61 results on '"Nebigil CG"'
Search Results
2. Therapeutic Potential of Targeting Prokineticin Receptors in Diseases.
- Author
-
Vincenzi M, Kremić A, Jouve A, Lattanzi R, Miele R, Benharouga M, Alfaidy N, Migrenne-Li S, Kanthasamy AG, Porcionatto M, Ferrara N, Tetko IV, Désaubry L, and Nebigil CG
- Subjects
- Humans, Receptors, G-Protein-Coupled metabolism, Peptides, Biomarkers, Neuropeptides metabolism, Neoplasms drug therapy
- Abstract
The prokineticins (PKs) were discovered approximately 20 years ago as small peptides inducing gut contractility. Today, they are established as angiogenic, anorectic, and proinflammatory cytokines, chemokines, hormones, and neuropeptides involved in variety of physiologic and pathophysiological pathways. Their altered expression or mutations implicated in several diseases make them a potential biomarker. Their G-protein coupled receptors, PKR1 and PKR2, have divergent roles that can be therapeutic target for treatment of cardiovascular, metabolic, and neural diseases as well as pain and cancer. This article reviews and summarizes our current knowledge of PK family functions from development of heart and brain to regulation of homeostasis in health and diseases. Finally, the review summarizes the established roles of the endogenous peptides, synthetic peptides and the selective ligands of PKR1 and PKR2, and nonpeptide orthostatic and allosteric modulator of the receptors in preclinical disease models. The present review emphasizes the ambiguous aspects and gaps in our knowledge of functions of PKR ligands and elucidates future perspectives for PK research. SIGNIFICANCE STATEMENT: This review provides an in-depth view of the prokineticin family and PK receptors that can be active without their endogenous ligand and exhibits "constitutive" activity in diseases. Their non- peptide ligands display promising effects in several preclinical disease models. PKs can be the diagnostic biomarker of several diseases. A thorough understanding of the role of prokineticin family and their receptor types in health and diseases is critical to develop novel therapeutic strategies with safety concerns., (Copyright © 2023 by The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
3. The flavagline FL3 interferes with the association of Annexin A2 with the eIF4F initiation complex and transiently stimulates the translation of annexin A2 mRNA.
- Author
-
Grindheim AK, Patil SS, Nebigil CG, Désaubry L, and Vedeler A
- Abstract
Introduction: Annexin A2 (AnxA2) plays a critical role in cell transformation, immune response, and resistance to cancer therapy. Besides functioning as a calcium- and lipidbinding protein, AnxA2 also acts as an mRNA-binding protein, for instance, by interacting with regulatory regions of specific cytoskeleton-associated mRNAs. Methods and Results: Nanomolar concentrations of FL3, an inhibitor of the translation factor eIF4A, transiently increases the expression of AnxA2 in PC12 cells and stimulates shortterm transcription/translation of anxA2 mRNA in the rabbit reticulocyte lysate. AnxA2 regulates the translation of its cognate mRNA by a feed-back mechanism, which can partly be relieved by FL3. Results obtained using the holdup chromatographic retention assay results suggest that AnxA2 interacts transiently with eIF4E (possibly eIF4G) and PABP in an RNA-independent manner while cap pulldown experiments indicate a more stable RNA-dependent interaction. Short-term (2 h) treatment of PC12 cells with FL3 increases the amount of eIF4A in cap pulldown complexes of total lysates, but not of the cytoskeletal fraction. AnxA2 is only present in cap analogue-purified initiation complexes from the cytoskeletal fraction and not total lysates confirming that AnxA2 binds to a specific subpopulation of mRNAs. Discussion: Thus, AnxA2 interacts with PABP1 and subunits of the initiation complex eIF4F, explaining its inhibitory effect on translation by preventing the formation of the full eIF4F complex. This interaction appears to be modulated by FL3. These novel findings shed light on the regulation of translation by AnxA2 and contribute to a better understanding of the mechanism of action of eIF4A inhibitors., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Grindheim, Patil, Nebigil, Désaubry and Vedeler.)
- Published
- 2023
- Full Text
- View/download PDF
4. Editorial: HF2Cancer: Exploring bidirectional interaction between cardiovascular diseases and cancer.
- Author
-
Nebigil CG and Chan MWY
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
5. Development of fluorizoline analogues as prohibitin ligands that modulate C-RAF signaling, p21 expression and melanogenesis.
- Author
-
Chouha N, Abou-Hamdan H, Yurugi H, Yoshii R, Ii H, Najem A, Ghanem GE, Nakata S, Rajalingam K, Peng Y, Wang D, Nebigil CG, and Désaubry L
- Subjects
- Cyclin-Dependent Kinase Inhibitor p21 metabolism, HeLa Cells, Humans, Ligands, Melanins metabolism, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-raf metabolism, Proto-Oncogene Proteins c-raf pharmacology, Repressor Proteins, Transcription Factors metabolism, Apoptosis, Prohibitins
- Abstract
Fluorizoline is a cytotoxic trifluorothiazoline that targets the scaffold proteins prohibitins-1 and -2 (PHB1/2) to inhibit the kinase C-RAF and promote the expression of the cyclin-dependent kinase inhibitor p21 to induce cancer cell death. In melanocytes, fluorizoline also induces the synthesis of melanin. Herein we report the first structural requirement of fluorizoline analogues for these activities. We identified in particular some compounds that display enhanced anti-C-RAF and anti-MEK activities, and a higher cytotoxicity in HeLa cells compared to fluorizoline. These results provide a foundation for further optimization of PHB ligands for the treatment of cancers. We also discovered an analogue of fluorizoline that displays pharmacological effects opposed to those of fluorizoline and that can be used as a chemical tool to explore PHB signaling in cancers and other diseases., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022. Published by Elsevier Masson SAS.)
- Published
- 2022
- Full Text
- View/download PDF
6. Evidence for reciprocal network interactions between injured hearts and cancer.
- Author
-
Guler MN, Tscheiller NM, Sabater-Molina M, Gimeno JR, and Nebigil CG
- Abstract
Heart failure (HF) and cancer are responsible for 50% of all deaths in middle-aged people. These diseases are tightly linked, which is supported by recent epidemiological studies and case control studies, demonstrating that HF patients have a higher risk to develop cancer such as lung and breast cancer. For HF patients, a one-size-fits-all clinical management strategy is not effective and patient management represents a major economical and clinical burden. Anti-cancer treatments-mediated cardiotoxicity, leading to HF have been extensively studied. However, recent studies showed that even before the initiation of cancer therapy, cancer patients presented impairments in the cardiovascular functions and exercise capacity. Thus, the optimal cardioprotective and surveillance strategies should be applied to cancer patients with pre-existing HF. Recently, preclinical studies addressed the hypothesis that there is bilateral interaction between cardiac injury and cancer development. Understanding of molecular mechanisms of HF-cancer interaction can define the profiles of bilateral signaling networks, and identify the disease-specific biomarkers and possibly therapeutic targets. Here we discuss the shared pathological events, and some treatments of cancer- and HF-mediated risk incidence. Finally, we address the evidences on bilateral connection between cardiac injury (HF and early cardiac remodeling) and cancer through secreted factors (secretoms)., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Guler, Tscheiller, Sabater-Molina, Gimeno and Nebigil.)
- Published
- 2022
- Full Text
- View/download PDF
7. Updates on Anticancer Therapy-Mediated Vascular Toxicity and New Horizons in Therapeutic Strategies.
- Author
-
Hsu PY, Mammadova A, Benkirane-Jessel N, Désaubry L, and Nebigil CG
- Abstract
Vascular toxicity is a frequent adverse effect of current anticancer chemotherapies and often results from endothelial dysfunction. Vascular endothelial growth factor inhibitors (VEGFi), anthracyclines, plant alkaloids, alkylating agents, antimetabolites, and radiation therapy evoke vascular toxicity. These anticancer treatments not only affect tumor vascularization in a beneficial manner, they also damage ECs in the heart. Cardiac ECs have a vital role in cardiovascular functions including hemostasis, inflammatory and coagulation responses, vasculogenesis, and angiogenesis. EC damage can be resulted from capturing angiogenic factors, inhibiting EC proliferation, survival and signal transduction, or altering vascular tone. EC dysfunction accounts for the pathogenesis of myocardial infarction, atherothrombosis, microangiopathies, and hypertension. In this review, we provide a comprehensive overview of the effects of chemotherapeutic agents on vascular toxicity leading to hypertension, microvascular rarefaction thrombosis and atherosclerosis, and affecting drug delivery. We also describe the potential therapeutic approaches such as vascular endothelial growth factor (VEGF)-B and prokineticin receptor-1 agonists to maintain endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor effectiveness., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Hsu, Mammadova, Benkirane-Jessel, Désaubry and Nebigil.)
- Published
- 2021
- Full Text
- View/download PDF
8. Pressure Overload-Mediated Sustained PKR2 (Prokineticin-2 Receptor) Signaling in Cardiomyocytes Contributes to Cardiac Hypertrophy and Endotheliopathies.
- Author
-
Demir F, Urayama K, Audebrand A, Toprak-Semiz A, Steenman M, Kurose H, and Nebigil CG
- Subjects
- Activins genetics, Activins metabolism, Animals, Cardiomegaly genetics, Cardiomegaly physiopathology, Endothelium, Vascular physiopathology, Gastrointestinal Hormones genetics, Heart Failure genetics, Heart Failure physiopathology, Humans, Mice, Mice, Transgenic, Neuropeptides genetics, Cardiomegaly metabolism, Endothelium, Vascular metabolism, Gastrointestinal Hormones metabolism, Heart Failure metabolism, Myocytes, Cardiac metabolism, Neuropeptides metabolism, Signal Transduction physiology
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
9. Correction: Nguyen T.L., et al. Role of Prokineticin Receptor-1 in Epicardial Progenitor Cells. J. Dev. Biol. 2013, 1 , 20-31.
- Author
-
Nguyen TL, Gasser A, and Nebigil CG
- Abstract
The authors wish to make the following corrections to this paper [...].
- Published
- 2020
- Full Text
- View/download PDF
10. SFPH proteins as therapeutic targets for a myriad of diseases.
- Author
-
Wang D, Tabti R, Elderwish S, Djehal A, Chouha N, Pinot F, Yu P, Nebigil CG, and Désaubry L
- Subjects
- Disease, Humans, Ligands, Molecular Structure, Small Molecule Libraries chemistry, Bacterial Proteins antagonists & inhibitors, Small Molecule Libraries pharmacology
- Abstract
The stomatin/prohibitin/flotillin/HflK/HflC (SPFH) domain is present in an evolutionarily conserved family of proteins that regulate a myriad of signaling pathways in archaea, bacteria and eukaryotes. The most studied SPFH proteins, prohibitins, have already been targeted by different families of small molecules to induce anticancer, cardioprotective, anti-inflammatory, antiviral, and antiosteoporotic activities. Ligands of other SPFH proteins have also been identified and shown to act as anesthetics, anti-allodynia, anticancer, and anti-inflammatory agents. These findings indicate that modulators of human or bacterial SPFH proteins can be developed to treat a wide variety of human disorders., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
11. Prokineticin signaling in heart-brain developmental axis: Therapeutic options for heart and brain injuries.
- Author
-
Désaubry L, Kanthasamy AG, and Nebigil CG
- Subjects
- Animals, Brain physiology, Heart physiology, Humans, Neurogenesis genetics, Receptors, G-Protein-Coupled drug effects, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled physiology, Brain growth & development, Brain Diseases genetics, Heart growth & development, Heart Diseases genetics, Vascular Endothelial Growth Factor, Endocrine-Gland-Derived genetics, Vascular Endothelial Growth Factor, Endocrine-Gland-Derived physiology
- Abstract
Heart and brain development occur simultaneously during the embryogenesis, and both organ development and injuries are interconnected. Early neuronal and cardiac injuries share mutual cellular events, such as angiogenesis and plasticity that could either delay disease progression or, in the long run, result in detrimental health effects. For this reason, the common mechanisms provide a new and previously undervalued window of opportunity for intervention. Because angiogenesis, cardiogenesis and neurogenesis are essential for the development and regeneration of the heart and brain, we discuss therein the role of prokineticin as an angiogenic neuropeptide in heart-brain development and injuries. We focus on the role of prokineticin signaling and the effect of drugs targeting prokineticin receptors in neuroprotection and cardioprotection, with a special emphasis on heart failure, neurodegenerativParkinson's disease and ischemic heart and brain injuries. Indeed, prokineticin triggers common pro-survival signaling pathway in heart and brain. Our review aims at stimulating researchers and clinicians in neurocardiology to focus on the role of prokineticin signaling in the reciprocal interaction between heart and brain. We hope to facilitate the discovery of new treatment strategies, acting in both heart and brain degenerative diseases., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
12. Flavaglines as natural products targeting eIF4A and prohibitins: From traditional Chinese medicine to antiviral activity against coronaviruses.
- Author
-
Nebigil CG, Moog C, Vagner S, Benkirane-Jessel N, Smith DR, and Désaubry L
- Subjects
- Animals, COVID-19, Eukaryotic Initiation Factor-4A drug effects, Humans, Medicine, Chinese Traditional, Pandemics, Prohibitins, Repressor Proteins drug effects, Aglaia chemistry, Antiviral Agents therapeutic use, Biological Products therapeutic use, Coronavirus Infections drug therapy, Eukaryotic Initiation Factor-4A genetics, Pneumonia, Viral drug therapy, Repressor Proteins genetics
- Abstract
Flavaglines are cyclopenta[b]benzofurans found in plants of the genus Aglaia, several species of which are used in traditional Chinese medicine. These compounds target the initiation factor of translation eIF4A and the scaffold proteins prohibitins-1 and 2 (PHB1/2) to exert various pharmacological activities, including antiviral effects against several types of viruses, including coronaviruses. This review is focused on the antiviral effects of flavaglines and their therapeutic potential against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Masson SAS. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
13. Prohibitin ligands: a growing armamentarium to tackle cancers, osteoporosis, inflammatory, cardiac and neurological diseases.
- Author
-
Wang D, Tabti R, Elderwish S, Abou-Hamdan H, Djehal A, Yu P, Yurugi H, Rajalingam K, Nebigil CG, and Désaubry L
- Subjects
- Gene Expression, Heart Diseases metabolism, Heart Diseases therapy, Humans, Neoplasms metabolism, Neoplasms pathology, Nervous System Diseases metabolism, Nervous System Diseases pathology, Osteoporosis metabolism, Osteoporosis pathology, Prohibitins, Protein Processing, Post-Translational, Repressor Proteins chemistry, Repressor Proteins genetics, Signal Transduction, Heart Diseases pathology, Ligands, Neoplasms therapy, Nervous System Diseases therapy, Osteoporosis therapy, Repressor Proteins metabolism
- Abstract
Over the last three decades, the scaffold proteins prohibitins-1 and -2 (PHB1/2) have emerged as key signaling proteins regulating a myriad of signaling pathways in health and diseases. Small molecules targeting PHBs display promising effects against cancers, osteoporosis, inflammatory, cardiac and neurodegenerative diseases. This review provides an updated overview of the various classes of PHB ligands, with an emphasis on their mechanism of action and therapeutic potential. We also describe how these ligands have been used to explore PHB signaling in different physiological and pathological settings.
- Published
- 2020
- Full Text
- View/download PDF
14. Editorial: Emerging Challenges of Cardiovascular and Metabolic Dysfunctions in Cardio-Oncology: From Bench to Bedside.
- Author
-
Nebigil CG, Chan MWY, and Rassaf T
- Published
- 2020
- Full Text
- View/download PDF
15. Targeting GPCRs Against Cardiotoxicity Induced by Anticancer Treatments.
- Author
-
Audebrand A, Désaubry L, and Nebigil CG
- Abstract
Novel anticancer medicines, including targeted therapies and immune checkpoint inhibitors, have greatly improved the management of cancers. However, both conventional and new anticancer treatments induce cardiac adverse effects, which remain a critical issue in clinic. Cardiotoxicity induced by anti-cancer treatments compromise vasospastic and thromboembolic ischemia, dysrhythmia, hypertension, myocarditis, and cardiac dysfunction that can result in heart failure. Importantly, none of the strategies to prevent cardiotoxicity from anticancer therapies is completely safe and satisfactory. Certain clinically used cardioprotective drugs can even contribute to cancer induction. Since G protein coupled receptors (GPCRs) are target of forty percent of clinically used drugs, here we discuss the newly identified cardioprotective agents that bind GPCRs of adrenalin, adenosine, melatonin, ghrelin, galanin, apelin, prokineticin and cannabidiol. We hope to provoke further drug development studies considering these GPCRs as potential targets to be translated to treatment of human heart failure induced by anticancer drugs., (Copyright © 2020 Audebrand, Désaubry and Nebigil.)
- Published
- 2020
- Full Text
- View/download PDF
16. Discovery of 3,3'-pyrrolidinyl-spirooxindoles as cardioprotectant prohibitin ligands.
- Author
-
Elderwish S, Audebrand A, Nebigil CG, and Désaubry L
- Subjects
- Apoptosis drug effects, Cardiotonic Agents chemical synthesis, Cardiotonic Agents chemistry, Cell Line, Cell Survival drug effects, Dose-Response Relationship, Drug, Doxorubicin antagonists & inhibitors, Doxorubicin pharmacology, Humans, Ligands, Molecular Structure, Myocytes, Cardiac metabolism, Oxindoles chemical synthesis, Oxindoles chemistry, Prohibitins, Repressor Proteins metabolism, Spiro Compounds chemical synthesis, Spiro Compounds chemistry, Structure-Activity Relationship, Cardiotonic Agents pharmacology, Drug Discovery, Myocytes, Cardiac drug effects, Oxindoles pharmacology, Repressor Proteins antagonists & inhibitors, Spiro Compounds pharmacology
- Abstract
The scaffold proteins prohibitins-1 and 2 (PHB1/2) play many important roles in coordinating many cell signaling pathways and represent emerging targets in cardiology and oncology. We previously reported that a family of natural products derivatives, flavaglines, binds to PHB1/2 to exert cardioprotectant and anti-cancer effects. However, flavaglines also target the initiation factor of translation eIF4A, which doesn't contribute to cardioprotection and may even induce some adverse effects. Herein, we report the development of a convenient and robust synthesis of the new PHB2 ligand 2'-phenylpyrrolidinyl-spirooxindole, and its analogues. We discovered that these compounds displays cardioprotective effect against doxorubicin mediated cardiotoxicity and uncovered the structural requirement for this activity. We identified in particular some analogues that are more cardioprotectant than flavaglines. Pull-down experiments demonstrated that these compounds bind not only to PHB2 but also PHB1. These novel PHB ligands may provide the basis for the development of new drugs candidates to protect the heart against the adverse effects of anticancer treatments., (Copyright © 2019. Published by Elsevier Masson SAS.)
- Published
- 2020
- Full Text
- View/download PDF
17. Prokineticin Receptor-1 Signaling Inhibits Dose- and Time-Dependent Anthracycline-Induced Cardiovascular Toxicity Via Myocardial and Vascular Protection.
- Author
-
Gasser A, Chen YW, Audebrand A, Daglayan A, Charavin M, Escoubet B, Karpov P, Tetko I, Chan MWY, Cardinale D, Désaubry L, and Nebigil CG
- Abstract
Objectives: This study investigated how different concentrations of doxorubicin (DOX) can affect the function of cardiac cells. This study also examined whether activation of prokineticin receptor (PKR)-1 by a nonpeptide agonist, IS20, prevents DOX-induced cardiovascular toxicity in mouse models., Background: High prevalence of heart failure during and following cancer treatments remains a subject of intense research and therapeutic interest., Methods: This study used cultured cardiomyocytes, endothelial cells (ECs), and epicardium-derived progenitor cells (EDPCs) for in vitro assays, tumor-bearing models, and acute and chronic toxicity mouse models for in vivo assays ., Results: Brief exposure to cardiomyocytes with high-dose DOX increased the accumulation of reactive oxygen species (ROS) by inhibiting a detoxification mechanism via stabilization of cytoplasmic nuclear factor, erythroid 2. Prolonged exposure to medium-dose DOX induced apoptosis in cardiomyocytes, ECs, and EDPCs. However, low-dose DOX promoted functional defects without inducing apoptosis in EDPCs and ECs. IS20 alleviated detrimental effects of DOX in cardiac cells by activating the serin threonin protein kinase B (Akt) or mitogen-activated protein kinase pathways. Genetic or pharmacological inactivation of PKR1 subdues these effects of IS20. In a chronic mouse model of DOX cardiotoxicity, IS20 normalized an elevated serum marker of cardiotoxicity and vascular and EDPC deficits, attenuated apoptosis and fibrosis, and improved the survival rate and cardiac function. IS20 did not interfere with the cytotoxicity or antitumor effects of DOX in breast cancer lines or in a mouse model of breast cancer, but it did attenuate the decreases in left ventricular diastolic volume induced by acute DOX treatment., Conclusions: This study identified the molecular and cellular signature of dose-dependent, DOX-mediated cardiotoxicity and provided evidence that PKR-1 is a promising target to combat cardiotoxicity of cancer treatments., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
18. The role of GPCR signaling in cardiac Epithelial to Mesenchymal Transformation (EMT).
- Author
-
Nebigil CG and Désaubry L
- Subjects
- Animals, Fetal Heart metabolism, Fetal Heart pathology, Fibrosis, Heart Defects, Congenital pathology, Humans, Morphogenesis, Myocardial Infarction pathology, Myocardium pathology, Epithelial-Mesenchymal Transition, Heart Defects, Congenital metabolism, Myocardial Infarction metabolism, Myocardium metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction
- Abstract
Congenital heart disease is the most common birth defect, affecting 1.35 million newborns every year. Heart failure is a primary cause of late morbidity and mortality after myocardial infarction. Heart development is involved in several rounds of epithelial-to-mesenchymal transition (EMT) and mesenchymal-to-epithelial transition (MET). Errors in these processes contribute to congenital heart disease, and exert deleterious effects on the heart and circulation after myocardial infarction. The identification of factors that are involved in heart development and disease, and the development of new approaches for the treatment of these disorders are of great interest. G protein coupled receptors (GPCRs) comprise 40% of clinically used drug targets, and their signaling are vital components of the heart during development, cardiac repair and in cardiac disease pathogenesis. This review focuses on the importance of EMT program in the heart, and outlines the newly identified GPCRs as potential therapeutic targets of reprogramming EMT to support cardiac cell fate during heart development and after myocardial infarction. More specifically we discuss prokineticin, serotonin, sphingosine-1-phosphate and apelin receptors in heart development and diseases. Further understanding of the regulation of EMT/MET by GPCRs during development and in the adult hearts can provide the following clinical exploitation of these pathways., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
19. Updates in Anthracycline-Mediated Cardiotoxicity.
- Author
-
Nebigil CG and Désaubry L
- Abstract
Cardiotoxicity is one of the main adverse effects of chemotheraphy, affecting the completion of cancer therapies and the short- and long-term quality of life. Anthracyclines are currently used to treat many cancers, including the various forms of leukemia, lymphoma, melanoma, uterine, breast, and gastric cancers. World Health Organization registered anthracyclines in the list of essential medicines. However, anthracyclines display a major cardiotoxicity that can ultimately culminate in congestive heart failure. Taking into account the growing rate of cancer survivorship, the clinical significance of anthracycline cardiotoxicity is an emerging medical issue. In this review, we focus on the key progenitor cells and cardiac cells (cardiomyocytes, fibroblasts, and vascular cells), focusing on the signaling pathways involved in cellular damage, and the clinical biomarkers in anthracycline-mediated cardiotoxicity.
- Published
- 2018
- Full Text
- View/download PDF
20. Emergence of cardio-oncology.
- Author
-
Nebigil CG and Désaubry L
- Subjects
- Antineoplastic Agents therapeutic use, Cardiology standards, Cardiotoxicity, Certification, Heart Diseases chemically induced, Heart Diseases therapy, Humans, Medical Oncology standards, Neoplasms complications, Neoplasms drug therapy, Patient Care Team, Antineoplastic Agents adverse effects, Cardiology trends, Heart Diseases etiology, Medical Oncology trends
- Abstract
Cardio-oncology is a new discipline that focuses on understanding, detection, monitoring and treating cardiovascular disease during and after cancer treatment. The development of this emerging field is based on an interdisciplinary collaboration between cardiology and oncology researchers and clinicians. Cardio-oncology aims at identifying how cancer therapies impact cardiovascular homeostasis, particular risk factors, diagnostic biomarkers and novel therapeutic approaches to help to effectively detect, prevent and cure the cardiotoxicity., (Copyright © 2018 Académie Nationale de Pharmacie. Published by Elsevier Masson SAS. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
21. A Prokineticin-Driven Epigenetic Switch Regulates Human Epicardial Cell Stemness and Fate.
- Author
-
Qureshi R, Kindo M, Boulberdaa M, von Hunolstein JJ, Steenman M, and Nebigil CG
- Subjects
- Cell Differentiation physiology, Epigenesis, Genetic, Epithelial-Mesenchymal Transition, Gastrointestinal Hormones genetics, Histone Demethylases metabolism, Humans, Neuropeptides genetics, Nuclear Proteins metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Stem Cells cytology, Stem Cells metabolism, Endothelial Cells cytology, Endothelial Cells metabolism, Gastrointestinal Hormones metabolism, Neuropeptides metabolism, Pericardium cytology, Pericardium metabolism
- Abstract
Epicardial adipose tissues (EATs) and vascular tissues may both belong to the mesoepithelial lineage that develops from epicardium-derived progenitor cells (EPDCs) in developing and injured hearts. Very little is known of the molecular mechanisms of EPDC contribution in EAT development and neovascularization in adult heart, which the topic remains a subject of intense therapeutic interest and scientific debate. Here we studied the epigenetic control of stemness and anti-adipogenic and pro-vasculogenic fate of human EPDCs (hEPDCs), through investigating an angiogenic hormone, prokineticin-2 (PK2) signaling via its receptor PKR1. We found that hEPDCs spontaneously undergoes epithelial-to-mesenchymal transformation (EMT), and are not predestined for the vascular lineages. However, PK2 via a histone demethylase KDM6A inhibits EMT, and induces asymmetric division, leading to self-renewal and formation of vascular and epithelial/endothelial precursors with angiogenic potential capable of differentiating into vascular smooth muscle and endothelial cells. PK2 upregulates and activates KDM6A to inhibit repressive histone H3K27me3 marks on promoters of vascular genes (Flk-1 and SM22α) involved in vascular lineage commitment and maturation. In PK2-mediated anti-adipogenic signaling, KDM6A stabilizes and increases cytoplasmic β-catenin levels to repress peroxisome proliferator-activated receptor-γ expression and activity. Our findings offer additional molecular targets to manipulate hEPDCs-involved tissue repair/regeneration in cardiometabolic and ischemic heart diseases. Stem Cells 2018;36:1589-1602., (© AlphaMed Press 2018.)
- Published
- 2018
- Full Text
- View/download PDF
22. Targeting prohibitin with small molecules to promote melanogenesis and apoptosis in melanoma cells.
- Author
-
Djehal A, Krayem M, Najem A, Hammoud H, Cresteil T, Nebigil CG, Wang D, Yu P, Bentouhami E, Ghanem GE, and Désaubry L
- Subjects
- Antineoplastic Agents chemistry, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Humans, Melanocytes drug effects, Melanoma pathology, Molecular Structure, Prohibitins, Repressor Proteins chemistry, Small Molecule Libraries chemistry, Structure-Activity Relationship, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, Apoptosis drug effects, Melanoma drug therapy, Repressor Proteins pharmacology, Small Molecule Libraries pharmacology
- Abstract
Prohibitins 1 and 2 (PHB1/2) are scaffold proteins that are involved in both melanogenesis and oncogenic pathways. We hypothesized that a PHB1 ligand, melanogenin, may display anti-cancer effects in addition to its known melanogenic activity in melanocytes. Here, we disclose a convenient synthesis of melanogenin, and its analogs. We found that, among 57 new melanogenin analogs, two (Mel9 and Mel41) significantly promoted both melanogenesis in melanocytes by activating one of the PHB2-interacting proteins, microtubule-associated protein light chain 3 (LC3), and upregulating the expression of microphthalmia associated transcription factor (MITF). These analogs also activate ERK. Besides, in addition to their promelanogenic activities, we uncovered that melanogenin and its active analogs induce apoptosis in several cancer cell lines, including melanoma cells, and that this effect is caused by an inhibition of AKT survival pathway. Our findings present a new putative function for PHBs as regulators of LC3/ERK/MITF melanogenic signaling, and suggest that Mel9 and Mel41 may provide the basis for the development of new drugs candidates to treat melanoma and other types of cancers., (Copyright © 2018 Elsevier Masson SAS. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
23. Prokineticin receptor-1-dependent paracrine and autocrine pathways control cardiac tcf21 + fibroblast progenitor cell transformation into adipocytes and vascular cells.
- Author
-
Qureshi R, Kindo M, Arora H, Boulberdaa M, Steenman M, and Nebigil CG
- Subjects
- Adipocytes metabolism, Adipose Tissue metabolism, Animals, Blood Cells metabolism, Cell Lineage, Diet, High-Fat, Fibroblasts metabolism, Gene Expression Regulation, Mice, Inbred C57BL, Mice, Transgenic, Models, Biological, Myocytes, Cardiac metabolism, Neovascularization, Physiologic, PPAR gamma genetics, PPAR gamma metabolism, Pericardium metabolism, Signal Transduction, Stem Cells metabolism, Adipocytes cytology, Autocrine Communication, Basic Helix-Loop-Helix Transcription Factors metabolism, Blood Cells cytology, Cell Transdifferentiation, Fibroblasts cytology, Paracrine Communication, Receptors, G-Protein-Coupled metabolism
- Abstract
Cardiac fat tissue volume and vascular dysfunction are strongly associated, accounting for overall body mass. Despite its pathophysiological significance, the origin and autocrine/paracrine pathways that regulate cardiac fat tissue and vascular network formation are unclear. We hypothesize that adipocytes and vasculogenic cells in adult mice hearts may share a common cardiac cells that could transform into adipocytes or vascular lineages, depending on the paracrine and autocrine stimuli. In this study utilizing transgenic mice overexpressing prokineticin receptor (PKR1) in cardiomyocytes, and tcf21ERT-cre
TM -derived cardiac fibroblast progenitor (CFP)-specific PKR1 knockout mice (PKR1tcf-/- ), as well as FACS-isolated CFPs, we showed that adipogenesis and vasculogenesis share a common CFPs originating from the tcf21+ epithelial lineage. We found that prokineticin-2 is a cardiomyocyte secretome that controls CFP transformation into adipocytes and vasculogenic cells in vivo and in vitro. Upon HFD exposure, PKR1tcf-/- mice displayed excessive fat deposition in the atrioventricular groove, perivascular area, and pericardium, which was accompanied by an impaired vascular network and cardiac dysfunction. This study contributes to the cardio-obesity field by demonstrating that PKR1 via autocrine/paracrine pathways controls CFP-vasculogenic- and CFP-adipocyte-transformation in adult heart. Our study may open up new possibilities for the treatment of metabolic cardiac diseases and atherosclerosis.- Published
- 2017
- Full Text
- View/download PDF
24. Prokineticin Is a New Linker between Obesity and Cardiovascular Diseases.
- Author
-
Nebigil CG
- Abstract
Obesity is a fast growing epidemic event worldwide. Fatness is associated with a number of comorbidities, including cardiovascular diseases (CVDs). Although obesity can be heredity in 30-70% cases, the environmental contributions also play an important role in the increasing prevalence of obesity. The relationship between development of obesity and CVD is poorly characterized. Obesity and CVD can also be resulted from a common mechanism such as metabolic, inflammatory, and neurohormonal changes. Prokineticins are defined as cytokines (immunoregulatory proteins), adipokines (adipocyte-secreted hormone), angiogenic (increasing vessel formation), or aneroxic (lowering food intake) hormones. Prokineticin-mediated signaling plays a key role in the development of obesity and CVD. Two forms of prokineticins exist in circulation and in various tissues including the brain, heart, kidney, and adipose. Prokineticins act on the two G protein-coupled receptors, namely, PKR1 and PKR2. Prokineticin-2 (PK2) via PKR1 receptor controls food intake and prevents adipose tissue expansion. The anti-adipocyte effect of PKR1 signaling is due to suppression of preadipocyte proliferation and differentiation capacity into adipocytes. PK2/PKR1 signaling promotes transcapillary passages of insulin and increases insulin sensitivity. It also plays an important role in the heart and kidney development and functions. Here, we discuss PK2 as a new adipocytokine in the association between obesity and CVD. We also highlight targeting PKR1 can be a new approach to treat obesity and CVD.
- Published
- 2017
- Full Text
- View/download PDF
25. Updates on Endothelial Functions of Proangiogenic Prokineticin.
- Author
-
Nebigil CG
- Subjects
- Animals, Cardiovascular Diseases metabolism, Cardiovascular Diseases physiopathology, Cell Differentiation, Cell Movement, Cells, Cultured, Humans, Hypertension metabolism, Hypertension physiopathology, Kidney Diseases metabolism, Kidney Diseases physiopathology, Mice, Molecular Biology, Myocytes, Cardiac metabolism, Renal Circulation physiology, Sensitivity and Specificity, Coronary Circulation physiology, Endothelium, Vascular metabolism, Endothelium, Vascular physiology, Receptors, G-Protein-Coupled metabolism, Receptors, Peptide metabolism
- Published
- 2016
- Full Text
- View/download PDF
26. Prokineticin receptor 1 is required for mesenchymal-epithelial transition in kidney development.
- Author
-
Arora H, Boulberdaa M, Qureshi R, Bitirim V, Messadeq N, Dolle P, and Nebigil CG
- Subjects
- Animals, Apoptosis, Cell Proliferation, Embryo, Mammalian metabolism, Embryonic Development, Epithelial-Mesenchymal Transition genetics, Mice, Mice, Knockout, Mutation, Neovascularization, Physiologic, Receptors, G-Protein-Coupled genetics, Epithelial-Mesenchymal Transition physiology, Gene Expression Regulation, Developmental physiology, Receptors, G-Protein-Coupled metabolism
- Abstract
Identification of factors regulating renal development is important to understand the pathogenesis of congenital kidney diseases. Little is known about the molecular mechanism of renal development and functions triggered by the angiogenic hormone prokineticin-2 and its receptor, PKR1. Utilizing the Gata5 (G5)-Cre and Wilms tumor 1 (Wt1)(GFP)cre transgenic lines, we generated mutant mice with targeted PKR1 gene disruptions in nephron progenitors. These mutant mice exhibited partial embryonic and postnatal lethality. Kidney developmental defects in PKR(G5-/-) mice are manifested in the adult stage as renal atrophy with glomerular defects, nephropathy, and uremia. PKR1(Wt1-/-) embryos exhibit hypoplastic kidneys with premature glomeruli and necrotic nephrons as a result of impaired proliferation and increased apoptosis in Wt1(+) renal mesenchymal cells. PKR1 regulates renal mesenchymal-epithelial transition (MET) that is involved in formation of renal progenitors, regulating glomerulogenesis toward forming nephrons during kidney development. In the isolated embryonic Wt1(+) renal cells, overexpression or activation of PKR1 promotes MET defined by the transition from elongated cell to octagonal cell morphology, and alteration of the expression of MET markers via activating NFATc3 signaling. Together, these results establish PKR1 via NFATc3 as a crucial modifier of MET processing to the development of nephron. Our study should facilitate new therapeutic opportunities in human renal disorders.-Arora, H., Boulberdaa, M., Qureshi, R., Bitirim, V., Messadeq, N., Dolle, P., Nebigil, C. G. Prokineticin receptor 1 is required for mesenchymal-epithelial transition in kidney development., (© FASEB.)
- Published
- 2016
- Full Text
- View/download PDF
27. Prokineticin receptor-1 signaling promotes Epicardial to Mesenchymal Transition during heart development.
- Author
-
Arora H, Boulberdaa M, Qureshi R, Bitirim V, Gasser A, Messaddeq N, Dolle P, and Nebigil CG
- Subjects
- Animals, Mice, Receptors, G-Protein-Coupled genetics, Epithelial-Mesenchymal Transition, Gastrointestinal Hormones metabolism, Heart embryology, Neuropeptides metabolism, Pericardium embryology, Receptors, G-Protein-Coupled metabolism, Signal Transduction
- Abstract
The epicardium plays an essential role in coronary artery formation and myocardial development. However, signals controlling the developing epicardium and epicardial-mesenchymal transition (EMT) in the normal and diseased adult heart are studied less rigorously. Here we investigated the role of angiogenic hormone, prokineticin-2 and its receptor PKR1 in the epicardium of developing and adult heart. Genetic ablation of PKR1 in epicardium leads to partial embryonic and postnatal lethality with abnormal heart development. Cardiac developmental defects are manifested in the adult stage as ischemic cardiomyopathy with systolic dysfunction. We discovered that PKR1 regulates epicardial-mesenchymal transition (EMT) for epicardial-derived progenitor cell (EPDC), formation. This event affects at least three consequential steps during heart development: (i) EPDC and cardiomyocyte proliferation involved in thickening of an outer compact ventricular chamber wall, (ii) rhythmicity, (iii) formation of coronary circulation. In isolated embryonic EPDCs, overexpression or activation of PKR1 alters cell morphology and EMT markers via activating Akt signaling. Lack of PKR1 signal in epicardium leads to defective heart development and underlies the origin of congenital heart disease in adult mice. Our mice provide genetic models for congenital dysfunction of the heart and should facilitate studies of both pathogenesis and therapy of cardiac disorders in humans.
- Published
- 2016
- Full Text
- View/download PDF
28. FL3, a Synthetic Flavagline and Ligand of Prohibitins, Protects Cardiomyocytes via STAT3 from Doxorubicin Toxicity.
- Author
-
Qureshi R, Yildirim O, Gasser A, Basmadjian C, Zhao Q, Wilmet JP, Désaubry L, and Nebigil CG
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Apoptosis drug effects, Blotting, Western, Cardiotoxicity, Cells, Cultured, Immunoenzyme Techniques, Immunoprecipitation, Ligands, Mice, Mitochondria drug effects, Mitochondria metabolism, Mitochondria pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Phosphorylation drug effects, Prohibitins, RNA, Small Interfering genetics, Rats, Repressor Proteins antagonists & inhibitors, Repressor Proteins genetics, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor genetics, Benzofurans pharmacology, Cardiotonic Agents pharmacology, Doxorubicin pharmacology, Myocytes, Cardiac drug effects, Repressor Proteins metabolism, STAT3 Transcription Factor metabolism
- Abstract
Aims: The clinical use of doxorubicin for the treatment of cancer is limited by its cardiotoxicity. Flavaglines are natural products that have both potent anticancer and cardioprotective properties. A synthetic analog of flavaglines, FL3, efficiently protects mice from the cardiotoxicity of doxorubicin. The mechanism underlying this cardioprotective effect has yet to be elucidated., Methods and Results: Here, we show that FL3 binds to the scaffold proteins prohibitins (PHBs) and thus promotes their translocation to mitochondria in the H9c2 cardiomyocytes. FL3 induces heterodimerization of PHB1 with STAT3, thereby ensuring cardioprotection from doxorubicin toxicity. This interaction is associated with phosphorylation of STAT3. A JAK2 inhibitor, WP1066, suppresses both the phosphorylation of STAT3 and the protective effect of FL3 in cardiomyocytes. The involvement of PHBs in the FL3-mediated cardioprotection was confirmed by means of small interfering RNAs (siRNAs) targeting PHB1 and PHB2. The siRNA knockdown of PHBs inhibits both phosphorylation of STAT3 and the cardioprotective effect of FL3., Conclusion: Activation of mitochondrial STAT3/PHB1 complex by PHB ligands may be a new strategy against doxorubicin-induced cardiotoxicity and possibly other cardiac problems.
- Published
- 2015
- Full Text
- View/download PDF
29. Can prokineticin prevent obesity and insulin resistance?
- Author
-
Von Hunolstein JJ and Nebigil CG
- Subjects
- Animals, Appetite Depressants therapeutic use, Eating drug effects, Humans, Anti-Obesity Agents therapeutic use, Insulin Resistance, Obesity prevention & control, Receptors, G-Protein-Coupled therapeutic use, Receptors, Peptide therapeutic use
- Abstract
Purpose of Review: Because of its increasing prevalence and morbi-mortality, obesity is a major health problem. Obesity etiology includes a combination of excess dietary calories and decreased physical activity, coupled with either predisposing genetic factors or metabolic disorders such as insulin resistance. Adipose tissue secretes several metabolically important proteins known as 'adipokines' that play a major role in obesity and insulin resistance. High levels of a newly identified group of adipokines, called prokineticins, have been found in obese adipose tissues. Prokineticins are peptide hormones released principally from macrophages and reproductive organs. They act on the G protein-coupled receptors PKR1 and PKR2. This review aims to provide an overview of current knowledge of the role of prokineticins and their receptors in the development of obesity and insulin resistance., Recent Findings: The principal biological effect of prokineticins in the central nervous system is the control of food intake. Nevertheless, peripheral biological effects of prokineticin are associated with increasing insulin sensitivity and suppressing the adipose tissue expansion., Summary: We outline the biological significance of the central and peripheral effects of prokineticins, and the potential of their receptors as targets for the treatment of obesity and insulin resistance.
- Published
- 2015
- Full Text
- View/download PDF
30. Discovery and cardioprotective effects of the first non-Peptide agonists of the G protein-coupled prokineticin receptor-1.
- Author
-
Gasser A, Brogi S, Urayama K, Nishi T, Kurose H, Tafi A, Ribeiro N, Désaubry L, and Nebigil CG
- Subjects
- Animals, Benzamides pharmacology, Benzamides therapeutic use, Binding Sites, Blood Pressure drug effects, CHO Cells, Calcium metabolism, Cells, Cultured, Computational Biology, Cricetinae, Cricetulus, Disease Models, Animal, Echocardiography, Endothelial Cells cytology, Endothelial Cells metabolism, Male, Mice, Mice, Inbred C57BL, Microscopy, Confocal, Molecular Docking Simulation, Myocardial Infarction drug therapy, Myocardial Infarction metabolism, Myocardial Infarction pathology, Peptides chemistry, Protective Agents pharmacology, Protective Agents therapeutic use, Pyridines pharmacology, Pyridines therapeutic use, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism, Signal Transduction drug effects, Benzamides chemistry, Protective Agents chemistry, Pyridines chemistry, Receptors, G-Protein-Coupled agonists
- Abstract
Prokineticins are angiogenic hormones that activate two G protein-coupled receptors: PKR1 and PKR2. PKR1 has emerged as a critical mediator of cardiovascular homeostasis and cardioprotection. Identification of non-peptide PKR1 agonists that contribute to myocardial repair and collateral vessel growth hold promises for treatment of heart diseases. Through a combination of in silico studies, medicinal chemistry, and pharmacological profiling approaches, we designed, synthesized, and characterized the first PKR1 agonists, demonstrating their cardioprotective activity against myocardial infarction (MI) in mice. Based on high throughput docking protocol, 250,000 compounds were computationally screened for putative PKR1 agonistic activity, using a homology model, and 10 virtual hits were pharmacologically evaluated. One hit internalizes PKR1, increases calcium release and activates ERK and Akt kinases. Among the 30 derivatives of the hit compound, the most potent derivative, IS20, was confirmed for its selectivity and specificity through genetic gain- and loss-of-function of PKR1. Importantly, IS20 prevented cardiac lesion formation and improved cardiac function after MI in mice, promoting proliferation of cardiac progenitor cells and neovasculogenesis. The preclinical investigation of the first PKR1 agonists provides a novel approach to promote cardiac neovasculogenesis after MI.
- Published
- 2015
- Full Text
- View/download PDF
31. Discovery of GPCR ligands for probing signal transduction pathways.
- Author
-
Brogi S, Tafi A, Désaubry L, and Nebigil CG
- Abstract
G protein-coupled receptors (GPCRs) are seven integral transmembrane proteins that are the primary targets of almost 30% of approved drugs and continue to represent a major focus of pharmaceutical research. All of GPCR targeted medicines were discovered by classical medicinal chemistry approaches. After the first GPCR crystal structures were determined, the docking screens using these structures lead to discovery of more novel and potent ligands. There are over 360 pharmaceutically relevant GPCRs in the human genome and to date about only 30 of structures have been determined. For these reasons, computational techniques such as homology modeling and molecular dynamics simulations have proven their usefulness to explore the structure and function of GPCRs. Furthermore, structure-based drug design and in silico screening (High Throughput Docking) are still the most common computational procedures in GPCRs drug discovery. Moreover, ligand-based methods such as three-dimensional quantitative structure-selectivity relationships, are the ideal molecular modeling approaches to rationalize the activity of tested GPCR ligands and identify novel GPCR ligands. In this review, we discuss the most recent advances for the computational approaches to effectively guide selectivity and affinity of ligands. We also describe novel approaches in medicinal chemistry, such as the development of biased agonists, allosteric modulators, and bivalent ligands for class A GPCRs. Furthermore, we highlight some knockout mice models in discovering biased signaling selectivity.
- Published
- 2014
- Full Text
- View/download PDF
32. Cancer wars: natural products strike back.
- Author
-
Basmadjian C, Zhao Q, Bentouhami E, Djehal A, Nebigil CG, Johnson RA, Serova M, de Gramont A, Faivre S, Raymond E, and Désaubry LG
- Abstract
Natural products have historically been a mainstay source of anticancer drugs, but in the 90's they fell out of favor in pharmaceutical companies with the emergence of targeted therapies, which rely on antibodies or small synthetic molecules identified by high throughput screening. Although targeted therapies greatly improved the treatment of a few cancers, the benefit has remained disappointing for many solid tumors, which revitalized the interest in natural products. With the approval of rapamycin in 2007, 12 novel natural product derivatives have been brought to market. The present review describes the discovery and development of these new anticancer drugs and highlights the peculiarities of natural product and new trends in this exciting field of drug discovery.
- Published
- 2014
- Full Text
- View/download PDF
33. Prokineticin receptor 1 as a novel suppressor of preadipocyte proliferation and differentiation to control obesity.
- Author
-
Szatkowski C, Vallet J, Dormishian M, Messaddeq N, Valet P, Boulberdaa M, Metzger D, Chambon P, and Nebigil CG
- Subjects
- 3T3-L1 Cells, Abdominal Fat pathology, Adipogenesis, Animals, Cell Proliferation, Diabetes Mellitus pathology, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Adipocytes metabolism, Adipocytes pathology, Cell Differentiation, Obesity pathology, Receptors, G-Protein-Coupled metabolism
- Abstract
Background: Adipocyte renewal from preadipocytes occurs throughout the lifetime and contributes to obesity. To date, little is known about the mechanisms that control preadipocyte proliferation and differentiation. Prokineticin-2 is an angiogenic and anorexigenic hormone that activate two G protein-coupled receptors (GPCRs): PKR1 and PKR2. Prokineticin-2 regulates food intake and energy metabolism via central mechanisms (PKR2). The peripheral effect of prokineticin-2 on adipocytes/preadipocytes has not been studied yet., Methodology/principal Findings: Since adipocytes and preadipocytes express mainly prokineticin receptor-1 (PKR1), here, we explored the role of PKR1 in adipose tissue expansion, generating PKR1-null (PKR1(-/-)) and adipocyte-specific (PKR1(ad-/-)) mutant mice, and using murine and human preadipocyte cell lines. Both PKR1(-/-) and PKR1(ad-/-) had excessive abdominal adipose tissue, but only PKR1(-/-) mice showed severe obesity and diabetes-like syndrome. PKR1(ad-/-)) mice had increased proliferating preadipocytes and newly formed adipocyte levels, leading to expansion of adipose tissue. Using PKR1-knockdown in 3T3-L1 preadipocytes, we show that PKR1 directly inhibits preadipocyte proliferation and differentiation. These PKR1 cell autonomous actions appear targeted at preadipocyte cell cycle regulatory pathways, through reducing cyclin D, E, cdk2, c-Myc levels., Conclusions/significance: These results suggest PKR1 to be a crucial player in the preadipocyte proliferation and differentiation. Our data should facilitate studies of both the pathogenesis and therapy of obesity in humans.
- Published
- 2013
- Full Text
- View/download PDF
34. Prokineticin receptor-1 is a new regulator of endothelial insulin uptake and capillary formation to control insulin sensitivity and cardiovascular and kidney functions.
- Author
-
Dormishian M, Turkeri G, Urayama K, Nguyen TL, Boulberdaa M, Messaddeq N, Renault G, Henrion D, and Nebigil CG
- Subjects
- Animals, Cell Proliferation, Endothelium, Vascular cytology, Male, Mice, Mice, Transgenic, Capillaries growth & development, Cardiovascular Physiological Phenomena, Endothelium, Vascular metabolism, Heart physiology, Insulin metabolism, Insulin Resistance physiology, Receptors, G-Protein-Coupled physiology
- Abstract
Background: Reciprocal relationships between endothelial dysfunction and insulin resistance result in a vicious cycle of cardiovascular, renal, and metabolic disorders. The mechanisms underlying these impairments are unclear. The peptide hormones prokineticins exert their angiogenic function via prokineticin receptor-1 (PKR1). We explored the extent to which endothelial PKR1 contributes to expansion of capillary network and the transcapillary passage of insulin into the heart, kidney, and adipose tissues, regulating organ functions and metabolism in a specific mice model., Methods and Results: By combining cellular studies and studies in endothelium-specific loss-of-function mouse model (ec-PKR1-/-), we showed that a genetically induced PKR1 loss in the endothelial cells causes the impaired capillary formation and transendothelial insulin delivery, leading to insulin resistance and cardiovascular and renal disorders. Impaired insulin delivery in endothelial cells accompanied with defective expression and activation of endothelial nitric oxide synthase in the ec-PKR1-/- aorta, consequently diminishing endothelium-dependent relaxation. Despite having a lean body phenotype, ec-PKR1-/- mice exhibited polyphagia, polydipsia, polyurinemia, and hyperinsulinemia, which are reminiscent of human lipodystrophy. High plasma free fatty acid levels and low leptin levels further contribute to the development of insulin resistance at the later age. Peripheral insulin resistance and ectopic lipid accumulation in mutant skeletal muscle, heart, and kidneys were accompanied by impaired insulin-mediated Akt signaling in these organs. The ec-PKR1-/- mice displayed myocardial fibrosis, low levels of capillary formation, and high rates of apoptosis, leading to diastolic dysfunction. Compact fibrotic glomeruli and high levels of phosphate excretion were found in mutant kidneys. PKR1 restoration in ec-PKR1-/- mice reversed the decrease in capillary recruitment and insulin uptake and improved heart and kidney function and insulin resistance., Conclusions: We show a novel role for endothelial PKR1 signaling in cardiac, renal, and metabolic functions by regulating transendothelial insulin uptake and endothelial cell proliferation. Targeting endothelial PKR1 may serve as a therapeutic strategy for ameliorating these disorders.
- Published
- 2013
- Full Text
- View/download PDF
35. Role of Prokineticin Receptor-1 in Epicardial Progenitor Cells.
- Author
-
Nguyen TL, Gasser A, and Nebigil CG
- Abstract
G protein-coupled receptors (GPCRs) form a large class of seven transmembrane (TM) domain receptors. The use of endogenous GPCR ligands to activate the stem cell maintenance or to direct cell differentiation would overcome many of the problems currently encountered in the use of stem cells, such as rapid in vitro differentiation and expansion or rejection in clinical applications. This review focuses on the definition of a new GPCR signaling pathway activated by peptide hormones, called "prokineticins", in epicardium-derived cells (EPDCs) . Signaling via prokineticin-2 and its receptor, PKR1, is required for cardiomyocyte survival during hypoxic stress. The binding of prokineticin-2 to PKR1 induces proliferation, migration and angiogenesis in endothelial cells. The expression of prokineticin and PKR1 increases during cardiac remodeling after myocardial infarction. Gain of function of PKR1 in the adult mouse heart revealed that cardiomyocyte-PKR1 signaling activates EPDCs in a paracrine fashion, thereby promoting de novo vasculogenesis. Transient PKR1 gene therapy after myocardial infarction in mice decreases mortality and improves heart function by promoting neovascularization, protecting cardiomyocytes and mobilizing WT1
+ cells. Furthermore, PKR1 signaling promotes adult EPDC proliferation and differentiation to adopt endothelial and smooth muscle cell fate, for the induction of de novo vasculogenesis. PKR1 is expressed in the proepicardium and epicardial cells derived from mice kidneys. Loss of PKR1 causes deficits in EPDCs in the neonatal mice hearts and kidneys and impairs vascularization and heart and kidney function. Taken together, these data indicate a novel role for PKR1 in heart-kidney complex via EPDCs.- Published
- 2013
- Full Text
- View/download PDF
36. Prohibitin ligands in cell death and survival: mode of action and therapeutic potential.
- Author
-
Thuaud F, Ribeiro N, Nebigil CG, and Désaubry L
- Subjects
- Animals, Cell Death drug effects, Cell Survival drug effects, Disease, Drug Discovery, Humans, Ligands, Prohibitins, Repressor Proteins metabolism
- Abstract
Prohibitins (PHBs) are scaffold proteins that modulate many signaling pathways controlling cell survival, metabolism, and inflammation. Several drugs that target PHBs have been identified and evaluated for various clinical applications. Preclinical and clinical studies indicate that these PHB ligands may be useful in oncology, cardiology, and neurology, as well as against obesity. This review covers the physiological role of PHBs in health and diseases and current developments concerning PHB ligands., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Flavaglines as potent anticancer and cytoprotective agents.
- Author
-
Ribeiro N, Thuaud F, Bernard Y, Gaiddon C, Cresteil T, Hild A, Hirsch EC, Michel PP, Nebigil CG, and Désaubry L
- Subjects
- Animals, Antibiotics, Antineoplastic toxicity, Antineoplastic Agents toxicity, Antineoplastic Agents, Phytogenic chemical synthesis, Apoptosis drug effects, Benzofurans pharmacology, Cardiotonic Agents chemical synthesis, Cells, Cultured, Cisplatin toxicity, Disease Models, Animal, Dopaminergic Neurons drug effects, Doxorubicin toxicity, Humans, Mice, Models, Molecular, Molecular Structure, Myocytes, Cardiac cytology, Neoplasms pathology, Parkinson Disease drug therapy, Rats, Structure-Activity Relationship, Antineoplastic Agents, Phytogenic pharmacology, Benzofurans chemistry, Biological Products pharmacology, Cardiotonic Agents pharmacology, Cytoprotection drug effects, Myocytes, Cardiac drug effects, Neoplasms drug therapy
- Abstract
Flavaglines represent a family of plant natural products that display potent anticancer, cardioprotective, and neuroprotective activities. Novel flavagline derivatives were synthesized and examined for their cytotoxicity on a panel of human cancer cell lines, their cardioprotection against doxorubicin-induced apoptosis in cardiomyocytes, and their neuroprotection in culture models of Parkinson's disease and cisplatin-induced neurotoxicity. The structural requirements of flavaglines for cardio- and neuroprotection were for the first time unraveled and appeared to be slightly different from those for cytotoxicity on cancer cells. We provide also the first evidence that flavaglines may alleviate cisplatin-induced neurotoxicity, suggesting a prophylactic potential of these compounds to prevent this frequently encountered adverse effect of cancer chemotherapies.
- Published
- 2012
- Full Text
- View/download PDF
38. Prokineticin receptor 1 (PKR1) signalling in cardiovascular and kidney functions.
- Author
-
Boulberdaa M, Urayama K, and Nebigil CG
- Subjects
- Animals, Coronary Vessels metabolism, Coronary Vessels physiopathology, Endothelial Cells metabolism, Humans, Kidney blood supply, Kidney physiopathology, Kidney Diseases metabolism, Kidney Diseases physiopathology, Mice, Mice, Transgenic, Muscle, Smooth, Vascular metabolism, Myocardial Infarction metabolism, Myocardial Infarction physiopathology, Myocytes, Smooth Muscle metabolism, Receptors, G-Protein-Coupled genetics, Stem Cells metabolism, Gastrointestinal Hormones metabolism, Kidney metabolism, Myocytes, Cardiac metabolism, Neovascularization, Physiologic, Neuropeptides metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction
- Abstract
Prokineticins (PK1 and PK2) are peptide hormones that exert their biological activity via two common G-protein-coupled receptors: prokineticin receptor (PKR) 1 and 2. Their physiology was originally explored mostly in the context of angiogenic actions in the reproductive tract and gut motility. Since autocrine and paracrine loops have been established between PK2 and PKR1 in the heart, in this review we focus on the PK2/PKR1 signalling in the functions of the heart and kidney. PKR1 signalling is required for cardiomyocyte survival and angiogenesis. In the mouse model of myocardial infarction, intracardiac transient PKR1 transfection protects the structure and function of the heart. Gain- and loss-of-function studies reveal that PKR1 in mouse heart up-regulates its own ligand and PK2, which in turn acts as a paracrine signal and promotes epicardin-positive progenitor cell differentiation into a vasculogenic cell type. Transgenic mice over-expressing PKR1 in cardiomyocytes exhibit increased neovascularization. Loss of PKR1 causes structural and functional changes in the heart and kidney. In isolated epicardin-positive progenitor cells from the kidney, PK2, acting via PKR1, stimulates differentiation of these progenitor cells into endothelial and smooth muscle cells. Taken together, these data show that PK2/PKR1 is involved in postnatal cardiac and renal neovascularization. The knowledge gained from these studies should facilitate the discovery of therapeutic interventions in heart and kidney diseases targeting PKR1.
- Published
- 2011
- Full Text
- View/download PDF
39. Genetic inactivation of prokineticin receptor-1 leads to heart and kidney disorders.
- Author
-
Boulberdaa M, Turkeri G, Urayama K, Dormishian M, Szatkowski C, Zimmer L, Messaddeq N, Laugel V, Dollé P, and Nebigil CG
- Subjects
- Aging, Animals, Apoptosis, Cell Differentiation, Cell Proliferation, Cells, Cultured, Genetic Predisposition to Disease, Heart Diseases metabolism, Heart Diseases pathology, Heart Diseases physiopathology, Kidney pathology, Kidney Diseases metabolism, Kidney Diseases pathology, Kidney Diseases physiopathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitochondria, Heart metabolism, Mitochondria, Heart pathology, Myocardium pathology, Neovascularization, Physiologic, Phenotype, Receptors, G-Protein-Coupled metabolism, Signal Transduction, Stem Cells metabolism, Stem Cells pathology, Ventricular Function, Left, Gene Silencing, Heart Diseases genetics, Kidney metabolism, Kidney Diseases genetics, Myocardium metabolism, Receptors, G-Protein-Coupled genetics
- Abstract
Objective: Prokineticins are potent angiogenic hormones that use 2 receptors, prokineticin receptor-1 (PKR1) and PKR2, with important therapeutic use in anticancer therapy. Observations of cardiac and renal toxicity in cancer patients treated with antiangiogenic compounds led us to explore how PKR1 signaling functioned in heart and kidney in vivo., Methods and Results: We generated mice with a conditional disruption of the PKR1 gene. We observed that PKR1 loss led to cardiomegaly, severe interstitial fibrosis, and cardiac dysfunction under stress conditions, accompanied by renal tubular dilation, reduced glomerular capillaries, urinary phosphate excretion, and proteinuria at later ages. Abnormal mitochondria and increased apoptosis were evident in both organs. Perturbation of capillary angiogenesis in both organs was restored at the adult stage potentially via upregulation of hypoxia-inducible factor-1 and proangiogenic factors. Compensatory mechanism could not revoke the epicardial and glomerular capillary networks, because of increased apoptosis and reduced progenitor cell numbers, consistent with an endogenous role of PKR1 signaling in stimulating epicardin+ progenitor cell proliferation and differentiation., Conclusions: Here, we showed for the first time that the loss of PKR1 causes renal and cardiac structural and functional changes because of deficits in survival signaling, mitochondrial, and progenitor cell functions in found both organs.
- Published
- 2011
- Full Text
- View/download PDF
40. Flavaglines alleviate doxorubicin cardiotoxicity: implication of Hsp27.
- Author
-
Bernard Y, Ribeiro N, Thuaud F, Türkeri G, Dirr R, Boulberdaa M, Nebigil CG, and Désaubry L
- Subjects
- Animals, Apoptosis drug effects, Benzofurans chemistry, Cardiotonic Agents chemistry, Cardiotonic Agents pharmacology, Culture Media, Serum-Free, Cytoprotection drug effects, Fibrosis, Male, Mice, Mice, Inbred BALB C, Phosphorylation drug effects, Benzofurans pharmacology, Doxorubicin adverse effects, HSP27 Heat-Shock Proteins metabolism, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology
- Abstract
Background: Despite its effectiveness in the treatment of various cancers, the use of doxorubicin is limited by a potentially fatal cardiomyopathy. Prevention of this cardiotoxicity remains a critical issue in clinical oncology. We hypothesized that flavaglines, a family of natural compounds that display potent neuroprotective effects, may also alleviate doxorubicin-induced cardiotoxicity., Methodology/principal Findings: Our in vitro data established that a pretreatment with flavaglines significantly increased viability of doxorubicin-injured H9c2 cardiomyocytes as demonstrated by annexin V, TUNEL and active caspase-3 assays. We demonstrated also that phosphorylation of the small heat shock protein Hsp27 is involved in the mechanism by which flavaglines display their cardioprotective effect. Furthermore, knocking-down Hsp27 in H9c2 cardiomyocytes completely reversed this cardioprotection. Administration of our lead compound (FL3) to mice attenuated cardiomyocyte apoptosis and cardiac fibrosis, as reflected by a 50% decrease of mortality., Conclusions/significance: These results suggest a prophylactic potential of flavaglines to prevent doxorubicin-induced cardiac toxicity.
- Published
- 2011
- Full Text
- View/download PDF
41. Divergent roles of prokineticin receptors in the endothelial cells: angiogenesis and fenestration.
- Author
-
Guilini C, Urayama K, Turkeri G, Dedeoglu DB, Kurose H, Messaddeq N, and Nebigil CG
- Subjects
- Animals, Cell Line, Cell Movement, Cell Proliferation, Cells, Cultured, Endothelium, Vascular cytology, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, Humans, Membrane Proteins metabolism, Mice, Mitogen-Activated Protein Kinase Kinases physiology, Models, Animal, Phosphoproteins metabolism, Proto-Oncogene Proteins c-akt physiology, Receptors, G-Protein-Coupled genetics, Receptors, Peptide genetics, Signal Transduction physiology, Zonula Occludens-1 Protein, Endothelium, Vascular physiology, Neovascularization, Physiologic physiology, Receptors, G-Protein-Coupled physiology, Receptors, Peptide physiology
- Abstract
Prokineticins are secreted peptides that activate two G protein-coupled receptors: PKR1 and PKR2. Prokineticins induce angiogenesis and fenestration, but the cognate receptors involved in these functions are unknown. We hypothesized a role for prokineticin receptor signaling pathways and expression profiles in determining the selective effects of prokineticins on coronary endothelial cells (H5V). Activation of the PKR1/MAPK/Akt signaling pathway stimulates proliferation, migration, and angiogenesis in H5V cells, in which PKR1 predominates over PKR2. PKR1 was colocalized with Galpha(11) and was internalized following the stimulation of these cells with prokineticin-2. Knock down of PKR1 or Galpha(11) expression in H5V cells effectively inhibited prokineticin-2-induced vessel formation and MAPK/Akt activation, indicating a role for PKR1/Galpha(11) in this process. However, in conditions in which PKR2 predominated over PKR1, these cells displayed a fenestrated endothelial cell phenotype. H5V cells overexpressing PKR2 displayed large numbers of multivesicular bodies and caveolar clusters and a disruption of the distribution of zonula occluden-1 tight junction protein. Prokineticin-2 induced the colocalization of PKR2 with Galpha(12), and activated Galpha(12), which bound to zonula occluden-1 to trigger the degradation of this protein in these cells. Prokineticin-2 induced the formation of vessel-like structures by human aortic endothelial cells expressing only PKR1, and disorganized the tight junctions in human hepatic sinusoidal endothelial cells expressing only PKR2, confirming the divergent roles of these receptors. Our findings show the functional characteristics of coronary endothelial cells depend on the expression of PKR1 and PKR2 levels and the divergent signaling pathways used by these receptors.
- Published
- 2010
- Full Text
- View/download PDF
42. Synthetic analogue of rocaglaol displays a potent and selective cytotoxicity in cancer cells: involvement of apoptosis inducing factor and caspase-12.
- Author
-
Thuaud F, Bernard Y, Türkeri G, Dirr R, Aubert G, Cresteil T, Baguet A, Tomasetto C, Svitkin Y, Sonenberg N, Nebigil CG, and Désaubry L
- Subjects
- Active Transport, Cell Nucleus, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Apoptosis, Benzofurans chemistry, Benzofurans pharmacology, Cell Line, Tumor, Cell Nucleus metabolism, Cell Proliferation drug effects, Cell Survival drug effects, Doxorubicin pharmacology, Drug Resistance, Neoplasm, Drug Screening Assays, Antitumor, Drug Synergism, Endoplasmic Reticulum metabolism, G2 Phase drug effects, Humans, Stereoisomerism, Structure-Activity Relationship, Antineoplastic Agents chemical synthesis, Apoptosis Inducing Factor metabolism, Benzofurans chemical synthesis, Caspase 12 metabolism
- Abstract
Flavaglines constitute a family of natural anticancer compounds. We present here 3 (FL3), the first synthetic flavagline that inhibits cell proliferation and viability (IC(50) approximately 1 nM) at lower doses than did the parent compound, racemic rocaglaol. Compound 3 enhanced doxorubicin cytotoxicity in HepG2 cells and retained its potency against adriamycin-resistant cell lines without inducing cardiomyocyte toxicity. Compound 3 induced apoptosis of HL60 and Hela cells by triggering the translocation of Apoptosis Inducing Factor (AIF) and caspase-12 to the nucleus. A fluorescent conjugate of 3 accumulated in endoplasmic reticulum (ER), suggesting that flavaglines bind to their target in the ER, where it triggers a cascade of events that leads to the translocation of AIF and caspase-12 to the nucleus and probably inhibition of eIF4A. Our studies highlight structural features critical to their antineoplastic potential and suggest that these compounds would retain their activity in cells refractory to caspase activation.
- Published
- 2009
- Full Text
- View/download PDF
43. Prokineticin receptors in cardiovascular function: foe or friend?
- Author
-
Nebigil CG
- Subjects
- Animals, Cardiomegaly metabolism, Cardiomyopathy, Dilated metabolism, Cell Differentiation, Gastrointestinal Hormones metabolism, Genetic Therapy, Heart Diseases genetics, Heart Diseases physiopathology, Heart Diseases therapy, Humans, Myocardial Infarction metabolism, Neovascularization, Physiologic, Neuropeptides metabolism, Receptors, G-Protein-Coupled genetics, Receptors, Peptide metabolism, Stem Cells metabolism, Vascular Endothelial Growth Factor, Endocrine-Gland-Derived metabolism, Endothelial Cells metabolism, Heart Diseases metabolism, Myocytes, Cardiac metabolism, Receptors, G-Protein-Coupled metabolism, Signal Transduction
- Abstract
Prokineticins are small secreted bioactive peptides, comprising two classes: prokineticin-1 and prokineticin-2. They exert their biological activity by binding to two G-protein-coupled receptors: prokineticin receptor (PKR) 1 and 2. Recent data have demonstrated that PKR1 induces postnatal neovasculogenesis by activating adult epicardial-derived progenitor cell differentiation, whereas myocardial PRK2 signaling confers detrimental actions on cardiomyocytes, leading to dilated cardiomyopathy and release of an unknown paracrine factor to induce capillary fenestration and vascular leakage. The knowledge gained from these studies leads to a model in which PKR1 and PKR2 signaling exert opposing actions in heart physiology and pathophysiology and facilitate the discovery of specific agonists and antagonists targeting PKR1 and PKR2 for possible use in treatment of cardiovascular diseases.
- Published
- 2009
- Full Text
- View/download PDF
44. Transgenic myocardial overexpression of prokineticin receptor-2 (GPR73b) induces hypertrophy and capillary vessel leakage.
- Author
-
Urayama K, Dedeoglu DB, Guilini C, Frantz S, Ertl G, Messaddeq N, and Nebigil CG
- Subjects
- Animals, Arterioles pathology, Arterioles physiopathology, Blood Pressure physiology, Capillary Leak Syndrome metabolism, Capillary Leak Syndrome pathology, Cardiomegaly metabolism, Cardiomegaly pathology, Cell Membrane Permeability physiology, Cells, Cultured, Coronary Vessels pathology, Coronary Vessels physiopathology, Disease Models, Animal, Endothelium, Vascular pathology, Endothelium, Vascular physiopathology, Heart Rate physiology, Mice, Mice, Transgenic, Myocardium pathology, Myocytes, Cardiac pathology, Myocytes, Cardiac ultrastructure, Receptors, G-Protein-Coupled genetics, Signal Transduction physiology, Capillary Leak Syndrome etiology, Cardiomegaly etiology, Myocardium metabolism, Myocytes, Cardiac metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Aims: Prokineticins are small secreted bioactive molecules. They exert their biological activity by binding to two G protein-coupled receptors. Previously, we have shown that the overexpression of prokineticin receptor-1 (PKR1) in transgenic (TG) mouse hearts induced neovascularization. Since PKR1 and PKR2 are 85% identical and expressed in cardiovascular tissues, we hypothesized that PKR2 may also contribute to cardiomyocyte growth and vascularization., Methods and Results: We have generated TG mice overexpressing PKR2 in cardiomyocytes. TG mice exhibit increased hypertrophic gene expression and heart-to-body weight ratio accompanied by an increased length of cardiomyocytes at the age of 12 weeks. Increased left ventricular end-systolic and diastolic diameters without cardiac dysfunction at the age of 24 weeks indicate that TG mice have an eccentric hypertrophy with compensated cardiac function. Quantitative morphological analysis showed that TG hearts have a normal microvessel density and number of branch points. However, they exhibit increased abnormal endothelial cell shape and ultrastructure, changed cellular distribution of a tight junction protein zona occludens-1 (ZO-1), and vascular leakage in heart without a rise of angiogenic factor levels at early and late age. The application of media conditioned by H9c2 cardioblast cells overexpressing PKR2 significantly induced impaired ZO-1 localization in H5V endothelial cells, mimicking the TG model., Conclusion: These findings provide the first genetic evidence that cardiomyocyte PKR2 signalling leads to eccentric hypertrophy in an autocrine regulation and impaired endothelial integrity in a paracrine regulation without inducing angiogenesis. These TG mice may provide a new genetic model for heart diseases.
- Published
- 2009
- Full Text
- View/download PDF
45. Prokineticin receptor-1 induces neovascularization and epicardial-derived progenitor cell differentiation.
- Author
-
Urayama K, Guilini C, Turkeri G, Takir S, Kurose H, Messaddeq N, Dierich A, and Nebigil CG
- Subjects
- Animals, Cell Movement physiology, Cell Proliferation, Coculture Techniques, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Gastrointestinal Hormones genetics, Gastrointestinal Hormones metabolism, Mice, Mice, Transgenic, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Neuropeptides genetics, Neuropeptides metabolism, Pericardium metabolism, Protein Kinase C genetics, Signal Transduction genetics, Signal Transduction physiology, Stem Cells metabolism, Up-Regulation, Cell Differentiation physiology, Neovascularization, Physiologic physiology, Pericardium pathology, Protein Kinase C metabolism, Stem Cells pathology
- Abstract
Objective: Identification of novel factors that contribute to myocardial repair and collateral vessel growth hold promise for treatment of heart diseases. We have shown that transient prokineticin receptor-1 (PKR1) gene transfer protects the heart against myocardial infarction in a mouse model. Here, we investigated the role of excessive PKR1 signaling in heart., Methods and Results: Transgenic mice overexpressing PKR1 in cardiomyocytes displayed no spontaneous abnormalities in cardiomyocytes but showed an increased number of epicardial-derived progenitor cells (EPDCs), capillary density, and coronary arterioles. Coculturing EPDCs with H9c2 cardiomyoblasts overexpressing PKR1 promotes EPDC differentiation into endothelial and smooth muscle cells, mimicking our transgenic model. Overexpressing PKR1 in H9c2 cardiomyoblasts or in transgenic hearts upregulated prokineticin-2 levels. Exogenous prokineticin-2 induces significant outgrowth from neonatal and adult epicardial explants, promoting EPDC differentiation. These prokineticin-2 effects were abolished in cardiac explants from mice with PKR1-null mutation. Reduced capillary density and prokineticin-2 levels in PKR1-null mutant hearts supports the hypothesis of an autocrine/paracrine loop between PKR1 and prokineticin-2., Conclusions: Cardiomyocyte-PKR1 signaling upregulates its own ligand prokineticin-2 that acts as a paracrine factor, triggering EPDCs proliferation/differentiation. This study provides a novel insight for possible therapeutic strategies aiming at restoring pluripotency of adult EPDCs to promote neovasculogenesis by induction of cardiomyocyte PKR1 signaling.
- Published
- 2008
- Full Text
- View/download PDF
46. The prokineticin receptor-1 (GPR73) promotes cardiomyocyte survival and angiogenesis.
- Author
-
Urayama K, Guilini C, Messaddeq N, Hu K, Steenman M, Kurose H, Ert G, and Nebigil CG
- Subjects
- Adenoviridae genetics, Animals, Apoptosis, Cell Hypoxia, Cells, Cultured, Embryo, Mammalian, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Genetic Therapy, In Situ Hybridization, Male, Mice, Mice, Inbred C57BL, Myocardial Infarction metabolism, Myocardial Infarction prevention & control, Myocardial Ischemia metabolism, Myocardial Reperfusion Injury metabolism, Myocardial Reperfusion Injury prevention & control, Myocardium metabolism, Myocardium pathology, Myocytes, Cardiac cytology, Proto-Oncogene Proteins c-akt metabolism, RNA Probes, RNA, Small Interfering pharmacology, Rats, Receptors, G-Protein-Coupled antagonists & inhibitors, Receptors, G-Protein-Coupled genetics, Receptors, Peptide genetics, Receptors, Peptide metabolism, Heart physiology, Myocardial Ischemia prevention & control, Myocytes, Cardiac metabolism, Neovascularization, Pathologic, Receptors, G-Protein-Coupled metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Prokineticins are potent angiogenic factors that bind to two G protein-coupled receptors to initiate their biological effects. We hypothesize that prokineticin receptor-1 (PKR1/GPR73) signaling may contribute to cardiomyocyte survival or repair in myocardial infarction. Since we showed that prokineticin-2 and PKR1 are expressed in adult mouse heart and cardiac cells, we investigated the role of prokineticin-2 on capillary endothelial cell and cardiomyocyte function. In cultured cardiac endothelial cells, prokineticin-2 or overexpression of PKR1 induces vessel-like formation without increasing VEGF levels. In cardiomyocytes and H9c2 cells, prokineticin-2 or overexpressing PKR1 activates Akt to protect cardiomyocytes against oxidative stress. The survival and angiogenesis promoting effects of prokineticin-2 in cardiac cells were completely reversed by siRNA-PKR1, indicating PKR1 involvement. We thus, further investigated whether intramyocardial gene transfer of DNA encoding PKR1 may rescue the myocardium against myocardial infarction in mouse model. Transient PKR1 gene transfer after coronary ligation reduces mortality and preserves left ventricular function by promoting neovascularization and protecting cardiomyocytes without altering VEGF levels. In human end-stage failing heart samples, reduced PKR1 and prokineticin-2 transcripts and protein levels implicate a more important role for prokineticin-2/PKR1 signaling in heart. Our results suggest that PKR1 may represent a novel therapeutic target to limit myocardial injury following ischemic events.
- Published
- 2007
- Full Text
- View/download PDF
47. Involvement of the serotonin 5-HT2B receptor in cardiac hypertrophy linked to sympathetic stimulation: control of interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha cytokine production by ventricular fibroblasts.
- Author
-
Jaffré F, Callebert J, Sarre A, Etienne N, Nebigil CG, Launay JM, Maroteaux L, and Monassier L
- Subjects
- Adrenergic beta-1 Receptor Antagonists, Adrenergic beta-2 Receptor Antagonists, Adrenergic beta-Agonists toxicity, Animals, Cardiomegaly chemically induced, Cardiomegaly etiology, Cardiomegaly genetics, Cardiomegaly prevention & control, Cells, Cultured drug effects, Cells, Cultured metabolism, Drug Evaluation, Preclinical, Fibroblasts drug effects, Gene Expression Regulation drug effects, Gene Expression Regulation physiology, Heart Ventricles cytology, Imidazoles pharmacology, Interleukin-1 biosynthesis, Interleukin-1 blood, Interleukin-1 genetics, Interleukin-6 biosynthesis, Interleukin-6 blood, Interleukin-6 genetics, Isoproterenol toxicity, Mice, Mice, Knockout, Propanolamines pharmacology, Pyridines pharmacology, Quinolines therapeutic use, Receptor, Serotonin, 5-HT2B deficiency, Receptor, Serotonin, 5-HT2B genetics, Receptors, Adrenergic, beta-1 analysis, Receptors, Adrenergic, beta-2 analysis, Serotonin 5-HT2 Receptor Antagonists, Serotonin Antagonists pharmacology, Sympathetic Nervous System drug effects, Sympathomimetics toxicity, Tumor Necrosis Factor-alpha analysis, Tumor Necrosis Factor-alpha biosynthesis, Tumor Necrosis Factor-alpha genetics, Cardiomegaly physiopathology, Fibroblasts metabolism, Indoles pharmacology, Indoles therapeutic use, Myocytes, Cardiac cytology, Pyridines therapeutic use, Quinolines pharmacology, Receptor, Serotonin, 5-HT2B physiology, Serotonin Antagonists therapeutic use, Sympathetic Nervous System physiopathology
- Abstract
Background: The serotonergic 5-HT2B receptor regulates cardiomyocyte development and growth. A putative contribution of this receptor to fibroblast-dependent cardiac function has not been identified., Methods and Results: By mimicking sympathetic stimulation with chronic isoproterenol perfusion in vivo, we found that mice developed a cardiac hypertrophy, which was prevented by exposure to the 5-HT2B receptor antagonists SB206553 or SB215505 or in 5-HT2B receptor-knockout mice. The isoproterenol-induced hypertrophy was associated with an increase in the plasma levels of interleukin-1beta and tumor necrosis factor-alpha but not interleukin-6. In contrast, the plasma isoproterenol-induced cytokine increase was not observed in either 5-HT2B receptor-mutant or wild-type mice perfused with isoproterenol+SB206553. We demonstrated that stimulation of wild-type cardiac fibroblasts by isoproterenol markedly increased the production of the interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha cytokines. Strikingly, we found that this isoproterenol-induced cytokine production was abolished by SB206553 or in 5-HT2B receptor-knockout fibroblasts. Serotonin also stimulated production of the 3 cytokines in wild-type fibroblasts, which was effectively reduced in 5-HT2B receptor-knockout fibroblasts., Conclusions: Our results demonstrate for the first time that 5-HT2B receptors are essential for isoproterenol-induced cardiac hypertrophy, which involves the regulation of interleukin-6, interleukin-1beta, and tumor necrosis factor-alpha cytokine production by cardiac fibroblasts.
- Published
- 2004
- Full Text
- View/download PDF
48. Overexpression of the serotonin 5-HT2B receptor in heart leads to abnormal mitochondrial function and cardiac hypertrophy.
- Author
-
Nebigil CG, Jaffré F, Messaddeq N, Hickel P, Monassier L, Launay JM, and Maroteaux L
- Subjects
- Animals, Atrial Natriuretic Factor metabolism, Cardiomegaly diagnostic imaging, Cardiomegaly pathology, Cell Count, Cell Size, Disease Models, Animal, Echocardiography, Electron Transport Complex IV metabolism, GTP-Binding Protein alpha Subunits, Gq-G11, Gene Expression, Heart Ventricles pathology, Heterotrimeric GTP-Binding Proteins metabolism, Male, Mice, Mice, Transgenic, Mitochondria pathology, Mitochondria ultrastructure, Mitochondrial ADP, ATP Translocases metabolism, Myocardium pathology, Myocytes, Cardiac pathology, Myosin Heavy Chains metabolism, Organ Specificity, Receptor, Serotonin, 5-HT2B, Receptors, Serotonin genetics, Signal Transduction, Succinate Dehydrogenase metabolism, Cardiomegaly physiopathology, Mitochondria metabolism, Myocardium metabolism, Receptors, Serotonin biosynthesis
- Abstract
Background: Identification of factors regulating myocardial structure and function is important to understand the pathogenesis of heart disease. We previously reported that 5-HT2B receptor ablation in mice leads to dilated cardiomyopathy. In this study, we investigated the pathological consequence of overexpressing 5-HT2B receptors in heart in vivo., Methods and Results: We have generated transgenic mice overexpressing the Gq-coupled 5-HT2B receptor specifically in heart. We found that overexpression of 5-HT2B receptor in heart leads to ventricular hypertrophy as the result of increased cell number and size. Increased atrial natriuretic peptide and myosin heavy chain expression demonstrated activation of the molecular program for cardiac hypertrophy. Echocardiographic analysis indicated the presence of thickened ventricular free wall without alteration of the systolic function, showing that transgenic mice have compensated hypertrophy. Electron microscopic analysis revealed structural abnormalities including mitochondrial proliferation, as also manifested by histological staining. Transgenic mouse heart displayed a specific reduction in the expression levels of the adenine nucleotide translocator associated to increase in the succinate dehydrogenase and cytochrome C oxidase mitochondrial activities., Conclusions: Our results constitute the first genetic evidence that overexpression of the 5-HT2B receptor in the heart leads to compensated hypertrophic cardiomyopathy associated with proliferation of the mitochondria. This observation suggests a role for mitochondria in the hypertrophic signaling that is regulated by serotonin. These transgenic mice provide a new genetic model for hypertrophic heart disease.
- Published
- 2003
- Full Text
- View/download PDF
49. Serotonin is a novel survival factor of cardiomyocytes: mitochondria as a target of 5-HT2B receptor signaling.
- Author
-
Nebigil CG, Etienne N, Messaddeq N, and Maroteaux L
- Subjects
- Adenine Nucleotide Translocator 1 metabolism, Animals, Apoptosis, Cardiomyopathy, Dilated etiology, Caspases metabolism, Cell Survival, Cells, Cultured, Culture Media, Serum-Free, Cytochrome c Group metabolism, Cytoprotection, Mice, Mice, Knockout, Mitochondria, Heart physiology, Mitochondria, Heart ultrastructure, Mitogen-Activated Protein Kinases metabolism, Myocytes, Cardiac cytology, Myocytes, Cardiac ultrastructure, NF-kappa B metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Receptor, Serotonin, 5-HT2B, bcl-2-Associated X Protein, Mitochondria, Heart metabolism, Myocytes, Cardiac metabolism, Protein Serine-Threonine Kinases, Proto-Oncogene Proteins c-bcl-2, Receptors, Serotonin metabolism, Serotonin pharmacology, Signal Transduction
- Abstract
Identification of factors regulating cardiomyocyte survival and growth is important to understand the pathogenesis of congenital heart diseases. Little is known about the molecular mechanism of cardiac functions triggered by serotonin. The link between signaling circuitry of external stimuli and the mitochondrial apoptotic machinery is of wide interest in cardiac diseases. Using cultured cardiomyocytes and 5-hydroxytryptamine (5-HT)2B-receptor knockout mice as an animal model of dilated cardiomyopathy, for the first time we show that serotonin via the Gq-coupled 5-HT2B-receptor protect cardiomyocytes against serum deprivation-induced apoptosis as manifested by DNA fragmentation, nuclear chromatin condensation, and TUNEL labeling. Serotonin prevents cytochrome c release and caspase-9 and -3 activation after serum deprivation via cross-talks between phosphatidylinositol-3 kinase/Akt and extracellular signal-regulated kinase (ERK) 1/2 signaling pathways. Serotonin binding to 5-HT2B-receptor activates ERK kinases to inhibit Bax expression induced by serum deprivation. Serotonin via phosphatidylinositol-3 kinase/Akt can activate NF-kappaB that is required for the regulation of the mitochondrial adenine nucleotide translocator (ANT-1). Parallel to these observations, ultrastructural analysis in the 5-HT2B-receptor knockout mice heart revealed pronounced mitochondrial defects in addition to altered mitochondrial enzyme activities (cytochrome oxidase and succinate dehydrogenase) and ANT-1 and Bax expressions. These findings identify 5-HT as a novel survival factor targeting mitochondria in cardiomyocytes.
- Published
- 2003
- Full Text
- View/download PDF
50. Function of the serotonin 5-hydroxytryptamine 2B receptor in pulmonary hypertension.
- Author
-
Launay JM, Hervé P, Peoc'h K, Tournois C, Callebert J, Nebigil CG, Etienne N, Drouet L, Humbert M, Simonneau G, and Maroteaux L
- Subjects
- Animals, Blood Pressure, Cell Division, DNA biosynthesis, Dexfenfluramine metabolism, Dexfenfluramine pharmacology, Disease Models, Animal, Female, Humans, Hypertension, Pulmonary pathology, Hypoxia physiopathology, Lung blood supply, Lung metabolism, Lung pathology, Male, Mice, Organ Culture Techniques, Pulmonary Artery metabolism, Pulmonary Artery pathology, Pyrimidines pharmacology, Receptor, Serotonin, 5-HT2B, Receptors, Serotonin genetics, Serotonin metabolism, Serotonin Antagonists metabolism, Serotonin Receptor Agonists pharmacology, Vasoconstriction, Hypertension, Pulmonary metabolism, Receptors, Serotonin metabolism
- Abstract
Primary pulmonary hypertension is a progressive and often fatal disorder in humans that results from an increase in pulmonary blood pressure associated with abnormal vascular proliferation. Dexfenfluramine increases the risk of pulmonary hypertension in humans, and its active metabolite is a selective serotonin 5-hydroxytryptamine 2B (5-HT(2B)) receptor agonist. Thus, we investigated the contribution of the 5-HT(2B)receptor to the pathogenesis of pulmonary hypertension. Using the chronic-hypoxic-mouse model of pulmonary hypertension, we found that the hypoxia-dependent increase in pulmonary blood pressure and lung remodeling are associated with an increase in vascular proliferation, elastase activity and transforming growth factor-beta levels, and that these parameters are potentiated by dexfenfluramine treatment. In contrast, hypoxic mice with genetically or pharmacologically inactive 5-HT(2B)receptors manifested no change in any of these parameters. In both humans and mice, pulmonary hypertension is associated with a substantial increase in 5-HT(2B) receptor expression in pulmonary arteries. These data show that activation of 5-HT(2B) receptors is a limiting step in the development of pulmonary hypertension.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.