role in the Renin-Angiotensin System, being involved in several physiological events including blood pressure control and electrolyte balance. The AT1R is a member of the G protein coupled receptors (GPCR) family, classically known to couple Gαq and engage β-arrestin recruitment. Both G protein and arrestin signaling pathways are involved in modulation of different downstream kinases. A previous study reported that mutations in the AT1R (A244S and I103T-A244S) were positively correlated with higher risk of atrial fibrillation in men. Based on that report, we aimed to investigate if these mutations, including I103T only, could affect AT1R signal transduction profile, and consequently, implicate in atrial fibrillation outcome. To address that, we engineered an AT1R carrying the above-mentioned mutations, and functionally evaluated different signaling pathways. Phosphokinase profiler array to assess the mutations downstream effects on kinases and kinase substrates phosphorylation levels was used. Our results show that the I103T-A244S mutant receptor presents decreased β-arrestin 2 recruitment, which could lead to a harmful condition of sustained Gαq signaling. Moreover, the phosphokinase profiler array revealed that the same mutation led to downstream modulation of kinase pathways that are linked to physiological responses such as fibrous tissue formation, apoptosis and cell proliferation.
Collapse
Affiliation(s)
- Sarah C Simões
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - André L Balico-Silva
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - Lucas T Parreiras-E-Silva
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - André L B Bitencourt
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine and Institute for Research in Immunology and Cancer, University of Montréal, Montréal, QC, Canada
| | - Claudio M Costa-Neto
- Ribeirao Preto Medical School, Department of Biochemistry and Immunology, University of São Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
12
|
Guo J, Otis JM, Suciu SK, Catalano C, Xing L, Constable S, Wachten D, Gupton S, Lee J, Lee A, Blackley KH, Ptacek T, Simon JM, Schurmans S, Stuber GD, Caspary T, Anton ES. Primary Cilia Signaling Promotes Axonal Tract Development and Is Disrupted in Joubert Syndrome-Related Disorders Models. Dev Cell 2019; 51:759-774.e5. [PMID: 31846650 PMCID: PMC6953258 DOI: 10.1016/j.devcel.2019.11.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/08/2019] [Accepted: 11/10/2019] [Indexed: 12/18/2022]
Abstract
Appropriate axonal growth and connectivity are essential for functional wiring of the brain. Joubert syndrome-related disorders (JSRD), a group of ciliopathies in which mutations disrupt primary cilia function, are characterized by axonal tract malformations. However, little is known about how cilia-driven signaling regulates axonal growth and connectivity. We demonstrate that the deletion of related JSRD genes, Arl13b and Inpp5e, in projection neurons leads to de-fasciculated and misoriented axonal tracts. Arl13b deletion disrupts the function of its downstream effector, Inpp5e, and deregulates ciliary-PI3K/AKT signaling. Chemogenetic activation of ciliary GPCR signaling and cilia-specific optogenetic modulation of downstream second messenger cascades (PI3K, AKT, and AC3) commonly regulated by ciliary signaling receptors induce rapid changes in axonal dynamics. Further, Arl13b deletion leads to changes in transcriptional landscape associated with dysregulated PI3K/AKT signaling. These data suggest that ciliary signaling acts to modulate axonal connectivity and that impaired primary cilia signaling underlies axonal tract defects in JSRD.
Collapse
Affiliation(s)
- Jiami Guo
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA; Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA.
| | - James M Otis
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sarah K Suciu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christy Catalano
- Hotchkiss Brain Institute and the Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, USA
| | - Lei Xing
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Sandii Constable
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dagmar Wachten
- Biophysical Imaging, Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Stephanie Gupton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Janice Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Amelia Lee
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Katherine H Blackley
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Travis Ptacek
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jeremy M Simon
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Stephane Schurmans
- Laboratory of Functional Genetics, GIGA Research Center, University of Liège, Liège, Belgium
| | - Garret D Stuber
- Center for the Neurobiology of Addiction, Pain and Emotion, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| |
Collapse
|
13
|
Villegas-Comonfort S, Guzmán-Silva A, Romero-Ávila MT, Takei Y, Tsujimoto G, Hirasawa A, García-Sáinz JA. Receptor tyrosine kinase activation induces free fatty acid 4 receptor phosphorylation, β-arrestin interaction, and internalization. Eur J Pharmacol 2019; 855:267-275. [PMID: 31078517 DOI: 10.1016/j.ejphar.2019.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/12/2019] [Accepted: 05/06/2019] [Indexed: 12/30/2022]
Abstract
FFA4 (Free Fatty Acid receptor 4, previously known as GPR120) is a G protein-coupled receptor that acts as a sensor of long-chain fatty acids, modulates metabolism, and whose dysfunction participates in endocrine disturbances. FFA4 is known to be phosphorylated and internalized in response to agonists and protein kinase C activation. In this paper report the modulation of this fatty acid receptor by activation of receptor tyrosine kinases. Cell-activation with growth factors (insulin, epidermal growth factor, insulin-like growth factor-I, and platelet-derived growth factor) increases FFA4 phosphorylation in a time- and concentration-dependent fashion. This effect was blocked by inhibitors of protein kinase C and phosphoinositide 3-kinase, suggesting the involvement of these kinases in it. FFA4 phosphorylation did not alter agonist-induced FFA4 calcium signaling, but was associated with decreased ERK 1/2 phosphorylation. In addition, insulin, insulin-like growth factor-I, epidermal growth factor, and to a lesser extent, platelet-derived growth factor, induce receptor internalization. This action of insulin, insulin-like growth factor I, and epidermal growth factor was blocked by inhibitors of protein kinase C and phosphoinositide 3-kinase. Additionally, cell treatment with these growth factors induced FFA4-β-arrestin coimmunoprecipitation. Our results evidenced cross-talk between receptor tyrosine kinases and FFA4 and suggest roles of protein kinase C and phosphoinositide 3-kinase in such a functional interaction.
Collapse
Affiliation(s)
- Sócrates Villegas-Comonfort
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico
| | - Alejandro Guzmán-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico
| | - M Teresa Romero-Ávila
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico
| | - Yoshinori Takei
- Graduate School of Pharmaceutical Sciences, Kyoto University: Sakyo-ku, Kyoto, 606-8501, Japan
| | - Gozoh Tsujimoto
- Graduate School of Pharmaceutical Sciences, Kyoto University: Sakyo-ku, Kyoto, 606-8501, Japan
| | - Akira Hirasawa
- Graduate School of Pharmaceutical Sciences, Kyoto University: Sakyo-ku, Kyoto, 606-8501, Japan
| | - J Adolfo García-Sáinz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico.
| |
Collapse
|
14
|
Chavez-Abiega S, Goedhart J, Bruggeman FJ. Physical biology of GPCR signalling dynamics inferred from fluorescence spectroscopy and imaging. Curr Opin Struct Biol 2019; 55:204-211. [PMID: 31319372 DOI: 10.1016/j.sbi.2019.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/30/2019] [Accepted: 05/08/2019] [Indexed: 12/20/2022]
Abstract
The physical biology of G protein-coupled receptor (GPCR) signalling can be inferred from imaging of single molecules and single living cells. In this opinion paper, we highlight recent developments in technologies to study GPCR signalling in vitro and in cyto. We start from mobility and localisation characteristics of single receptors in membranes. Subsequently, we discuss the kinetics of shifts in receptor-conformation equilibrium due to allosteric binding events and G protein activation. We continue with recent insights into downstream signalling and the role of delayed negative feedback to suppress GPCR signalling. Finally, we discuss new strategies to reveal how the multiplex signalling responses of cells to ligand mixtures, mediated by their entire receptor arsenal, can be disentangled, using single-cell data.
Collapse
Affiliation(s)
- Sergei Chavez-Abiega
- Section Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, NL-1098 XH, Amsterdam, The Netherlands; Section Systems Bioinformatics, Amsterdam Institute for Molecules, Medicines and Systems, VU University, De Boelelaan 1085, NL-1081 HV, Amsterdam, The Netherlands
| | - Joachim Goedhart
- Section Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, NL-1098 XH, Amsterdam, The Netherlands
| | - Frank Johannes Bruggeman
- Section Systems Bioinformatics, Amsterdam Institute for Molecules, Medicines and Systems, VU University, De Boelelaan 1085, NL-1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
van der Werf A, de van der Schueren MAE, Rovithi M, Labots M, Hashemi SMS, Langius JAE, Verheul HMW. Taste Alterations During Treatment With Protein Kinase Inhibitors: A Pilot Study. J Pain Symptom Manage 2018; 56:e1-e4. [PMID: 30025935 DOI: 10.1016/j.jpainsymman.2018.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Anne van der Werf
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; Department of Nutrition and Dietetics, Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Marian A E de van der Schueren
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands; Department of Nutrition and Health, Faculty of Health and Social studies, HAN University of Applied Sciences, Nijmegen, The Netherlands
| | - Maria Rovithi
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Mariette Labots
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Sayed M S Hashemi
- Department of Pulmonology, VU University Medical Center, Amsterdam, The Netherlands
| | - Jacqueline A E Langius
- Department of Nutrition and Dietetics, Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands; Department of Nutrition and Dietetics, Faculty of Health, Nutrition and Sport, The Hague University of Applied Sciences, The Hague, The Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Expanding neuropeptide signalling by multiplying receptor functional states and sub-cellular locations. Cell Tissue Res 2018; 375:49-56. [DOI: 10.1007/s00441-018-2923-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022]
|
17
|
Hung WT, Navakanitworakul R, Khan T, Zhang P, Davis JS, McGinnis LK, Christenson LK. Stage-specific follicular extracellular vesicle uptake and regulation of bovine granulosa cell proliferation. Biol Reprod 2018; 97:644-655. [PMID: 29025042 DOI: 10.1093/biolre/iox106] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/25/2017] [Indexed: 12/24/2022] Open
Abstract
Follicular fluid within ovarian antral follicles contains numerous factors, which influence the development of a healthy oocyte including nucleic acids, steroids, proteins, and extracellular vesicles (EVs). Current evidence indicates that follicular EVs promote changes in cellular gene expression and support cumulus-oocyte complex expansion in vitro. In this study, we found EVs from different sized follicles differentially stimulate granulosa cell proliferation and this could be explained by both the differential contents associated, on or within the vesicles and by the preferential uptake of EVs dependent on follicle size from which they were isolated. Antibody array and inhibitor studies indicated that the Src, PI3K/Akt, and MAPK signaling pathways mediate the stimulatory effects of EVs on granulosa cell proliferation. This study demonstrates for the first time that EVs isolated from follicular fluid are capable of stimulating granulosa cell proliferation and that this stimulatory response is associated with the size of antral follicle from which the EVs originated. The study further also provides the first evidence that vesicles released by small antral follicles are preferentially taken up when compared to those isolated from large follicles, suggesting that vesicular surface proteins change during follicular maturation.
Collapse
Affiliation(s)
- Wei-Ting Hung
- Department Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Raphatphorn Navakanitworakul
- Department Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Tarique Khan
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Pan Zhang
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA
| | - Lynda K McGinnis
- Department Obstetrics and Gynecology, University of Southern California, Norris Cancer Center, Los Angeles, California, USA
| | - Lane K Christenson
- Department Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
18
|
Yen HY, Hoi KK, Liko I, Hedger G, Horrell MR, Song W, Wu D, Heine P, Warne T, Lee Y, Carpenter B, Plückthun A, Tate CG, Sansom MSP, Robinson CV. PtdIns(4,5)P 2 stabilizes active states of GPCRs and enhances selectivity of G-protein coupling. Nature 2018; 559:423-427. [PMID: 29995853 PMCID: PMC6059376 DOI: 10.1038/s41586-018-0325-6] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 05/14/2018] [Indexed: 11/21/2022]
Abstract
G-protein-coupled receptors (GPCRs) are involved in many physiological processes and are therefore key drug targets1. Although detailed structural information is available for GPCRs, the effects of lipids on the receptors, and on downstream coupling of GPCRs to G proteins are largely unknown. Here we use native mass spectrometry to identify endogenous lipids bound to three class A GPCRs. We observed preferential binding of phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2) over related lipids and confirm that the intracellular surface of the receptors contain hotspots for PtdIns(4,5)P2 binding. Endogenous lipids were also observed bound directly to the trimeric Gαsβγ protein complex of the adenosine A2A receptor (A2AR) in the gas phase. Using engineered Gα subunits (mini-Gαs, mini-Gαi and mini-Gα12)2, we demonstrate that the complex of mini-Gαs with the β1 adrenergic receptor (β1AR) is stabilized by the binding of two PtdIns(4,5)P2 molecules. By contrast, PtdIns(4,5)P2 does not stabilize coupling between β1AR and other Gα subunits (mini-Gαi or mini-Gα12) or a high-affinity nanobody. Other endogenous lipids that bind to these receptors have no effect on coupling, highlighting the specificity of PtdIns(4,5)P2. Calculations of potential of mean force and increased GTP turnover by the activated neurotensin receptor when coupled to trimeric Gαiβγ complex in the presence of PtdIns(4,5)P2 provide further evidence for a specific effect of PtdIns(4,5)P2 on coupling. We identify key residues on cognate Gα subunits through which PtdIns(4,5)P2 forms bridging interactions with basic residues on class A GPCRs. These modulating effects of lipids on receptors suggest consequences for understanding function, G-protein selectivity and drug targeting of class A GPCRs.
Collapse
MESH Headings
- Animals
- GTP-Binding Protein alpha Subunits, Gi-Go/metabolism
- GTP-Binding Protein alpha Subunits, Gs/metabolism
- Heterotrimeric GTP-Binding Proteins/metabolism
- Humans
- Molecular Dynamics Simulation
- Phosphatidylinositol 4,5-Diphosphate/metabolism
- Protein Stability
- Rats
- Receptors, Adrenergic, alpha-2/chemistry
- Receptors, Adrenergic, alpha-2/genetics
- Receptors, Adrenergic, alpha-2/metabolism
- Receptors, Adrenergic, beta-1/chemistry
- Receptors, Adrenergic, beta-1/genetics
- Receptors, Adrenergic, beta-1/metabolism
- Receptors, G-Protein-Coupled/chemistry
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Neurotensin/chemistry
- Receptors, Neurotensin/genetics
- Receptors, Neurotensin/metabolism
- Single-Chain Antibodies/chemistry
- Single-Chain Antibodies/metabolism
- Substrate Specificity
- Turkeys
Collapse
Affiliation(s)
- Hsin-Yung Yen
- Chemical Research Laboratory, University of Oxford, Oxford, UK
- OMass Technologies, Kidlington, UK
| | - Kin Kuan Hoi
- Chemical Research Laboratory, University of Oxford, Oxford, UK
| | - Idlir Liko
- Chemical Research Laboratory, University of Oxford, Oxford, UK
- OMass Technologies, Kidlington, UK
| | - George Hedger
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | - Wanling Song
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Di Wu
- Chemical Research Laboratory, University of Oxford, Oxford, UK
| | - Philipp Heine
- Biochemisches Institut, Universität Zürich, Zurich, Switzerland
| | - Tony Warne
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Yang Lee
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Byron Carpenter
- MRC Laboratory of Molecular Biology, Cambridge, UK
- Warwick Integrative Synthetic Biology Centre, School of Life Sciences, The University of Warwick, Coventry, UK
| | | | | | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, UK.
| | | |
Collapse
|
19
|
Abstract
Diabetes is a major risk factor for the development of heart failure. One of the hallmarks of diabetes is insulin resistance associated with hyperinsulinemia. The literature shows that insulin and adrenergic signaling is intimately linked to each other; however, whether and how insulin may modulate cardiac adrenergic signaling and cardiac function remains unknown. Notably, recent studies have revealed that insulin receptor and β2 adrenergic receptor (β2AR) forms a membrane complex in animal hearts, bringing together the direct contact between 2 receptor signaling systems, and forming an integrated and dynamic network. Moreover, insulin can drive cardiac adrenergic desensitization via protein kinase A and G protein-receptor kinases phosphorylation of the β2AR, which compromises adrenergic regulation of cardiac contractile function. In this review, we will explore the current state of knowledge linking insulin and G protein-coupled receptor signaling, especially β-adrenergic receptor signaling in the heart, with emphasis on molecular insights regarding its role in diabetic cardiomyopathy.
Collapse
|
20
|
Eibl G, Rozengurt E. KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol 2017; 54:50-62. [PMID: 29079305 DOI: 10.1016/j.semcancer.2017.10.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/22/2017] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be a lethal disease with no efficacious treatment modalities. The incidence of PDAC is expected to increase, at least partially because of the obesity epidemic. Increased efforts to prevent or intercept this disease are clearly needed. Mutations in KRAS are initiating events in pancreatic carcinogenesis supported by genetically engineered mouse models of the disease. However, oncogenic KRAS is not entirely sufficient for the development of fully invasive PDAC. Additional genetic mutations and/or environmental, nutritional, and metabolic stressors, e.g. inflammation and obesity, are required for efficient PDAC formation with activation of KRAS downstream effectors. Multiple factors "upstream" of KRAS associated with obesity, including insulin resistance, inflammation, changes in gut microbiota and GI peptides, can enhance/modulate downstream signals. Multiple signaling networks and feedback loops "downstream" of KRAS have been described that respond to obesogenic diets. We propose that KRAS mutations potentiate a signaling network that is promoted by environmental factors. Specifically, we envisage that KRAS mutations increase the intensity and duration of the growth-promoting signaling network. As the transcriptional activator YAP plays a critical role in the network, we conclude that the rationale for targeting the network (at different points), e.g. with FDA approved drugs such as statins and metformin, is therefore compelling.
Collapse
Affiliation(s)
- Guido Eibl
- Departments of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, CA, United States.
| | - Enrique Rozengurt
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
21
|
Alfonzo-Méndez MA, Alcántara-Hernández R, García-Sáinz JA. Novel Structural Approaches to Study GPCR Regulation. Int J Mol Sci 2016; 18:E27. [PMID: 28025563 PMCID: PMC5297662 DOI: 10.3390/ijms18010027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Upon natural agonist or pharmacological stimulation, G protein-coupled receptors (GPCRs) are subjected to posttranslational modifications, such as phosphorylation and ubiquitination. These posttranslational modifications allow protein-protein interactions that turn off and/or switch receptor signaling as well as trigger receptor internalization, recycling or degradation, among other responses. Characterization of these processes is essential to unravel the function and regulation of GPCR. METHODS In silico analysis and methods such as mass spectrometry have emerged as novel powerful tools. Both approaches have allowed proteomic studies to detect not only GPCR posttranslational modifications and receptor association with other signaling macromolecules but also to assess receptor conformational dynamics after ligand (agonist/antagonist) association. RESULTS this review aims to provide insights into some of these methodologies and to highlight how their use is enhancing our comprehension of GPCR function. We present an overview using data from different laboratories (including our own), particularly focusing on free fatty acid receptor 4 (FFA4) (previously known as GPR120) and α1A- and α1D-adrenergic receptors. From our perspective, these studies contribute to the understanding of GPCR regulation and will help to design better therapeutic agents.
Collapse
Affiliation(s)
- Marco A Alfonzo-Méndez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| | - Rocío Alcántara-Hernández
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| | - J Adolfo García-Sáinz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| |
Collapse
|
22
|
Histamine prevents radiation-induced mesenchymal changes in breast cancer cells. Pharmacol Res 2016; 111:731-739. [DOI: 10.1016/j.phrs.2016.07.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/09/2016] [Accepted: 07/25/2016] [Indexed: 11/19/2022]
|
23
|
Atef ME, Anand-Srivastava MB. Role of PKCδ in Enhanced Expression of Gqα/PLCβ1 Proteins and VSMC Hypertrophy in Spontaneously Hypertensive Rats. PLoS One 2016; 11:e0157955. [PMID: 27379421 PMCID: PMC4933357 DOI: 10.1371/journal.pone.0157955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 06/06/2016] [Indexed: 02/07/2023] Open
Abstract
Gqα signaling has been implicated in cardiac hypertrophy. In addition, angiotensin II (Ang II) was also shown to induce its hypertrophic effect through Gqα and PKCδ activation. We recently showed the role of enhanced expression of Gqα/PLCβ1 proteins in vascular smooth muscle cell (VSMC) hypertrophy, however, the role of PKCδ in VSMC hypertrophy in animal model is still lacking. The present study was therefore undertaken to examine the role of PKCδ and the associated signaling mechanisms in VSMC hypertrophy using 16-week-old spontaneously hypertensive rats (SHR). VSMC from 16-week-old SHR exhibited enhanced phosphorylation of PKCδ-Tyr311 and increased protein synthesis, marker of hypertrophy, as compared to WKY rats which was attenuated by rottlerin, an inhibitor of PKCδ. In addition, knocking down of PKCδ by PKCδ-siRNA also attenuated enhanced protein synthesis in VSMC from SHR. Furthermore, rottlerin attenuated the increased production of superoxide anion, NAD(P)H oxidase activity, increased expression of Gqα, phospholipase C (PLC)β1, insulin like growth factor-1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) proteins in VSMC from SHR. In addition, the enhanced phosphorylation of c-Src, PKCδ-Tyr311, IGF-1R, EGFR and ERK1/2 exhibited by VSMC from SHR was also attenuated by rottlerin. These results suggest that VSMC from SHR exhibit enhanced activity of PKCδ and that PKCδ is the upstream molecule of reactive oxygen species (ROS) and contributes to the enhanced expression of Gqα and PLCβ1 proteins and resultant VSMC hypertrophy involving c-Src, growth factor receptor transactivation and MAP kinase signaling.
Collapse
MESH Headings
- Acetophenones/pharmacology
- Animals
- Benzopyrans/pharmacology
- Blotting, Western
- Cells, Cultured
- Enzyme Inhibitors/pharmacology
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Hypertrophy
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NADPH Oxidases/metabolism
- Phospholipase C beta/metabolism
- Phosphorylation/drug effects
- Protein Kinase C-delta/genetics
- Protein Kinase C-delta/metabolism
- Proto-Oncogene Proteins pp60(c-src)/metabolism
- RNA Interference
- Rats, Inbred SHR
- Rats, Inbred WKY
- Receptors, Growth Factor/metabolism
- Species Specificity
- Superoxides/metabolism
- Tyrosine/genetics
- Tyrosine/metabolism
Collapse
Affiliation(s)
- Mohammed Emehdi Atef
- Department of Molecular and Integrative Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Madhu B. Anand-Srivastava
- Department of Molecular and Integrative Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
24
|
Chen H, Zeng Q, Yao C, Cai Z, Wei T, Huang Z, Su J. Src family tyrosine kinase inhibitors suppress Nav1.1 expression in cultured rat spiral ganglion neurons. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2016; 202:185-93. [PMID: 26790420 DOI: 10.1007/s00359-016-1066-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 12/22/2015] [Accepted: 01/01/2016] [Indexed: 11/25/2022]
Abstract
Src family kinases regulate neuronal voltage-gated Na(+) channels, which generate action potentials. The mechanisms of action, however, remain poorly understood. The aim of the present study was to further elucidate the effects of Src family kinases on Nav1.1 mRNA and protein expression in spiral ganglion neurons. Immunofluorescence staining techniques detected Nav1.1 expression in the spiral ganglion neurons. Additionally, quantitative PCR and western blot techniques were used to analyze Nav1.1 mRNA and protein expression, respectively, in spiral ganglion neurons following exposure to Src family kinase inhibitors PP2 (1 and 10 μM) and SU6656 (0.1 and 1 μM) for different lengths of time (6 and 24 h). In the spiral ganglion neurons, Nav1.1 protein expression was detected in the somas and axons. The Src family kinase inhibitors PP2 and SU6665 significantly decreased Nav1.1 mRNA and protein expression (p < 0.05), respectively, in the spiral ganglion neurons, and changes in expression were not dependent on time or dose (p > 0.05).
Collapse
Affiliation(s)
- Huiying Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qingjiao Zeng
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chen Yao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zheng Cai
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Tingjia Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zhihui Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiping Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
25
|
Banga JP, Moshkelgosha S, Berchner-Pfannschmidt U, Eckstein A. Modeling Graves' Orbitopathy in Experimental Graves' Disease. Horm Metab Res 2015; 47:797-803. [PMID: 26287396 DOI: 10.1055/s-0035-1555956] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Graves' orbitopathy (GO), also known as thyroid eye disease is an inflammatory disease of the orbital tissue of the eye that arises as a consequence of autoimmune thyroid disease. The central feature of the disease is the production of antibodies to the thyrotropin hormone receptor (TSHR) that modulate the function of the receptor leading to autoimmune hyperthyroidism and GO. Over the years, all viable preclinical models of Graves' disease have been incomplete and singularly failed to progress in the treatment of orbital complications. A new mouse model of GO based upon immunogenic presentation of human TSHR A-subunit plasmid by close field electroporation is shown to lead to induction of prolonged functional antibodies to TSHR resulting in chronic disease with subsequent progression to GO. The stable preclinical GO model exhibited pathologies reminiscent of human disease characterized by orbital remodeling by inflammation and adipogenesis. Inflammatory lesions characterized by CD3+ T cells and macrophages were localized in the orbital muscle tissue. This was accompanied by extensive adipogenesis of orbital fat in some immune animals. Surprisingly, other signs of orbital involvement were reminiscent of eyelid inflammation involving chemosis, with dilated and congested orbital blood vessels. More recently, the model is replicated in the author's independent laboratories. The pre-clinical model will provide the basis to study the pathogenic and regulatory roles of immune T and B cells and their subpopulations to understand the initiation, pathophysiology, and progression of GO.
Collapse
Affiliation(s)
- J P Banga
- Faculty of Life Sciences & Medicine, King's College London, The Rayne Institute, London, UK
| | - S Moshkelgosha
- Faculty of Life Sciences & Medicine, King's College London, The Rayne Institute, London, UK
| | | | - A Eckstein
- Department of Ophthalmology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
26
|
Olianas MC, Dedoni S, Onali P. Antidepressants activate the lysophosphatidic acid receptor LPA(1) to induce insulin-like growth factor-I receptor transactivation, stimulation of ERK1/2 signaling and cell proliferation in CHO-K1 fibroblasts. Biochem Pharmacol 2015; 95:311-23. [PMID: 25888927 DOI: 10.1016/j.bcp.2015.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Different lines of evidence indicate that the lysophosphatidic acid (LPA) receptor LPA1 is involved in neurogenesis, synaptic plasticity and anxiety-related behavior, but little is known on whether this receptor can be targeted by neuropsychopharmacological agents. The present study investigated the effects of different antidepressants on LPA1 signaling. We found that in Chinese hamster ovary (CHO)-K1 fibroblasts expressing endogenous LPA1 tricyclic and tetracyclic antidepressants and fluoxetine induced the phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2) and CREB. This response was antagonized by either LPA1 blockade with Ki16425 and AM966 or knocking down LPA1 with siRNA. Antidepressants induced ERK1/2 phosphorylation in human embryonic kidney (HEK)-293 cells overexpressing LPA1, but not in wild-type cells. In PathHunter™ assay measuring receptor-β-arrestin interaction, amitriptyline, mianserin and fluoxetine failed to induce activation of LPA2 and LPA3 stably expressed in CHO-K1 cells. ERK1/2 stimulation by antidepressants and LPA was suppressed by pertussis toxin and inhibition of Src, phosphatidylinositol-3 kinase and insulin-like growth factor-I receptor (IGF-IR) activities. Antidepressants and LPA induced tyrosine phosphorylation of IGF-IR and insulin receptor-substrate-1 through LPA1 and Src. Prolonged exposure of CHO-K1 fibroblasts to either mianserin, mirtazapine or LPA enhanced cell proliferation as indicated by increased [(3)H]-thymidine incorporation and Ki-67 immunofluorescence. This effect was inhibited by blockade of LPA1- and ERK1/2 activity. These data provide evidence that different antidepressants induce LPA1 activation, leading to receptor tyrosine kinase transactivation, stimulation of ERK1/2 signaling and enhanced cell proliferation.
Collapse
Affiliation(s)
- Maria C Olianas
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato Cagliari, Italy
| | - Simona Dedoni
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato Cagliari, Italy
| | - Pierluigi Onali
- Laboratory of Cellular and Molecular Pharmacology, Section of Neurosciences and Clinical Pharmacology, Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato Cagliari, Italy.
| |
Collapse
|
27
|
Yost EA, Hynes TR, Hartle CM, Ott BJ, Berlot CH. Inhibition of G-protein βγ signaling enhances T cell receptor-stimulated interleukin 2 transcription in CD4+ T helper cells. PLoS One 2015; 10:e0116575. [PMID: 25629163 PMCID: PMC4309538 DOI: 10.1371/journal.pone.0116575] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 12/10/2014] [Indexed: 01/23/2023] Open
Abstract
G-protein-coupled receptor (GPCR) signaling modulates the expression of cytokines that are drug targets for immune disorders. However, although GPCRs are common targets for other diseases, there are few GPCR-based pharmaceuticals for inflammation. The purpose of this study was to determine whether targeting G-protein βγ (Gβγ) complexes could provide a useful new approach for modulating interleukin 2 (IL-2) levels in CD4+ T helper cells. Gallein, a small molecule inhibitor of Gβγ, increased levels of T cell receptor (TCR)-stimulated IL-2 mRNA in primary human naïve and memory CD4+ T helper cells and in Jurkat human CD4+ leukemia T cells. Gβ1 and Gβ2 mRNA accounted for >99% of Gβ mRNA, and small interfering RNA (siRNA)-mediated silencing of Gβ1 but not Gβ2 enhanced TCR-stimulated IL-2 mRNA increases. Blocking Gβγ enhanced TCR-stimulated increases in IL-2 transcription without affecting IL-2 mRNA stability. Blocking Gβγ also enhanced TCR-stimulated increases in nuclear localization of nuclear factor of activated T cells 1 (NFAT1), NFAT transcriptional activity, and levels of intracellular Ca2+. Potentiation of IL-2 transcription required continuous Gβγ inhibition during at least two days of TCR stimulation, suggesting that induction or repression of additional signaling proteins during T cell activation and differentiation might be involved. The potentiation of TCR-stimulated IL-2 transcription that results from blocking Gβγ in CD4+ T helper cells could have applications for autoimmune diseases.
Collapse
Affiliation(s)
- Evan A. Yost
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, 17822-2623, United States of America
| | - Thomas R. Hynes
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, 17822-2623, United States of America
| | - Cassandra M. Hartle
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, 17822-2623, United States of America
| | - Braden J. Ott
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, 17822-2623, United States of America
| | - Catherine H. Berlot
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, 17822-2623, United States of America
- * E-mail:
| |
Collapse
|
28
|
Papagiannouli F, Lohmann I. Stage-specific control of stem cell niche architecture in the Drosophila testis by the posterior Hox gene Abd-B. Comput Struct Biotechnol J 2015; 13:122-30. [PMID: 25750700 PMCID: PMC4348433 DOI: 10.1016/j.csbj.2015.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/12/2015] [Accepted: 01/15/2015] [Indexed: 11/30/2022] Open
Abstract
A fundamental question in biology is how complex structures are maintained after their initial specification. We address this question by reviewing the role of the Hox gene Abd-B in Drosophila testis organogenesis, which proceeds through embryonic, larval and pupal stages to reach maturation in adult stages. The data presented in this review highlight a cell- and stage-specific function of Abd-B, since the mechanisms regulating stem cell niche positioning and architecture at different stages seem to be different despite the employment of similar factors. In addition to its described role in the male embryonic gonads, sustained activity of Abd-B in the pre-meiotic germline spermatocytes during larval stages is required to maintain the architecture of the stem cell niche by regulating βPS-integrin localization in the neighboring somatic cyst cells. Loss of Abd-B is associated with cell non-autonomous effects within the niche, leading to a dramatic reduction of pre-meiotic cell populations in adult testes. Identification of Abd-B target genes revealed that Abd-B mediates its effects by controlling the activity of the sevenless ligand Boss via its direct targets Src42A and Sec63. During adult stages, when testis morphogenesis is completed with the addition of the acto-myosin sheath originating from the genital disc, stem cell niche positioning and integrity are regulated by Abd-B activity in the acto-myosin sheath whereas integrin acts in an Abd-B independent way. It seems that the occurrence of new cell types and cell interactions in the course of testis organogenesis made it necessary to adapt the system to the new cellular conditions by reusing the same players for testis stem cell niche positioning in an alternative manner.
Collapse
Affiliation(s)
- Fani Papagiannouli
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany
| | - Ingrid Lohmann
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany
| |
Collapse
|
29
|
Yan C, Du H. Lysosomal acid lipase is critical for myeloid-derived suppressive cell differentiation, development, and homeostasis. World J Immunol 2014; 4:42-51. [DOI: 10.5411/wji.v4.i2.42] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/02/2014] [Accepted: 06/18/2014] [Indexed: 02/05/2023] Open
Abstract
Lysosomal acid lipase (LAL) cleaves cholesteryl esters (CE) and triglycerides (TG) to generate cholesterol and free fatty acid in lysosomes of cells. The downstream metabolic products of fatty acids are ligands for activation of peroxisome proliferator-activated receptor gamma (PPARγ). Accumulation of CEs and TGs is resulted from lack of functional LAL in lysosomes of cells, especially in myeloid cells. One characteristic phenotype in LAL knock-out (lal-/-) mice is systemic elevation of myeloid-derived suppressive cells (MDSCs). MDSCs infiltrate into multiple distal organs, alter T cell development, and suppress T cell proliferation and lymphokine production in lal-/- mice, which lead to severe pathogeneses in multiple organs. The gene transcriptional profile analysis in MDSCs from the bone marrow has identified multiple defects responsible for MDSCs malformation and malfunction in lal-/- mice, including G protein signaling, cell cycles, glycolysis metabolism, mitochondrial bioenergetics, mTOR pathway etc. In a separate gene transcriptional profile analysis in the lung of lal-/- mice, matrix metalloproteinase 12 (MMP12) and apoptosis inhibitor 6 (Api6) are highly overexpressed due to lack of ligand synthesis for PPARγ. PPARγ negatively regulates MMP12 and Api6. Blocking the PPAR signaling by overexpression of a dominant negative PPARγ (dnPPARγ) form, or overexpressing MMP12 or Api6 in myeloid or lung epithelial cells in inducible transgenic mouse models results in elevated MDSCs and inflammation-induced tumorigenesis. These studies demonstrate that LAL and its downstream effectors are critical for MDSCs development, differentiation and malfunction.
Collapse
|
30
|
Rivat C, Sebaihi S, Van Steenwinckel J, Fouquet S, Kitabgi P, Pohl M, Melik Parsadaniantz S, Reaux-Le Goazigo A. Src family kinases involved in CXCL12-induced loss of acute morphine analgesia. Brain Behav Immun 2014; 38:38-52. [PMID: 24263070 DOI: 10.1016/j.bbi.2013.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/30/2013] [Accepted: 11/09/2013] [Indexed: 11/19/2022] Open
Abstract
Functional interactions between the chemokine receptor CXCR4 and opioid receptors have been reported in the brain, leading to a decreased morphine analgesic activity. However the cellular mechanisms responsible for this loss of opioid analgesia are largely unknown. Here we examined whether Src family-kinases (SFK)-linked mechanisms induced by CXCR4 contributed to the loss of acute morphine analgesia and could represent a new physiological anti-opioid signaling pathway. In this way, we showed by immunohistochemistry and western blot that CXCL12 rapidly activated SFK phosphorylation in vitro in primary cultured lumbar rat dorsal root ganglia (DRG) but also in vivo in the DRG and the spinal cord. We showed that SFK activation occurred in a sub population of sensory neurons, in spinal microglia but also in spinal nerve terminals expressing mu-(MOR) and delta-opioid (DOR) receptor. In addition we described that CXCR4 is detected in MOR- and DOR-immunoreactive neurons in the DRG and spinal cord. In vivo, we demonstrated that an intrathecal administration of CXCL12 (1μg) significantly attenuated the subcutaneous morphine (4mg/kg) analgesia. Conversely, pretreatment with a potent CXCR4 antagonist (5μg) significantly enhanced morphine analgesia. Similar effects were obtained after an intrathecal injection of a specific SFK inhibitor, PP2 (10μg). Furthermore, PP2 abrogated CXCL12-induced decrease in morphine analgesia by suppressing SFK activation in the spinal cord. In conclusion, our data highlight that CXCL12-induced loss of acute morphine analgesia is linked to Src family kinases activation.
Collapse
Affiliation(s)
- Cyril Rivat
- Institut des Neurosciences, INSERM U1051, 34091 Montpellier, France
| | - Soumia Sebaihi
- Pain Group, CRICM, Université Pierre et Marie Curie, UMRS 975, Paris, France
| | - Juliette Van Steenwinckel
- Pathophysiology and Neuroprotection in Brain Damage, INSERM UMR 676, Université Paris 7, Hôpital Robert Debré, Paris, France
| | - Stéphane Fouquet
- Centre de Recherche Institut de la Vision, Université Pierre et Marie Curie, INSERM, UMR_S968, CNRS, UMR_7210, Paris, France
| | - Patrick Kitabgi
- Pain Group, CRICM, Université Pierre et Marie Curie, UMRS 975, Paris, France
| | - Michel Pohl
- Centre de Psychiatrie et Neurosciences, INSERM UMR894, Equipe: Douleurs, Neuroinflammation et Stress, 75014 Paris, France
| | - Stéphane Melik Parsadaniantz
- Centre de Recherche Institut de la Vision, Université Pierre et Marie Curie, INSERM, UMR_S968, CNRS, UMR_7210, Paris, France
| | - Annabelle Reaux-Le Goazigo
- Centre de Recherche Institut de la Vision, Université Pierre et Marie Curie, INSERM, UMR_S968, CNRS, UMR_7210, Paris, France.
| |
Collapse
|
31
|
Papagiannouli F, Schardt L, Grajcarek J, Ha N, Lohmann I. The Hox gene Abd-B controls stem cell niche function in the Drosophila testis. Dev Cell 2014; 28:189-202. [PMID: 24480643 DOI: 10.1016/j.devcel.2013.12.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 09/06/2013] [Accepted: 12/23/2013] [Indexed: 01/20/2023]
Abstract
Proper niche architecture is critical for stem cell function, yet only few upstream regulators are known. Here, we report that the Hox transcription factor Abdominal-B (Abd-B), active in premeiotic spermatocytes of Drosophila testes, is essential for positioning the niche to the testis anterior by regulating integrin in neighboring somatic cyst cells. Abd-B also non-cell-autonomously controls critical features within the niche, including centrosome orientation and division rates of germline stem cells. By using genome-wide binding studies, we find that Abd-B mediates its effects on integrin localization by directly controlling at multiple levels the signaling activity of the Sev ligand Boss via its direct targets src42A and sec63, two genes involved in protein trafficking and recycling. Our data show that Abd-B, through local signaling between adjucent cell types, provides positional cues for integrin localization, which is critical for placement of the distant stem cell niche and stem cell activity.
Collapse
Affiliation(s)
- Fani Papagiannouli
- Centre for Organismal Studies (COS) Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany.
| | - Lisa Schardt
- Centre for Organismal Studies (COS) Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany; Deutsches Krebsforschungszentrum (DKFZ), D-69120 Heidelberg, Germany
| | - Janin Grajcarek
- Centre for Organismal Studies (COS) Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany
| | - Nati Ha
- Centre for Organismal Studies (COS) Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany
| | - Ingrid Lohmann
- Centre for Organismal Studies (COS) Heidelberg, Heidelberg University, D-69120 Heidelberg, Germany.
| |
Collapse
|
32
|
Organista-Juárez D, Carretero-Ortega J, Vicente-Fermín O, Vázquez-Victorio G, Sosa-Garrocho M, Vázquez-Prado J, Macías-Silva M, Reyes-Cruz G. Calcium-sensing receptor inhibits TGF-β-signaling by decreasing Smad2 phosphorylation. IUBMB Life 2013; 65:1035-42. [DOI: 10.1002/iub.1232] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 11/12/2013] [Indexed: 12/13/2022]
Affiliation(s)
- Diana Organista-Juárez
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Jorge Carretero-Ortega
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Onasis Vicente-Fermín
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Genaro Vázquez-Victorio
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - José Vázquez-Prado
- Department of Pharmacology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México; México D.F. 04510 Mexico
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology; Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional; Apartado postal 14-740 México D.F. 07000 Mexico
| |
Collapse
|
33
|
On eukaryotic intelligence: signaling system's guidance in the evolution of multicellular organization. Biosystems 2013; 114:8-24. [PMID: 23850535 DOI: 10.1016/j.biosystems.2013.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 06/28/2013] [Accepted: 06/30/2013] [Indexed: 12/11/2022]
Abstract
Communication with the environment is an essential characteristic of the living cell, even more when considering the origins and evolution of multicellularity. A number of changes and tinkering inventions were necessary in the evolutionary transition between prokaryotic and eukaryotic cells, which finally made possible the appearance of genuine multicellular organisms. In the study of this process, however, the transformations experimented by signaling systems themselves have been rarely object of analysis, obscured by other more conspicuous biological traits: incorporation of mitochondria, segregated nucleus, introns/exons, flagellum, membrane systems, etc. Herein a discussion of the main avenues of change from prokaryotic to eukaryotic signaling systems and a review of the signaling resources and strategies underlying multicellularity will be attempted. In the expansion of prokaryotic signaling systems, four main systemic resources were incorporated: molecular tools for detection of solutes, molecular tools for detection of solvent (Donnan effect), the apparatuses of cell-cycle control, and the combined system endocytosis/cytoskeleton. The multiple kinds of enlarged, mixed pathways that emerged made possible the eukaryotic revolution in morphological and physiological complexity. The massive incorporation of processing resources of electro-molecular nature, derived from the osmotic tools counteracting the Donnan effect, made also possible the organization of a computational tissue with huge information processing capabilities: the nervous system. In the central nervous systems of vertebrates, and particularly in humans, neurons have achieved both the highest level of molecular-signaling complexity and the highest degree of information-processing adaptability. Theoretically, it can be argued that there has been an accelerated pace of evolutionary change in eukaryotic signaling systems, beyond the other general novelties introduced by eukaryotic cells in their handling of DNA processes. Under signaling system's guidance, the whole processes of transcription, alternative splicing, mobile elements, and other elements of domain recombination have become closely intertwined and have propelled the differentiation capabilities of multicellular tissues and morphologies. An amazing variety of signaling and self-construction strategies have emerged out from the basic eukaryotic design of multicellular complexity, in millions and millions of new species evolved. This design can also be seen abstractly as a new kind of quasi-universal problem-solving 'engine' implemented at the biomolecular scale-providing the fundamentals of eukaryotic 'intelligence'. Analyzing in depth the problem-solving intelligence of eukaryotic cells would help to establish an integrative panorama of their information processing organization, and of their capability to handle the morphological and physiological complexity associated. Whether an informational updating of the venerable "cell theory" is feasible or not, becomes, at the time being - right in the middle of the massive data deluge/revolution from omic disciplines - a matter to careful consider.
Collapse
|
34
|
Abstract
CONTEXT The use of kinase inhibitors (KIs) in the treatment of cancer has become increasingly common, and practitioners must be familiar with endocrine-related side effects associated with these agents. This review provides an update to the clinician regarding the management of potential endocrinological effects of KIs. EVIDENCE ACQUISITION PubMed was employed to identify relevant manuscripts. A review of the literature was conducted, and data were summarized and incorporated. EVIDENCE SYNTHESIS KIs, including small molecule KIs and monoclonal antibodies directed against kinases, have emerged over the past decade as an important class of anticancer agents. KIs specifically interfere with signaling pathways that are dysregulated in certain types of cancers and also target common mechanisms of growth, invasion, metastasis, and angiogenesis. Currently, at least 20 KIs are approved as cancer therapeutics. However, KIs may affect a broad spectrum of targets and may have additional, unidentified mechanisms of action at the cellular level due to overlap between signaling pathways in the tumor cell and endocrine system. Recent reports in the literature have identified side effects associated with KIs, including alterations in thyroid function, bone metabolism, linear growth, gonadal function, fetal development, adrenal function, and glucose metabolism. CONCLUSIONS Clinicians need to monitor the thyroid functions of patients on KIs. In addition, bone density and vitamin D status should be assessed. Special care should be taken to follow linear growth and development in children taking these agents. Clinicians should counsel patients appropriately on the potential adverse effects of KIs on fetal development.
Collapse
Affiliation(s)
- Maya B Lodish
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, and the Pediatric Endocrinology Inter-Institute Training Program, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
35
|
Alonso V, Friedman PA. Minireview: ubiquitination-regulated G protein-coupled receptor signaling and trafficking. Mol Endocrinol 2013; 27:558-72. [PMID: 23471539 DOI: 10.1210/me.2012-1404] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest and most diverse superfamily of membrane proteins and mediate most cellular responses to hormones and neurotransmitters. Posttranslational modifications are considered the main regulators of all GPCRs. In addition to phosphorylation, glycosylation, and palmitoylation, increasing evidence as reviewed here reveals that ubiquitination also regulates the magnitude and temporospatial aspects of GPCR signaling. Posttranslational protein modification by ubiquitin is a key molecular mechanism governing proteins degradation. Ubiquitination mediates the covalent conjugation of ubiquitin, a highly conserved polypeptide of 76 amino acids, to protein substrates. This process is catalyzed by 3 enzymes acting in tandem: an E1, ubiquitin-activating enzyme; an E2, ubiquitin-carrying enzyme; and an E3, ubiquitin ligase. Ubiquitination is counteracted by deubiquitinating enzymes that deconjugate ubiquitin-modified proteins and rescue the substrate from proteasomal degradation. Although ubiquitination is known to target many GPCRs for lysosomal or proteasomal degradation, emerging findings define novel roles for the basal status of ubiquitination and for rapid deubiquitination and transubiquitination controlling cell surface expression and cellular responsiveness of some GPCRs. In this review, we highlight the classical and novel roles of ubiquitin in the regulation of GPCR function, signaling, and trafficking.
Collapse
Affiliation(s)
- Verónica Alonso
- Institute of Applied Molecular Medicine, San Pablo-CEU University School of Medicine, Madrid, 28668, Spain
| | | |
Collapse
|
36
|
Novel tyrosine kinase signaling pathways: implications in vascular remodeling. Curr Opin Nephrol Hypertens 2012; 21:122-7. [PMID: 22240445 DOI: 10.1097/mnh.0b013e3283503ce9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW This review summarizes the recent advances in molecular mechanisms by which five classes of receptor tyrosine kinases (RTKs) contribute to vascular remodeling. RECENT FINDINGS Recent findings have expanded our knowledge regarding RTK regulation. In particular, G-protein-coupled receptors, mineralocorticoid receptors, mechanical and oxidative stresses transactivate RTKs. These receptors are highly interactive with many downstream targets (including tyrosine kinases and other RTKs) and function as key regulatory nodes in a dynamic signaling network. Interactions between vascular and nonvascular (immune and neuronal) cells are controlled by RTKs in vascular remodeling. Inhibition of RTKs could be an advantageous therapeutic strategy for vascular disorders. SUMMARY RTK-dependent signaling is important for regulation of key functions during vascular remodeling. However, current challenges are related to integration of the data on multiple RTKs in vascular pathology.
Collapse
|
37
|
Kunal SB, Killivalavan A, Medhamurthy R. Involvement of Src family of kinases and cAMP phosphodiesterase in the luteinizing hormone/chorionic gonadotropin receptor-mediated signaling in the corpus luteum of monkey. Reprod Biol Endocrinol 2012; 10:25. [PMID: 22455442 PMCID: PMC3353251 DOI: 10.1186/1477-7827-10-25] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/29/2012] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In higher primates, during non-pregnant cycles, it is indisputable that circulating LH is essential for maintenance of corpus luteum (CL) function. On the other hand, during pregnancy, CL function gets rescued by the LH analogue, chorionic gonadotropin (CG). The molecular mechanisms involved in the control of luteal function during spontaneous luteolysis and rescue processes are not completely understood. Emerging evidence suggests that LH/CGR activation triggers proliferation and transformation of target cells by various signaling molecules as evident from studies demonstrating participation of Src family of tyrosine kinases (SFKs) and MAP kinases in hCG-mediated actions in Leydig cells. Since circulating LH concentration does not vary during luteal regression, it was hypothesized that decreased responsiveness of luteal cells to LH might occur due to changes in LH/CGR expression dynamics, modulation of SFKs or interference with steroid biosynthesis. METHODS Since, maintenance of structure and function of CL is dependent on the presence of functional LH/CGR its expression dynamics as well as mRNA and protein expressions of SFKs were determined throughout the luteal phase. Employing well characterized luteolysis and CL rescue animal models, activities of SFKs, cAMP phosphodiesterase (cAMP-PDE) and expression of SR-B1 (a membrane receptor associated with trafficking of cholesterol ester) were examined. Also, studies were carried out to investigate the mechanisms responsible for decline in progesterone biosynthesis in CL during the latter part of the non-pregnant cycle. RESULTS AND DISCUSSION The decreased responsiveness of CL to LH during late luteal phase could not be accounted for by changes in LH/CGR mRNA levels, its transcript variants or protein. Results obtained employing model systems depicting different functional states of CL revealed increased activity of SFKs [pSrc (Y-416)] and PDE as well as decreased expression of SR-B1 correlating with initiation of spontaneous luteolysis. However, CG, by virtue of its heroic efforts, perhaps by inhibition of SFKs and PDE activation, prevents CL from undergoing regression during pregnancy. CONCLUSIONS The results indicated participation of activated Src and increased activity of cAMP-PDE in the control of luteal function in vivo. That the exogenous hCG treatment caused decreased activation of Src and cAMP-PDE activity with increased circulating progesterone might explain the transient CL rescue that occurs during early pregnancy.
Collapse
Affiliation(s)
- Shah B Kunal
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Asaithambi Killivalavan
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Rudraiah Medhamurthy
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
38
|
Yan C, Ding X, Dasgupta N, Wu L, Du H. Gene profile of myeloid-derived suppressive cells from the bone marrow of lysosomal acid lipase knock-out mice. PLoS One 2012; 7:e30701. [PMID: 22383970 PMCID: PMC3288004 DOI: 10.1371/journal.pone.0030701] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/28/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Lysosomal acid lipase (LAL) controls development and homeostasis of myeloid lineage cells. Loss of the lysosomal acid lipase (LAL) function leads to expansion of myeloid-derived suppressive cells (MDSCs) that cause myeloproliferative neoplasm. METHODOLOGY/PRINCIPAL FINDINGS Affymetrix GeneChip microarray analysis identified detailed intrinsic defects in Ly6G(+) myeloid lineage cells of LAL knock-out (lal-/-) mice. Ingenuity Pathway Analysis revealed activation of the mammalian target of rapamycin (mTOR) signaling, which functions as a nutrient/energy/redox sensor, and controls cell growth, cell cycle entry, cell survival, and cell motility. Loss of the LAL function led to major alteration of large GTPase and small GTPase signal transduction pathways. lal-/- Ly6G(+) myeloid cells in the bone marrow showed substantial increase of cell proliferation in association with up-regulation of cyclin and cyclin-dependent kinase (cdk) genes. The epigenetic microenvironment was significantly changed due to the increased expression of multiple histone cluster genes, centromere protein genes and chromosome modification genes. Gene expression of bioenergetic pathways, including glycolysis, aerobic glycolysis, mitochondrial oxidative phosphorylation, and respiratory chain proteins, was also increased, while the mitochondrial function was impaired in lal-/- Ly6G(+) myeloid cells. The concentration of reactive oxygen species (ROS) was significantly increased accompanied by up-regulation of nitric oxide/ROS production genes in these cells. CONCLUSIONS/SIGNIFICANCE This comprehensive gene profile study for the first time identifies and defines important gene pathways involved in the myeloid lineage cells towards MDSCs using lal-/- mouse model.
Collapse
Affiliation(s)
- Cong Yan
- The Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xinchun Ding
- The Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Nupur Dasgupta
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Lingyan Wu
- The Center for Immunobiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Hong Du
- IU Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| |
Collapse
|
39
|
Butcher AJ, Kong KC, Prihandoko R, Tobin AB. Physiological role of G-protein coupled receptor phosphorylation. Handb Exp Pharmacol 2012:79-94. [PMID: 22222696 DOI: 10.1007/978-3-642-23274-9_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It is now well established that G-protein coupled receptors (GPCRs) are hyper-phosphorylated following agonist occupation usually at serine and threonine residues contained on the third intracellular loop and C-terminal tail. After some 2 decades of intensive research, the nature of protein kinases involved in this process together with the signalling consequences of receptor phosphorylation has been firmly established. The major challenge that the field currently faces is placing all this information within a physiological context and determining to what extent does phosphoregulation of GPCRs impact on whole animal responses. In this chapter, we address this issue by describing how GPCR phosphorylation might vary depending on the cell type in which the receptor is expressed and how this might be employed to drive selective regulation of physiological responses.
Collapse
Affiliation(s)
- Adrian J Butcher
- Department of Cell Physiology and Pharmacology, University of Leicester, Hodgkin Building, Lancaster Road, Leicester, LE1 9HN, UK
| | | | | | | |
Collapse
|
40
|
Olianas MC, Dedoni S, Onali P. δ-Opioid receptors stimulate GLUT1-mediated glucose uptake through Src- and IGF-1 receptor-dependent activation of PI3-kinase signalling in CHO cells. Br J Pharmacol 2011; 163:624-37. [PMID: 21250979 DOI: 10.1111/j.1476-5381.2011.01234.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Although opioids have been reported to affect glucose homeostasis, relatively little is known on the role of δ-opioid receptors. We have investigated the regulation of glucose transport by human δ-opioid receptors expressed in Chinese hamster ovary cells. EXPERIMENTAL APPROACH The uptake of [(3)H]-2-deoxy-D-glucose and 3-O-[methyl-[(3)H]]-D-glucose in response to δ-opioid receptor ligands and the expression of GLUT1, GLUT3 and GLUT4 glucose transporters were examined. Moreover, the effects of intracellular signal transduction inhibitors on δ-opioid receptor-regulated [(3)H]-2-deoxy-D-glucose uptake and protein phosphorylation were investigated. KEY RESULTS Activation of δ-opioid receptors rapidly stimulated [(3)H]-2-deoxy-D-glucose and 3-O-[methyl-[(3)H]]-D-glucose uptakes, which were blocked by the GLUT inhibitors cytochalasin B and phloretin. The stimulation of [(3)H]-2-deoxy-D-glucose uptake that occurred without a change in plasma membrane GLUT1 - required the coupling to G(i) /G(o) proteins - was independent of cAMP and extracellular signal-regulated protein kinases, and was suppressed by blockade of Src and insulin-like growth factor-1 receptor (IGF-1R) tyrosine kinases. Inhibition of phosphatidylinositol 3-kinase (PI3K) by wortmannin or LY294002 and by PI3Kα, but not γ, isoform-selective inhibitors greatly reduced the δ-opioid receptor stimulation of glucose uptake. Moreover, the response was attenuated by overexpressing a dominant-negative kinase-deficient Akt form and by chemical inhibition of Akt. Stimulation of δ-opioid receptors increased protein kinase Cζ/λ (PKCζ/λ) phosphorylation and a selective PKCζ/λ inhibitor slightly reduced opioid stimulation of glucose uptake. CONCLUSIONS AND IMPLICATIONS δ-Opioid receptors stimulated glucose transport probably by enhancing GLUT1 intrinsic activity through a signalling cascade involving G(i)/G(o), Src, IGF-1R, PI3Kα, Akt and, to a minor extent, PKCζ/λ. This effect may contribute to the opioid regulation of glucose homeostasis in physio-pathological conditions.
Collapse
Affiliation(s)
- Maria C Olianas
- Section of Biochemical Pharmacology, Department of Neuroscience, University of Cagliari, Monserrato, Italy
| | | | | |
Collapse
|
41
|
Colín-Santana CC, Avendaño-Vázquez SE, Alcántara-Hernández R, García-Sáinz JA. EGF and angiotensin II modulate lysophosphatidic acid LPA(1) receptor function and phosphorylation state. Biochim Biophys Acta Gen Subj 2011; 1810:1170-7. [PMID: 21914461 DOI: 10.1016/j.bbagen.2011.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 08/23/2011] [Accepted: 08/28/2011] [Indexed: 10/17/2022]
Abstract
BACKGROUND Lysophosphatidic acid (LPA) is a local mediator that exerts its actions through G protein coupled receptors. Knowledge on the regulation of such receptors is scarce to date. Here we show that bidirectional cross-talk exits between LPA(1) and EGF receptors. METHODS C9 cells expressing LPA(1) receptor fussed to the enhanced green fluorescent protein were used. We studied intracellular calcium concentration, Akt/PKB phosphorylation, LPA(1) and EGF receptor phosphorylation. RESULTS EGF diminished LPA-mediated intracellular calcium response and induced LPA(1) receptor phosphorylation, which was sensitive to protein kinase C inhibitors. Angiotensin II and LPA induced EGF receptor transactivation as evidenced by Akt/PKB phosphorylation through metalloproteinase-catalyzed membrane shedding of heparin-binding EGF and autocrine/paracrine activation of EGF receptors. This process was found to be of major importance in angiotensin II-induced LPA(1) receptor phosphorylation. Attempts to define a role for EGF receptor transactivation in homologous LPA(1) receptor desensitization and phosphorylation suggested that G protein-coupled receptor kinases are the major players in this process, overshadowing other events. CONCLUSIONS EGF receptors and LPA(1) receptors are engaged in an intense liaison, in that EGF receptors are capable of modulating LPA(1) receptor function through phosphorylation cascades. EGF transactivation plays a dual role: it mediates some LPA actions, and it modulates LPA(1) receptor function in inhibitory fashion. GENERAL SIGNIFICANCE EGF and LPA receptors coexist in many cell types and play key roles in maintaining the delicate equilibrium that we call health and in the pathogenesis of many diseases. The intense cross-talk described here has important physiological and pathophysiological implications.
Collapse
Affiliation(s)
- Christian C Colín-Santana
- Departmento de Biología Celular y Desarrollo, Instituto de Fisiologíca Celular, Universidad Naxional Autónoma de México, México
| | | | | | | |
Collapse
|
42
|
Perrault R, Wright B, Storie B, Hatherell A, Zahradka P. Tyrosine kinase-independent activation of extracellular-regulated kinase (ERK) 1/2 by the insulin-like growth factor-1 receptor. Cell Signal 2011; 23:739-46. [DOI: 10.1016/j.cellsig.2010.12.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/22/2010] [Indexed: 10/18/2022]
|
43
|
Endothelin-1 stimulation of proteoglycan synthesis in vascular smooth muscle is mediated by endothelin receptor transactivation of the transforming growth factor-[beta] type I receptor. J Cardiovasc Pharmacol 2011; 56:360-8. [PMID: 20625315 DOI: 10.1097/fjc.0b013e3181ee6811] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We utilized human vascular smooth muscle cells to address the question if a G-protein-coupled receptor, the endothelin (ET) receptor, could transactivate a serine/threonine kinase receptor, specifically the transforming growth factor (TGF)-[beta] receptor, T[beta]RI. Functionality of the interaction was addressed by studying endothelin-1-stimulated proteoglycan synthesis. Signaling molecules were assessed by Western blotting and proteoglycan synthesis by [35S]sulfate and 35S-met/cys incorporation and molecular size by SDS-PAGE. Endothelin-1 treatment led to a time- and concentration-dependent increase in cytosolic phosphoSmad2C, which was inhibited by the mixed endothelin receptor antagonist bosentan and the T[beta]RI antagonist SB431542. Endothelin-1 treatment led to a time-dependent increase in nuclear phosphoSmad2C. Endothelin-1-stimulated proteoglycan synthesis was partially inhibited (40%) by SB431542 and completely blocked by bosentan. The effect of endothelin-1 to stimulate an increase in glycosaminoglycan size on biglycan was also blocked in a concentration-dependent manner by SB431542. These data extend the current paradigm of G-protein coupled receptor signaling to include the transactivation of the serine kinase receptor for TGF-[beta] (T[beta]RI). This response should be considered in the context of response to endothelin-1, and the options for therapeutically targeting endothelin-1 are accordingly broadened to include downstream signaling otherwise associated with TGF-[beta] receptor activation.
Collapse
|
44
|
García-Sáinz JA, Romero-Ávila MT, Medina LDC. Dissecting how receptor tyrosine kinases modulate G protein-coupled receptor function. Eur J Pharmacol 2010; 648:1-5. [PMID: 20828551 DOI: 10.1016/j.ejphar.2010.08.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Revised: 08/30/2010] [Accepted: 08/31/2010] [Indexed: 02/06/2023]
Abstract
Receptor tyrosine kinases and G protein-coupled receptors modulate physiological processes and are also involved in the pathogenesis of some diseases. These receptors have intense bidirectional crosstalks leading to interactions in their signaling pathways and also modulation of the receptors themselves. In some cases, the receptor tyrosine kinases phosphorylate G protein-coupled receptors whereas in others phosphoinositide 3-kinase, protein kinase B and protein kinase C are key elements in these crosstalks. Two paracrine/ autocrine processes also participate, i.e., epidermal growth factor transactivation and sphingosine 1-phosphate generation and signaling. G proteins seem to mediate actions of receptor tyrosine kinases, but how this takes place is far from completely understood; some models are presented. Recent data indicate that the mitogen activated protein kinase cascade also mediate crosstalks. In the present perspective these processes are outlined using information from receptors that have been intensively studied, and important gaps in our knowledge are indicated.
Collapse
Affiliation(s)
- J Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, México D.F. 04510.
| | | | | |
Collapse
|
45
|
Abstract
Targeted therapy in oncology consists of drugs that specifically interfere with abnormal signaling pathways that are dysregulated in cancer cells. Tyrosine kinase inhibitors (TKIs) take advantage of unique oncogenes that are activated in certain types of cancer, and also target common mechanisms of growth, invasion, metastasis, and angiogenesis. However, many kinase inhibitors for cancer therapy are somewhat nonselective, and most have additional mechanisms of action at the cellular level, which are not completely understood. The use of these agents has increased our knowledge of important side effects, of which the practicing clinician must be aware. Recently, proposed endocrine-related side effects of these agents include alterations in thyroid function, bone metabolism, linear growth, gonadal function, fetal development, and glucose metabolism, and adrenal function. This review summarizes the most recent data on the endocrine side effects of TKIs.
Collapse
Affiliation(s)
- Maya B Lodish
- Section on Endocrinology Genetics, Program on Developmental Endocrinology Genetics, Eunice Kennedy Shriver NICHD Pediatric Endocrinology Inter-Institute Training Program, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | | |
Collapse
|
46
|
Burch ML, Ballinger ML, Yang SNY, Getachew R, Itman C, Loveland K, Osman N, Little PJ. Thrombin stimulation of proteoglycan synthesis in vascular smooth muscle is mediated by protease-activated receptor-1 transactivation of the transforming growth factor beta type I receptor. J Biol Chem 2010; 285:26798-26805. [PMID: 20571025 DOI: 10.1074/jbc.m109.092767] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growth factors modify the structure of the glycosaminoglycan (GAG) chains on biglycan leading to enhanced LDL binding. G-protein receptor-coupled agonists such as thrombin, signal changes the structure of proteoglycans produced by vascular smooth muscle cells (VSMCs). One component of classical G-protein-coupled receptor (GPCR) signaling invokes transactivation of protein tyrosine kinase receptors such as the epidermal growth factor receptor. Serine/threonine receptor growth factors such as transforming growth factor-(TGF)-beta are potent activators of proteoglycan synthesis. We have used the model of proteoglycan synthesis to demonstrate that the signaling paradigm of GPCR signaling can be extended to include the transactivation of serine/threonine receptor, specifically the TGF-beta type I receptor (TbetaRI) also known as activin-like kinase (ALK) V. Thrombin stimulated elongation of GAG chains and increased proteoglycan core protein expression and these responses were blocked by the TbetaRI antagonist, SB431542 and TbetaRI siRNA knockdown, as well as several protease-activated receptor (PAR)-1 antagonists. The canonical downstream response to TGF-beta is increased C-terminal phosphorylation of the transcription factor Smad2 generating phospho-Smad2C (phosphorylation of Smad2 C-terminal region). Thrombin stimulated increased phospho-Smad2C levels, and the response was blocked by SB431542 and JNJ5177094. The proteolytically inactive thrombin mimetic thrombin-receptor activating peptide also stimulated an increase in cytosolic phospho-Smad2C. Signaling pathways for growth factor regulated proteoglycan synthesis represent therapeutic targets for the prevention of atherosclerosis, but the novel finding of a GPCR-mediated transactivation of a serine/threonine growth factor receptor almost certainly has implications well beyond the synthesis of proteoglycans.
Collapse
Affiliation(s)
- Micah L Burch
- Diabetes and Cell Biology Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, 3004 Victoria, Australia; Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Hospital, Monash University, Melbourne, 3004 Victoria, Australia
| | - Mandy L Ballinger
- Diabetes and Cell Biology Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, 3004 Victoria, Australia
| | - Sundy N Y Yang
- Diabetes and Cell Biology Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, 3004 Victoria, Australia
| | - Robel Getachew
- Diabetes and Cell Biology Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, 3004 Victoria, Australia
| | - Catherine Itman
- Departments of Biochemistry & Molecular Biology and Anatomy & Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Kate Loveland
- Departments of Biochemistry & Molecular Biology and Anatomy & Developmental Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Narin Osman
- Diabetes and Cell Biology Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, 3004 Victoria, Australia; Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Hospital, Monash University, Melbourne, 3004 Victoria, Australia
| | - Peter J Little
- Diabetes and Cell Biology Laboratory, BakerIDI Heart and Diabetes Institute, Melbourne, 3004 Victoria, Australia; Departments of Medicine and Immunology, Central and Eastern Clinical School, Alfred Hospital, Monash University, Melbourne, 3004 Victoria, Australia.
| |
Collapse
|
47
|
Distrutti E, Cipriani S, Renga B, Mencarelli A, Migliorati M, Cianetti S, Fiorucci S. Hydrogen sulphide induces micro opioid receptor-dependent analgesia in a rodent model of visceral pain. Mol Pain 2010; 6:36. [PMID: 20540729 PMCID: PMC2908066 DOI: 10.1186/1744-8069-6-36] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 06/11/2010] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Hydrogen sulphide (H2S) is a gaseous neuro-mediator that exerts analgesic effects in rodent models of visceral pain by activating KATP channels. A body of evidence support the notion that KATP channels interact with endogenous opioids. Whether H2S-induced analgesia involves opioid receptors is unknown. METHODS The perception of painful sensation induced by colorectal distension (CRD) in conscious rats was measured by assessing the abdominal withdrawal reflex. The contribution of opioid receptors to H2S-induced analgesia was investigated by administering rats with selective mu, kappa and delta opioid receptor antagonists and antisenses. To investigate whether H2S causes mu opioid receptor (MOR) transactivation, the neuronal like cells SKNMCs were challenged with H2S in the presence of MOR agonist (DAMGO) or antagonist (CTAP). MOR activation and phosphorylation, its association to beta arrestin and internalization were measured. RESULTS H2S exerted a potent analgesic effects on CRD-induced pain. H2S-induced analgesia required the activation of the opioid system. By pharmacological and molecular analyses, a robust inhibition of H2S-induced analgesia was observed in response to central administration of CTAP and MOR antisense, while kappa and delta receptors were less involved. H2S caused MOR transactivation and internalization in SKNMCs by a mechanism that required AKT phosphorylation. MOR transactivation was inhibited by LY294002, a PI3K inhibitor, and glibenclamide, a KATP channels blocker. CONCLUSIONS This study provides pharmacological and molecular evidence that antinociception exerted by H2S in a rodent model of visceral pain is modulated by the transactivation of MOR. This observation provides support for development of new pharmacological approaches to visceral pain.
Collapse
Affiliation(s)
- Eleonora Distrutti
- S.C. di Gastroenterologia, Azienda Ospedaliera di Perugia, Perugia, Italia.
| | | | | | | | | | | | | |
Collapse
|
48
|
Strachan RT, Allen JA, Sheffler DJ, Roth BL. p90 Ribosomal S6 kinase 2, a novel GPCR kinase, is required for growth factor-mediated attenuation of GPCR signaling. Biochemistry 2010; 49:2657-71. [PMID: 20136148 DOI: 10.1021/bi901921k] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The 5-hydroxytryptamine 2A (5-HT(2A)) receptor is a member of the G protein-coupled receptor superfamily (GPCR) and plays a key role in transducing a variety of cellular signals elicited by serotonin (5-HT; 5-hydroxytryptamine) in both peripheral and central tissues. Recently, we discovered that the ERK/MAPK effector p90 ribosomal S6 kinase 2 (RSK2) phosphorylates the 5-HT(2A) receptor and attenuates 5-HT(2A) receptor signaling. This raised the intriguing possibility of a regulatory paradigm whereby receptor tyrosine kinases (RTKs) attenuate GPCR signaling (i.e., "inhibitory cross-talk") by activating RSK2 [Strachan et al. (2009) J. Biol. Chem. 284, 5557-5573]. We report here that activation of multiple endogenous RTKs such as the epidermal growth factor receptor (EGFR), the platelet-derived growth factor receptor (PDGFR), and ErbB4 significantly attenuates 5-HT(2A) receptor signaling in a variety of cell types including mouse embryonic fibroblasts (MEFs), mouse vascular smooth muscle cells (mVSMCs), and primary cortical neurons. Importantly, genetic deletion of RSK2 completely prevented signal attenuation, thereby suggesting that RSK2 is a critical mediator of inhibitory cross-talk between RTKs and 5-HT(2A) receptors. We also discovered that P2Y purinergic receptor signaling was similarly attenuated following EGFR activation. By directly testing multiple endogenous growth factors/RTK pathways and multiple Gq-coupled GPCRs, we have now established a cellular mechanism whereby RTK signaling cascades act via RSK2 to attenuate GPCR signaling. Given the pervasiveness of growth factor signaling, this novel regulatory mechanism has the potential to explain how 5-HT(2A) receptors are regulated in vivo, with potential implications for human diseases in which 5-HT(2A) or RTK activity is altered (e.g., neuropsychiatric and neurodevelopmental disorders).
Collapse
Affiliation(s)
- Ryan T Strachan
- Department of Biochemistry, Case Western Reserve University Medical School, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
49
|
Shi M, Liu D, Duan H, Qian L, Wang L, Niu L, Zhang H, Yong Z, Gong Z, Song L, Yu M, Hu M, Xia Q, Shen B, Guo N. The β2-adrenergic receptor and Her2 comprise a positive feedback loop in human breast cancer cells. Breast Cancer Res Treat 2010; 125:351-62. [PMID: 20237834 DOI: 10.1007/s10549-010-0822-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 02/24/2010] [Indexed: 01/16/2023]
Abstract
In this study, β2-AR level was found to be up-regulated in MCF-7 cells overexpressing Her2 (MCF-7/Her2). Correlation of β2-AR level with Her2 status was demonstrated in breast cancer tissue samples. Constitutive phosphorylation of ERK, mRNA expression up-regulation of catecholamine-synthesis enzymes, and increased epinephrine release were detected in MCF-7/Her2 cells. β2-AR expression induced by epinephrine and involvement of ERK signaling were validated. The data indicate that Her2 overexpression and excessive phosphorylation of ERK cause epinephrine autocrine release from breast cancer cells, resulting in up-regulation of β2-AR expression. The data also showed that catecholamine prominently stimulated Her2 mRNA expression and promoter activity. The activation and nuclear translocation of STAT3 triggered by isoproterenol were observed. Enhanced binding activities of STAT3 to the Her2 promoter after isoproterenol stimulation were verified. Using STAT3 shRNA and dominant negative STAT3 mutant, the role of STAT3 in isoproterenol-induced Her2 expression was further confirmed. The data support a model where β2-AR and Her2 comprise a positive feedback loop in human breast cancer cells.
Collapse
Affiliation(s)
- Ming Shi
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Cruse G, Yang W, Duffy SM, Chachi L, Leyland M, Amrani Y, Bradding P. Counterregulation of beta(2)-adrenoceptor function in human mast cells by stem cell factor. J Allergy Clin Immunol 2009; 125:257-63.e1-5. [PMID: 19864009 DOI: 10.1016/j.jaci.2009.08.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 07/10/2009] [Accepted: 08/05/2009] [Indexed: 02/01/2023]
Abstract
BACKGROUND Mast cells contribute to the pathophysiology of asthma with the sustained release of both preformed and newly generated mediators in response to allergens and other diverse stimuli. Stem cell factor (SCF) is the key human mast cell growth factor, but also primes mast cells for mediator release. SCF expression is markedly increased in asthmatic airways. Short-acting beta(2)-adrenoceptor drugs such as albuterol inhibit human lung mast cell (HLMC) degranulation in vitro in the absence of SCF, but their effect in the presence of SCF is not known. OBJECTIVE The aim of this study was to elucidate the effects of albuterol on HLMC function in the presence of SCF. METHODS Mediator release and K(Ca)3.1 ion channel activity were analyzed in purified HLMC. Intracellular signalling and beta(2)-adrenoceptor phosphorylation and internalization were analyzed in the HMC-1 human mast cell line. RESULTS beta(2)-Adrenoceptor agonist-dependent inhibition of K(Ca)3.1 ion channels and HLMC mediator release was markedly attenuated in the presence of SCF. Remarkably, albuterol actually potentiated IgE-induced histamine release in a dose-dependent manner when both SCF and IgE were present. These effects were related to the SCF-dependent phosphorylation of Tyr350 on the beta(2)-adrenoceptor with immediate uncoupling of the receptor followed by receptor internalization. CONCLUSION The potentially beneficial effects of beta(2)-adrenoceptor agonists in asthmatic airways may be blunted as a result of the high concentrations of SCF present.
Collapse
Affiliation(s)
- Glenn Cruse
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|