1
|
Moreno-Vargas LM, Prada-Gracia D. Exploring the Chemical Features and Biomedical Relevance of Cell-Penetrating Peptides. Int J Mol Sci 2024; 26:59. [PMID: 39795918 PMCID: PMC11720145 DOI: 10.3390/ijms26010059] [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: 10/23/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/13/2025] Open
Abstract
Cell-penetrating peptides (CPPs) are a diverse group of peptides, typically composed of 4 to 40 amino acids, known for their unique ability to transport a wide range of substances-such as small molecules, plasmid DNA, small interfering RNA, proteins, viruses, and nanoparticles-across cellular membranes while preserving the integrity of the cargo. CPPs exhibit passive and non-selective behavior, often requiring functionalization or chemical modification to enhance their specificity and efficacy. The precise mechanisms governing the cellular uptake of CPPs remain ambiguous; however, electrostatic interactions between positively charged amino acids and negatively charged glycosaminoglycans on the membrane, particularly heparan sulfate proteoglycans, are considered the initial crucial step for CPP uptake. Clinical trials have highlighted the potential of CPPs in diagnosing and treating various diseases, including cancer, central nervous system disorders, eye disorders, and diabetes. This review provides a comprehensive overview of CPP classifications, potential applications, transduction mechanisms, and the most relevant algorithms to improve the accuracy and reliability of predictions in CPP development.
Collapse
|
2
|
Nappi F. P2Y12 Receptor Inhibitor for Antiaggregant Therapies: From Molecular Pathway to Clinical Application. Int J Mol Sci 2024; 25:7575. [PMID: 39062819 PMCID: PMC11277343 DOI: 10.3390/ijms25147575] [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: 06/09/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Platelets play a significant role in hemostasis, forming plugs at sites of vascular injury to limit blood loss. However, if platelet activation is not controlled, it can lead to thrombotic events, such as myocardial infarction and stroke. To prevent this, antiplatelet agents are used in clinical settings to limit platelet activation in patients at risk of arterial thrombotic events. However, their use can be associated with a significant risk of bleeding. An enhanced comprehension of platelet signaling mechanisms should facilitate the identification of safer targets for antiplatelet therapy. Over the past decade, our comprehension of the breadth and intricacy of signaling pathways that orchestrate platelet activation has expanded exponentially. Several recent studies have provided further insight into the regulation of platelet signaling events and identified novel targets against which to develop novel antiplatelet agents. Antiplatelet drugs are essential in managing atherothrombotic vascular disease. The current antiplatelet therapy in clinical practice is limited in terms of safety and efficacy. Novel compounds have been developed in response to patient variability and resistance to aspirin and/or clopidogrel. Recent studies based on randomized controlled trials and systematic reviews have definitively demonstrated the role of antiplatelet therapy in reducing the risk of cardiovascular events. Antiplatelet therapy is the recommended course of action for patients with established atherosclerosis. These studies compared monotherapy with a P2Y12 inhibitor versus aspirin for secondary prevention. However, in patients undergoing percutaneous coronary intervention, it is still unclear whether the efficacy of P2Y12 inhibitor monotherapy after a short course of dual antiplatelet therapy depends on the type of P2Y12 inhibitor. This paper focuses on the advanced-stage evaluation of several promising antiplatelet drugs.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
3
|
Smadja DM, Rossi E, Haviari S, Bieche I, Cras A, Gaussem P. Thrombin receptor PAR1 silencing in endothelial colony-forming cells modifies stemness and vasculogenic properties. J Thromb Haemost 2023; 21:3640-3648. [PMID: 37678550 DOI: 10.1016/j.jtha.2023.08.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The involvement of thrombin receptor PAR1 in blood vessel development has been largely demonstrated in knockout mice; however, its implication in adult mouse angiogenesis seems very moderate. OBJECTIVES We aimed to explore the potential relationships between PAR1, stemness, and angiogenic properties of human endothelial colony-forming cells (ECFCs). METHODS AND RESULTS PAR1 activation on ECFCs using the selective PAR1-activating peptide induced a significant decrease in CD133 expression (RTQ-PCR analysis). In line, silencing of PAR1 gene expression with siRNA increased CD133 mRNA as well as intracellular CD133 protein expression. To confirm the link between CD133 and PAR1, we explored the association between PAR1 and CD133 levels in fast and slow fibroblasts prone to reprogramming. An imbalance between PAR1 and CD133 levels was evidenced, with a decreased expression of PAR1 in fast reprogramming fibroblasts expressing a high CD133 level. Regarding in vitro ECFC angiogenic properties, PAR1 silencing with specific siRNA induced cell proliferation evidenced by the overexpression of Ki67. However, it did not impact migration properties nor ECFC adhesion on smooth muscle cells or human arterial endothelial cells. In a mouse model of hind-limb ischemia, PAR1 silencing in ECFCs significantly increased postischemic revascularization compared to siCtrl-ECFCs along with a significant increase in cutaneous blood flows (P < .0001), microvessel density (P = .02), myofiber regeneration (P < .0001), and human endothelial cell incorporation in muscle (P < .0001). CONCLUSION In conclusion, our work describes for the first time a link between PAR1, stemness, and vasculogenesis in human ECFCs.
Collapse
Affiliation(s)
- David M Smadja
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, France; Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France.
| | - Elisa Rossi
- Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France
| | - Skerdi Haviari
- Université Paris-Cité, INSERM UMR-S 1137 (IAME), Paris, France; Département Épidémiologie Biostatistiques et Recherche Clinique, AP-HP, Hôpital Bichat, Paris, France
| | | | - Audrey Cras
- Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France; Cell Therapy Unit, AP-HP, Saint Louis Hospital, Paris, France
| | - Pascale Gaussem
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, France; Université Paris-Cité, INSERM UMR-S 1140, Innovative Therapies in Haemostasis, Paris, France
| |
Collapse
|
4
|
Fletcher EK, Ngwenyama N, Nguyen N, Turner SE, Covic L, Alcaide P, Kuliopulos A. Suppression of Heart Failure With PAR1 Pepducin Technology in a Pressure Overload Model in Mice. Circ Heart Fail 2023; 16:e010621. [PMID: 37477012 PMCID: PMC10592519 DOI: 10.1161/circheartfailure.123.010621] [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: 02/16/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND PAR1 (protease-activated receptor-1) contributes to acute thrombosis, but it is not clear whether the receptor is involved in deleterious inflammatory and profibrotic processes in heart failure. Here, we employ the pepducin technology to determine the effects of targeting PAR1 in a mouse heart failure with reduced ejection fraction model. METHODS After undergoing transverse aortic constriction pressure overload or sham surgery, C57BL/6J mice were randomized to daily sc PZ-128 pepducin or vehicle, and cardiac function, inflammation, fibrosis, and molecular analyses conducted at 7 weeks RESULTS: After 7 weeks of transverse aortic constriction, vehicle mice had marked increases in macrophage/monocyte infiltration and fibrosis of the left ventricle as compared with Sham mice. PZ-128 treatment significantly suppressed the inflammatory cell infiltration and cardiac fibrosis. Despite no effect on myocyte cell hypertrophy, PZ-128 afforded a significant reduction in overall left ventricle weight and completely protected against the transverse aortic constriction-induced impairments in left ventricle ejection fraction. PZ-128 significantly suppressed transverse aortic constriction-induced increases in an array of genes involved in myocardial stress, fibrosis, and inflammation. CONCLUSIONS The PZ-128 pepducin is highly effective in protecting against cardiac inflammation, fibrosis, and loss of left ventricle function in a mouse model.
Collapse
Affiliation(s)
- Elizabeth K Fletcher
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Njabulo Ngwenyama
- Department of Immunology, Tufts University School of Medicine, Boston, MA (N. Ngwenyama, P.A.)
| | - Nga Nguyen
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Susan E Turner
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA (N. Ngwenyama, P.A.)
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Department of Medicine, Division of Hematology-Oncology, Tufts Medical Center (E.K.F., N. Nguyen, S.E.T., L.C., A.K.)
| |
Collapse
|
5
|
Gawaz M, Geisler T, Borst O. Current concepts and novel targets for antiplatelet therapy. Nat Rev Cardiol 2023; 20:583-599. [PMID: 37016032 DOI: 10.1038/s41569-023-00854-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
Platelets have a crucial role in haemostasis and atherothrombosis. Pharmacological control of platelet hyper-reactivity has become a cornerstone in the prevention of thrombo-ischaemic complications in atherosclerotic diseases. Current antiplatelet therapies substantially improve clinical outcomes in patients with coronary artery disease, but at the cost of increased risk of bleeding. Beyond their role in thrombosis, platelets are known to regulate inflammatory (thrombo-inflammatory) and microcirculatory pathways. Therefore, controlling platelet hyper-reactivity might have implications for both tissue inflammation (myocardial ischaemia) and vascular inflammation (vulnerable plaque formation) to prevent atherosclerosis. In this Review, we summarize the pathophysiological role of platelets in acute myocardial ischaemia, vascular inflammation and atherosclerotic progression. Furthermore, we highlight current clinical concepts of antiplatelet therapy that have contributed to improving patient care and have facilitated more individualized therapy. Finally, we discuss novel therapeutic targets and compounds for antiplatelet therapy that are currently in preclinical development, some of which have a more favourable safety profile than currently approved drugs with regard to bleeding risk. These novel antiplatelet targets might offer new strategies to treat cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Gawaz
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany.
| | - Tobias Geisler
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
6
|
Sun Z, Huang J, Fishelson Z, Wang C, Zhang S. Cell-Penetrating Peptide-Based Delivery of Macromolecular Drugs: Development, Strategies, and Progress. Biomedicines 2023; 11:1971. [PMID: 37509610 PMCID: PMC10377493 DOI: 10.3390/biomedicines11071971] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Cell-penetrating peptides (CPPs), developed for more than 30 years, are still being extensively studied due to their excellent delivery performance. Compared with other delivery vehicles, CPPs hold promise for delivering different types of drugs. Here, we review the development process of CPPs and summarize the composition and classification of the CPP-based delivery systems, cellular uptake mechanisms, influencing factors, and biological barriers. We also summarize the optimization routes of CPP-based macromolecular drug delivery from stability and targeting perspectives. Strategies for enhanced endosomal escape, which prolong its half-life in blood, improved targeting efficiency and stimuli-responsive design are comprehensively summarized for CPP-based macromolecule delivery. Finally, after concluding the clinical trials of CPP-based drug delivery systems, we extracted the necessary conditions for a successful CPP-based delivery system. This review provides the latest framework for the CPP-based delivery of macromolecular drugs and summarizes the optimized strategies to improve delivery efficiency.
Collapse
Affiliation(s)
- Zhe Sun
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin 300072, China
| | - Zvi Fishelson
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chenhui Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
7
|
Tantry US, Duhan S, Navarese E, Ramotowski B, Kundan P, Bliden KP, Gurbel P. An update on novel therapies for treating patients with arterial thrombosis. Expert Rev Hematol 2023; 16:593-605. [PMID: 37335893 DOI: 10.1080/17474086.2023.2227788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
INTRODUCTION Antithrombotic therapy field is undergoing rapid and significant changes during the past decade. In addition to new therapeutic strategies with existing targets, investigators are exploring the potential use of new targets to address unmet needs to treat patients with arterial diseases. AREAS COVERED We aim to provide an update on and a comprehensive review of the antithrombic agents that are being explored in patients with arterial diseases. We discuss latest developments with respect to upstream antiplatelet agents, and collagen and thrombin pathway inhibitors. We searched PubMed databases for English language articles using keywords: antiplatelet agents, thrombin pathway inhibitors, collagen receptors, arterial disease. EXPERT OPINION Despite implementation of potent P2Y12 inhibitors, there are numerous unmet needs in the treatment of arterial diseases including ceiling effect of currently available antiplatelet agents along with and an elevated risk of bleeding. The latter observations encouraged investigators to explore new targets that can attenuate the generation of platelet-fibrin clot formation and subsequent ischemic event occurrences with minimal effect on bleeding. These targets include collagen receptors on platelets and thrombin generation including FXa, FXIa, and FXIIa. In addition, investigators are studying novel antiplatelet agents/strategies to facilitate upstream therapy in high-risk patients.
Collapse
Affiliation(s)
- Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Sanchit Duhan
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Eliano Navarese
- Interventional Cardiology and Cardiovascular Medicine Research, Department of Cardiology and Internal Medicine, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Bogumil Ramotowski
- Department of Cardiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Parshotam Kundan
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Kevin P Bliden
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Paul Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, Baltimore, MD, USA
| |
Collapse
|
8
|
Künze G, Isermann B. Targeting biased signaling by PAR1: function and molecular mechanism of parmodulins. Blood 2023; 141:2675-2684. [PMID: 36952648 PMCID: PMC10646804 DOI: 10.1182/blood.2023019775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 03/25/2023] Open
Abstract
The G protein-coupled receptor (GPCR) protease-activated receptor 1 (PAR1) is a therapeutic target that was originally pursued with the aim of restricting platelet activation and the burden of cardiovascular diseases. In clinical studies, the use of orthosteric PAR1 inhibitors was associated with an increased risk of hemorrhage, including intracranial hemorrhage. Because (1) PAR1 is expressed by various cell types, including endothelial cells, (2) conveys in mice a physiological indispensable function for vascular development during embryogenesis, and (3) is subject to biased signaling dependent on the activating proteases, orthosteric PAR1 inhibition may be associated with unwanted side effects. Alternatively, the protease-activated protein C (aPC) and its variants can promote valuable anti-inflammatory signaling via PAR1. Most recently, small molecule allosteric modulators of PAR1 signaling, called parmodulins, have been developed. Parmodulins inhibit coagulation and platelet activation yet maintain cytoprotective effects typically provoked by PAR1 signaling upon the activation by aPC. In this study, we review the discovery of parmodulins and their preclinical data, summarize the current knowledge about their mode of action, and compare the structural interaction of parmodulin and PAR1 with that of other intracellularly binding allosteric GPCR modulators. Thus, we highlight the pharmaceutical potential and challenges associated with the future development of parmodulins.
Collapse
Affiliation(s)
- Georg Künze
- Institute for Drug Discovery, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital, Leipzig, Germany
| |
Collapse
|
9
|
Okyere AD, Song J, Patwa V, Carter RL, Enjamuri N, Lucchese AM, Ibetti J, de Lucia C, Schumacher SM, Koch WJ, Cheung JY, Benovic JL, Tilley DG. Pepducin ICL1-9-Mediated β2-Adrenergic Receptor-Dependent Cardiomyocyte Contractility Occurs in a G i Protein/ROCK/PKD-Sensitive Manner. Cardiovasc Drugs Ther 2023; 37:245-256. [PMID: 34997361 PMCID: PMC9262991 DOI: 10.1007/s10557-021-07299-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 01/14/2023]
Abstract
PURPOSE β-Adrenergic receptors (βAR) are essential targets for the treatment of heart failure (HF); however, chronic use of βAR agonists as positive inotropes to increase contractility in a Gs protein-dependent manner is associated with increased mortality. Alternatively, we previously reported that allosteric modulation of β2AR with the pepducin intracellular loop (ICL)1-9 increased cardiomyocyte contractility in a β-arrestin (βarr)-dependent manner, and subsequently showed that ICL1-9 activates the Ras homolog family member A (RhoA). Here, we aimed to elucidate both the proximal and downstream signaling mediators involved in the promotion of cardiomyocyte contractility in response to ICL1-9. METHODS We measured adult mouse cardiomyocyte contractility in response to ICL1-9 or isoproterenol (ISO, as a positive control) alone or in the presence of inhibitors of various potential components of βarr- or RhoA-dependent signaling. We also assessed the contractile effects of ICL1-9 on cardiomyocytes lacking G protein-coupled receptor (GPCR) kinase 2 (GRK2) or 5 (GRK5). RESULTS Consistent with RhoA activation by ICL1-9, both Rho-associated protein kinase (ROCK) and protein kinase D (PKD) inhibition were able to attenuate ICL1-9-mediated contractility, as was inhibition of myosin light chain kinase (MLCK). While neither GRK2 nor GRK5 deletion impacted ICL1-9-mediated contractility, pertussis toxin attenuated the response, suggesting that ICL1-9 promotes downstream RhoA-dependent signaling in a Gi protein-dependent manner. CONCLUSION Altogether, our study highlights a novel signaling modality that may offer a new approach to the promotion, or preservation, of cardiac contractility during HF via the allosteric regulation of β2AR to promote Gi protein/βarr-dependent activation of RhoA/ROCK/PKD signaling.
Collapse
Affiliation(s)
- Ama Dedo Okyere
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Jianliang Song
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Viren Patwa
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Rhonda L Carter
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Nitya Enjamuri
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Anna Maria Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Jessica Ibetti
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Claudio de Lucia
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
- Instituti Clinici Scientifici Maugeri di Telese Terme, Telese Terme, Italy
| | - Sarah M Schumacher
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Joseph Y Cheung
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Douglas G Tilley
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Room 945A MERB, 3500 N. Broad St, Philadelphia, PA, 19140, USA.
| |
Collapse
|
10
|
Shpakov AO. Allosteric Regulation of G-Protein-Coupled Receptors: From Diversity of Molecular Mechanisms to Multiple Allosteric Sites and Their Ligands. Int J Mol Sci 2023; 24:6187. [PMID: 37047169 PMCID: PMC10094638 DOI: 10.3390/ijms24076187] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Allosteric regulation is critical for the functioning of G protein-coupled receptors (GPCRs) and their signaling pathways. Endogenous allosteric regulators of GPCRs are simple ions, various biomolecules, and protein components of GPCR signaling (G proteins and β-arrestins). The stability and functional activity of GPCR complexes is also due to multicenter allosteric interactions between protomers. The complexity of allosteric effects caused by numerous regulators differing in structure, availability, and mechanisms of action predetermines the multiplicity and different topology of allosteric sites in GPCRs. These sites can be localized in extracellular loops; inside the transmembrane tunnel and in its upper and lower vestibules; in cytoplasmic loops; and on the outer, membrane-contacting surface of the transmembrane domain. They are involved in the regulation of basal and orthosteric agonist-stimulated receptor activity, biased agonism, GPCR-complex formation, and endocytosis. They are targets for a large number of synthetic allosteric regulators and modulators, including those constructed using molecular docking. The review is devoted to the principles and mechanisms of GPCRs allosteric regulation, the multiplicity of allosteric sites and their topology, and the endogenous and synthetic allosteric regulators, including autoantibodies and pepducins. The allosteric regulation of chemokine receptors, proteinase-activated receptors, thyroid-stimulating and luteinizing hormone receptors, and beta-adrenergic receptors are described in more detail.
Collapse
Affiliation(s)
- Alexander O Shpakov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
11
|
Miceli G, Basso MG, Rizzo G, Pintus C, Tuttolomondo A. The Role of the Coagulation System in Peripheral Arterial Disease: Interactions with the Arterial Wall and Its Vascular Microenvironment and Implications for Rational Therapies. Int J Mol Sci 2022; 23:14914. [PMID: 36499242 PMCID: PMC9739112 DOI: 10.3390/ijms232314914] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Peripheral artery disease (PAD) is a clinical manifestation of atherosclerotic disease with a large-scale impact on the economy and global health. Despite the role played by platelets in the process of atherogenesis being well recognized, evidence has been increasing on the contribution of the coagulation system to the atherosclerosis formation and PAD development, with important repercussions for the therapeutic approach. Histopathological analysis and some clinical studies conducted on atherosclerotic plaques testify to the existence of different types of plaques. Likely, the role of coagulation in each specific type of plaque can be an important determinant in the histopathological composition of atherosclerosis and in its future stability. In this review, we analyze the molecular contribution of inflammation and the coagulation system on PAD pathogenesis, focusing on molecular similarities and differences between atherogenesis in PAD and coronary artery disease (CAD) and discussing the possible implications for current therapeutic strategies and future perspectives accounting for molecular inflammatory and coagulation targets. Understanding the role of cross-talking between coagulation and inflammation in atherosclerosis genesis and progression could help in choosing the right patients for future dual pathway inhibition strategies, where an antiplatelet agent is combined with an anticoagulant, whose role, despite pathophysiological premises and trials' results, is still under debate.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| |
Collapse
|
12
|
Barriuso I, Worner F, Vilahur G. Novel Antithrombotic Agents in Ischemic Cardiovascular Disease: Progress in the Search for the Optimal Treatment. J Cardiovasc Dev Dis 2022; 9:397. [PMID: 36421932 PMCID: PMC9699470 DOI: 10.3390/jcdd9110397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 09/10/2024] Open
Abstract
Ischemic cardiovascular diseases have a high incidence and high mortality worldwide. Therapeutic advances in the last decades have reduced cardiovascular mortality, with antithrombotic therapy being the cornerstone of medical treatment. Yet, currently used antithrombotic agents carry an inherent risk of bleeding associated with adverse cardiovascular outcomes and mortality. Advances in understanding the pathophysiology of thrombus formation have led to the discovery of new targets and the development of new anticoagulants and antiplatelet agents aimed at preventing thrombus stabilization and growth while preserving hemostasis. In the following review, we will comment on the key limitation of the currently used antithrombotic regimes in ischemic heart disease and ischemic stroke and provide an in-depth and state-of-the-art overview of the emerging anticoagulant and antiplatelet agents in the pipeline with the potential to improve clinical outcomes.
Collapse
Affiliation(s)
- Ignacio Barriuso
- Hospital Universitario Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, 25198 Lleida, Spain
- Institut de Recerca, Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
- Department of Medicine, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Fernando Worner
- Hospital Universitario Arnau de Vilanova, Institut de Recerca Biomèdica de Lleida, 25198 Lleida, Spain
| | - Gemma Vilahur
- Institut de Recerca, Hospital Santa Creu i Sant Pau, IIB Sant Pau, 08025 Barcelona, Spain
- Centro de Investigaciones Biomédicas En Red de enfermedades CardioVasculares (CiberCV), 28029 Madrid, Spain
| |
Collapse
|
13
|
Xu H, Tilley DG. Pepducin-mediated G Protein-Coupled Receptor Signaling in the Cardiovascular System. J Cardiovasc Pharmacol 2022; 80:378-385. [PMID: 35170495 PMCID: PMC9365886 DOI: 10.1097/fjc.0000000000001236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/29/2022] [Indexed: 01/31/2023]
Abstract
ABSTRACT Pepducins are small-lipidated peptides designed from the intracellular loops of G protein-coupled receptors (GPCRs) that act in an allosteric manner to modulate the activity of GPCRs. Over the past 2 decades, pepducins have progressed initially from pharmacologic tools used to manipulate GPCR activity in an orthosteric site-independent manner to compounds with therapeutic potential that have even been used safely in phase 1 and 2 clinical trials in human subjects. The effect of pepducins at their cognate receptors has been shown to vary between antagonist, partial agonist, and biased agonist outcomes in various primary and clonal cell systems, with even small changes in amino acid sequence altering these properties and their receptor selectivity. To date, pepducins designed from numerous GPCRs have been studied for their impact on pathologic conditions, including cardiovascular diseases such as thrombosis, myocardial infarction, and atherosclerosis. This review will focus in particular on pepducins designed from protease-activated receptors, C-X-C motif chemokine receptors, formyl peptide receptors, and the β2-adrenergic receptor. We will discuss the historic context of pepducin development for each receptor, as well as the structural, signaling, pathophysiologic consequences, and therapeutic potential for each pepducin class.
Collapse
Affiliation(s)
- Heli Xu
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | | |
Collapse
|
14
|
Wang L, Wang X, Guo E, Mao X, Miao S. Emerging roles of platelets in cancer biology and their potential as therapeutic targets. Front Oncol 2022; 12:939089. [PMID: 35936717 PMCID: PMC9355257 DOI: 10.3389/fonc.2022.939089] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
The main role of platelets is to control bleeding and repair vascular damage via thrombosis. They have also been implicated to promote tumor metastasis through platelet-tumor cell interactions. Platelet-tumor cell interactions promote tumor cell survival and dissemination in blood circulation. Tumor cells are known to induce platelet activation and alter platelet RNA profiles. Liquid biopsies based on tumor-educated platelet biomarkers can detect tumors and correlate with prognosis, personalized therapy, treatment monitoring, and recurrence prediction. Platelet-based strategies for cancer prevention and tumor-targeted therapy include developing drugs that target platelet receptors, interfere with the release of platelet particles, inhibit platelet-specific enzymes, and utilize platelet-derived “nano-platelets” as a targeted drug delivery platform for tumor therapy. This review elaborates on platelet-tumor cell interactions and the molecular mechanisms and discusses future research directions for platelet-based liquid biopsy techniques and platelet-targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Erliang Guo
- Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xionghui Mao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| |
Collapse
|
15
|
Lan Z, Zhang Y, Sun Y, Wang L, Huang Y, Cao H, Wang S, Meng J. Identifying of Anti-Thrombin Active Components From Curcumae Rhizoma by Affinity-Ultrafiltration Coupled With UPLC-Q-Exactive Orbitrap/MS. Front Pharmacol 2021; 12:769021. [PMID: 34955839 PMCID: PMC8703108 DOI: 10.3389/fphar.2021.769021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/11/2021] [Indexed: 01/14/2023] Open
Abstract
Recent studies concerning products that originate from natural plants have sought to clarify active ingredients, which both explains the mechanisms of the function and aids in quality control during production. As a traditional functional plant, Curcumae Rhizoma (CR) has been proven to be effective in promoting blood circulation and removing blood stasis. However, the components that play a role in its huge compound library are still unclear. The present study aimed to develop a high-throughput screening method to identify thrombin inhibitors in CR and validate them by in vitro and in vivo experiments. The effect of CR on thrombin in HUVECs cells was determined by ELISA, then an affinity-ultrafiltration-UPLC-Q-Exactive Orbitrap/MS approach was applied. Agatroban and adenosine were used as positive and negative drugs respectively to verify the reliability of the established method. The in vitro activity of the compounds was determined by specific substrate S-2238. The in vivo effect of the active ingredients was determined using zebrafish. Molecular docking was used to understand the internal interactions between compounds and enzymes. ELISA results showed that CR had an inhibitory effect on thrombin. The screening method established in this paper is reliable, by which a total of 15 active compounds were successfully identified. This study is the first to report that C7, 8, and 11 have in vitro thrombin-inhibitory activity and significantly inhibit thrombosis in zebrafish models at a safe dose. Molecular docking studies were employed to analyze the possible active binding sites, with the results suggesting that compound 16 is likely a better thrombin inhibitor compared with the other compounds. Based on the affinity-ultrafiltration-UPLC-Q-Exactive Orbitrap/MS approach, a precisely targeted therapy method using bio-active compounds from CR might be successfully established, which also provides a valuable reference for targeted therapy, mechanism exploration, and the quality control of traditional herbal medicine.
Collapse
Affiliation(s)
- Zhenwei Lan
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica, State Administration of Traditional Chinese Medicine (TCM), Engineering Technology Research Center for Chinese Materia Medica Quality of Universities in Guangdong Province, Guangzhou, China
| | - Ying Zhang
- College of Pharmacy, Jinan University, Research Center for Traditional Chinese Medicine of Lingnan, Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China
| | - Yue Sun
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica, State Administration of Traditional Chinese Medicine (TCM), Engineering Technology Research Center for Chinese Materia Medica Quality of Universities in Guangdong Province, Guangzhou, China
| | - Lvhong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica, State Administration of Traditional Chinese Medicine (TCM), Engineering Technology Research Center for Chinese Materia Medica Quality of Universities in Guangdong Province, Guangzhou, China
| | - Yuting Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica, State Administration of Traditional Chinese Medicine (TCM), Engineering Technology Research Center for Chinese Materia Medica Quality of Universities in Guangdong Province, Guangzhou, China
| | - Hui Cao
- College of Pharmacy, Jinan University, Research Center for Traditional Chinese Medicine of Lingnan, Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, China
| | - Shumei Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica, State Administration of Traditional Chinese Medicine (TCM), Engineering Technology Research Center for Chinese Materia Medica Quality of Universities in Guangdong Province, Guangzhou, China
| | - Jiang Meng
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica, State Administration of Traditional Chinese Medicine (TCM), Engineering Technology Research Center for Chinese Materia Medica Quality of Universities in Guangdong Province, Guangzhou, China
| |
Collapse
|
16
|
Bisht B, Madhavan N. Quick Access to High-Purity Peptide Drugs Bradykinin, Leuprolide Analogue, 2(PZ-128), and Rapastinel with Minimal Reagents. J Org Chem 2021; 86:17667-17672. [PMID: 34823358 DOI: 10.1021/acs.joc.1c01906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Peptide drugs bradykinin, a leuprolide analogue, 2(PZ-128), and rapastinel are synthesized in 56-77% yield using heating-assisted liquid-phase peptide synthesis on a soluble polynorbornene support. These drugs of commercial utility and complex structures are obtained in 2-5.5 h with no epimerization and >95% purity using only 1.2 equivalents of amino acids and coupling reagents. The peptide yield and purity are comparable or superior to the reported methods.
Collapse
Affiliation(s)
- Babita Bisht
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra 400076, India
| | - Nandita Madhavan
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra 400076, India
| |
Collapse
|
17
|
Imidazo[1,2-b]pyridazine as privileged scaffold in medicinal chemistry: An extensive review. Eur J Med Chem 2021; 226:113867. [PMID: 34607244 DOI: 10.1016/j.ejmech.2021.113867] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Abstract
Imidazo[1,2-b]pyridazine scaffold represents an important class of heterocyclic nucleus which provides various bioactives molecules. Among them, the successful kinase inhibitor ponatinib led to a resurgence of interest in exploring new imidazo[1,2-b]pyridazine-containing derivatives for their putative therapeutic applications in medicine. This present review intends to provide a state-of-the-art of this framework in medicinal chemistry from 1966 to nowadays, unveiling different aspects of its structure-activity relationships (SAR). This extensive literature surveil may guide medicinal chemists for the quest of novel imidazo[1,2-b]pyridazine compounds with enhanced pharmacokinetics profile and efficiency.
Collapse
|
18
|
Jourdi G, Lordkipanidzé M, Philippe A, Bachelot-Loza C, Gaussem P. Current and Novel Antiplatelet Therapies for the Treatment of Cardiovascular Diseases. Int J Mol Sci 2021; 22:ijms222313079. [PMID: 34884884 PMCID: PMC8658271 DOI: 10.3390/ijms222313079] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 12/14/2022] Open
Abstract
Over the last decades, antiplatelet agents, mainly aspirin and P2Y12 receptor antagonists, have significantly reduced morbidity and mortality associated with arterial thrombosis. Their pharmacological characteristics, including pharmacokinetic/pharmacodynamics profiles, have been extensively studied, and a significant number of clinical trials assessing their efficacy and safety in various clinical settings have established antithrombotic efficacy. Notwithstanding, antiplatelet agents carry an inherent risk of bleeding. Given that bleeding is associated with adverse cardiovascular outcomes and mortality, there is an unmet clinical need to develop novel antiplatelet therapies that inhibit thrombosis while maintaining hemostasis. In this review, we present the currently available antiplatelet agents, with a particular focus on their targets, pharmacological characteristics, and patterns of use. We will further discuss the novel antiplatelet therapies in the pipeline, with the goal of improved clinical outcomes among patients with atherothrombotic diseases.
Collapse
Affiliation(s)
- Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: (G.J.); (P.G.)
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, QC H1T 1C8, Canada;
- Faculty of Pharmacy, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Aurélien Philippe
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Christilla Bachelot-Loza
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
| | - Pascale Gaussem
- INSERM, Innovations Thérapeutiques en Hémostase, Université de Paris, F-75006 Paris, France; (A.P.); (C.B.-L.)
- Service d’Hématologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
- Correspondence: (G.J.); (P.G.)
| |
Collapse
|
19
|
Lipids in Pathophysiology and Development of the Membrane Lipid Therapy: New Bioactive Lipids. MEMBRANES 2021; 11:membranes11120919. [PMID: 34940418 PMCID: PMC8708953 DOI: 10.3390/membranes11120919] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Membranes are mainly composed of a lipid bilayer and proteins, constituting a checkpoint for the entry and passage of signals and other molecules. Their composition can be modulated by diet, pathophysiological processes, and nutritional/pharmaceutical interventions. In addition to their use as an energy source, lipids have important structural and functional roles, e.g., fatty acyl moieties in phospholipids have distinct impacts on human health depending on their saturation, carbon length, and isometry. These and other membrane lipids have quite specific effects on the lipid bilayer structure, which regulates the interaction with signaling proteins. Alterations to lipids have been associated with important diseases, and, consequently, normalization of these alterations or regulatory interventions that control membrane lipid composition have therapeutic potential. This approach, termed membrane lipid therapy or membrane lipid replacement, has emerged as a novel technology platform for nutraceutical interventions and drug discovery. Several clinical trials and therapeutic products have validated this technology based on the understanding of membrane structure and function. The present review analyzes the molecular basis of this innovative approach, describing how membrane lipid composition and structure affects protein-lipid interactions, cell signaling, disease, and therapy (e.g., fatigue and cardiovascular, neurodegenerative, tumor, infectious diseases).
Collapse
|
20
|
Michael E, Covic L, Kuliopulos A. Lipopeptide Pepducins as Therapeutic Agents. Methods Mol Biol 2021; 2383:307-333. [PMID: 34766299 DOI: 10.1007/978-1-0716-1752-6_21] [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] [Indexed: 03/29/2023]
Abstract
Pepducins are lipidated peptides that target the intracellular loops of G protein-coupled receptors (GPCRs) in order to modulate transmembrane signaling to internally located effectors. With a wide array of potential activities ranging from partial, biased, or full agonism to antagonism, pepducins represent a versatile class of compounds that can be used to potentially treat diverse human diseases or be employed as novel tools to probe complex mechanisms of receptor activation and signaling in cells and in animals. Here, we describe a number of different pepducins including an advanced compound, PZ-128, that has successfully progressed through phase 2 clinical trials in cardiac patients demonstrating safety and efficacy in suppressing myonecrosis and arterial thrombosis.
Collapse
Affiliation(s)
- Emily Michael
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Lidija Covic
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Athan Kuliopulos
- Center of Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
21
|
Olson KM, Traynor JR, Alt A. Allosteric Modulator Leads Hiding in Plain Site: Developing Peptide and Peptidomimetics as GPCR Allosteric Modulators. Front Chem 2021; 9:671483. [PMID: 34692635 PMCID: PMC8529114 DOI: 10.3389/fchem.2021.671483] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
Allosteric modulators (AMs) of G-protein coupled receptors (GPCRs) are desirable drug targets because they can produce fewer on-target side effects, improved selectivity, and better biological specificity (e.g., biased signaling or probe dependence) than orthosteric drugs. An underappreciated source for identifying AM leads are peptides and proteins-many of which were evolutionarily selected as AMs-derived from endogenous protein-protein interactions (e.g., transducer/accessory proteins), intramolecular receptor contacts (e.g., pepducins or extracellular domains), endogenous peptides, and exogenous libraries (e.g., nanobodies or conotoxins). Peptides offer distinct advantages over small molecules, including high affinity, good tolerability, and good bioactivity, and specific disadvantages, including relatively poor metabolic stability and bioavailability. Peptidomimetics are molecules that combine the advantages of both peptides and small molecules by mimicking the peptide's chemical features responsible for bioactivity while improving its druggability. This review 1) discusses sources and strategies to identify peptide/peptidomimetic AMs, 2) overviews strategies to convert a peptide lead into more drug-like "peptidomimetic," and 3) critically analyzes the advantages, disadvantages, and future directions of peptidomimetic AMs. While small molecules will and should play a vital role in AM drug discovery, peptidomimetics can complement and even exceed the advantages of small molecules, depending on the target, site, lead, and associated factors.
Collapse
Affiliation(s)
- Keith M. Olson
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
| | - John R. Traynor
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Andrew Alt
- Department of Pharmacology and Edward F Domino Research Center, University of Michigan, Ann Arbor, MI, United States
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
22
|
Nassour H, Hoang TA, Martin RD, Dallagnol JCC, Billard É, Létourneau M, Novellino E, Carotenuto A, Allen BG, Tanny JC, Fournier A, Hébert TE, Chatenet D. Lipidated peptides derived from intracellular loops 2 and 3 of the urotensin II receptor act as biased allosteric ligands. J Biol Chem 2021; 297:101057. [PMID: 34389356 PMCID: PMC8424217 DOI: 10.1016/j.jbc.2021.101057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 01/14/2023] Open
Abstract
Over the last decade, the urotensinergic system, composed of one G protein-coupled receptor and two endogenous ligands, has garnered significant attention as a promising new target for the treatment of various cardiovascular diseases. Indeed, this system is associated with various biomarkers of cardiovascular dysfunctions and is involved in changes in cardiac contractility, fibrosis and hypertrophy contributing, like the angiotensinergic system, to the pathogenesis and progression of heart failure. Significant investment has been made toward the development of clinically relevant UT ligands for therapeutic intervention, but with little or no success to date. This system therefore remains to be therapeutically exploited. Pepducins and other lipidated peptides have been used as both mechanistic probes and potential therapeutics; therefore, pepducins derived from the human urotensin II receptor might represent unique tools to generate signaling bias and study hUT signaling networks. Two hUT-derived pepducins, derived from the second and the third intracellular loop of the receptor (hUT-Pep2 and [Trp1, Leu2]hUT-Pep3, respectively) were synthesized and pharmacologically characterized. Our results demonstrated that hUT-Pep2 and [Trp1, Leu2]hUT-Pep3 acted as biased ago-allosteric modulators, triggered ERK1/2 phosphorylation and to a lesser extent, IP1 production and stimulated cell proliferation yet were devoid of contractile activity. Interestingly, both hUT-derived pepducins were able to modulate human urotensin II (hUII)- and urotensin II-related peptide (URP)-mediated contraction albeit to different extents. These new derivatives represent unique tools to reveal the intricacies of hUT signaling and also a novel avenue for the design of allosteric ligands selectively targeting hUT signaling potentially.
Collapse
Affiliation(s)
- Hassan Nassour
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada
| | - Tuan Anh Hoang
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada
| | - Juliana C C Dallagnol
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada; Department of Medicine, Université de Montreal, Montreal Heart Institute, Montreal, Québec, Canada
| | - Étienne Billard
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada
| | - Myriam Létourneau
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada
| | - Ettore Novellino
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Alfonso Carotenuto
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Bruce G Allen
- Department of Medicine, Université de Montreal, Montreal Heart Institute, Montreal, Québec, Canada
| | - Jason C Tanny
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada
| | - Alain Fournier
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Santé Biotechnologie, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, Québec, Canada.
| |
Collapse
|
23
|
Fletcher EK, Wang Y, Flynn LK, Turner SE, Rade JJ, Kimmelstiel CD, Gurbel PA, Bliden KP, Covic L, Kuliopulos A. Deficiency of MMP1a (Matrix Metalloprotease 1a) Collagenase Suppresses Development of Atherosclerosis in Mice: Translational Implications for Human Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2021; 41:e265-e279. [PMID: 33761760 PMCID: PMC8062306 DOI: 10.1161/atvbaha.120.315837] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Elizabeth K Fletcher
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Yanling Wang
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Laura K Flynn
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Susan E Turner
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Jeffrey J Rade
- Interventional Cardiology, Division of Cardiology, University of Massachusetts Memorial Medical Center, University of Massachusetts Medical School, Worcester (J.J.R.)
| | - Carey D Kimmelstiel
- Adult Interventional Cardiology, Division of Cardiology, Tufts Medical Center, Boston, MA (C.D.K.)
| | - Paul A Gurbel
- Inova Center for Thrombosis Research and Translational Medicine, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
- Sinai Hospital of Baltimore, MD (P.A.G., K.P.B.)
| | - Kevin P Bliden
- Inova Center for Thrombosis Research and Translational Medicine, Inova Fairfax Hospital, Falls Church, VA (P.A.G., K.P.B.)
- Sinai Hospital of Baltimore, MD (P.A.G., K.P.B.)
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| | - Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Division of Hematology-Oncology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (E.K.F., Y.W., L.K.F, S.E.T., L.C., A.K.)
| |
Collapse
|
24
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|
25
|
Mimicry of Dopamine 1 Receptor Signaling with Cell-Penetrating Peptides. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-020-10066-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AbstractIn this study, through the use of protein mimicry, a peptide was developed to activate the dopamine 1 receptor signaling pathway from the inside of the cell and in absence of the natural extracellular ligand. The sequence was initially derived from the intracellular interaction site between the activated receptor and the alpha domain of its associated G-protein and subsequently modified to increase its cell-penetrating properties. The peptide was then synthesized via solid phase peptide synthesis, purified and tested on cell models. This novel lipopeptide proved to be capable of efficiently ubiquitously penetrating the cell without the need for transfection agents or chiral recognition by specific pathways. Furthermore, the peptide induced the cellular response normally achieved through the activation of the receptor in cells that had not been treated with the natural ligand. The peptide could work as a candidate substitute to l-DOPA, leading the way for a peptides-based treatment for Parkinson’s disease.
Collapse
|
26
|
Han X, Nieman MT, Kerlin BA. Protease-activated receptors: An illustrated review. Res Pract Thromb Haemost 2020; 5:17-26. [PMID: 33537526 PMCID: PMC7845062 DOI: 10.1002/rth2.12454] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 01/04/2023] Open
Abstract
Proteases are important regulators of cell behavior, survival, and apoptosis. They communicate to cells directly through a special class of G‐protein–coupled receptors known as protease‐activated receptors (PARs). N‐terminal PAR proteolysis unmasks a neo‐N‐terminus, which serves as a tethered ligand to activate PARs. Using this unique irreversible activation mechanism, PARs relay information across cell membranes. The year 2020 is the 30th year since discovery of the first member of this family, PAR1. In this illustrated review, we highlight achievements in the PAR field over the past 3 decades. Additionally, the known expression profiles of PARs in human tissues and across species are portrayed. We also illustrate the tethered ligand activation mechanism, which is unique to PARs, and PAR regulatory mechanisms. PAR1 was originally named “thrombin receptor” because thrombin was the first protease identified to activate PAR1. However, over the past 30 years, a growing number of proteases have been found to cleave PARs and trigger differential downstream signaling depending on cleavage site, cell type, and species. We exemplify the diversity of PAR1‐mediated signaling outcomes in platelets and endothelial cells as pertinent examples to the hemostasis, thrombosis, and vascular biology fields. Further, the termination and regulation of PAR signaling via endocytosis and currently available pharmacologic approaches are depicted. We conclude with portrayal of clinically translational aspects of PAR biology including pharmacologic manipulation and single‐nucleotide polymorphisms.
Collapse
Affiliation(s)
- Xu Han
- Department of Pharmacology Case Western Reserve University Cleveland OH USA
| | - Marvin T Nieman
- Department of Pharmacology Case Western Reserve University Cleveland OH USA
| | - Bryce A Kerlin
- Center for Clinical and Translational Research Abigail Wexner Research Institute at Nationwide Children's Hospital Columbus OH USA.,Department of Pediatrics The Ohio State University College of Medicine Columbus OH USA
| |
Collapse
|
27
|
Kuliopulos A, Gurbel PA, Rade JJ, Kimmelstiel CD, Turner SE, Bliden KP, Fletcher EK, Cox DH, Covic L. PAR1 (Protease-Activated Receptor 1) Pepducin Therapy Targeting Myocardial Necrosis in Coronary Artery Disease and Acute Coronary Syndrome Patients Undergoing Cardiac Catheterization: A Randomized, Placebo-Controlled, Phase 2 Study. Arterioscler Thromb Vasc Biol 2020; 40:2990-3003. [PMID: 33028101 PMCID: PMC7682800 DOI: 10.1161/atvbaha.120.315168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/25/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Arterial thrombosis leading to ischemic injury worsens the prognosis of many patients with cardiovascular disease. PZ-128 is a first-in-class pepducin that reversibly inhibits PAR1 (protease-activated receptor 1) on platelets and other vascular cells by targeting the intracellular surface of the receptor. The TRIP-PCI (Thrombin Receptor Inhibitory Pepducin in Percutaneous Coronary Intervention) trial was conducted to assess the safety and efficacy of PZ-128 in patients undergoing cardiac catheterization with intent to perform percutaneous coronary intervention. Approach and Results: In this randomized, double-blind, placebo-controlled, phase 2 trial, 100 patients were randomly assigned (2:1) to receive PZ-128 (0.3 or 0.5 mg/kg), or placebo in a 2-hour infusion initiated just before the start of cardiac catheterization, on top of standard oral antiplatelet therapy. Rates of the primary end point of bleeding were not different between the combined PZ-128 doses (1.6%, 1/62) and placebo group (0%, 0/35). The secondary end points of major adverse coronary events at 30 and 90 days did not significantly differ but were numerically lower in the PZ-128 groups (0% and 2% in the PZ-128 groups, 6% and 6% with placebo, p=0.13, p=0.29, respectively). In the subgroup of patients with elevated baseline cardiac troponin I, the exploratory end point of 30-day major adverse coronary events + myocardial injury showed 83% events in the placebo group versus 31% events in the combined PZ-128 drug groups, an adjusted relative risk of 0.14 (95% CI, 0.02-0.75); P=0.02. CONCLUSIONS In this first-in-patient experience, PZ-128 added to standard antiplatelet therapy appeared to be safe, well tolerated, and potentially reduced periprocedural myonecrosis, thus providing the basis for further clinical trials. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02561000.
Collapse
Affiliation(s)
- Athan Kuliopulos
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (A.K., S.E.T., E.K.F., D.H.C., L.C.)
| | - Paul A. Gurbel
- Inova Center for Thrombosis Research and Translational Medicine, Inova Fairfax Hospital, Falls Church, VA and Sinai Hospital of Baltimore, MD (P.A.G., K.P.B.)
| | - Jeffrey J. Rade
- Division of Cardiology, Department of Medicine, University of Massachusetts Memorial Medical Center, University of Massachusetts Medical School, Worcester (J.J.R)
| | - Carey D. Kimmelstiel
- Division of Cardiology, Department of Medicine, Tufts Medical Center, Boston, MA (C.D.K.)
| | - Susan E. Turner
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (A.K., S.E.T., E.K.F., D.H.C., L.C.)
| | - Kevin P. Bliden
- Inova Center for Thrombosis Research and Translational Medicine, Inova Fairfax Hospital, Falls Church, VA and Sinai Hospital of Baltimore, MD (P.A.G., K.P.B.)
| | - Elizabeth K. Fletcher
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (A.K., S.E.T., E.K.F., D.H.C., L.C.)
| | - Daniel H. Cox
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (A.K., S.E.T., E.K.F., D.H.C., L.C.)
| | - Lidija Covic
- Center for Hemostasis and Thrombosis Research, Tufts Medical Center, Tufts University School of Medicine, Boston, MA (A.K., S.E.T., E.K.F., D.H.C., L.C.)
| |
Collapse
|
28
|
Liu S, Li S, Yuan D, Wang E, Xie R, Zhang W, Kong Y, Zhu X. Protease activated receptor 4 (PAR4) antagonists: Research progress on small molecules in the field of antiplatelet agents. Eur J Med Chem 2020; 209:112893. [PMID: 33049608 DOI: 10.1016/j.ejmech.2020.112893] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/27/2022]
Abstract
Protease activated receptor 4 (PAR4) is a key target in antiplatelet medication to reduce the risk of heart attack and thrombotic complications in stroke. PAR4 antagonists can prevent harmful and stable thrombus growth while retaining initial thrombus formation by acting on the late diffusion stage of platelet activation, which may provide a safer alternative than other antiplatelet agents. Currently, research on PAR4 antagonists is of increasing interest in the field of antiplatelet agents. This article provides an overview of the discovery and development of small-molecule antagonists of PAR4 as novel antiplatelet agents, including structure-activity relationship (SAR) analysis, progress of structure and bioassay optimization, and the latest structural and/or clinical information of representative small-molecule antagonists of PAR4.
Collapse
Affiliation(s)
- Shangde Liu
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shanshan Li
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Duo Yuan
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Enmao Wang
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Roujie Xie
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Weiqi Zhang
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yi Kong
- School of Life & Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Xiong Zhu
- Institute of Medicinal & Chemistry, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
29
|
Han X, Nieman MT. The domino effect triggered by the tethered ligand of the protease activated receptors. Thromb Res 2020; 196:87-98. [PMID: 32853981 DOI: 10.1016/j.thromres.2020.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/23/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022]
Abstract
Protease activated receptors (PARs) are G-protein coupled receptors (GPCRs) that have a unique activation mechanism. Unlike other GPCRs that can be activated by free ligands, under physiological conditions, PARs are activated by the tethered ligand, which is a part of their N-terminus that is unmasked by proteolysis. It has been 30 years since the first member of the family, PAR1, was identified. In this review, we will discuss this unique tethered ligand mediate receptor activation of PARs in detail: how they interact with the proteases, the complex structural rearrangement of the receptors upon activation, and the termination of the signaling. We also summarize the structural studies of the PARs and how single nucleotide polymorphisms impact the receptor reactivity. Finally, we review the current strategies for inhibiting PAR function with therapeutic targets for anti-thrombosis. The focus of this review is PAR1 and PAR4 as they are the thrombin signal mediators on human platelets and therapeutics targets. We also include the structural studies of PAR2 as it informs the mechanism of action for PARs in general.
Collapse
Affiliation(s)
- Xu Han
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Marvin T Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
30
|
Poole LG, Pant A, Cline‐Fedewa HM, Williams KJ, Copple BL, Palumbo JS, Luyendyk JP. Liver fibrosis is driven by protease-activated receptor-1 expressed by hepatic stellate cells in experimental chronic liver injury. Res Pract Thromb Haemost 2020; 4:906-917. [PMID: 32685902 PMCID: PMC7354391 DOI: 10.1002/rth2.12403] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Blood coagulation protease activity is proposed to drive hepatic fibrosis through activation of protease-activated receptors (PARs). Whole-body PAR-1 deficiency reduces experimental hepatic fibrosis, and in vitro studies suggest a potential contribution by PAR-1 expressed by hepatic stellate cells. However, owing to a lack of specific tools, the cell-specific role of PAR-1 in experimental hepatic fibrosis has never been formally investigated. Using a novel mouse expressing a conditional PAR-1 allele, we tested the hypothesis that PAR-1 expressed by hepatic stellate cells contributes to hepatic fibrosis. METHODS PAR-1flox/flox mice were crossed with mice expressing Cre recombinase controlled by the lecithin retinol acyltransferase (LRAT) promoter, which induces recombination in hepatic stellate cells. Male PAR-1flox/flox/LRATCre and PAR-1flox/flox mice were challenged twice weekly with carbon tetrachloride (CCl4, 1 mL/kg i.p.) for 6 weeks to induce liver fibrosis. RESULTS PAR-1 mRNA levels were reduced (>95%) in hepatic stellate cells isolated from PAR-1flox/flox/LRATCre mice. Hepatic stellate cell activation was evident in CCl4-challenged PAR-1flox/flox mice, indicated by increased α-smooth muscle actin labeling and induction of several profibrogenic genes. CCl4-challenged PAR-1flox/flox mice displayed robust hepatic collagen deposition, indicated by picrosirius red staining and type I collagen immunolabeling. Notably, stellate cell activation and collagen deposition were significantly reduced (>30%) in PAR-1flox/flox/LRATCre mice. Importantly, the reduction in liver fibrosis was not a consequence of reduced acute CCl4 hepatotoxicity in PAR-1flox/flox/LRATCre mice. CONCLUSIONS The results constitute the first direct experimental evidence that PAR-1 expressed by stellate cells directly promotes their profibrogenic phenotype and hepatic fibrosis in vivo.
Collapse
Affiliation(s)
- Lauren G. Poole
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Asmita Pant
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Holly M. Cline‐Fedewa
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Kurt J. Williams
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
| | - Bryan L. Copple
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| | - Joseph S. Palumbo
- Cancer and Blood Diseases InstituteCincinnati Children’s Hospital Medical Center and the University of Cincinnati College of MedicineCincinnatiOHUSA
| | - James P. Luyendyk
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| |
Collapse
|
31
|
Rehman A, Baloch NUA, Morrow JP, Pacher P, Haskó G. Targeting of G-protein coupled receptors in sepsis. Pharmacol Ther 2020; 211:107529. [PMID: 32197794 PMCID: PMC7388546 DOI: 10.1016/j.pharmthera.2020.107529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/11/2022]
Abstract
The Third International Consensus Definitions (Sepsis-3) define sepsis as life-threatening multi-organ dysfunction caused by a dysregulated host response to infection. Sepsis can progress to septic shock-an even more lethal condition associated with profound circulatory, cellular and metabolic abnormalities. Septic shock remains a leading cause of death in intensive care units and carries a mortality of almost 25%. Despite significant advances in our understanding of the pathobiology of sepsis, therapeutic interventions have not translated into tangible differences in the overall outcome for patients. Clinical trials of antagonists of various pro-inflammatory mediators in sepsis have been largely unsuccessful in the past. Given the diverse physiologic roles played by G-protein coupled receptors (GPCR), modulation of GPCR signaling for the treatment of sepsis has also been explored. Traditional pharmacologic approaches have mainly focused on ligands targeting the extracellular domains of GPCR. However, novel techniques aimed at modulating GPCR intracellularly through aptamers, pepducins and intrabodies have opened a fresh avenue of therapeutic possibilities. In this review, we summarize the diverse roles played by various subfamilies of GPCR in the pathogenesis of sepsis and identify potential targets for pharmacotherapy through these novel approaches.
Collapse
Affiliation(s)
- Abdul Rehman
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Noor Ul-Ain Baloch
- Department of Medicine, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - John P Morrow
- Department of Medicine, Columbia University, New York City, NY, United States
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York City, NY, United States.
| |
Collapse
|
32
|
El Alaoui MZ, Guy A, Khalki L, Limami Y, Benomar A, Zaid N, Cherrah Y, Mekhfi H, Cadi R, Zaid Y. [Current antiplatelet agents, new inhibitors and therapeutic targets]. Med Sci (Paris) 2020; 36:348-357. [PMID: 32356711 DOI: 10.1051/medsci/2020061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular diseases are the leading cause of deaths in the world. Platelets play a major role in the occurrence of these diseases and the development of antiplatelet drugs is a priority in the fight against cardiovascular diseases-associated mortality. Aspirin and thienopyridine-based P2Y12 inhibitors are the main drugs currently used. These molecules target the initiation of platelets activation and are responsible for a moderate inhibitory action. Other antiplatelet agents, as glycoprotein (GP) IIb/IIIa antagonists, inhibit platelet aggregation independently of initial activation-associated pathways, but are responsible for increased hemorrhagic events. Regarding each antiplatelet agent's specific characteristics, the prescription of these drugs must take into account the type of cardiovascular event, the age of the patient, the past medical history, and the potential hemorrhagic adverse events. Thus, there is a need for the development of new molecules with a more targeted effect, maintaining optimal efficiency but with a reduction of the hemorrhagic risk, which is the principal limitation of these treatments.
Collapse
Affiliation(s)
| | - Alexandre Guy
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Maroc
| | - Loubna Khalki
- Research Center of Mohammed VI University of Health Sciences, Casablanca, Maroc
| | - Youness Limami
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Ali Benomar
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Nabil Zaid
- Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Maroc
| | - Yahia Cherrah
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc
| | - Hassan Mekhfi
- Laboratory of Physiology, Genetic and Ethnopharmacology, Faculty of Sciences, Mohammed the First University, Oujda, Maroc
| | - Rachida Cadi
- Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Maroc
| | - Younes Zaid
- Research Center of Abulcasis University of Health Sciences, Rabat, Maroc - Faculty of Sciences, Department of Biology, Mohammed V University, Rabat, Maroc
| |
Collapse
|
33
|
Davenport AP, Scully CCG, de Graaf C, Brown AJH, Maguire JJ. Advances in therapeutic peptides targeting G protein-coupled receptors. Nat Rev Drug Discov 2020; 19:389-413. [PMID: 32494050 DOI: 10.1038/s41573-020-0062-z] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 02/06/2023]
Abstract
Dysregulation of peptide-activated pathways causes a range of diseases, fostering the discovery and clinical development of peptide drugs. Many endogenous peptides activate G protein-coupled receptors (GPCRs) - nearly 50 GPCR peptide drugs have been approved to date, most of them for metabolic disease or oncology, and more than 10 potentially first-in-class peptide therapeutics are in the pipeline. The majority of existing peptide therapeutics are agonists, which reflects the currently dominant strategy of modifying the endogenous peptide sequence of ligands for peptide-binding GPCRs. Increasingly, novel strategies are being employed to develop both agonists and antagonists, to both introduce chemical novelty and improve drug-like properties. Pharmacodynamic improvements are evolving to allow biasing ligands to activate specific downstream signalling pathways, in order to optimize efficacy and reduce side effects. In pharmacokinetics, modifications that increase plasma half-life have been revolutionary. Here, we discuss the current status of the peptide drugs targeting GPCRs, with a focus on evolving strategies to improve pharmacokinetic and pharmacodynamic properties.
Collapse
Affiliation(s)
- Anthony P Davenport
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | | | | | | | - Janet J Maguire
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| |
Collapse
|
34
|
Brouillette RL, Besserer-Offroy É, Mona CE, Chartier M, Lavenus S, Sousbie M, Belleville K, Longpré JM, Marsault É, Grandbois M, Sarret P. Cell-penetrating pepducins targeting the neurotensin receptor type 1 relieve pain. Pharmacol Res 2020; 155:104750. [PMID: 32151680 DOI: 10.1016/j.phrs.2020.104750] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 02/12/2020] [Accepted: 03/05/2020] [Indexed: 01/29/2023]
Abstract
Pepducins are cell-penetrating, membrane-tethered lipopeptides designed to target the intracellular region of a G protein-coupled receptor (GPCR) in order to allosterically modulate the receptor's signaling output. In this proof-of-concept study, we explored the pain-relief potential of a pepducin series derived from the first intracellular loop of neurotensin receptor type 1 (NTS1), a class A GPCR that mediates many of the effects of the neurotensin (NT) tridecapeptide, including hypothermia, hypotension and analgesia. We used BRET-based biosensors to determine the pepducins' ability to engage G protein signaling pathways associated with NTS1 activation. We observed partial Gαq and Gα13 activation at a 10 μM concentration, indicating that these pepducins may act as allosteric agonists of NTS1. Additionally, we used surface plasmon resonance (SPR) as a label-free assay to monitor pepducin-induced responses in CHO-K1 cells stably expressing hNTS1. This whole-cell integrated assay enabled us to subdivide our pepducin series into three profile response groups. In order to determine the pepducins' antinociceptive potential, we then screened the series in an acute pain model (tail-flick test) by measuring tail withdrawal latencies to a thermal nociceptive stimulus, following intrathecal (i.t.) pepducin administration (275 nmol/kg). We further evaluated promising pepducins in a tonic pain model (formalin test), as well as in neuropathic (Chronic Constriction Injury) and inflammatory (Complete Freund's Adjuvant) chronic pain models. We report one pepducin, PP-001, that consistently reduced rat nociceptive behaviors, even in chronic pain paradigms. Finally, we designed a TAMRA-tagged version of PP-001 and found by confocal microscopy that the pepducin reached the rat dorsal root ganglia post i.t. injection, thus potentially modulating the activity of NTS1 at this location to produce its analgesic effect. Altogether, these results suggest that NTS1-derived pepducins may represent a promising strategy in pain-relief.
Collapse
Affiliation(s)
- Rebecca L Brouillette
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Élie Besserer-Offroy
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada.
| | - Christine E Mona
- Ahmanson Translational Theranostic Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| | - Magali Chartier
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Sandrine Lavenus
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Marc Sousbie
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Karine Belleville
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Éric Marsault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Michel Grandbois
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada; Institut de pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
35
|
Therapeutic strategies for thrombosis: new targets and approaches. Nat Rev Drug Discov 2020; 19:333-352. [PMID: 32132678 DOI: 10.1038/s41573-020-0061-0] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2020] [Indexed: 12/19/2022]
Abstract
Antiplatelet agents and anticoagulants are a mainstay for the prevention and treatment of thrombosis. However, despite advances in antithrombotic therapy, a fundamental challenge is the side effect of bleeding. Improved understanding of the mechanisms of haemostasis and thrombosis has revealed new targets for attenuating thrombosis with the potential for less bleeding, including glycoprotein VI on platelets and factor XIa of the coagulation system. The efficacy and safety of new agents are currently being evaluated in phase III trials. This Review provides an overview of haemostasis and thrombosis, details the current landscape of antithrombotic agents, addresses challenges with preventing thromboembolic events in patients at high risk and describes the emerging therapeutic strategies that may break the inexorable link between antithrombotic therapy and bleeding risk.
Collapse
|
36
|
Taylor RE, Zahid M. Cell Penetrating Peptides, Novel Vectors for Gene Therapy. Pharmaceutics 2020; 12:E225. [PMID: 32138146 PMCID: PMC7150854 DOI: 10.3390/pharmaceutics12030225] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022] Open
Abstract
Cell penetrating peptides (CPPs), also known as protein transduction domains (PTDs), first identified ~25 years ago, are small, 6-30 amino acid long, synthetic, or naturally occurring peptides, able to carry variety of cargoes across the cellular membranes in an intact, functional form. Since their initial description and characterization, the field of cell penetrating peptides as vectors has exploded. The cargoes they can deliver range from other small peptides, full-length proteins, nucleic acids including RNA and DNA, liposomes, nanoparticles, and viral particles as well as radioisotopes and other fluorescent probes for imaging purposes. In this review, we will focus briefly on their history, classification system, and mechanism of transduction followed by a summary of the existing literature on use of CPPs as gene delivery vectors either in the form of modified viruses, plasmid DNA, small interfering RNA, oligonucleotides, full-length genes, DNA origami or peptide nucleic acids.
Collapse
Affiliation(s)
- Rebecca E. Taylor
- Mechanical Engineering, Biomedical Engineering and Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Maliha Zahid
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
37
|
Tscharre M, Michelson AD, Gremmel T. Novel Antiplatelet Agents in Cardiovascular Disease. J Cardiovasc Pharmacol Ther 2020; 25:191-200. [DOI: 10.1177/1074248419899314] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Antiplatelet therapy reduces atherothrombotic risk and has therefore become a cornerstone in the treatment of cardiovascular disease. Aspirin, adenosine diphosphate P2Y12 receptor antagonists, glycoprotein IIb/IIIa inhibitors, and the thrombin receptor blocker vorapaxar are effective antiplatelet agents but significantly increase the risk of bleeding. Moreover, atherothrombotic events still impair the prognosis of many patients with cardiovascular disease despite established antiplatelet therapy. Over the last years, advances in the understanding of thrombus formation and hemostasis led to the discovery of various new receptors and signaling pathways of platelet activation. As a consequence, many new antiplatelet agents with high antithrombotic efficacy and supposedly only moderate effects on regular hemostasis have been developed and yielded promising results in preclinical and early clinical studies. Although their long journey from animal studies to randomized clinical trials and finally administration in daily clinical routine has just begun, some of the new agents may in the future become meaningful additions to the pharmacological armamentarium in cardiovascular disease.
Collapse
Affiliation(s)
- Maximilian Tscharre
- Department of Internal Medicine, Cardiology and Nephrology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
- Institute of Vascular Medicine and Cardiac Electrophysiology, Karl Landsteiner Society, St Poelten, Austria
| | - Alan D. Michelson
- Center for Platelet Research Studies, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Thomas Gremmel
- Department of Internal Medicine, Cardiology and Nephrology, Landesklinikum Wiener Neustadt, Wiener Neustadt, Austria
- Institute of Vascular Medicine and Cardiac Electrophysiology, Karl Landsteiner Society, St Poelten, Austria
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Rout A, Sukhi A, Chaudhary R, Bliden KP, Tantry US, Gurbel PA. Investigational drugs in phase II clinical trials for acute coronary syndromes. Expert Opin Investig Drugs 2020; 29:33-47. [DOI: 10.1080/13543784.2020.1708324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Amit Rout
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridgehealth, Baltimore, MD, USA
| | - Ajaypaul Sukhi
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridgehealth, Baltimore, MD, USA
| | - Rahul Chaudhary
- Division of Hospital Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kevin P Bliden
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridgehealth, Baltimore, MD, USA
| | - Udaya S Tantry
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridgehealth, Baltimore, MD, USA
| | - Paul A Gurbel
- Sinai Center for Thrombosis Research, Sinai Hospital of Baltimore, LifeBridgehealth, Baltimore, MD, USA
| |
Collapse
|
39
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
40
|
Retamal JS, Ramírez-García PD, Shenoy PA, Poole DP, Veldhuis NA. Internalized GPCRs as Potential Therapeutic Targets for the Management of Pain. Front Mol Neurosci 2019; 12:273. [PMID: 31798411 PMCID: PMC6874167 DOI: 10.3389/fnmol.2019.00273] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/28/2019] [Indexed: 01/14/2023] Open
Abstract
Peripheral and central neurons in the pain pathway are well equipped to detect and respond to extracellular stimuli such as pro-inflammatory mediators and neurotransmitters through the cell surface expression of receptors that can mediate rapid intracellular signaling. Following injury or infection, activation of cell surface G protein-coupled receptors (GPCRs) initiates cell signaling processes that lead to the generation of action potentials in neurons or inflammatory responses such as cytokine secretion by immune cells. However, it is now appreciated that cell surface events alone may not be sufficient for all receptors to generate their complete signaling repertoire. Following an initial wave of signaling at the cell surface, active GPCRs can engage with endocytic proteins such as the adaptor protein β-arrestin (βArr) to promote clathrin-mediated internalization. Classically, βArr-mediated internalization of GPCRs was hypothesized to terminate signaling, yet for multiple GPCRs known to contribute to pain, it has been demonstrated that endocytosis can also promote a unique "second wave" of signaling from intracellular membranes, including those of endosomes and the Golgi, that is spatiotemporally distinct from initial cell-surface events. In the context of pain, understanding the cellular and molecular mechanisms that drive spatiotemporal signaling of GPCRs is invaluable for understanding how pain occurs and persists, and how current analgesics achieve efficacy or promote side-effects. This review article discusses the importance of receptor localization for signaling outcomes of pro- and anti-nociceptive GPCRs, and new analgesic opportunities emerging through the development of "location-biased" ligands that favor binding with intracellular GPCR populations.
Collapse
Affiliation(s)
- Jeffri S Retamal
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Paulina D Ramírez-García
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Priyank A Shenoy
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| |
Collapse
|
41
|
Slack MA, Gordon SM. Protease Activity in Vascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:e210-e218. [PMID: 31553665 PMCID: PMC6764587 DOI: 10.1161/atvbaha.119.312413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 01/13/2023]
Affiliation(s)
- Megan A. Slack
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Scott M. Gordon
- Saha Cardiovascular Research Center and Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
42
|
Xu J, Khan AR, Fu M, Wang R, Ji J, Zhai G. Cell-penetrating peptide: a means of breaking through the physiological barriers of different tissues and organs. J Control Release 2019; 309:106-124. [PMID: 31323244 DOI: 10.1016/j.jconrel.2019.07.020] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022]
Abstract
The selective infiltration of cell membranes and tissue barriers often blocks the entry of most active molecules. This natural defense mechanism prevents the invasion of exogenous substances and limits the therapeutic value of most available molecules. Therefore, it is particularly important to find appropriate ways of membrane translocation and therapeutic agent delivery to its target site. Cell penetrating peptides (CPPs) are a group of short peptides harnessed in this condition, possessing a significant capacity for membrane transduction and could be exploited to transfer various biologically active cargoes into the cells. Since their discovery, CPPs have been employed for delivery of a wide variety of therapeutic molecules to treat various disorders including cranial nerve involvement, ocular inflammation, myocardial ischemia, dermatosis and cancer. The promising results of CPPs-derived therapeutics in various tumor models demonstrated a potential and worthwhile scope of CPPs in chemotherapy. This review describes the detailed description of CPPs and CPPs-assisted molecular delivery against various tissues and organs disorders. An emphasis is focused on summarizing the novel insights and achievements of CPPs in surmounting the natural membrane barriers during the last 5 years.
Collapse
Affiliation(s)
- Jiangkang Xu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Abdur Rauf Khan
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Manfei Fu
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Rujuan Wang
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China
| | - Guangxi Zhai
- School of Pharmaceutical Sciences, Key Laboratory of Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, China.
| |
Collapse
|
43
|
Poujol de Molliens M, Jamadagni P, Létourneau M, Devost D, Hébert TE, Patten SA, Fournier A, Chatenet D. Design and biological assessment of membrane-tethering neuroprotective peptides derived from the pituitary adenylate cyclase-activating polypeptide type 1 receptor. Biochim Biophys Acta Gen Subj 2019; 1863:129398. [PMID: 31306709 DOI: 10.1016/j.bbagen.2019.07.007] [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: 05/02/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The pituitary adenylate cyclase-activating polypeptide (PACAP) type 1 receptor (PAC1), a class B G protein-coupled receptor (GPCR), has emerged as a promising target for treating neurodegenerative conditions. Unfortunately, despite years of research, no PAC1-specific agonist has been discovered, as activity on two other GPCRs, VPAC1 and VPAC2, is retained with current analogs. Cell signaling is related to structural modifications in the intracellular loops (ICLs) of GPCRs. Thus, we hypothesized that peptides derived from the ICLs (called pepducins) of PAC1 might initiate, as allosteric ligands, signaling cascades after recognition of the parent receptor and modulation of its conformational landscape. METHODS Three pepducins were synthesized and evaluated for their ability to 1) promote cell survival; 2) stimulate various signaling pathways associated with PAC1 activation; 3) modulate selectively PAC1, VPAC1 or VPAC2 activation; and 4) sustain mobility and prevent death of dopaminergic neurons in a zebrafish model of neurodegeneration. RESULTS Assays demonstrated that these molecules promote SH-SY5Y cell survival, a human neuroblastoma cell line expressing PAC1, and activate signaling via Gαs and Gαq, with distinct potencies and efficacies. Also, PAC1-Pep1 and PAC1-Pep2 activated selectively PAC1-mediated Gαs stimulation. Finally, experiments, using a zebrafish neurodegeneration model, showed a neuroprotective action with all three pepducins and in particular, revealed the ability of PAC1-Pep1 and PAC1-Pep3 to preserve fish mobility and tyrosine hydroxylase expression in the brain. CONCLUSION We have developed the first neuroprotective pepducins derived from PAC1, a class B GPCR. GENERAL SIGNIFICANCE PAC1-derived pepducins represent attractive templates for the development of innovative neuroprotecting molecules.
Collapse
Affiliation(s)
- Mathilde Poujol de Molliens
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada; Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - Priyanka Jamadagni
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Université du Québec, Ville de Laval, QC, Canada
| | - Myriam Létourneau
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada; Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Shunmoogum A Patten
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Université du Québec, Ville de Laval, QC, Canada
| | - Alain Fournier
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Laboratoire d'études moléculaires et pharmacologiques des peptides (LEMPP), Université du Québec, Ville de Laval, QC, Canada
| | - David Chatenet
- Institut National de la Recherche Scientifique, Centre Armand-Frappier, Groupe de Recherche en Ingénierie des Peptides et en Pharmacothérapie (GRIPP), Université du Québec, Ville de Laval, QC, Canada.
| |
Collapse
|
44
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
45
|
Habault J, Poyet JL. Recent Advances in Cell Penetrating Peptide-Based Anticancer Therapies. Molecules 2019; 24:E927. [PMID: 30866424 PMCID: PMC6429072 DOI: 10.3390/molecules24050927] [Citation(s) in RCA: 215] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 12/12/2022] Open
Abstract
Cell-penetrating-peptides (CPPs) are small amino-acid sequences characterized by their ability to cross cellular membranes. They can transport various bioactive cargos inside cells including nucleic acids, large proteins, and other chemical compounds. Since 1988, natural and synthetic CPPs have been developed for applications ranging from fundamental to applied biology (cell imaging, gene editing, therapeutics delivery). In recent years, a great number of studies reported the potential of CPPs as carriers for the treatment of various diseases. Apart from a good efficacy due to a rapid and potent delivery, a crucial advantage of CPP-based therapies is the peptides low toxicity compared to most drug carriers. On the other hand, they are quite unstable and lack specificity. Higher specificity can be obtained using a cell-specific CPP to transport the therapeutic agent or using a non-specific CPP to transport a cargo with a targeted activity. CPP-cargo complexes can also be conjugated to another moiety that brings cell- or tissue-specificity. Studies based on all these approaches are showing promising results. Here, we focus on recent advances in the potential usage of CPPs in the context of cancer therapy, with a particular interest in CPP-mediated delivery of anti-tumoral proteins.
Collapse
Affiliation(s)
- Justine Habault
- INSERM U976, Institut de Recherche St Louis, 1 avenue Claude Vellefaux, 75010 Paris, France.
- Université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France.
| | - Jean-Luc Poyet
- INSERM U976, Institut de Recherche St Louis, 1 avenue Claude Vellefaux, 75010 Paris, France.
- Université Paris Diderot, Sorbonne Paris Cité, 75013 Paris, France.
- c-Dithem, Inserm Consortium for Discovery and Innovation in Therapy and Medicine, 75013 Paris, France.
| |
Collapse
|
46
|
Isono A, Tsuji M, Sanada Y, Matsushita A, Masunaga S, Hirayama T, Nagasawa H. Design, Synthesis, and Evaluation of Lipopeptide Conjugates of Mercaptoundecahydrododecaborate for Boron Neutron Capture Therapy. ChemMedChem 2019; 14:823-832. [DOI: 10.1002/cmdc.201800793] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/23/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Aoi Isono
- Laboratory of Pharmaceutical and Medicinal ChemistryGifu Pharmaceutical University Daigaku-nishi 1-25-4 Gifu-city Gifu 501-1196 Japan
| | - Mieko Tsuji
- Laboratory of Pharmaceutical and Medicinal ChemistryGifu Pharmaceutical University Daigaku-nishi 1-25-4 Gifu-city Gifu 501-1196 Japan
| | - Yu Sanada
- Particle Radiation BiologyInstitute for Integrated Radiation and Nuclear ScienceKyoto University 2-1010 Asashiro-Nishi Kumatori-cho, Sennan-gun Osaka 590-0494 Japan
| | - Akari Matsushita
- Laboratory of Pharmaceutical and Medicinal ChemistryGifu Pharmaceutical University Daigaku-nishi 1-25-4 Gifu-city Gifu 501-1196 Japan
| | - Shinichiro Masunaga
- Particle Radiation BiologyInstitute for Integrated Radiation and Nuclear ScienceKyoto University 2-1010 Asashiro-Nishi Kumatori-cho, Sennan-gun Osaka 590-0494 Japan
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal ChemistryGifu Pharmaceutical University Daigaku-nishi 1-25-4 Gifu-city Gifu 501-1196 Japan
| | - Hideko Nagasawa
- Laboratory of Pharmaceutical and Medicinal ChemistryGifu Pharmaceutical University Daigaku-nishi 1-25-4 Gifu-city Gifu 501-1196 Japan
| |
Collapse
|
47
|
Zhao Y, Zheng Y, Liu X, Luo Q, Wu D, Liu X, Zou L. Inhibiting trophoblast PAR-1 overexpression suppresses sFlt-1-induced anti-angiogenesis and abnormal vascular remodeling: a possible therapeutic approach for preeclampsia. Mol Hum Reprod 2019; 24:158-169. [PMID: 29325127 DOI: 10.1093/molehr/gax068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 01/04/2018] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Is it possible to improve vascular remodeling by inhibiting the excessive expression of protease-activated receptor 1 (PAR-1) in trophoblast of abnormal placenta? SUMMARY ANSWER Inhibition of trophoblast PAR-1 overexpression may promote placental angiogenesis and vascular remodeling, offering an alternative therapeutic approach for preeclampsia. WHAT IS KNOWN ALREADY PAR-1 is high-affinity receptor of thrombin. Thrombin increases sFlt-1 secretion in trophoblast via the activation of PAR-1. It is reported that the expression of both thrombin and PAR-1 expression are increased in placentas of preeclampsia patients compared with normal placentas. STUDY DESIGN, SIZE, DURATION Trophoblast cells were transfected with PAR-1 short hairpin RNA (shRNA) or PAR-1 overexpression plasmids in vitro. Tube formation assays and a villus-decidua co-culture system were used to study the effect of PAR-1 inhibition on placental angiogenesis and vascular remodeling, respectively. Placentas from rats with preeclampsia were transfected with PAR-1 shRNA to confirm the effect of inhibiting PAR-1 overexpression in placenta. PARTICIPANTS/MATERIALS, SETTING, METHODS The trophoblast cell line HTR-8/SVneo was transfected with PAR-1 shRNA or PAR-1 overexpression plasmids. After 48 h, supernatant was collected and the level of sFlt-1 secretion was measured by ELISA. Human umbilical cord epithelial cells and a villus-decidua co-culture system were treated with conditioned media to study the effect of PAR-1 inhibition on tube formation and villi vascular remodeling. A preeclampsia rat model was established by intraperitoneal injection of L-NAME. Plasmids were injected into the placenta of the preeclampsia rats and systolic blood pressure was measured on Days 15 and 19. The effect of different treatments was evaluated by proteinuria, placental weights, fetal weights and fetal numbers in study and control groups. The level of serum sFlt-1 in rats with preeclampsia was also measured. Changes in the placenta microvessels were studied by histopathological staining. MAIN RESULTS AND THE ROLE OF CHANCE PAR-1 shRNA inhibited PAR-1 expression and significantly suppressed sFlt-1 expression in trophoblasts. Soluble Flt-1 level in the supernatant was suppressed by PAR-1 inhibition plasmid transfection and increased by PAR-1 overexpression plasmids (46.93 ± 5.22 vs. 25.21 ± 4.18 vs. 67.84 ± 3.58 ng/ml, P < 0.01). Tube formation assays showed that conditioned media from shPAR-1 transfected cells resulted in an increase in the total number of branching points compared with that of blank controls (P < 0.05). The villus-decidua co-culture system confirmed down-regulation of PAR-1 was conducive to angiogenesis and vascular remodeling. Transfecting placenta with PAR-1 shRNA plasmids improved placental vascular development and ameliorated the symptoms of preeclampsia in rats. After treatment with shRNA, blood pressure was controlled (140.83 ± 1.08 vs. 123.6 ± 1.47 mmHg, P < 0.001) and proteinuria levels were decreased (4.48 ± 0.36 vs. 2.64 ± 0.25 μg/μl, P < 0.01). sFlt-1 protein levels were significantly higher in preeclampsia group than in the control group (1.44 ± 0.33 vs. 2.92 ± 0.85 ng/ml, P < 0.001), but was reduced (0.92 ± 0.06 ng/ml, vs. PE, P < 0.001) in the treatment group. The histopathological changes of the placental microvessels showed that in the preeclampsia group, the number of blood vessels was reduced, while in treatment group, the placental microvasculature was improved (P < 0.001). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Despite our promising results, the evaluation of kidney damage was studied only by proteinuria measurement. Histochemistry of kidney damage will be supplemented in a further study. WIDER IMPLICATIONS OF THE FINDINGS The data showed that inhibition of trophoblast PAR-1 overexpression may promote placental angiogenesis and vascular remodeling, potentially offering an alternative therapeutic approach for preeclampsia. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from the National Natural Science Foundation of China (Grant Nos. 81100442 and 81771605 for Y.Z. and 81179584 for L.Z.) and the Hubei Province Health and Family Planning Scientific Research Project (Grant No. WJ2017 M093 for Y.Z.). The authors declare that there is no conflict of interest.
Collapse
Affiliation(s)
- Yin Zhao
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - YanFang Zheng
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - XiaoXia Liu
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - QingQing Luo
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - Di Wu
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - XiaoPing Liu
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| | - Li Zou
- Obstetrics and Gynaecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Street, Wuhan 430022, China
| |
Collapse
|
48
|
Majewski MW, Gandhi DM, Rosas R, Kodali R, Arnold LA, Dockendorff C. Design and Evaluation of Heterobivalent PAR1-PAR2 Ligands as Antagonists of Calcium Mobilization. ACS Med Chem Lett 2019; 10:121-126. [PMID: 30655958 DOI: 10.1021/acsmedchemlett.8b00538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/03/2018] [Indexed: 12/28/2022] Open
Abstract
A novel class of bivalent ligands targeting putative protease-activated receptor (PAR) heteromers has been prepared based upon reported antagonists for the subtypes PAR1 and PAR2. Modified versions of the PAR1 antagonist RWJ-58259 containing alkyne adapters were connected via cycloaddition reactions to azide-capped polyethylene glycol (PEG) spacers attached to imidazopyridazine-based PAR2 antagonists. Initial studies of the PAR1-PAR2 antagonists indicated that they inhibited G alpha q-mediated calcium mobilization in endothelial and cancer cells driven by both PAR1 and PAR2 agonists. Compounds of this novel class hold promise for the prevention of restenosis, cancer cell metastasis, and other proliferative disorders.
Collapse
Affiliation(s)
- Mark W. Majewski
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, Wisconsin 53201-1881, United States
| | - Disha M. Gandhi
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, Wisconsin 53201-1881, United States
| | - Ricardo Rosas
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, Wisconsin 53201-1881, United States
| | - Revathi Kodali
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin, Milwaukee, Wisconsin 53211, United States
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin, Milwaukee, Wisconsin 53211, United States
| | - Chris Dockendorff
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, Wisconsin 53201-1881, United States
| |
Collapse
|
49
|
Grover SP, Bergmeier W, Mackman N. Platelet Signaling Pathways and New Inhibitors. Arterioscler Thromb Vasc Biol 2019; 38:e28-e35. [PMID: 29563117 DOI: 10.1161/atvbaha.118.310224] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Steven P Grover
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill
| | - Wolfgang Bergmeier
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine (S.P.G., N.M.) and McAllister Heart Institute and Department of Biochemistry and Biophysics (W.B.), University of North Carolina at Chapel Hill.
| |
Collapse
|
50
|
Schneditz G, Elias JE, Pagano E, Zaeem Cader M, Saveljeva S, Long K, Mukhopadhyay S, Arasteh M, Lawley TD, Dougan G, Bassett A, Karlsen TH, Kaser A, Kaneider NC. GPR35 promotes glycolysis, proliferation, and oncogenic signaling by engaging with the sodium potassium pump. Sci Signal 2019; 12:12/562/eaau9048. [PMID: 30600262 DOI: 10.1126/scisignal.aau9048] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The sodium potassium pump (Na/K-ATPase) ensures the electrochemical gradient of a cell through an energy-dependent process that consumes about one-third of regenerated ATP. We report that the G protein-coupled receptor GPR35 interacted with the α chain of Na/K-ATPase and promotes its ion transport and Src signaling activity in a ligand-independent manner. Deletion of Gpr35 increased baseline Ca2+ to maximal levels and reduced Src activation and overall metabolic activity in macrophages and intestinal epithelial cells (IECs). In contrast, a common T108M polymorphism in GPR35 was hypermorphic and had the opposite effects to Gpr35 deletion on Src activation and metabolic activity. The T108M polymorphism is associated with ulcerative colitis and primary sclerosing cholangitis, inflammatory diseases with a high cancer risk. GPR35 promoted homeostatic IEC turnover, whereas Gpr35 deletion or inhibition by a selective pepducin prevented inflammation-associated and spontaneous intestinal tumorigenesis in mice. Thus, GPR35 acts as a central signaling and metabolic pacesetter, which reveals an unexpected role of Na/K-ATPase in macrophage and IEC biology.
Collapse
Affiliation(s)
- Georg Schneditz
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK.,Norwegian PSC Research Center, Department of Transplantation Medicine and Institute of Clinical Medicine, Oslo University Hospital and University of Oslo, 0027 Oslo, Norway
| | - Joshua E Elias
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ester Pagano
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK.,Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - M Zaeem Cader
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Svetlana Saveljeva
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Kathleen Long
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
| | - Subhankar Mukhopadhyay
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK.,MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London SE1 9RT, UK
| | | | | | - Gordon Dougan
- Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK
| | | | - Tom H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine and Institute of Clinical Medicine, Oslo University Hospital and University of Oslo, 0027 Oslo, Norway
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Nicole C Kaneider
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK.
| |
Collapse
|