1
|
Stefan K, Namasivayam V, Stefan SM. Computer-aided pattern scoring - A multitarget dataset-driven workflow to predict ligands of orphan targets. Sci Data 2024; 11:530. [PMID: 38783061 PMCID: PMC11116543 DOI: 10.1038/s41597-024-03343-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
The identification of lead molecules and the exploration of novel pharmacological drug targets are major challenges of medical life sciences today. Genome-wide association studies, multi-omics, and systems pharmacology steadily reveal new protein networks, extending the known and relevant disease-modifying proteome. Unfortunately, the vast majority of the disease-modifying proteome consists of 'orphan targets' of which intrinsic ligands/substrates, (patho)physiological roles, and/or modulators are unknown. Undruggability is a major challenge in drug development today, and medicinal chemistry efforts cannot keep up with hit identification and hit-to-lead optimization studies. New 'thinking-outside-the-box' approaches are necessary to identify structurally novel and functionally distinctive ligands for orphan targets. Here we present a unique dataset that includes critical information on the orphan target ABCA1, from which a novel cheminformatic workflow - computer-aided pattern scoring (C@PS) - for the identification of novel ligands was developed. Providing a hit rate of 95.5% and molecules with high potency and molecular-structural diversity, this dataset represents a suitable template for general deorphanization studies.
Collapse
Affiliation(s)
- Katja Stefan
- University of Oslo and Oslo University Hospital, Department of Pathology, Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
- University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck Institute of Experimental Dermatology, Medical Systems Biology Division, Medicinal Chemistry and Systems Polypharmacology, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Vigneshwaran Namasivayam
- University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck Institute of Experimental Dermatology, Medical Systems Biology Division, Medicinal Chemistry and Systems Polypharmacology, Ratzeburger Allee 160, 23538, Lübeck, Germany.
- University of Bonn, Pharmaceutical Institute, Department of Pharmaceutical and Cellbiological Chemistry, An der Immenburg 4, 53121, Bonn, Germany.
| | - Sven Marcel Stefan
- University of Oslo and Oslo University Hospital, Department of Pathology, Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway.
- University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck Institute of Experimental Dermatology, Medical Systems Biology Division, Medicinal Chemistry and Systems Polypharmacology, Ratzeburger Allee 160, 23538, Lübeck, Germany.
- Medical University of Lublin, Department of Biopharmacy, Chodzki 4a, 20-093, Lublin, Poland.
| |
Collapse
|
2
|
Adamczuk K, Ngo TH, Czapiński J, Rivero-Müller A. Glycoprotein-glycoprotein Receptor Binding Detection Using Bioluminescence Resonance Energy Transfer. Endocrinology 2024; 165:bqae052. [PMID: 38679471 DOI: 10.1210/endocr/bqae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
The glycoprotein receptors, members of the large G protein-coupled receptor family, are characterized by a large extracellular domains responsible for binding their glycoprotein hormones. Hormone-receptor interactions are traditionally analyzed by ligand-binding assays, most often using radiolabeling but also by thermal shift assays. Despite their high sensitivity, these assays require appropriate laboratory conditions and, often, purified plasma cell membranes, which do not provide information on receptor localization or activity because the assays typically focus on measuring binding only. Here, we apply bioluminescence resonance energy transfer in living cells to determine hormone-receptor interactions between a Gaussia luciferase (Gluc)-luteinizing hormone/chorionic gonadotropin receptor (LHCGR) fusion and its ligands (human chorionic gonadotropin or LH) fused to the enhanced green fluorescent protein. The Gluc-LHCGR, as well as other Gluc-G protein-coupled receptors such as the somatostatin and the C-X-C motif chemokine receptors, is expressed on the plasma membrane, where luminescence activity is equal to membrane receptor expression, and is fully functional. The chimeric enhanced green fluorescent protein-ligands are properly secreted from cells and able to bind and activate the wild-type LHCGR as well as the Gluc-LHCGR. Finally, bioluminescence resonance energy transfer was used to determine the interactions between clinically relevant mutations of the hormones and the LHCGR that show that this bioassay provides a fast and effective, safe, and cost-efficient tool to assist the molecular characterization of mutations in either the receptor or ligand and that it is compatible with downstream cellular assays to determine receptor activation/function.
Collapse
Affiliation(s)
- Kamila Adamczuk
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Thu Ha Ngo
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Jakub Czapiński
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
3
|
Paramonov VM, Gerstenberg M, Sahlgren C, Lindén M, Rivero-Müller A. In vitro Targetability Validation of Peptide-Functionalized Mesoporous Silica Nanoparticles in the Presence of Serum Proteins. Front Chem 2020; 8:603616. [PMID: 33282845 PMCID: PMC7691633 DOI: 10.3389/fchem.2020.603616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/12/2020] [Indexed: 11/13/2022] Open
Abstract
Demonstration of receptor-mediated targeting of nanoparticles to specific organs and/or cell types is an integral aim in many bionanomedicine development projects. However, engagement of targeted receptors with ligands on nanocarriers, which is the cornerstone of the active targeting concept, is challenging to study under biologically relevant conditions and thus often stays overlooked. In this work, we utilize an in-house established bioassay for in vitro targetability validation of mesoporous silica nanoparticles (MSNs), functionalized with high-affinity peptide ligands to somatostatin receptors via protective group chemistry, ensuring the correct orientation of the peptide's pharmacophore. We demonstrate that targeted nanoparticles, but not scrambled peptide-decorated counterparts, specifically engage the targeted receptors in living cells in culture media containing serum protein. The importance of being able to exclude false positives originating from the premature detachment of targeting peptides from the MSNs is highlighted.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | | | - Cecilia Sahlgren
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mika Lindén
- Department of Inorganic Chemistry II, Ulm University, Ulm, Germany
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
4
|
Paramonov VM, Sahlgren C, Rivero-Müller A, Pulliainen AT. iGIST-A Kinetic Bioassay for Pertussis Toxin Based on Its Effect on Inhibitory GPCR Signaling. ACS Sens 2020; 5:3438-3448. [PMID: 33147407 PMCID: PMC7706119 DOI: 10.1021/acssensors.0c01340] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Detection of pertussis toxin (PTX) activity is instrumental for the development and manufacturing of pertussis vaccines. These quality and safety measures require thousands of mice annually. Here, we describe Interference in Gαi-mediated Signal Transduction (iGIST), an animal-free kinetic bioassay for detection of PTX, by measuring its effect on inhibitory G protein-coupled receptor (GPCR) signaling. PTX ADP-ribosylates inhibitory α-subunits of the heterotrimeric G proteins, thereby perturbing the inhibitory GPCR signaling. iGIST is based on HEK293 cells coexpressing a somatostatin receptor 2 (SSTR2), which is an inhibitory GPCR controllable by a high-affinity agonist octreotide; and a luminescent 3'5'-cyclic adenosine monophosphate (cAMP) probe. iGIST has a low sensitivity threshold in the pg/mL range of PTX, surpassing by 100-fold in a parallel analysis the currently used in vitro end-point technique to detect PTX, the cluster formation assay (CFA) in Chinese hamster ovary cells. iGIST also detects PTX in complex samples, i.e., a commercial PTX-toxoid-containing pertussis vaccine that was spiked with an active PTX. iGIST has an objective digital readout and is observer independent, offering prospects for automation. iGIST emerges as a promising animal-free alternative to detect PTX activity in the development and manufacturing of pertussis vaccines. iGIST is also expected to facilitate basic PTX research, including identification and characterization of novel compounds interfering with PTX.
Collapse
Affiliation(s)
- Valeriy M. Paramonov
- Institute of Biomedicine, Research Unit for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, FI-20500 Turku, Finland
| | - Cecilia Sahlgren
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, FI-20500 Turku, Finland
- Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Unit for Integrative Physiology and Pharmacology, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Arto T. Pulliainen
- Institute of Biomedicine, Research Unit for Infection and Immunity, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| |
Collapse
|
5
|
High-Throughput Fluorescence Assays for Ion Channels and GPCRs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:27-72. [DOI: 10.1007/978-3-030-12457-1_3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
6
|
Casarini L, Santi D, Brigante G, Simoni M. Two Hormones for One Receptor: Evolution, Biochemistry, Actions, and Pathophysiology of LH and hCG. Endocr Rev 2018; 39:549-592. [PMID: 29905829 DOI: 10.1210/er.2018-00065] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/08/2018] [Indexed: 01/03/2023]
Abstract
LH and chorionic gonadotropin (CG) are glycoproteins fundamental to sexual development and reproduction. Because they act on the same receptor (LHCGR), the general consensus has been that LH and human CG (hCG) are equivalent. However, separate evolution of LHβ and hCGβ subunits occurred in primates, resulting in two molecules sharing ~85% identity and regulating different physiological events. Pituitary, pulsatile LH production results in an ~90-minute half-life molecule targeting the gonads to regulate gametogenesis and androgen synthesis. Trophoblast hCG, the "pregnancy hormone," exists in several isoforms and glycosylation variants with long half-lives (hours) and angiogenic potential and acts on luteinized ovarian cells as progestational. The different molecular features of LH and hCG lead to hormone-specific LHCGR binding and intracellular signaling cascades. In ovarian cells, LH action is preferentially exerted through kinases, phosphorylated extracellular-regulated kinase 1/2 (pERK1/2) and phosphorylated AKT (also known as protein kinase B), resulting in irreplaceable proliferative/antiapoptotic signals and partial agonism on progesterone production in vitro. In contrast, hCG displays notable cAMP/protein kinase A (PKA)-mediated steroidogenic and proapoptotic potential, which is masked by estrogen action in vivo. In vitro data have been confirmed by a large data set from assisted reproduction, because the steroidogenic potential of hCG positively affects the number of retrieved oocytes, and LH affects the pregnancy rate (per oocyte number). Leydig cell in vitro exposure to hCG results in qualitatively similar cAMP/PKA and pERK1/2 activation compared with LH and testosterone. The supposed equivalence of LH and hCG has been disproved by such data, highlighting their sex-specific functions and thus deeming it an oversight caused by incomplete understanding of clinical data.
Collapse
Affiliation(s)
- Livio Casarini
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniele Santi
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Giulia Brigante
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy.,Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria, Modena, Italy
| |
Collapse
|
7
|
Paramonov VM, Desai D, Kettiger H, Mamaeva V, Rosenholm JM, Sahlgren C, Rivero-Müller A. Targeting Somatostatin Receptors By Functionalized Mesoporous Silica Nanoparticles - Are We Striking Home? Nanotheranostics 2018; 2:320-346. [PMID: 30148051 PMCID: PMC6107779 DOI: 10.7150/ntno.23826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/25/2018] [Indexed: 12/02/2022] Open
Abstract
The concept of delivering nanoformulations to desired tissues by means of targeting membrane receptors of high local abundance by ligands anchored to the nanocarrier has gained a lot of attention over the last decade. Currently, there is no unanimous opinion on whether surface functionalization of nanocarriers by targeting ligands translates into any real benefit in terms of pharmacokinetics or treatment outcomes. Having examined the published nanocarriers designed to engage with somatostatin receptors, we realized that in the majority of cases targetability claims were not supported by solid evidence of targeting ligand-targeted receptor coupling, which is the very crux of a targetability concept. Here, we present an approach to characterize targetability of mesoporous silica-based nanocarriers functionalized with ligands of somatostatin receptors. The targetability proof in our case comes from a functional assay based on a genetically-encoded cAMP probe, which allows for real-time capture of receptor activation in living cells, triggered by targeting ligands on nanoparticles. We elaborate on the development and validation of the assay, highlighting the power of proper functional tests in the characterization pipeline of targeted nanoformulations.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Helene Kettiger
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Veronika Mamaeva
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Cecilia Sahlgren
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Poland
| |
Collapse
|
8
|
Szymańska K, Kałafut J, Przybyszewska A, Paziewska B, Adamczuk G, Kiełbus M, Rivero-Müller A. FSHR Trans-Activation and Oligomerization. Front Endocrinol (Lausanne) 2018; 9:760. [PMID: 30619090 PMCID: PMC6301190 DOI: 10.3389/fendo.2018.00760] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/30/2018] [Indexed: 12/12/2022] Open
Abstract
Follicle stimulating hormone (FSH) plays a key role in human reproduction through, among others, induction of spermatogenesis in men and production of estrogen in women. The function FSH is performed upon binding to its cognate receptor-follicle-stimulating hormone receptor (FSHR) expressed on the surface of target cells (granulosa and Sertoli cells). FSHR belongs to the family of G protein-coupled receptors (GPCRs), a family of receptors distinguished by the presence of various signaling pathway activation as well as formation of cross-talking aggregates. Until recently, it was claimed that the FSHR occurred naturally as a monomer, however, the crystal structure as well as experimental evidence have shown that FSHR both self-associates and forms heterodimers with the luteinizing hormone/chorionic gonadotropin receptor-LHCGR. The tremendous gain of knowledge is also visible on the subject of receptor activation. It was once thought that activation occurs only as a result of ligand binding to a particular receptor, however there is mounting evidence of trans-activation as well as biased signaling between GPCRs. Herein, we describe the mechanisms of aforementioned phenomena as well as briefly describe important experiments that contributed to their better understanding.
Collapse
Affiliation(s)
- Kamila Szymańska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Alicja Przybyszewska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Beata Paziewska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Medical University of Lublin, Lublin, Poland
| | - Michał Kiełbus
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| | - Adolfo Rivero-Müller
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
- *Correspondence: Adolfo Rivero-Müller ;
| |
Collapse
|
9
|
Prashanth JR, Hasaballah N, Vetter I. Pharmacological screening technologies for venom peptide discovery. Neuropharmacology 2017; 127:4-19. [PMID: 28377116 DOI: 10.1016/j.neuropharm.2017.03.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/11/2017] [Accepted: 03/31/2017] [Indexed: 01/13/2023]
Abstract
Venomous animals occupy one of the most successful evolutionary niches and occur on nearly every continent. They deliver venoms via biting and stinging apparatuses with the aim to rapidly incapacitate prey and deter predators. This has led to the evolution of venom components that act at a number of biological targets - including ion channels, G-protein coupled receptors, transporters and enzymes - with exquisite selectivity and potency, making venom-derived components attractive pharmacological tool compounds and drug leads. In recent years, plate-based pharmacological screening approaches have been introduced to accelerate venom-derived drug discovery. A range of assays are amenable to this purpose, including high-throughput electrophysiology, fluorescence-based functional and binding assays. However, despite these technological advances, the traditional activity-guided fractionation approach is time-consuming and resource-intensive. The combination of screening techniques suitable for miniaturization with sequence-based discovery approaches - supported by advanced proteomics, mass spectrometry, chromatography as well as synthesis and expression techniques - promises to further improve venom peptide discovery. Here, we discuss practical aspects of establishing a pipeline for venom peptide drug discovery with a particular emphasis on pharmacology and pharmacological screening approaches. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Jutty Rajan Prashanth
- Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd, The University of Queensland, St Lucia, Qld 4072, Australia
| | - Nojod Hasaballah
- Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd, The University of Queensland, St Lucia, Qld 4072, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, 306 Carmody Rd, The University of Queensland, St Lucia, Qld 4072, Australia; School of Pharmacy, 20 Cornwall St, Woolloongabba, Qld 4102, Australia.
| |
Collapse
|
10
|
Potorac I, Rivero-Müller A, Trehan A, Kiełbus M, Jozwiak K, Pralong F, Hafidi A, Thiry A, Ménagé JJ, Huhtaniemi I, Beckers A, Daly AF. A vital region for human glycoprotein hormone trafficking revealed by an LHB mutation. J Endocrinol 2016; 231:197-207. [PMID: 27656125 DOI: 10.1530/joe-16-0384] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/21/2016] [Indexed: 11/08/2022]
Abstract
Glycoprotein hormones are complex hormonally active macromolecules. Luteinizing hormone (LH) is essential for the postnatal development and maturation of the male gonad. Inactivating Luteinizing hormone beta (LHB) gene mutations are exceptionally rare and lead to hypogonadism that is particularly severe in males. We describe a family with selective LH deficiency and hypogonadism in two brothers. DNA sequencing of LHB was performed and the effects of genetic variants on hormone function and secretion were characterized by mutagenesis studies, confocal microscopy and functional assays. A 20-year-old male from a consanguineous family had pubertal delay, hypogonadism and undetectable LH. A homozygous c.118_120del (p.Lys40del) mutation was identified in the patient and his brother, who subsequently had the same phenotype. Treatment with hCG led to pubertal development, increased circulating testosterone and spermatogenesis. Experiments in HeLa cells revealed that the mutant LH is retained intracellularly and showed diffuse cytoplasmic distribution. The mutated LHB heterodimerizes with the common alpha-subunit and can activate its receptor. Deletion of flanking glutamic acid residues at positions 39 and 41 impair LH to a similar extent as deletion of Lys40. This region is functionally important across all heterodimeric glycoprotein hormones, because deletion of the corresponding residues in hCG, follicle-stimulating hormone and thyroid-stimulating hormone beta-subunits also led to intracellular hormone retention. This novel LHB mutation results in hypogonadism due to intracellular sequestration of the hormone and reveals a discrete region in the protein that is crucial for normal secretion of all human glycoprotein hormones.
Collapse
Affiliation(s)
- Iulia Potorac
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| | - Adolfo Rivero-Müller
- Department of PhysiologyInstitute of Biomedicine, University of Turku, Turku, Finland
- Faculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, Finland
- Department of Biochemistry and Molecular BiologyMedical University of Lublin, Lublin, Poland
| | - Ashutosh Trehan
- Department of PhysiologyInstitute of Biomedicine, University of Turku, Turku, Finland
| | - Michał Kiełbus
- Department of Biochemistry and Molecular BiologyMedical University of Lublin, Lublin, Poland
| | - Krzysztof Jozwiak
- Laboratory of Medicinal Chemistry and NeuroengineeringMedical University of Lublin, Lublin, Poland
| | - Francois Pralong
- Service of EndocrinologyDiabetology and Metabolism, Department of Medicine, CHU Vaudois, Lausanne, Switzerland
| | - Aicha Hafidi
- Department of Diabetology and Metabolic DiseasesCentre Hospitalier Universitaire Ibn Sina, Rabat, Morocco
| | - Albert Thiry
- Department of PathologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| | | | - Ilpo Huhtaniemi
- Department of PhysiologyInstitute of Biomedicine, University of Turku, Turku, Finland
- Department of Surgery and CancerInstitute of Reproductive and Developmental Biology, Hammersmith Campus, Imperial College London, London, UK
| | - Albert Beckers
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| | - Adrian F Daly
- Department of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, Liège, Belgium
| |
Collapse
|
11
|
Boularan C, Gales C. Cardiac cAMP: production, hydrolysis, modulation and detection. Front Pharmacol 2015; 6:203. [PMID: 26483685 PMCID: PMC4589651 DOI: 10.3389/fphar.2015.00203] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/03/2015] [Indexed: 01/04/2023] Open
Abstract
Cyclic adenosine 3′,5′-monophosphate (cAMP) modulates a broad range of biological processes including the regulation of cardiac myocyte contractile function where it constitutes the main second messenger for β-adrenergic receptors' signaling to fulfill positive chronotropic, inotropic and lusitropic effects. A growing number of studies pinpoint the role of spatial organization of the cAMP signaling as an essential mechanism to regulate cAMP outcomes in cardiac physiology. Here, we will briefly discuss the complexity of cAMP synthesis and degradation in the cardiac context, describe the way to detect it and review the main pharmacological arsenal to modulate its availability.
Collapse
Affiliation(s)
- Cédric Boularan
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier Toulouse, France
| | - Céline Gales
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier Toulouse, France
| |
Collapse
|
12
|
Paramonov VM, Mamaeva V, Sahlgren C, Rivero-Müller A. Genetically-encoded tools for cAMP probing and modulation in living systems. Front Pharmacol 2015; 6:196. [PMID: 26441653 PMCID: PMC4569861 DOI: 10.3389/fphar.2015.00196] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/28/2015] [Indexed: 11/19/2022] Open
Abstract
Intracellular 3′-5′-cyclic adenosine monophosphate (cAMP) is one of the principal second messengers downstream of a manifold of signal transduction pathways, including the ones triggered by G protein-coupled receptors. Not surprisingly, biochemical assays for cAMP have been instrumental for basic research and drug discovery for decades, providing insights into cellular physiology and guiding pharmaceutical industry. However, despite impressive track record, the majority of conventional biochemical tools for cAMP probing share the same fundamental shortcoming—all the measurements require sample disruption for cAMP liberation. This common bottleneck, together with inherently low spatial resolution of measurements (as cAMP is typically analyzed in lysates of thousands of cells), underpin the ensuing limitations of the conventional cAMP assays: (1) genuine kinetic measurements of cAMP levels over time in a single given sample are unfeasible; (2) inability to obtain precise information on cAMP spatial distribution and transfer at subcellular levels, let alone the attempts to pinpoint dynamic interactions of cAMP and its effectors. At the same time, tremendous progress in synthetic biology over the recent years culminated in drastic refinement of our toolbox, allowing us not only to bypass the limitations of conventional assays, but to put intracellular cAMP life-span under tight control—something, that seemed scarcely attainable before. In this review article we discuss the main classes of modern genetically-encoded tools tailored for cAMP probing and modulation in living systems. We examine the capabilities and weaknesses of these different tools in the context of their operational characteristics and applicability to various experimental set-ups involving living cells, providing the guidance for rational selection of the best tools for particular needs.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Department of Physiology, Institute of Biomedicine, University of Turku , Turku, Finland ; Turku Center for Biotechnology, University of Turku and Åbo Akademi University , Turku, Finland
| | - Veronika Mamaeva
- Department of Clinical Science, University of Bergen , Bergen, Norway
| | - Cecilia Sahlgren
- Turku Center for Biotechnology, University of Turku and Åbo Akademi University , Turku, Finland ; Department of Biomedical Engineering, Eindhoven University of Technology , Eindhoven, Netherlands
| | - Adolfo Rivero-Müller
- Department of Physiology, Institute of Biomedicine, University of Turku , Turku, Finland ; Faculty of Natural Sciences and Technology, Åbo Akademi University , Turku, Finland ; Department of Biochemistry and Molecular Biology, Medical University of Lublin , Lublin, Poland
| |
Collapse
|
13
|
Rivero-Müller A, Potorac I, Pintiaux A, Daly AF, Thiry A, Rydlewski C, Nisolle M, Parent AS, Huhtaniemi I, Beckers A. A novel inactivating mutation of the LH/chorionic gonadotrophin receptor with impaired membrane trafficking leading to Leydig cell hypoplasia type 1. Eur J Endocrinol 2015; 172:K27-36. [PMID: 25795638 DOI: 10.1530/eje-14-1095] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 03/20/2015] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The LH/chorionic gonadotrophin receptor (LHCGR) is a G protein-coupled receptor (GPCR) that plays a central role in male sexual differentiation, regulation of ovarian follicular maturation, ovulation and maintenance of corpus luteum and pregnancy, as well as maintenance of testicular testosterone production. Mutations in the LHCGR gene are very rare. The aim of this work was to study the clinical and molecular characteristics of a rare familial LHCGR mutation. METHODS Five affected members of a family, including a phenotypically female, but genotypically male (46,XY), patient with Leydig cell hypoplasia type 1 and four genotypically female siblings with reproductive abnormalities, were studied genetically. Cell trafficking studies as well as signalling studies of mutated receptor were performed. RESULTS The five affected patients were all homozygous for a novel mutation in the LHCGR gene, a deletion of guanine in position 1850 (1850delG). This resulted in a frameshift affecting most of the C-terminal intracellular domain. In vitro studies demonstrated that the 1850delG receptor was completely incapable of transit to the cell membrane, becoming trapped within the endoplasmic reticulum. This could not be rescued by small-molecule agonist treatment or stimulated intracellularly by co-expression of a yoked human chorionic gonadotrophin. CONCLUSIONS This novel LHCGR mutation leads to complete inactivation of the LHCGR receptor due to trafficking and signalling abnormalities, which improves our understanding of the impact of the affected structural domain on receptor trafficking and function.
Collapse
Affiliation(s)
- Adolfo Rivero-Müller
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFa
| | - Iulia Potorac
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Axelle Pintiaux
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Adrian F Daly
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Albert Thiry
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Catherine Rydlewski
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Michelle Nisolle
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Anne-Simone Parent
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Ilpo Huhtaniemi
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| | - Albert Beckers
- Department of PhysiologyInstitute for Biomedicine, University of Turku, Turku, FinlandDepartment of EndocrinologyCentre Hospitalier Universitaire de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumFaculty of Natural Sciences and TechnologyÅbo Akademi University, Turku, FinlandDepartment of Biochemistry and Molecular BiologyMedical University of Lublin, 20-093 Lublin, PolandDepartment of Surgery and CancerImperial College London, Institute of Reproductive and Developmental Biology, Hammersmith Campus, London, UKDepartments of GynecologyAnatomopathologyCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, BelgiumDepartment of Medical GeneticsErasme Hospital, Brussels, BelgiumDepartment of PediatricsCHU de Liège, Université de Liège, Domaine Universitaire du Sart-Tilman, 4000 Liège, Belgium
| |
Collapse
|