1
|
Ham H, Jing H, Lamborn IT, Kober MM, Koval A, Berchiche YA, Anderson DE, Druey KM, Mandl JN, Isidor B, Ferreira CR, Freeman AF, Ganesan S, Karsak M, Mustillo PJ, Teo J, Zolkipli-Cunningham Z, Chatron N, Lecoquierre F, Oler AJ, Schmid JP, Kuhns DB, Xu X, Hauck F, Al-Herz W, Wagner M, Terhal PA, Muurinen M, Barlogis V, Cruz P, Danielson J, Stewart H, Loid P, Rading S, Keren B, Pfundt R, Zarember KA, Vill K, Potocki L, Olivier KN, Lesca G, Faivre L, Wong M, Puel A, Chou J, Tusseau M, Moutsopoulos NM, Matthews HF, Simons C, Taft RJ, Soldatos A, Masle-Farquhar E, Pittaluga S, Brink R, Fink DL, Kong HH, Kabat J, Kim WS, Bierhals T, Meguro K, Hsu AP, Gu J, Stoddard J, Banos-Pinero B, Slack M, Trivellin G, Mazel B, Soomann M, Li S, Watts VJ, Stratakis CA, Rodriguez-Quevedo MF, Bruel AL, Lipsanen-Nyman M, Saultier P, Jain R, Lehalle D, Torres D, Sullivan KE, Barbarot S, Neu A, Duffourd Y, Similuk M, McWalter K, Blanc P, Bézieau S, Jin T, Geha RS, Casanova JL, Makitie OM, Kubisch C, Edery P, Christodoulou J, Germain RN, Goodnow CC, Sakmar TP, Billadeau DD, Küry S, Katanaev VL, Zhang Y, et alHam H, Jing H, Lamborn IT, Kober MM, Koval A, Berchiche YA, Anderson DE, Druey KM, Mandl JN, Isidor B, Ferreira CR, Freeman AF, Ganesan S, Karsak M, Mustillo PJ, Teo J, Zolkipli-Cunningham Z, Chatron N, Lecoquierre F, Oler AJ, Schmid JP, Kuhns DB, Xu X, Hauck F, Al-Herz W, Wagner M, Terhal PA, Muurinen M, Barlogis V, Cruz P, Danielson J, Stewart H, Loid P, Rading S, Keren B, Pfundt R, Zarember KA, Vill K, Potocki L, Olivier KN, Lesca G, Faivre L, Wong M, Puel A, Chou J, Tusseau M, Moutsopoulos NM, Matthews HF, Simons C, Taft RJ, Soldatos A, Masle-Farquhar E, Pittaluga S, Brink R, Fink DL, Kong HH, Kabat J, Kim WS, Bierhals T, Meguro K, Hsu AP, Gu J, Stoddard J, Banos-Pinero B, Slack M, Trivellin G, Mazel B, Soomann M, Li S, Watts VJ, Stratakis CA, Rodriguez-Quevedo MF, Bruel AL, Lipsanen-Nyman M, Saultier P, Jain R, Lehalle D, Torres D, Sullivan KE, Barbarot S, Neu A, Duffourd Y, Similuk M, McWalter K, Blanc P, Bézieau S, Jin T, Geha RS, Casanova JL, Makitie OM, Kubisch C, Edery P, Christodoulou J, Germain RN, Goodnow CC, Sakmar TP, Billadeau DD, Küry S, Katanaev VL, Zhang Y, Lenardo MJ, Su HC. Germline mutations in a G protein identify signaling cross-talk in T cells. Science 2024; 385:eadd8947. [PMID: 39298586 PMCID: PMC11811912 DOI: 10.1126/science.add8947] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/15/2023] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
Humans with monogenic inborn errors responsible for extreme disease phenotypes can reveal essential physiological pathways. We investigated germline mutations in GNAI2, which encodes Gαi2, a key component in heterotrimeric G protein signal transduction usually thought to regulate adenylyl cyclase-mediated cyclic adenosine monophosphate (cAMP) production. Patients with activating Gαi2 mutations had clinical presentations that included impaired immunity. Mutant Gαi2 impaired cell migration and augmented responses to T cell receptor (TCR) stimulation. We found that mutant Gαi2 influenced TCR signaling by sequestering the guanosine triphosphatase (GTPase)-activating protein RASA2, thereby promoting RAS activation and increasing downstream extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)-AKT S6 signaling to drive cellular growth and proliferation.
Collapse
Affiliation(s)
- Hyoungjun Ham
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic; Rochester, MN, USA
| | - Huie Jing
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Ian T. Lamborn
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| | - Megan M. Kober
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Alexey Koval
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva; 1211 Geneva, Switzerland
| | - Yamina A. Berchiche
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University; New York, NY, USA
| | - D. Eric Anderson
- Advanced Mass Spectrometry Facility, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH; Bethesda, MD 20892, USA
| | - Kirk M. Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Judith N. Mandl
- Lymphocyte Biology Section, Laboratory of Immune System Biology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, Service de Génétique Médicale; F-44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax; F-44000 Nantes, France
| | - Carlos R. Ferreira
- Skeletal Genomics Unit, Metabolic Medicine Branch, DIR, National Human Genome Research Institute (NHGRI), NIH; Bethesda, MD, USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, DIR, NIAID, NIH; Bethesda, MD 20892, USA
| | - Meliha Karsak
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
| | - Peter J. Mustillo
- Nationwide Children’s Hospital; Columbus, OH, USA
- The Ohio State University College of Medicine; Columbus, OH, USA
| | - Juliana Teo
- Department of Haematology, The Children’s Hospital Westmead; Sydney, New South Wales, Australia
| | - Zarazuela Zolkipli-Cunningham
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Nicolas Chatron
- Service de Génétique, Hospices Civils de Lyon; Lyon, France
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène; 69008 Lyon, France
| | - François Lecoquierre
- Univ Rouen Normandie, Inserm U12045 and CHU Rouen, Department of Genetics and Reference Center for Developmental Disorders; FHU-G4 Génomique, F-76000, Rouen, France
| | - Andrew J. Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology (OCICB), NIAID, NIH; Bethesda, MD, USA
| | - Jana Pachlopnik Schmid
- Division of Immunology, University Children’s Hospital Zurich; Zurich, Switzerland
- Pediatric Immunology, University of Zurich; Zurich, Switzerland
| | - Douglas B. Kuhns
- Neutrophil Monitoring Lab, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research; Frederick, MD, USA
| | - Xuehua Xu
- Chemotaxis Signal Section, Laboratory of Immunogenetics, DIR, NIAID, NIH; Rockville, MD, USA
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, Ludwig-Maximilians-Universität (LMU); Munich, Germany
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University; Kuwait City, Kuwait
- Department of Pediatrics, Al-Sabah Hospital; Kuwait City, Kuwait
| | - Matias Wagner
- Institute of Human Genetics, Technical University Munich, School of Medicine and Health; Munich, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München; Neuherberg, Germany
- Department of Pediatrics, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, University Hospital of Munich; Munich, Germany
| | - Paulien A. Terhal
- Division of Laboratories, Pharmacy and Biomedical Genetics, University Medical Centre Utrecht; 3584EA Utrecht, the Netherlands
| | - Mari Muurinen
- Folkhälsan Research Center, Genetics Research Program; Helsinki, Finland
- Children’s Hospital, University of Helsinki and Helsinki University Hospital; Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki; Helsinki, Finland
| | - Vincent Barlogis
- APHM, La Timone Children’s Hospital, Department of Pediatric Hematology, Immunology, and Oncology; Marseille, France
- Aix Marseille University, EA 3279 Research Unit; Marseille, France
| | - Phillip Cruz
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology (OCICB), NIAID, NIH; Bethesda, MD, USA
| | - Jeffrey Danielson
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Helen Stewart
- Oxford Centre for Genomic Medicine, Nuffield Orthopaedic Centre, Oxford University Hospitals, NHS Foundation Trust; Headington, Oxford OX3 7HE, UK
| | - Petra Loid
- Folkhälsan Research Center, Genetics Research Program; Helsinki, Finland
- Children’s Hospital, University of Helsinki and Helsinki University Hospital; Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki; Helsinki, Finland
| | - Sebastian Rading
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
| | - Boris Keren
- Genetic Departement, Assistance Publique - Hôpitaux de Paris.Sorbonne University; Paris, France
- SeqOIA Laboratory, FMG2025, Paris; France
| | - Rolph Pfundt
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center; Nijmegen, The Netherlands
| | - Kol A. Zarember
- Laboratory of Clinical Immunology and Microbiology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Katharina Vill
- LMU University Hospital I Department of Pediatrics I Division of Pediatric Neurology I MUC iSPZ Hauner - Munich University Center for Children with Medical and Developmental Complexity I Dr. von Hauner Children’s Hospital; Munich, Germany
| | - Lorraine Potocki
- Department of Molecular and Human Genetics, Baylor College of Medicine; Houston, Texas, USA
- Texas Children’s Hospital; Houston, Texas, USA
| | - Kenneth N. Olivier
- Pulmonary Branch, Division of Intramural Research, DIR, National Heart Lung and Blood Institute (NHLBI), NIH; Bethesda, MD, USA
| | - Gaetan Lesca
- Service de Génétique, Hospices Civils de Lyon; Lyon, France
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène; 69008 Lyon, France
| | - Laurence Faivre
- UMR1231 GAD, Inserm, Université Bourgogne-Franche Comté; Dijon, France
- Centre de Génétique et Centre de Référence “Anomalies du Développement et Syndromes Malformatifs de l’Inter-région Est”, FHU TRANSLAD, CHU Dijon Bourgogne; Dijon, France
| | - Melanie Wong
- Department of Allergy and Immunology, The Children’s Hospital at Westmead; Sydney, New South Wales, Australia
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University; New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale INSERM U1163; Paris, France
- University of Paris Cité, Imagine Institute; Paris, France
| | - Janet Chou
- Division of Immunology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School; Boston, MA, United States
| | - Maud Tusseau
- Genetics Department, Lyon University Hospital; Lyon, France
- The International Center of Research in Infectiology, Lyon University, INSERM U1111, CNRS UMR 5308, ENS, UCBL; Lyon, France
| | - Niki M. Moutsopoulos
- Oral Immunity and Infection Section, DIR, National Institute of Dental and Craniofacial Research (NIDCR), NIH; Bethesda, MD, USA
| | - Helen F. Matthews
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Cas Simons
- Centre for Population Genomics, Garvan Institute of Medical Research and UNSW Sydney; Sydney, NSW, Australia
- Murdoch Children’s Research Institute; Melbourne, Victoria, Australia
| | - Ryan J. Taft
- Institute for Molecular Bioscience, University of Queensland; St. Lucia, Queensland, Australia
- Illumina, Inc, San Diego; CA, USA
| | - Ariane Soldatos
- National Institute of Neurological Disorders and Stroke (NINDS), NIH; Bethesda, MD, USA
| | - Etienne Masle-Farquhar
- Immunogenomics Laboratory, Garvan Institute of Medical Research; Sydney, New South Wales, Australia
- School of Clinical Medicine, UNSW Sydney; Sydney, NSW, Australia
| | - Stefania Pittaluga
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH; Bethesda, MD, USA
| | - Robert Brink
- St Vincent’s Clinical School, UNSW; Sydney, NSW, Australia
- B cell Biology Laboratory, Garvan Institute of Medical Research; Sydney, New South Wales, Australia
| | - Danielle L. Fink
- Neutrophil Monitoring Lab, Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research; Frederick, MD, USA
| | - Heidi H. Kong
- Cutaneous Microbiome and Inflammation Section, Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH; Bethesda, MD, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, DIR, NIAID, NIH; Bethesda, MD 20892, USA
| | - Woo Sung Kim
- Chemotaxis Signal Section, Laboratory of Immunogenetics, DIR, NIAID, NIH; Rockville, MD, USA
| | - Tatjana Bierhals
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
| | - Kazuyuki Meguro
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Amy P. Hsu
- Laboratory of Clinical Immunology and Microbiology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Jingwen Gu
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology (OCICB), NIAID, NIH; Bethesda, MD, USA
| | - Jennifer Stoddard
- Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH; Bethesda, MD, USA
| | - Benito Banos-Pinero
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust; Oxford, Oxfordshire, UK
| | - Maria Slack
- Division of Allergy and Immunology, Department of Pediatrics, University of Rochester Medical Center and Golisano Children’s Hospital; Rochester, NY, USA
| | - Giampaolo Trivellin
- Section on Endocrinology & Genetics (SEGEN), DIR, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH; Bethesda, MD, USA
| | - Benoît Mazel
- Centre de Génétique et Centre de Référence “Anomalies du Développement et Syndromes Malformatifs de l’Inter-région Est”, FHU TRANSLAD, CHU Dijon Bourgogne; Dijon, France
- Centre de Référence Déficiences Intellectuelles de Causes Rares, CHU Dijon Bourgogne; Dijon, France
| | - Maarja Soomann
- Division of Immunology, University Children’s Hospital Zurich; Zurich, Switzerland
| | - Samuel Li
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology (OCICB), NIAID, NIH; Bethesda, MD, USA
| | - Val J. Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University; West Lafayette, IN, USA
| | - Constantine A. Stratakis
- Section on Endocrinology & Genetics (SEGEN), DIR, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH; Bethesda, MD, USA
| | | | - Ange-Line Bruel
- UMR1231 GAD, Inserm, Université Bourgogne-Franche Comté; Dijon, France
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU TRANSLAD; CHU Dijon Bourgogne, Dijon, France
| | - Marita Lipsanen-Nyman
- Children’s Hospital, University of Helsinki and Helsinki University Hospital; Helsinki, Finland
| | - Paul Saultier
- APHM, La Timone Children’s Hospital, Department of Pediatric Hematology, Immunology, and Oncology; Marseille, France
- Aix Marseille University, INSERM; INRAe, C2VN, Marseille, France
| | - Rashmi Jain
- Clinical Immunology, Oxford University Hospitals NHS Foundation Trust; Oxford, OX3 9DU, UK
| | - Daphne Lehalle
- AP-HP Sorbonne Université, UF de Génétique Clinique, Centre de Référence Maladies Rares des anomalies du développement et syndromes malformatifs, Hôpital Trousseau; Paris, France
| | - Daniel Torres
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Kathleen E. Sullivan
- Division of Allergy and Immunology, Children’s Hospital of Philadelphia; Philadelphia, PA, USA
| | - Sébastien Barbarot
- Nantes Université, Department of Dermatology, CHU Nantes, INRAE; UMR 1280, PhAN, F-44000 Nantes, France
| | - Axel Neu
- Department of Pediatrics, University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
| | - Yannis Duffourd
- UMR1231 GAD, Inserm, Université Bourgogne-Franche Comté; Dijon, France
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, FHU TRANSLAD; CHU Dijon Bourgogne, Dijon, France
| | - Morgan Similuk
- Centralized Sequencing Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | | | | | - Stéphane Bézieau
- Nantes Université, CHU Nantes, Service de Génétique Médicale; F-44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax; F-44000 Nantes, France
| | - Tian Jin
- Chemotaxis Signal Section, Laboratory of Immunogenetics, DIR, NIAID, NIH; Rockville, MD, USA
| | - Raif S. Geha
- Division of Immunology, Boston Children’s Hospital, Department of Pediatrics, Harvard Medical School; Boston, MA, United States
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University; New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale INSERM U1163; Paris, France
- University of Paris Cité, Imagine Institute; Paris, France
- Howard Hughes Medical Institute; New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children; Paris, France
| | - Outi M. Makitie
- Folkhälsan Research Center, Genetics Research Program; Helsinki, Finland
- Children’s Hospital, University of Helsinki and Helsinki University Hospital; Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki; Helsinki, Finland
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
- Martin Zeitz Center for Rare Diseases, University Medical Center Hamburg-Eppendorf; 20246 Hamburg, Germany
| | - Patrick Edery
- Service de Génétique, Hospices Civils de Lyon; Lyon, France
- Centre de Recherche en Neurosciences de Lyon, Inserm U1028, UMR CNRS 5292, Université Claude Bernard Lyon 1; Lyon, France
| | - John Christodoulou
- Murdoch Children’s Research Institute; Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne; Melbourne, Australia
- Specialty of Child & Adolescent Health, University of Sydney; Sydney, Australia
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Christopher C. Goodnow
- Immunogenomics Laboratory, Garvan Institute of Medical Research; Sydney, New South Wales, Australia
- Cellular Genomics Futures Institute; Sydney, NSW, Australia
| | - Thomas P. Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University; New York, NY, USA
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Neurogeriatrics; Stockholm, Sweden
| | - Daniel D. Billadeau
- Division of Oncology Research, Schulze Center for Novel Therapeutics, Mayo Clinic; Rochester, MN, USA
| | - Sébastien Küry
- Nantes Université, CHU Nantes, Service de Génétique Médicale; F-44000 Nantes, France
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax; F-44000 Nantes, France
| | - Vladimir L. Katanaev
- Department of Cell Physiology and Metabolism, Faculty of Medicine, Translational Research Center in Oncohaematology, University of Geneva; 1211 Geneva, Switzerland
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University; 690090 Vladivostok, Russia
| | - Yu Zhang
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Michael J. Lenardo
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, DIR, NIAID, NIH; Bethesda, MD, USA
| | - Helen C. Su
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), NIAID, NIH; Bethesda, MD, USA
- Clinical Genomics Program, DIR, NIAID, NIH; Bethesda, MD, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA, USA
| |
Collapse
|
2
|
Gray M, Nash KR, Yao Y. Adenylyl cyclase 2 expression and function in neurological diseases. CNS Neurosci Ther 2024; 30:e14880. [PMID: 39073001 PMCID: PMC11284242 DOI: 10.1111/cns.14880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
Adenylyl cyclases (Adcys) catalyze the formation of cAMP, a secondary messenger essential for cell survival and neurotransmission pathways in the CNS. Adcy2, one of ten Adcy isoforms, is highly expressed in the CNS. Abnormal Adcy2 expression and mutations have been reported in various neurological disorders in both rodents and humans. However, due to the lack of genetic tools, loss-of-function studies of Adcy2 are scarce. In this review, we summarize recent findings on Adcy2 expression and function in neurological diseases. Specifically, we first introduce the biochemistry, structure, and function of Adcy2 briefly. Next, the expression and association of Adcy2 in human patients and rodent models of neurodegenerative diseases (Alzheimer's disease and Parkinson's disease), psychiatric disorders (Tourette syndrome, schizophrenia, and bipolar disorder), and other neurological conditions (stress-associated disorders, stroke, epilepsy, and Lesch-Nyhan Syndrome) are elaborated. Furthermore, we discuss the pros and cons of current studies as well as key questions that need to be answered in the future. We hope to provide a focused review on Adcy2 that promotes future research in the field.
Collapse
Affiliation(s)
- Marsilla Gray
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Kevin R. Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of MedicineUniversity of South FloridaTampaFloridaUSA
| |
Collapse
|
3
|
Qin W, Zhang K, Yu Z, Liu H, Li H, Dong L, Han D, Li T. Molecular mechanism of Xiaohuoluo wan for rheumatoid arthritis by integrating in vitro and in vivo chemomics and network pharmacology. Biomed Chromatogr 2024; 38:e5801. [PMID: 38110193 DOI: 10.1002/bmc.5801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 12/20/2023]
Abstract
The cause of rheumatoid arthritis (RA) is unclear. Xiaohuoluo wan (XHLW) is a classical Chinese medicine that is particularly effective in the treatment of RA. Given the chemical composition of XHLW at the overall level has been little studied and the molecular mechanism for the treatment of RA is not clear, we searched for the potential active compounds of XHLW and explored their anti-inflammatory mechanism in the treatment of RA by flexibly integrating the high-resolution ultra-performance liquid chromatography-mass spectrometry (UPLC-MS)-based in vitro and in vivo chemomics, network pharmacology, and other means. The results of the study identified that the active compounds of XHLW, such as alkaloids, nucleosides, and fatty acids, may play an anti-inflammatory role by regulating key targets such as IL-2, STAT1, JAK3, and MAPK8, inducing immune response through IL-17 signaling pathway, T-cell receptor, FoxO, tumor necrosis factor (TNF), and so forth, inhibiting the release of inflammatory factors and resisting oxidative stress and other pathways to treat RA. The results of this study provide referable data for the screening of active compounds and the exploration of molecular mechanisms of XHLW in the treatment of RA.
Collapse
Affiliation(s)
- Wanli Qin
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Kai Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ziyang Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Heyuan Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hangyu Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ling Dong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Dongran Han
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tiangang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Oh M, Batty S, Banerjee N, Kim TH. High extracellular glucose promotes cell motility by modulating cell deformability and contractility via the cAMP-RhoA-ROCK axis in human breast cancer cells. Mol Biol Cell 2023; 34:ar79. [PMID: 37195739 PMCID: PMC10398875 DOI: 10.1091/mbc.e22-12-0560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/18/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023] Open
Abstract
The mechanical properties, or mechanotypes, of cells are largely determined by their deformability and contractility. The ability of cancer cells to deform and generate contractile force is critical in multiple steps of metastasis. Identifying soluble cues that regulate cancer cell mechanotypes and understanding the underlying molecular mechanisms regulating these cellular mechanotypes could provide novel therapeutic targets to prevent metastasis. Although a strong correlation between high glucose level and cancer metastasis has been demonstrated, the causality has not been elucidated, and the underlying molecular mechanisms remain largely unknown. In this study, using novel high-throughput mechanotyping assays, we show that human breast cancer cells become less deformable and more contractile with increased extracellular glucose levels (>5 mM). These altered cell mechanotypes are due to increased F-actin rearrangement and nonmuscle myosin II (NMII) activity. We identify the cAMP-RhoA-ROCK-NMII axis as playing a major role in regulating cell mechanotypes at high extracellular glucose levels, whereas calcium and myosin light-chain kinase (MLCK) are not required. The altered mechanotypes are also associated with increased cell migration and invasion. Our study identifies key components in breast cancer cells that convert high extracellular glucose levels into changes in cellular mechanotype and behavior relevant in cancer metastasis.
Collapse
Affiliation(s)
- Mijung Oh
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
| | - Skylar Batty
- Undergraduate Pipeline Network Summer Research Program, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721
| | - Nayan Banerjee
- School of Chemical Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, West Bengal, India
| | - Tae-Hyung Kim
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131
| |
Collapse
|
5
|
Gärtner C, Fallmann J, Stadler PF, Kaiser T, Berkemer SJ. Toward a Systematic Assessment of Sex Differences in Cystic Fibrosis. J Pers Med 2023; 13:924. [PMID: 37373913 DOI: 10.3390/jpm13060924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: Cystic fibrosis (CF) is a disease with well-documented clinical differences between female and male patients. However, this gender gap is very poorly studied at the molecular level. (2) Methods: Expression differences in whole blood transcriptomics between female and male CF patients are analyzed in order to determine the pathways related to sex-biased genes and assess their potential influence on sex-specific effects in CF patients. (3) Results: We identify sex-biased genes in female and male CF patients and provide explanations for some sex-specific differences at the molecular level. (4) Conclusion: Genes in key pathways associated with CF are differentially expressed between sexes, and thus may account for the gender gap in morbidity and mortality in CF.
Collapse
Affiliation(s)
- Christiane Gärtner
- Neuromorphic Information Processing, Institute of Computer Science, Leipzig University, Augustusplatz 10, D-04109 Leipzig, Germany
- Bioinformatics Group, Institute of Computer Science, Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107 Leipzig, Germany
- Academic Department of Laboratory Medicine, Microbiology and Pathobiochemistry, Medical School and University Medical Center East Westphalia-Lippe, Hospital Lippe, Bielefeld University, Röntgenstraße 18, D-32756 Detmold, Germany
| | - Jörg Fallmann
- Bioinformatics Group, Institute of Computer Science, Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107 Leipzig, Germany
| | - Peter F Stadler
- Bioinformatics Group, Institute of Computer Science, Interdisciplinary Center of Bioinformatics, Leipzig University, Härtelstraße 16-18, D-04107 Leipzig, Germany
| | - Thorsten Kaiser
- Academic Department of Laboratory Medicine, Microbiology and Pathobiochemistry, Medical School and University Medical Center East Westphalia-Lippe, Hospital Lippe, Bielefeld University, Röntgenstraße 18, D-32756 Detmold, Germany
| | - Sarah J Berkemer
- LIX CNRS UMR 7161, Ecole Polytechnique, Institut Polytechnique de Paris, 91120 Palaiseau, France
- Earth-Life Science Institute, Tokyo Institute of Technology, 2-12-1-I7E-318 Ookayama, Tokyo 152-8550, Japan
| |
Collapse
|
6
|
Cheng W, Wang X, Wu Y, Li W, Fu C, Zou L, Zhang J. Huanglian-Houpo extract attenuates DSS-induced UC mice by protecting intestinal mucosal barrier and regulating macrophage polarization. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116181. [PMID: 36738944 DOI: 10.1016/j.jep.2023.116181] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huanglian-Houpo Decoction (HLHP), a classical prescription, has been used to treat gastrointestinal diseases for hundreds of years in TCM. However, the effective constituents and underlying mechanisms of HLHP in the treatment of ulcerative colitis (UC) have not been fully investigated. AIM OF THE STUDY This study aimed to reveal the potential anti-UC mechanisms of 50% ethanol extraction of HL and HP (EHLHP), combining transcriptomes and network pharmacology, as well as the animal experiment verification. METHODS Primarily, we identified the chemical composition of EHLHP via UPLC-QE-MS analysis. A visualization network with components-targets-pathways on UC treatment were constructed using network pharmacology. And then, the transcriptomics sequencing method was applied to screen out the differentially expressed genes (DEGs) of EHLHP in the treatment of UC. The key targets and pathways of EHLHP were selected by the combination of the network pharmacology and transcriptomics results. Ultimately, the potential mechanisms of EHLHP on DSS-induced UC mice were verified. RESULTS A total of 34 components of EHLHP were identified by UPLC-QE-MS analysis. Combined with the analysis of network pharmacology and transcriptomics, there were 262 DEGs between the normal group and the model group, and 151 DEGs between the model group and the EHLHP group. At the same time, there are 79 interaction paths, such as PI3K-Akt signaling pathway, MAPK signaling pathway, etc. These results indicated that the anti-UC mechanisms would be involved in calcium signaling pathway, inflammatory signaling pathway (JAK-STAT, TNF-α, cGMP-PKG) and immune regulation (IL-17, B cell receptor). After 160 mg/kg and 320 mg/kg EHLHP were given to DSS induced UC mice, these typical symptoms could be significantly alleviated, such as the decrease of DAI value and inflammation level. The IHC staining results of ZO-1, Occludin and Claudin-1 suggested that the intestinal barrier of UC mice was enhanced by EHLHP. The expression of macrophages and immune cells in F4/80+, CD11c+, Gr-1+, NK1.1+ by FCM determination indicated that EHLHP could suppress UC by immunosuppression and macrophage polarization M1 to M2. CONCLUSION The potential mechanisms of HLHP extract on DSS-induced UC mice were revealed, by the prediction of integrated analysis of transcriptomes and network pharmacology, and subsequently animal test verification. It would provide a viable strategy to elucidate the mechanisms of TCM classical formula.
Collapse
Affiliation(s)
- Weijian Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wei Li
- School of Preclinical Medicine, Chengdu University, Chengdu, 610106, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Liang Zou
- School of Food and Biological Engineering Preclinical Medicine, Chengdu University, Chengdu, 610106, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
7
|
Ruskovska T, Morand C, Bonetti CI, Gebara KS, Cardozo Junior EL, Milenkovic D. Multigenomic modifications in human circulating immune cells in response to consumption of polyphenol-rich extract of yerba mate ( Ilex paraguariensis A. St.-Hil.) are suggestive of cardiometabolic protective effects. Br J Nutr 2023; 129:185-205. [PMID: 35373729 DOI: 10.1017/s0007114522001027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Mate is a traditional drink obtained from the leaves of yerba mate and rich in a diversity of plant bioactive compounds including polyphenols, particularly chlorogenic acids. Studies, even though limited, suggest that consumption of mate is associated with health effects, including prevention of cardiometabolic disorders. Molecular mechanisms underlying the potential health properties are still largely unknown, especially in humans. The aim of this study was to investigate nutrigenomic effects of mate consumption and identify regulatory networks potentially mediating cardiometabolic health benefits. Healthy middle-aged men at risk for CVD consumed a standardised mate extract or placebo for 4 weeks. Global gene expression, including protein coding and non-coding RNA profiles, was determined using microarrays. Biological function analyses were performed using integrated bioinformatic tools. Comparison of global gene expression profiles showed significant change following mate consumption with 2635 significantly differentially expressed genes, among which six are miRNA and 244 are lncRNA. Functional analyses showed that these genes are involved in regulation of cell interactions and motility, inflammation or cell signalling. Transcription factors, such as MEF2A, MYB or HNF1A, could have their activity modulated by mate consumption either by direct interaction with polyphenol metabolites or by interactions of metabolites with cell signalling proteins, like p38 or ERK1/2, that could modulate transcription factor activity and regulate expression of genes observed. Correlation analysis suggests that expression profile is inversely associated with gene expression profiles of patients with cardiometabolic disorders. Therefore, mate consumption may exert cardiometabolic protective effects by modulating gene expression towards a protective profile.
Collapse
Affiliation(s)
- Tatjana Ruskovska
- Faculty of Medical Sciences, Goce Delcev University, Stip, North Macedonia
| | - Christine Morand
- Human Nutrition Unit, Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Carla Indianara Bonetti
- Institute of Biological, Medical and Health Sciences, Universidade Paranaense, Av. Parigot de Souza, Toledo, PR, Brazil
| | - Karimi Sater Gebara
- Grande Dourados University Center, UNIGRAN, R. Balbina de Matos, Dourados, MS, Brazil
| | - Euclides Lara Cardozo Junior
- Institute of Biological, Medical and Health Sciences, Universidade Paranaense, Av. Parigot de Souza, Toledo, PR, Brazil
| | - Dragan Milenkovic
- Human Nutrition Unit, Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
- Department of Nutrition, University of California Davis, Davis, CA, USA
| |
Collapse
|
8
|
Calì B, Deygas M, Munari F, Marcuzzi E, Cassará A, Toffali L, Vetralla M, Bernard M, Piel M, Gagliano O, Mastrogiovanni M, Laudanna C, Elvassore N, Molon B, Vargas P, Viola A. Atypical CXCL12 signaling enhances neutrophil migration by modulating nuclear deformability. Sci Signal 2022; 15:eabk2552. [DOI: 10.1126/scisignal.abk2552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To reach inflamed tissues from the circulation, neutrophils must overcome physical constraints imposed by the tissue architecture, such as the endothelial barrier or the three-dimensional (3D) interstitial space. In these microenvironments, neutrophils are forced to migrate through spaces smaller than their own diameter. One of the main challenges for cell passage through narrow gaps is the deformation of the nucleus, the largest and stiffest organelle in cells. Here, we showed that chemokines, the extracellular signals that guide cell migration in vivo, modulated nuclear plasticity to support neutrophil migration in restricted microenvironments. Exploiting microfabricated devices, we found that the CXC chemokine CXCL12 enhanced the nuclear pliability of mouse bone marrow–derived neutrophils to sustain their migration in 3D landscapes. This previously uncharacterized function of CXCL12 was mediated by the atypical chemokine receptor ACKR3 (also known as CXCR7), required protein kinase A (PKA) activity, and induced chromatin compaction, which resulted in enhanced cell migration in 3D. Thus, we propose that chemical cues regulate the nuclear plasticity of migrating leukocytes to optimize their motility in restricted microenvironments.
Collapse
Affiliation(s)
- Bianca Calì
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France
| | - Mathieu Deygas
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France
- Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
| | - Fabio Munari
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Elisabetta Marcuzzi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Antonino Cassará
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Lara Toffali
- University of Verona, Department of Medicine, Division of General Pathology, Verona, Italy
| | - Massimo Vetralla
- Venetian Institute of Molecular Medicine, Padova, Italy
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Mathilde Bernard
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France
- Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
| | - Matthieu Piel
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France
- Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
| | - Onelia Gagliano
- Venetian Institute of Molecular Medicine, Padova, Italy
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Marta Mastrogiovanni
- Lymphocyte Cell Biology Unit, Department of Immunology, Institut Pasteur, INSERM-U1224, Ligue Nationale Contre le Cancer, Équipe Labellisée Ligue 2018, F-75015 Paris, France
- Sorbonne Université, Collège Doctoral, F-75005 Paris. France
| | - Carlo Laudanna
- University of Verona, Department of Medicine, Division of General Pathology, Verona, Italy
| | - Nicola Elvassore
- Venetian Institute of Molecular Medicine, Padova, Italy
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Barbara Molon
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Pablo Vargas
- Institut Curie, PSL Research University, CNRS, UMR 144, F-75005 Paris, France
- Institut Pierre-Gilles de Gennes, PSL Research University, F-75005 Paris, France
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| |
Collapse
|
9
|
Cytoskeleton Elements Contribute to Prion Peptide-Induced Endothelial Barrier Breakdown in a Blood–Brain Barrier In Vitro System. Int J Mol Sci 2022; 23:ijms232012126. [PMID: 36293002 PMCID: PMC9603506 DOI: 10.3390/ijms232012126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
The mechanisms involved in the interaction of PrP 106-126, a peptide corresponding to the prion protein amyloidogenic region, with the blood–brain barrier (BBB) were studied. PrP 106-126 treatment that was previously shown to impair BBB function, reduced cAMP levels in cultured brain endothelial cells, increased nitric oxide (NO) levels, and changed the activation mode of the small GTPases Rac1 (inactivation) and RhoA (activation). The latter are well established regulators of endothelial barrier properties that act via cytoskeletal elements. Indeed, liquid chromatography-mass spectrometry (LC-MS)-based proteomic profiling study revealed extensive changes in expression of cytoskeleton-related proteins. These results shed light on the nature of the interaction between the prion peptide PrP 106-126 and the BBB and emphasize the importance of the cytoskeleton in endothelium response to prion- induced stress.
Collapse
|
10
|
Palshikar MG, Palli R, Tyrell A, Maggirwar S, Schifitto G, Singh MV, Thakar J. Executable models of immune signaling pathways in HIV-associated atherosclerosis. NPJ Syst Biol Appl 2022; 8:35. [PMID: 36131068 PMCID: PMC9492768 DOI: 10.1038/s41540-022-00246-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Atherosclerosis (AS)-associated cardiovascular disease is an important cause of mortality in an aging population of people living with HIV (PLWH). This elevated risk has been attributed to viral infection, anti-retroviral therapy, chronic inflammation, and lifestyle factors. However, the rates at which PLWH develop AS vary even after controlling for length of infection, treatment duration, and for lifestyle factors. To investigate the molecular signaling underlying this variation, we sequenced 9368 peripheral blood mononuclear cells (PBMCs) from eight PLWH, four of whom have atherosclerosis (AS+). Additionally, a publicly available dataset of PBMCs from persons before and after HIV infection was used to investigate the effect of acute HIV infection. To characterize dysregulation of pathways rather than just measuring enrichment, we developed the single-cell Boolean Omics Network Invariant Time Analysis (scBONITA) algorithm. scBONITA infers executable dynamic pathway models and performs a perturbation analysis to identify high impact genes. These dynamic models are used for pathway analysis and to map sequenced cells to characteristic signaling states (attractor analysis). scBONITA revealed that lipid signaling regulates cell migration into the vascular endothelium in AS+ PLWH. Pathways implicated included AGE-RAGE and PI3K-AKT signaling in CD8+ T cells, and glucagon and cAMP signaling pathways in monocytes. Attractor analysis with scBONITA facilitated the pathway-based characterization of cellular states in CD8+ T cells and monocytes. In this manner, we identify critical cell-type specific molecular mechanisms underlying HIV-associated atherosclerosis using a novel computational method.
Collapse
Affiliation(s)
- Mukta G Palshikar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Alicia Tyrell
- University of Rochester Clinical & Translational Science Institute, Rochester, USA
| | - Sanjay Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Meera V Singh
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Juilee Thakar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, USA.
| |
Collapse
|
11
|
Zmajkovicova K, Pawar S, Maier-Munsa S, Maierhofer B, Wiest I, Skerlj R, Taveras AG, Badarau A. Genotype–phenotype correlations in WHIM syndrome: a systematic characterization of CXCR4WHIM variants. Genes Immun 2022; 23:196-204. [PMID: 36089616 PMCID: PMC9519442 DOI: 10.1038/s41435-022-00181-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 12/05/2022]
Abstract
Warts, hypogammaglobulinemia, infections, myelokathexis (WHIM) syndrome is a rare primary immunodeficiency predominantly caused by heterozygous gain-of-function mutations in CXCR4 C-terminus. We assessed genotype–phenotype correlations for known pathogenic CXCR4 variants and in vitro response of each variant to mavorixafor, an investigational CXCR4 antagonist. We used cell-based assays to analyze CXCL12-induced receptor trafficking and downstream signaling of 14 pathogenic CXCR4 variants previously identified in patients with WHIM syndrome. All CXCR4 variants displayed impaired receptor trafficking, hyperactive downstream signaling, and enhanced chemotaxis in response to CXCL12. Mavorixafor inhibited CXCL12-dependent signaling and hyperactivation in cells harboring CXCR4WHIM mutations. A strong correlation was found between CXCR4 internalization defect and severity of blood leukocytopenias and infection susceptibility, and between AKT activation and immunoglobulin A level and CD4+ T-cell counts. This study is the first to show WHIM syndrome clinical phenotype variability as a function of both CXCR4WHIM genotype diversity and associated functional dysregulation. Our findings suggest that CXCR4 internalization may be used to assess the pathogenicity of CXCR4 variants in vitro and also as a potential WHIM-related disease biomarker. The investigational CXCR4 antagonist mavorixafor inhibited CXCL12-dependent signaling in all tested CXCR4-variant cell lines at clinically relevant concentrations.
Collapse
|
12
|
Becker F, Kebschull L, Rieger C, Mohr A, Heitplatz B, Van Marck V, Hansen U, Ansari J, Reuter S, Strücker B, Pascher A, Brockmann JG, Castor T, Alexander JS, Gavins FNE. Bryostatin-1 Attenuates Ischemia-Elicited Neutrophil Transmigration and Ameliorates Graft Injury after Kidney Transplantation. Cells 2022; 11:cells11060948. [PMID: 35326400 PMCID: PMC8946580 DOI: 10.3390/cells11060948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 12/19/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is a form of sterile inflammation whose severity determines short- and long-term graft fates in kidney transplantation. Neutrophils are now recognized as a key cell type mediating early graft injury, which activates further innate immune responses and intensifies acquired immunity and alloimmunity. Since the macrolide Bryostatin-1 has been shown to block neutrophil transmigration, we aimed to determine whether these findings could be translated to the field of kidney transplantation. To study the effects of Bryostatin-1 on ischemia-elicited neutrophil transmigration, an in vitro model of hypoxia and normoxia was equipped with human endothelial cells and neutrophils. To translate these findings, a porcine renal autotransplantation model with eight hours of reperfusion was used to study neutrophil infiltration in vivo. Graft-specific treatment using Bryostatin-1 (100 nM) was applied during static cold storage. Bryostatin-1 dose-dependently blocked neutrophil activation and transmigration over ischemically challenged endothelial cell monolayers. When applied to porcine renal autografts, Bryostatin-1 reduced neutrophil graft infiltration, attenuated histological and ultrastructural damage, and improved renal function. Our novel findings demonstrate that Bryostatin-1 is a promising pharmacological candidate for graft-specific treatment in kidney transplantation, as it provides protection by blocking neutrophil infiltration and attenuating functional graft injury.
Collapse
Affiliation(s)
- Felix Becker
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Linus Kebschull
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Constantin Rieger
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Annika Mohr
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Barbara Heitplatz
- Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149 Münster, Germany; (B.H.); (V.V.M.)
| | - Veerle Van Marck
- Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149 Münster, Germany; (B.H.); (V.V.M.)
| | - Uwe Hansen
- Department of Molecular Medicine, Institute for Musculoskeletal Medicine, University Hospital Münster, 48149 Münster, Germany;
| | - Junaid Ansari
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA;
| | - Stefan Reuter
- Division of General Internal Medicine, Nephrology and Rheumatology, Department of Medicine D, University Hospital of Münster, 48149 Münster, Germany;
| | - Benjamin Strücker
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | - Jens G. Brockmann
- Department of General, Visceral and Transplant Surgery, University Hospital Münster, 48149 Münster, Germany; (F.B.); (L.K.); (C.R.); (A.M.); (B.S.); (A.P.); (J.G.B.)
| | | | - J. Steve Alexander
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA;
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
- Correspondence: (J.S.A.); (F.N.E.G.)
| | - Felicity N. E. Gavins
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge UB8 3PH, UK
- Correspondence: (J.S.A.); (F.N.E.G.)
| |
Collapse
|
13
|
Wu J, Wang K, Liu Q, Li Y, Huang Y, Liu Y, Cai J, Yin C, Li X, Yu H, Meng W, Wang H, Lu A, Li Y, Guan D. An Integrative Pharmacology Model for Decoding the Underlying Therapeutic Mechanisms of Ermiao Powder for Rheumatoid Arthritis. Front Pharmacol 2022; 13:801350. [PMID: 35281924 PMCID: PMC8905663 DOI: 10.3389/fphar.2022.801350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/04/2022] [Indexed: 12/17/2022] Open
Abstract
As a systemic inflammatory arthritis disease, rheumatoid arthritis (RA) is complex and hereditary. Traditional Chinese medicine (TCM) has evident advantages in treating complex diseases, and a variety of TCM formulas have been reported that have effective treatment on RA. Clinical and pharmacological studies showed that Ermiao Powder, which consists of Phellodendron amurense Rupr. (PAR) and Atractylodes lancea (Thunb.) DC. (ALD), can be used in the treatment of RA. Currently, most studies focus on the anti-inflammatory mechanism of PAR and ALD and are less focused on their coordinated molecular mechanism. In this research, we established an integrative pharmacological strategy to explore the coordinated molecular mechanism of the two herbs of Ermiao Powder in treating RA. To explore the potential coordinated mechanism of PAR and ALD, we firstly developed a novel mathematical model to calculate the contribution score of 126 active components and 85 active components, which contributed 90% of the total contribution scores that were retained to construct the coordinated functional space. Then, the knapsack algorithm was applied to identify the core coordinated functional components from the 85 active components. Finally, we obtained the potential coordinated functional components group (CFCG) with 37 components, including wogonin, paeonol, ethyl caffeate, and magnoflorine. Also, functional enrichment analysis was performed on the targets of CFCG to explore the potential coordinated molecular mechanisms of PAR and ALD. The results indicated that the CFCG could treat RA by coordinated targeting to the genes involved in immunity and inflammation-related signal pathways, such as phosphatidylinositol 3‑kinase/protein kinase B signaling pathway, mitogen-activated protein kinase signaling pathway, tumor necrosis factor signaling pathway, and nuclear factor-kappa B signaling pathway. The docking and in vitro experiments were used to predict the affinity and validate the effect of CFCG and further confirm the reliability of our method. Our integrative pharmacological strategy, including CFCG identification and verification, can provide the methodological references for exploring the coordinated mechanism of TCM in treating complex diseases and contribute to improving our understanding of the coordinated mechanism.
Collapse
Affiliation(s)
- Jie Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Kexin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Neurosurgery Institute, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qinwen Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingying Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Jieqi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Chuanhui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Xiaowei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Hailang Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Handuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Aiping Lu
- Institute of Integrated Bioinformedicine and Translational Science, Hong Kong Baptist University, Hong Kong, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Yazi Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Daogang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| |
Collapse
|
14
|
Ziegon L, Schlegel M. Netrin-1: A Modulator of Macrophage Driven Acute and Chronic Inflammation. Int J Mol Sci 2021; 23:275. [PMID: 35008701 PMCID: PMC8745333 DOI: 10.3390/ijms23010275] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
Netrins belong to the family of laminin-like secreted proteins, which guide axonal migration and neuronal growth in the developing central nervous system. Over the last 20 years, it has been established that netrin-1 acts as a chemoattractive or chemorepulsive cue in diverse biological processes far beyond neuronal development. Netrin-1 has been shown to play a central role in cell adhesion, cell migration, proliferation, and cell survival in neuronal and non-neuronal tissue. In this context, netrin-1 was found to orchestrate organogenesis, angiogenesis, tumorigenesis, and inflammation. In inflammation, as in neuronal development, netrin-1 plays a dichotomous role directing the migration of leukocytes, especially monocytes in the inflamed tissue. Monocyte-derived macrophages have long been known for a similar dual role in inflammation. In response to pathogen-induced acute injury, monocytes are rapidly recruited to damaged tissue as the first line of immune defense to phagocyte pathogens, present antigens to initiate the adaptive immune response, and promote wound healing in the resolution phase. On the other hand, dysregulated macrophages with impaired phagocytosis and egress capacity accumulate in chronic inflammation sites and foster the maintenance-and even the progression-of chronic inflammation. In this review article, we will highlight the dichotomous roles of netrin-1 and its impact on acute and chronic inflammation.
Collapse
Affiliation(s)
| | - Martin Schlegel
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany;
| |
Collapse
|
15
|
Oh K, Adnan M, Cho D. Uncovering Mechanisms of Zanthoxylum piperitum Fruits for the Alleviation of Rheumatoid Arthritis Based on Network Pharmacology. BIOLOGY 2021; 10:703. [PMID: 34439936 PMCID: PMC8389290 DOI: 10.3390/biology10080703] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 12/11/2022]
Abstract
Zanthoxylum piperitum fruits (ZPFs) have been demonstrated favorable clinical efficacy on rheumatoid arthritis (RA), but its compounds and mechanisms against RA have not been elucidated. This study was to investigate the compounds and mechanisms of ZPFs to alleviate RA via network pharmacology. The compounds from ZPFs were detected by gas chromatography-mass spectrometry (GC-MS) and screened to select drug-likeness compounds through SwissADME. Targets associated with bioactive compounds or RA were identified utilizing bioinformatics databases. The signaling pathways related to RA were constructed; interactions among targets; and signaling pathways-targets-compounds (STC) were analyzed by RPackage. Finally, a molecular docking test (MDT) was performed to validate affinity between targets and compounds on key signaling pathway(s). GC-MS detected a total of 85 compounds from ZPFs, and drug-likeness properties accepted all compounds. A total of 216 targets associated with compounds 3377 RA targets and 101 targets between them were finally identified. Then, a bubble chart exhibited that inactivation of MAPK (mitogen-activated protein kinase) and activation of PPAR (peroxisome proliferator-activated receptor) signaling pathway might be key pathways against RA. Overall, this work suggests that seven compounds from ZPFs and eight targets might be multiple targets on RA and provide integrated pharmacological evidence to support the clinical efficacy of ZPFs on RA.
Collapse
Affiliation(s)
| | | | - Dongha Cho
- Department of Bio-Health Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Korea; (K.O.); (M.A.)
| |
Collapse
|
16
|
Liu X, Wang Q, Zhu R. Association of GWAS-susceptibility loci with ischemic stroke recurrence in a Han Chinese population. J Gene Med 2020; 23:e3264. [PMID: 32840931 DOI: 10.1002/jgm.3264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/05/2020] [Accepted: 08/22/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recently, genome-wide association studies (GWAS) have found many new susceptible genetic variants for ischemic stroke (IS) occurrence. However,the roles of GWAS-susceptibility loci in stroke prognosis are just beginning. The present study aimed to examine whether these GWAS-linked loci polymorphisms are associated with ischemic stroke recurrence in a Chinese population. METHODS We genotyped six single nucleotide polymorphisms (SNPs) (9p21: rs2383207 and rs4977574; 12p13: rs12425791 and rs11833579; PDE4D: rs966221; and ALOX5AP: rs1050391) in four GWAS-reported ischemic stroke risk genes in 657 patients. RESULTS The risk of recurrent stroke was significantly associated with PDE4D rs966221 in the dominant model (p = 0.027)and recessive model (p = 0.027). Furthermore, Kaplan-Meier analyses indicated no significant difference in the rate of recurrent stroke among the three genotypes of other five SNPs. Cox regression analysis showed that the GA + GG genotype within the rs966221 polymorphism had a 1.399-fold risk for stoke recurrence (95% confidence interval = 1.038-1.886; p = 0.027). Stratified analysis revealed that the increased recurrence risk of PDE4D rs966221 was more prominent in the large artery atherosclerosis (LAA) subgroup. CONCLUSIONS The reults of the present study demonstrate that PDE4D rs966221 may be a valuable biomarker for predicting the recurrent risks of patient with the LAA-IS and adds to our knowledge of the genetic basis of recurrent stroke risk.
Collapse
Affiliation(s)
- Xu Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Qianwen Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Tibbo AJ, Baillie GS. Phosphodiesterase 4B: Master Regulator of Brain Signaling. Cells 2020; 9:cells9051254. [PMID: 32438615 PMCID: PMC7291338 DOI: 10.3390/cells9051254] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/25/2022] Open
Abstract
Phosphodiesterases (PDEs) are the only superfamily of enzymes that have the ability to break down cyclic nucleotides and, as such, they have a pivotal role in neurological disease and brain development. PDEs have a modular structure that allows targeting of individual isoforms to discrete brain locations and it is often the location of a PDE that shapes its cellular function. Many of the eleven different families of PDEs have been associated with specific diseases. However, we evaluate the evidence, which suggests the activity from a sub-family of the PDE4 family, namely PDE4B, underpins a range of important functions in the brain that positions the PDE4B enzymes as a therapeutic target for a diverse collection of indications, such as, schizophrenia, neuroinflammation, and cognitive function.
Collapse
|
18
|
Kim SH, Park BB, Hong SE, Ryu SR, Lee JH, Kim SH, Lee P, Cho EK, Moon C. Effects of 2-methoxy-1,4-naphthoquinone (MQ) on MCP-1 Induced THP-1 Migration. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2019. [DOI: 10.15324/kjcls.2019.51.2.245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Si Hyun Kim
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| | - Bo Bin Park
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| | - Sung Eun Hong
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| | - Sung Ryul Ryu
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| | - Jang Ho Lee
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| | - Sa Hyun Kim
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| | - Pyeongjae Lee
- School of Industrial Bio-Pharmaceutical Science, Semyung University, Jecheon, Korea
| | - Eun-Kyung Cho
- Department of Biomedical Laboratory Science, Kyungwoon University, Gumi, Korea
| | - Cheol Moon
- Department of Clinical Laboratory Science, Semyung University, Jecheon, Korea
| |
Collapse
|
19
|
Dunne AE, Kawamatawong T, Fenwick PS, Davies CM, Tullett H, Barnes PJ, Donnelly LE. Direct Inhibitory Effect of the PDE4 Inhibitor Roflumilast on Neutrophil Migration in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2019; 60:445-453. [DOI: 10.1165/rcmb.2018-0065oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Amy E. Dunne
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Theerasuk Kawamatawong
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter S. Fenwick
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ceri M. Davies
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Hannah Tullett
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter J. Barnes
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Louise E. Donnelly
- Airway Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
Umebayashi R, Uchida HA, Kakio Y, Subramanian V, Daugherty A, Wada J. Cilostazol Attenuates Angiotensin II-Induced Abdominal Aortic Aneurysms but Not Atherosclerosis in Apolipoprotein E-Deficient Mice. Arterioscler Thromb Vasc Biol 2018; 38:903-912. [PMID: 29437572 DOI: 10.1161/atvbaha.117.309707] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Abdominal aortic aneurysm (AAA) is a permanent dilation of the abdominal aorta associated with rupture, which frequently results in fatal consequences. AAA tissue is commonly characterized by localized structural deterioration accompanied with inflammation and profound accumulation of leukocytes, although the specific function of these cells is unknown. Cilostazol, a phosphodiesterase III inhibitor, is commonly used for patients with peripheral vascular disease or stroke because of its anti-platelet aggregation effect and anti-inflammatory effect, which is vasoprotective effect. In this study, we evaluated the effects of cilostazol on angiotensin II-induced AAA formation. APPROACH AND RESULTS Male apolipoprotein E-deficient mice were fed either normal diet or a diet containing cilostazol (0.1% wt/wt). After 1 week of diet consumption, mice were infused with angiotensin II (1000 ng/kg per minute) for 4 weeks. Angiotensin II infusion increased maximal diameters of abdominal aortas, whereas cilostazol administration significantly attenuated dilatation of abdominal aortas, thereby, reducing AAA incidence. Cilostazol also reduced macrophage accumulation, matrix metalloproteinases activation, and inflammatory gene expression in the aortic media. In cultured vascular endothelial cells, cilostazol reduced expression of inflammatory cytokines and adhesive molecules through activation of the cAMP-PKA (protein kinase A) pathway. CONCLUSIONS Cilostazol attenuated angiotensin II-induced AAA formation by its anti-inflammatory effect through phosphodiesterase III inhibition in the aortic wall. Cilostazol may be a promising new therapeutic option for AAAs.
Collapse
Affiliation(s)
- Ryoko Umebayashi
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Haruhito A Uchida
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington.
| | - Yuki Kakio
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Venkateswaran Subramanian
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Alan Daugherty
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| | - Jun Wada
- From the Department of Nephrology, Rheumatology, Endocrinology and Metabolism (R.U., H.A.U., Y.K., J.W.) and Department of Chronic Kidney Disease and Cardiovascular Disease (H.A.U.), Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan; and Saha Cardiovascular Research Center (V.S., A.D.) and Department of Physiology (V.S., A.D.), University of Kentucky, Lexington
| |
Collapse
|
21
|
Schmoch T, Uhle F, Siegler BH, Fleming T, Morgenstern J, Nawroth PP, Weigand MA, Brenner T. The Glyoxalase System and Methylglyoxal-Derived Carbonyl Stress in Sepsis: Glycotoxic Aspects of Sepsis Pathophysiology. Int J Mol Sci 2017; 18:E657. [PMID: 28304355 PMCID: PMC5372669 DOI: 10.3390/ijms18030657] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 01/08/2023] Open
Abstract
Sepsis remains one of the leading causes of death in intensive care units. Although sepsis is caused by a viral, fungal or bacterial infection, it is the dysregulated generalized host response that ultimately leads to severe dysfunction of multiple organs and death. The concomitant profound metabolic changes are characterized by hyperglycemia, insulin resistance, and profound transformations of the intracellular energy supply in both peripheral and immune cells. A further hallmark of the early phases of sepsis is a massive formation of reactive oxygen (ROS; e.g., superoxide) as well as nitrogen (RNS; e.g., nitric oxide) species. Reactive carbonyl species (RCS) form a third crucial group of highly reactive metabolites, which until today have been not the focus of interest in sepsis. However, we previously showed in a prospective observational clinical trial that patients suffering from septic shock are characterized by significant methylglyoxal (MG)-derived carbonyl stress, with the glyoxalase system being downregulated in peripheral blood mononuclear cells. In this review, we give a detailed insight into the current state of research regarding the metabolic changes that entail an increased MG-production in septicemia. Thus, we point out the special role of the glyoxalase system in the context of sepsis.
Collapse
Affiliation(s)
- Thomas Schmoch
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Benedikt H Siegler
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Jakob Morgenstern
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Peter P Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| | - Thorsten Brenner
- Department of Anesthesiology, Heidelberg University Hospital, 69120 Heidelberg, Germany.
| |
Collapse
|
22
|
Song YL, Wang CJ, Wu YP, Lin J, Wang PL, Du WL, Liu L, Lin JX, Wang YL, Wang YJ, Liu GF. Phosphodiesterase 4D polymorphisms associate with the short-term outcome in ischemic stroke. Sci Rep 2017; 7:42914. [PMID: 28225001 PMCID: PMC5320494 DOI: 10.1038/srep42914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/17/2017] [Indexed: 01/24/2023] Open
Abstract
It has been demonstrated that phosphodiesterase 4D (PDE4D) genetic polymorphism is associated with ischemic stroke. However, the association between PDE4D gene and prognosis after ischemic stroke remains unknown. We consecutively enrolled ischemic stroke patients admitted to Beijing Tiantan Hospital from October 2009 to December 2013. Clinical, laboratory and imaging data upon admission were collected. All patients were followed up 3 months after stroke onset. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the associations of genetic polymorphisms with 3-month outcome after ischemic stroke and different subtypes, under various genetic models. A total of 1447 patients were enrolled, and 3-month follow-up data were obtained from 1388 (95.92%). Multivariate regression analysis showed that SNP87 of PDE4D gene was associated with increased risk of unfavorable outcome after total ischemic stroke (OR = 1.47, 95%CI 1.12–1.93), as well as stroke due to large-artery atherosclerosis (OR = 1.49, 95%CI 1.04–2.11) and small-artery occlusion (OR = 1.76, 95%CI 1.05–2.96) under a recessive model. No association between SNP83 genotype and poor outcome was found. Overall, this study demonstrated that the TT genotype of SNP87 in PDE4D was associated with increased risk of poor outcome after total ischemic stroke, large-artery atherosclerosis and small-artery occlusion, in a Chinese population.
Collapse
Affiliation(s)
- Yan-Li Song
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Department of Neurology, Handan First Hospital, Handan, China
| | - Chun-Juan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yi-Ping Wu
- Department of Neurology, Handan First Hospital, Handan, China
| | - Jie Lin
- Department of Neurology, Handan First Hospital, Handan, China
| | - Peng-Lian Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Wan-Liang Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Li Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Jin-Xi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yi-Long Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yong-Jun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Gai-Fen Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|
23
|
The protective role of MLCP-mediated ERM dephosphorylation in endotoxin-induced lung injury in vitro and in vivo. Sci Rep 2016; 6:39018. [PMID: 27976727 PMCID: PMC5157034 DOI: 10.1038/srep39018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
The goal of this study was to investigate the role of MLC phosphatase (MLCP) in a LPS model of acute lung injury (ALI). We demonstrate that ectopic expression of a constitutively-active (C/A) MLCP regulatory subunit (MYPT1) attenuates the ability of LPS to increase endothelial (EC) permeability. Down-regulation of MYPT1 exacerbates LPS-induced expression of ICAM1 suggesting an anti-inflammatory role of MLCP. To determine whether MLCP contributes to LPS-induced ALI in vivo, we utilized a nanoparticle DNA delivery method to specifically target lung EC. Expression of a C/A MYPT1 reduced LPS-induced lung inflammation and vascular permeability. Further, increased expression of the CS1β (MLCP catalytic subunit) also reduced LPS-induced lung inflammation, whereas the inactive CS1β mutant increased vascular leak. We next examined the role of the cytoskeletal targets of MLCP, the ERM proteins (Ezrin/Radixin/Moesin), in mediating barrier dysfunction. LPS-induced increase in EC permeability was accompanied by PKC-mediated increase in ERM phosphorylation, which was more prominent in CS1β-depleted cells. Depletion of Moesin and Ezrin, but not Radixin attenuated LPS-induced increases in permeability. Further, delivery of a Moesin phospho-null mutant into murine lung endothelium attenuated LPS-induced lung inflammation and vascular leak suggesting that MLCP opposes LPS-induced ALI by mediating the dephosphorylation of Moesin and Ezrin.
Collapse
|
24
|
Tang W, Shen Z, Guo J, Sun S. Screening of long non-coding RNA and TUG1 inhibits proliferation with TGF-β induction in patients with COPD. Int J Chron Obstruct Pulmon Dis 2016; 11:2951-2964. [PMID: 27932875 PMCID: PMC5135066 DOI: 10.2147/copd.s109570] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective To evaluate differentially expressed long noncoding RNAs (lncRNAs) and the potential role of lncRNA TUG1 in patients with chronic obstructive pulmonary disease (COPD). Methods Total RNA was extracted from both COPD and non-COPD lung tissues, and microarray analysis was performed with 25,628 lncRNA probes and 20,106 mRNA probes. In addition, five up-regulated and five down-regulated lncRNAs were selected for identification using quantitative real-time polymerase chain reaction. COPD cell model was established by transforming growth factor β (TGF-β) treatment. Cell Counting Kit-8 assay was used to detect BEAS-2B and HFL1 cell proliferation after TUG-siRNA transfection with TGF-β treatment. In addition, the expression levels of α-SMA and fibronectin proteins were determined using Western blot in BEAS-2B and HFL1 cells after TUG-siRNA transfection with TGF-β treatment. Results There were 8,376 (32.7%) differentially expressed lncRNAs and 5,094 (25.3%) differentially expressed mRNAs in COPD lung tissues compared with non-COPD lung tissues. Five of the analyzed lncRNAs (BC038205, BC130595, TUG1, MEG3, and LOC646329) were markedly increased, while five lncRNAs (LOC729178, PLAC2, LOC339529, LINC00229, and SNHG5) were significantly decreased in COPD lung tissues compared with non-COPD lung tissues (n=20) (***P<0.001). Knockdown of lncRNA TUG1 promotes BEAS-2B and HFL1 cell proliferation after TGF-β treatment through inhibiting the expression levels of α-SMA and fibronectin. Conclusion Abundant, differentially expressed lncRNAs and mRNAs were identified by microarray analysis and these might play a partial or key role in the diagnosis of patients with COPD. LncRNA TUG1 may become a very important class of biomarker and may act as a potential diagnostic and therapeutic target for patients with COPD.
Collapse
Affiliation(s)
- Wenxiang Tang
- Department of Respiratory Medicine, The Third Xiangya Hospital of Central South University
| | - Zhenyu Shen
- Department of Respiratory Medicine, Xiangtan Central Hospital, Hunan, People's Republic of China
| | - Jiang Guo
- Department of Respiratory Medicine, Xiangtan Central Hospital, Hunan, People's Republic of China
| | - Shenghua Sun
- Department of Respiratory Medicine, The Third Xiangya Hospital of Central South University
| |
Collapse
|
25
|
Cheng H, Wu Z, He X, Liu Q, Jia H, Di Y, Ji Q. siRNA-mediated silencing of phosphodiesterase 4B expression affects the production of cytokines in endotoxin-stimulated primary cultured microglia. Exp Ther Med 2016; 12:2257-2264. [PMID: 27698721 DOI: 10.3892/etm.2016.3575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 05/19/2016] [Indexed: 11/06/2022] Open
Abstract
Phosphodiesterase 4 (PDE4) has four subtypes: PDE4A, PDE4B, PDE4C and PDE4D. The expression of PDE4 subtypes in microglial cells and the specific contribution of each subtype to inflammation remain unclear. In this study, the expression of PDE4 subtypes in primary microglial cells was assayed. Primary microglial cells were then transfected with specific small interfering RNA (siRNA) against each PDE4 subtype. PDE4 subtype A-D knockdown was confirmed by quantitative polymerase chain reaction. Secreted cytokines in the supernatant and intracellular cyclic adenosine monophosphate (cAMP) levels of transfected cells were measured. The effect of PDE4B siRNA on the activation of extracellular regulated protein kinase (ERK) induced by lipopolysaccharide (LPS) in microglia was further tested by western blotting. Results showed that the primary microglial cells expressed all four types of PDE4s at the protein level. Transfection with the four siRNAs inhibited PDE4 subtype A-D mRNA expression, respectively. In primary microglial cells, treatment with PDE4B siRNA significantly inhibited the expression of tumor necrosis factor-α and interleukin (IL)-1β, and enhanced the expression of cAMP, while siRNAs to other subtypes had no significant effects. However, none of the four siRNAs had any significant effect on the expression of IL-10. Furthermore, in the PDE4B group, the level of phosphorylated ERK was reduced. Among the four PDE4 subtypes, PDE4B plays an important role in regulating inflammatory responses in microglia, potentially through initially regulating the intracellular cAMP concentration.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Zhifang Wu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiaoyun He
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Hongbin Jia
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yan Di
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qing Ji
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
26
|
Newick K, O'Brien S, Sun J, Kapoor V, Maceyko S, Lo A, Puré E, Moon E, Albelda SM. Augmentation of CAR T-cell Trafficking and Antitumor Efficacy by Blocking Protein Kinase A Localization. Cancer Immunol Res 2016; 4:541-51. [PMID: 27045023 DOI: 10.1158/2326-6066.cir-15-0263] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 03/04/2016] [Indexed: 11/16/2022]
Abstract
Antitumor treatments based on the infusion of T cells expressing chimeric antigen receptors (CAR T cells) are still relatively ineffective for solid tumors, due to the presence of immunosuppressive mediators [such as prostaglandin E2 (PGE2) and adenosine] and poor T-cell trafficking. PGE2 and adenosine activate protein kinase A (PKA), which then inhibits T-cell receptor (TCR) activation. This inhibition process requires PKA to localize to the immune synapse via binding to the membrane protein ezrin. We generated CAR T cells that expressed a small peptide called the "regulatory subunit I anchoring disruptor" (RIAD) that inhibits the association of PKA with ezrin, thus blunting the negative effects of PKA on TCR activation. After exposure to PGE2 or adenosine in vitro, CAR-RIAD T cells showed increased TCR signaling, released more cytokines, and showed enhanced killing of tumor cells compared with CAR T cells. When injected into tumor-bearing mice, the antitumor efficacy of murine and human CAR-RIAD T cells was enhanced compared with that of CAR T cells, due to resistance to tumor-induced hypofunction and increased T-cell infiltration of established tumors. Subsequent in vitro assays showed that both mouse and human CAR-RIAD cells migrated more efficiently than CAR cells did in response to the chemokine CXCL10 and also had better adhesion to various matrices. Thus, the intracellular addition of the RIAD peptide to adoptively transferred CAR T cells augments their efficacy by increasing their effector function and by improving trafficking into tumor sites. This treatment strategy, therefore, shows potential clinical application for treating solid tumors. Cancer Immunol Res; 4(6); 541-51. ©2016 AACR.
Collapse
Affiliation(s)
- Kheng Newick
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shaun O'Brien
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jing Sun
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Veena Kapoor
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven Maceyko
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Albert Lo
- Department of Biomedical Sciences, School of Veterinary Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edmund Moon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven M Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
27
|
Kim MO, Ryu JM, Suh HN, Park SH, Oh YM, Lee SH, Han HJ. cAMP Promotes Cell Migration Through Cell Junctional Complex Dynamics and Actin Cytoskeleton Remodeling: Implications in Skin Wound Healing. Stem Cells Dev 2015; 24:2513-24. [PMID: 26192163 DOI: 10.1089/scd.2015.0130] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Stem cells have attracted great interest for their therapeutic capacity in tissue regeneration. Cyclic adenosine 3',5'-monophosphate (cAMP), existing in high concentration at wound sites, mediated various signaling pathways such as cytoskeleton dynamics, cell adhesion, and cell migration in stem cells, which suggest the critical roles of cAMP in the wound healing process through functional regulation of stem cells. However, the mechanisms behind the effect of cAMP on mouse embryonic stem cell (mESC) motility and its roles on skin wound healing remain to be fully elucidated. In the present study, 8-Bromo cAMP-treated mESCs showed significant wound closure and improved neovascularization. Moreover, 8-Bromo cAMP stimulated mESC migration into the wound bed. 8-Bromo cAMP also increased ESC motility in in vitro migration assay. 8-Bromo cAMP induced myosin light chain phosphorylation through Rac1 and Cdc42 signaling, which were involved in 8-Bromo cAMP-induced decrease in expression of junction proteins (connexin 43, E-cadherin, and occludin) at the plasma membrane. Subsequently, 8-Bromo cAMP induced the disruption of cell junctions (including gap junctions, adherens junctions, and tight junctions), which reduced the function of the gap junctions and cell adhesion. In addition, 8-Bromo cAMP-induced Rac1 and Cdc42 activation increased Arp3, TOCA, PAK, and N-WASP expression, but decreased cofilin phosphorylation level, which elicited actin cytoskeleton remodeling. In contrast to the control, 8-Bromo cAMP evoked a substantial migration of cells into the denuded area, which was blocked by the small interfering RNAs of the signaling pathway-related molecules or by inhibitors. In conclusion, cAMP enhanced the migration of mESCs through effective coordination of junctional disruption and actin cytoskeleton remodeling, which increased the wound healing capacity of ESCs.
Collapse
Affiliation(s)
- Mi Ok Kim
- 1 Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University , Seoul, Republic of Korea.,2 BK21 PLUS Creative Veterinary Research Center, Seoul National University , Seoul, Republic of Korea
| | - Jung Min Ryu
- 1 Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University , Seoul, Republic of Korea.,2 BK21 PLUS Creative Veterinary Research Center, Seoul National University , Seoul, Republic of Korea
| | - Han Na Suh
- 1 Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University , Seoul, Republic of Korea.,2 BK21 PLUS Creative Veterinary Research Center, Seoul National University , Seoul, Republic of Korea
| | - Soo Hyun Park
- 3 College of Veterinary Medicine, Chonnam National University , Gwangju, Republic of Korea
| | - Yeon-Mok Oh
- 4 Department of Pulmonary and Critical Care Medicine, and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Sang Hun Lee
- 5 Medical Science Research Institute, Soonchunhyang University Seoul Hospital , Seoul, Republic of Korea
| | - Ho Jae Han
- 1 Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University , Seoul, Republic of Korea.,2 BK21 PLUS Creative Veterinary Research Center, Seoul National University , Seoul, Republic of Korea
| |
Collapse
|
28
|
Muller WA. The regulation of transendothelial migration: new knowledge and new questions. Cardiovasc Res 2015; 107:310-20. [PMID: 25987544 PMCID: PMC4592322 DOI: 10.1093/cvr/cvv145] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 03/13/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022] Open
Abstract
Leucocyte transendothelial migration (TEM) involves a co-operative series of interactions between surface molecules on the leucocyte and cognate counter-ligands on the endothelial cell. These interactions set up a cascade of signalling events inside the endothelial cell that both allow for the junctions to loosen and for membrane to be recruited from the lateral border recycling compartment (LBRC). The LBRC is thought to provide an increased surface area and unligated receptors to the leucocyte to continue the process. The relative importance of the individual adhesion/signalling molecules that promote transmigration may vary depending on the type of leucocyte, the vascular bed, the inflammatory stimulus, and the stage of the inflammatory response. However, the molecular interactions between leucocyte and endothelial cell activate signalling pathways that disengage the adherens and tight junctions and recruit the LBRC to the site of transmigration. With the exception of disengaging the junctions, similar molecules and mechanisms promote transcellular migration as paracellular migration of leucocytes. This review will discuss the molecular interactions and signalling pathways that regulate transmigration, and the common themes that emerge from studying TEM of different leucocyte subsets under different inflammatory conditions. We will also raise some unanswered questions in need of future research.
Collapse
Affiliation(s)
- William A Muller
- Department of Pathology, Northwestern University Feinberg School of Medicine, Ward Building 3-140, 303 East Chicago Avenue, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Risøe PK, Rutkovskiy A, Ågren J, Kolseth IBM, Kjeldsen SF, Valen G, Vaage J, Dahle MK. Higher TNFα responses in young males compared to females are associated with attenuation of monocyte adenylyl cyclase expression. Hum Immunol 2015; 76:427-30. [PMID: 25959651 DOI: 10.1016/j.humimm.2015.03.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 04/24/2014] [Accepted: 03/31/2015] [Indexed: 01/20/2023]
Abstract
Tumor necrosis factor α (TNFα) expression is strongly attenuated by the intracellular signaling mediator cyclic adenosine monophosphate (cAMP), which is synthesized by adenylyl cyclase (AC) enzymes. We have compared AC regulation and TNFα production in male and female monocytes, and characterized the role of monocyte AC isoforms in TNFα regulation. Males and females, age groups 20-30 years and 50-70 years donated blood for this study. In lipopolysaccharide-stimulated blood from young male donors, we observed significantly higher TNFα responses (6h, p=0.03) compared to females of the same age, a difference not observed in the older donors. Rapid down-regulation of the monocyte AC isoforms AC4, AC7 and AC9 were observed in young males. AC-directed siRNA experiments in the human monocyte cell line THP-1 demonstrated that AC7 and AC9 knock-down significantly induced TNFα release (p=0.01 for both isoforms). These data indicate that the stronger TNFα-responses in young males may be partly associated with male-specific down-regulation of adenylyl cyclases.
Collapse
Affiliation(s)
- Petter K Risøe
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Arkady Rutkovskiy
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Joanna Ågren
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway; Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Ingrid B M Kolseth
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Signe Flood Kjeldsen
- Institute for Surgical Research, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Guro Valen
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Jarle Vaage
- Department of Emergency Medicine and Intensive Care, Institute of Clinical Medicine, Oslo University Hospital Ulleval, University of Oslo, Norway
| | - Maria K Dahle
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Norway; Section of Immunology, Norwegian Veterinary Institute, Oslo, Norway.
| |
Collapse
|
30
|
Li AQ, Zhao L, Zhou TF, Zhang MQ, Qin XM. Exendin-4 promotes endothelial barrier enhancement via PKA- and Epac1-dependent Rac1 activation. Am J Physiol Cell Physiol 2014; 308:C164-75. [PMID: 25377089 DOI: 10.1152/ajpcell.00249.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Among emerging antidiabetic agents, glucagon-like peptide-1 (GLP-1)-based therapies carry special cardiovascular implications, exerting both direct and indirect effects. The control of vascular permeability is of pivotal importance in vascular pathologies. The objective of the present study was to determine the effect of GLP-1 on endothelial barrier function and assess the underlying mechanism(s). Here we show for the first time that the stable GLP-1 analog exendin-4 attenuated the leakage of subcutaneous blood vessels in mice indexed by dye extravasation caused by injections of thrombin. Moreover, in cultured endothelial cells, exendin-4 significantly prevented the thrombin-induced FITC-dextran permeability of endothelial monolayers via GLP-1 receptor. Immunofluorescence microscopy reveals that exendin-4 abrogates detrimental effects of thrombin on VE-cadherin and the F-actin cytoskeleton, with decreased stress fiber and gap formation. Importantly, exendin-4 reduced thrombin-induced tyrosine phosphorylation of VE-cadherin at Y731 and Y658. Moreover, small GTPase Rac1 was significantly activated as a result of exendin-4 treatment. The efficacy of exendin-4 to counteract the barrier-compromising effect of thrombin was blunted when Rac1 was inactivated by Rac1 inhibitor NSC-23766. Inhibition of PKA activity or small-interfering RNA for exchange protein directly activated by cAMP 1 (Epac1) decreased exendin-4-induced Rac1 activation and barrier enhancement, indicating the participation of both PKA and Epac1 in the barrier-stabilizing effect of exendin-4 elicited on thrombin-impaired barrier function. Thus, our findings have uncovered an unpredicted role for exendin-4 in the coordination of vascular permeability and clarified the molecular underpinnings that contribute to barrier restoration initiated by exendin-4.
Collapse
Affiliation(s)
- Ai Q Li
- Institute of Cardiovascular Science, and Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Liang Zhao
- Institute of Cardiovascular Science, and Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Teng F Zhou
- Institute of Cardiovascular Science, and Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Meng Q Zhang
- Institute of Cardiovascular Science, and Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Xiao M Qin
- Institute of Cardiovascular Science, and Key Laboratory of Molecular Cardiovascular Science of Ministry of Education, Peking University Health Science Center, Beijing, China
| |
Collapse
|
31
|
Park TY, Baik EJ, Lee SH. Prostaglandin E₂-induced intercellular adhesion molecule-1 expression is mediated by cAMP/Epac signalling modules in bEnd.3 brain endothelial cells. Br J Pharmacol 2014; 169:604-18. [PMID: 23317035 DOI: 10.1111/bph.12103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 12/21/2012] [Accepted: 12/26/2012] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Prostaglandin E₂ (PGE₂) has been implicated in the regulation of adhesion molecules, leukocyte adhesion and infiltration into inflamed site. However, the underlying mechanism therein involved remains ill-defined. In this study, we explored its cellular mechanism of action in the regulation of the intercellular adhesion molecule-1 (ICAM-1) expression in the brain endothelial cells. EXPERIMENTAL APPROACH bEnd.3 cells, the murine cerebrovascular endothelial cell line and primary mouse brain endothelial cells were treated with PGE₂ with or without agonists/antagonists of PGE₂ receptors and associated signalling molecules. ICAM-1 expression, Akt phosphorylation and activity of NF-κB were determined by reverse transcription polymerase chain reaction (RT-PCR), immunoblot analysis, luciferase assay and immunocytochemistry. KEY RESULTS PGE₂ significantly up-regulated the expression of ICAM-1, which was blocked by EP4 antagonist (ONO-AE2-227) and knock-down of EP4. PGE₂ effects were mimicked by forskolin, dibutyryl cAMP (dbcAMP) and an exchange protein directly activated by cAMP (Epac) activator (8-Cpt-cAMP) but not a protein kinase A activator (N⁶-Bnz-cAMP). PGE₂-induced ICAM-1 expression was reduced by knock-down of Epac1. A PI3K specific inhibitor (LY294002), Akt inhibitor VIII (Akti) and NF-κB inhibitors (Bay-11-7082 and MG-132) attenuated the induction of ICAM-1 by PGE₂. PGE₂, dbcAMP and 8-Cpt-cAMP induced the phosphorylation of Akt, IκB kinase and IκBα and the translocation of p65 to the nucleus and increased NF-κB dependent reporter gene activity, which was diminished by Akti. CONCLUSION AND IMPLICATIONS Our findings suggest that PGE₂ induces ICAM-1 expression via EP4 receptor and Epac/Akt/NF-κB signalling pathway in bEnd.3 brain endothelial cells, supporting its pathophysiological role in brain inflammation.
Collapse
Affiliation(s)
- Tae Yeop Park
- Department of Physiology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | | | | |
Collapse
|
32
|
cAMP signalling in the vasculature: the role of Epac (exchange protein directly activated by cAMP). Biochem Soc Trans 2014; 42:89-97. [PMID: 24450633 DOI: 10.1042/bst20130253] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The second messenger cAMP plays a central role in mediating vascular smooth muscle relaxation in response to vasoactive transmitters and in strengthening endothelial cell-cell junctions that regulate the movement of solutes, cells and macromolecules between the blood and the surrounding tissue. The vasculature expresses three cAMP effector proteins: PKA (protein kinase A), CNG (cyclic-nucleotide-gated) ion channels, and the most recently discovered Epacs (exchange proteins directly activated by cAMP). Epacs are a family of GEFs (guanine-nucleotide-exchange factors) for the small Ras-related GTPases Rap1 and Rap2, and are being increasingly implicated as important mediators of cAMP signalling, both in their own right and in parallel with the prototypical cAMP target PKA. In the present paper, we review what is currently known about the role of Epac within blood vessels, particularly with regard to the regulation of vascular tone, endothelial barrier function and inflammation.
Collapse
|
33
|
Oldenburger A, Timens W, Bos S, Smit M, Smrcka AV, Laurent AC, Cao J, Hylkema M, Meurs H, Maarsingh H, Lezoualc'h F, Schmidt M. Epac1 and Epac2 are differentially involved in inflammatory and remodeling processes induced by cigarette smoke. FASEB J 2014; 28:4617-28. [PMID: 25103224 DOI: 10.1096/fj.13-248930] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cigarette smoke (CS) induces inflammatory responses characterized by increase of immune cells and cytokine release. Remodeling processes, such as mucus hypersecretion and extracellular matrix protein production, are also directly or indirectly induced by CS. Recently, we showed that activation of the exchange protein directly activated by cAMP (Epac) attenuates CS extract-induced interleukin (IL)-8 release from cultured airway smooth muscle cells. Using an acute, short-term model of CS exposure, we now studied the role of Epac1, Epac2, and the Epac effector phospholipase-Cε (PLCε) in airway inflammation and remodeling in vivo. Compared to wild-type mice exposed to CS, the number of total inflammatory cells, macrophages, and neutrophils and total IL-6 release was lower in Epac2(-/-) mice, which was also the case for neutrophils and IL-6 in PLCε(-/-) mice. Taken together, Epac2, acting partly via PLCε, but not Epac1, enhances CS-induced airway inflammation in vivo. In total lung homogenates of Epac1(-/-) mice, MUC5AC and matrix remodeling parameters (transforming growth factor-β1, collagen I, and fibronectin) were increased at baseline. Our findings suggest that Epac1 primarily is capable of inhibiting remodeling processes, whereas Epac2 primarily increases inflammatory processes in vivo.
Collapse
Affiliation(s)
- Anouk Oldenburger
- Department of Molecular Pharmacology and Groningen Research Institute for Asthma and COPD and
| | - Wim Timens
- Groningen Research Institute for Asthma and COPD and Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sophie Bos
- Department of Molecular Pharmacology and
| | | | - Alan V Smrcka
- Department of Pharmacology and Physiology, School of Medicine, University of Rochester, Rochester, NY, USA
| | - Anne-Coline Laurent
- Institut National de la Recherche Scientifique (INSERM), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Université de Toulouse III, Paul Sabatier, Toulouse, France; and
| | - Junjun Cao
- Groningen Research Institute for Asthma and COPD and Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Machteld Hylkema
- Groningen Research Institute for Asthma and COPD and Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Herman Meurs
- Department of Molecular Pharmacology and Groningen Research Institute for Asthma and COPD and
| | - Harm Maarsingh
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida, USA
| | - Frank Lezoualc'h
- Institut National de la Recherche Scientifique (INSERM), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France; Université de Toulouse III, Paul Sabatier, Toulouse, France; and
| | - Martina Schmidt
- Department of Molecular Pharmacology and Groningen Research Institute for Asthma and COPD and
| |
Collapse
|
34
|
Mizuno R, Kamioka Y, Kabashima K, Imajo M, Sumiyama K, Nakasho E, Ito T, Hamazaki Y, Okuchi Y, Sakai Y, Kiyokawa E, Matsuda M. In vivo imaging reveals PKA regulation of ERK activity during neutrophil recruitment to inflamed intestines. ACTA ACUST UNITED AC 2014; 211:1123-36. [PMID: 24842369 PMCID: PMC4042632 DOI: 10.1084/jem.20132112] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In vivo FRET demonstrates that ERK positively regulates the neutrophil recruitment cascade in the intestine by promoting adhesion and migration. Many chemical mediators regulate neutrophil recruitment to inflammatory sites. Although the actions of each chemical mediator have been demonstrated with neutrophils in vitro, how such chemical mediators act cooperatively or counteractively in vivo remains largely unknown. Here, by in vivo two-photon excitation microscopy with transgenic mice expressing biosensors based on Förster resonance energy transfer, we time-lapse–imaged the activities of extracellular signal–regulated kinase (ERK) and protein kinase A (PKA) in neutrophils in inflamed intestinal tissue. ERK activity in neutrophils rapidly increased during spreading on the endothelial cells and showed positive correlation with the migration velocity on endothelial cells or in interstitial tissue. Meanwhile, in the neutrophils migrating in the interstitial tissue, high PKA activity correlated negatively with migration velocity. In contradiction to previous in vitro studies that showed ERK activation by prostaglandin E2 (PGE2) engagement with prostaglandin receptor EP4, intravenous administration of EP4 agonist activated PKA, inhibited ERK, and suppressed migration of neutrophils. The opposite results were obtained using nonsteroidal antiinflammatory drugs (NSAIDs). Therefore, NSAID-induced enteritis may be caused at least partially by the inhibition of EP4 receptor signaling of neutrophils. Our results demonstrate that ERK positively regulates the neutrophil recruitment cascade by promoting adhesion and migration steps.
Collapse
Affiliation(s)
- Rei Mizuno
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, JapanDepartment of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Yuji Kamioka
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, JapanDepartment of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Kenji Kabashima
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Masamichi Imajo
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Kenta Sumiyama
- Division of Population Genetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - Eiji Nakasho
- Life & Industrial Products Development Department 1, R&D Division, Olympus Corporation, Hachioji-shi, Tokyo 192-8507, Japan
| | - Takeshi Ito
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Yoko Hamazaki
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Yoshihisa Okuchi
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, JapanDepartment of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Yoshiharu Sakai
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| | - Etsuko Kiyokawa
- Department of Oncologic Pathology, Kanazawa Medical University, Kanazawa, Ishikawa 920-0293, Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, JapanDepartment of Pathology and Biology of Diseases, Department of Gastrointestinal Surgery, Department of Dermatology, and Department of Immunology and Cell Biology, Graduate School of Medicine; Innovative Techno-Hub for Integrated Medical Bio-Imaging; and Laboratory of Bioimaging and Cell Signaling, Department of Molecular and System Biology, Graduate School of Biostudies; Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
35
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
36
|
Zimmerman NP, Roy I, Hauser AD, Wilson JM, Williams CL, Dwinell MB. Cyclic AMP regulates the migration and invasion potential of human pancreatic cancer cells. Mol Carcinog 2013; 54:203-15. [PMID: 24115212 DOI: 10.1002/mc.22091] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 08/30/2013] [Indexed: 12/14/2022]
Abstract
Aggressive dissemination and metastasis of pancreatic ductal adenocarcinoma (PDAC) results in poor prognosis and marked lethality. Rho monomeric G protein levels are increased in pancreatic cancer tissue. As the mechanisms underlying PDAC malignancy are little understood, we investigated the role for cAMP in regulating monomeric G protein regulated invasion and migration of pancreatic cancer cells. Treatment of PDAC cells with cAMP elevating agents that activate adenylyl cyclases, forskolin, protein kinase A (PKA), 6-Bnz-cAMP, or the cyclic nucleotide phosphodiesterase inhibitor cilostamide significantly decreased migration and Matrigel invasion of PDAC cell lines. Inhibition was dose-dependent and not significantly different between forskolin or cilostamide treatment. cAMP elevating drugs not only blocked basal migration, but similarly abrogated transforming-growth factor-β-directed PDAC cell migration and invasion. The inhibitory effects of cAMP were prevented by the pharmacological blockade of PKA. Drugs that increase cellular cAMP levels decreased levels of active RhoA or RhoC, with a concomitant increase in phosphorylated RhoA. Diminished Rho signaling was correlated with the appearance of thickened cortical actin bands along the perimeter of non-motile forskolin or cilostamide-treated cells. Decreased migration did not reflect alterations in cell growth or programmed cell death. Collectively these data support the notion that increased levels of cAMP specifically hinder PDAC cell motility through F-actin remodeling.
Collapse
Affiliation(s)
- Noah P Zimmerman
- Department of Microbiology and Molecular Genetics, The Medical College of Wisconsin Cancer Center, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226
| | | | | | | | | | | |
Collapse
|
37
|
García-Román J, Zentella-Dehesa A. Vascular permeability changes involved in tumor metastasis. Cancer Lett 2013; 335:259-69. [DOI: 10.1016/j.canlet.2013.03.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/05/2013] [Accepted: 03/06/2013] [Indexed: 12/12/2022]
|
38
|
Schmidt M, Dekker FJ, Maarsingh H. Exchange protein directly activated by cAMP (epac): a multidomain cAMP mediator in the regulation of diverse biological functions. Pharmacol Rev 2013; 65:670-709. [PMID: 23447132 DOI: 10.1124/pr.110.003707] [Citation(s) in RCA: 214] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Since the discovery nearly 60 years ago, cAMP is envisioned as one of the most universal and versatile second messengers. The tremendous feature of cAMP to tightly control highly diverse physiologic processes, including calcium homeostasis, metabolism, secretion, muscle contraction, cell fate, and gene transcription, is reflected by the award of five Nobel prizes. The discovery of Epac (exchange protein directly activated by cAMP) has ignited a new surge of cAMP-related research and has depicted novel cAMP properties independent of protein kinase A and cyclic nucleotide-gated channels. The multidomain architecture of Epac determines its activity state and allows cell-type specific protein-protein and protein-lipid interactions that control fine-tuning of pivotal biologic responses through the "old" second messenger cAMP. Compartmentalization of cAMP in space and time, maintained by A-kinase anchoring proteins, phosphodiesterases, and β-arrestins, contributes to the Epac signalosome of small GTPases, phospholipases, mitogen- and lipid-activated kinases, and transcription factors. These novel cAMP sensors seem to implement certain unexpected signaling properties of cAMP and thereby to permit delicate adaptations of biologic responses. Agonists and antagonists selective for Epac are developed and will support further studies on the biologic net outcome of the activation of Epac. This will increase our current knowledge on the pathophysiology of devastating diseases, such as diabetes, cognitive impairment, renal and heart failure, (pulmonary) hypertension, asthma, and chronic obstructive pulmonary disease. Further insights into the cAMP dynamics executed by the Epac signalosome will help to optimize the pharmacological treatment of these diseases.
Collapse
Affiliation(s)
- Martina Schmidt
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands.
| | | | | |
Collapse
|
39
|
Savill SA, Leitch HF, Harvey JN, Thomas TH. Functional structure of the promoter regions for the predominant low molecular weight isoforms of tropomyosin in human kidney cells. J Cell Biochem 2013; 113:3576-86. [PMID: 22740512 DOI: 10.1002/jcb.24236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
High and low molecular weight (LMW) tropomyosin isoforms, by regulation of actin filaments, have a major role in the regulation of cell behaviour. They affect malignant transformation, motility, differentiation, metastasis and cell membrane protein presentation. Expression of LMW isoforms from the TPM1 and TPM3 genes have an important role in these effects but the regulation of their expression is unknown. Luciferase assays on a progressively truncated 1.7 kb fragment upstream of the exon 1b translation start site in the TPM1 and TPM3 genes in HEK-293 cells showed upstream activation sequences in TPM1 between -152 and -139 bp and in TPM3 between -154 and -102 bp. The effect of mutating candidate transcription factor binding sites identified an AML1-like transcription factor binding site in TPM1 and a cAMP response element in TPM3. Downstream from the primary activation sequence in TPM1 was a repressor region corresponding to two Sp/KLF family binding GC boxes. Band shift assays confirmed that deletion of these sites altered transcription factor binding and ChIP assays confirmed the presence of AML1 and CREB at the TPM1 and TPM3 activation sequences in the respective promoters. Expression of LMW isoforms from TPM1 and TPM3 genes is regulated very differently. This facilitates regulation of the many cell processes involving these proteins. In situations where these proteins have a critical role, such as cancer metastasis, it also facilitates specific intervention.
Collapse
Affiliation(s)
- Stuart A Savill
- Diabetes and Endocrinology Research Group, College of Health and Behavioural Sciences, Wrexham Academic Unit, Bangor University, Bangor, UK.
| | | | | | | |
Collapse
|
40
|
Lemichez E, Gonzalez-Rodriguez D, Bassereau P, Brochard-Wyart F. Transcellular tunnel dynamics: Control of cellular dewetting by actomyosin contractility and I-BAR proteins. Biol Cell 2013. [PMID: 23189935 DOI: 10.1111/boc.201200063] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dewetting is the spontaneous withdrawal of a liquid film from a non-wettable surface by nucleation and growth of dry patches. Two recent reports now propose that the principles of dewetting explain the physical phenomena underpinning the opening of transendothelial cell macroaperture (TEM) tunnels, referred to as cellular dewetting. This was discovered by studying a group of bacterial toxins endowed with the property of corrupting actomyosin cytoskeleton contractility. For both liquid and cellular dewetting, the growth of holes is governed by a competition between surface forces and line tension. We also discuss how the dynamics of TEM opening and closure represent remarkable systems to investigate actin cytoskeleton regulation by sensors of plasma membrane curvature and investigate the impact on membrane tension and the role of TEM in vascular dysfunctions.
Collapse
Affiliation(s)
- Emmanuel Lemichez
- INSERM, U1065, Université de Nice-Sophia-Antipolis, Centre Méditerranéen de Médecine Moléculaire, C3M, Nice 06204, France.
| | | | | | | |
Collapse
|
41
|
Oldenburger A, Maarsingh H, Schmidt M. Multiple facets of cAMP signalling and physiological impact: cAMP compartmentalization in the lung. Pharmaceuticals (Basel) 2012; 5:1291-331. [PMID: 24281338 PMCID: PMC3816672 DOI: 10.3390/ph5121291] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/20/2022] Open
Abstract
Therapies involving elevation of the endogenous suppressor cyclic AMP (cAMP) are currently used in the treatment of several chronic inflammatory disorders, including chronic obstructive pulmonary disease (COPD). Characteristics of COPD are airway obstruction, airway inflammation and airway remodelling, processes encompassed by increased airway smooth muscle mass, epithelial changes, goblet cell and submucosal gland hyperplasia. In addition to inflammatory cells, airway smooth muscle cells and (myo)fibroblasts, epithelial cells underpin a variety of key responses in the airways such as inflammatory cytokine release, airway remodelling, mucus hypersecretion and airway barrier function. Cigarette smoke, being next to environmental pollution the main cause of COPD, is believed to cause epithelial hyperpermeability by disrupting the barrier function. Here we will focus on the most recent progress on compartmentalized signalling by cAMP. In addition to G protein-coupled receptors, adenylyl cyclases, cAMP-specific phospho-diesterases (PDEs) maintain compartmentalized cAMP signalling. Intriguingly, spatially discrete cAMP-sensing signalling complexes seem also to involve distinct members of the A-kinase anchoring (AKAP) superfamily and IQ motif containing GTPase activating protein (IQGAPs). In this review, we will highlight the interaction between cAMP and the epithelial barrier to retain proper lung function and to alleviate COPD symptoms and focus on the possible molecular mechanisms involved in this process. Future studies should include the development of cAMP-sensing multiprotein complex specific disruptors and/or stabilizers to orchestrate cellular functions. Compartmentalized cAMP signalling regulates important cellular processes in the lung and may serve as a therapeutic target.
Collapse
Affiliation(s)
- Anouk Oldenburger
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands.
| | | | | |
Collapse
|
42
|
Dekkers BGJ, Racké K, Schmidt M. Distinct PKA and Epac compartmentalization in airway function and plasticity. Pharmacol Ther 2012; 137:248-65. [PMID: 23089371 DOI: 10.1016/j.pharmthera.2012.10.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 10/09/2012] [Indexed: 12/15/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are obstructive lung diseases characterized by airway obstruction, airway inflammation and airway remodelling. Next to inflammatory cells and airway epithelial cells, airway mesenchymal cells, including airway smooth muscle cells and (myo)fibroblasts, substantially contribute to disease features by the release of inflammatory mediators, smooth muscle contraction, extracellular matrix deposition and structural changes in the airways. Current pharmacological treatment of both diseases intends to target the dynamic features of the endogenous intracellular suppressor cyclic AMP (cAMP). This review will summarize our current knowledge on cAMP and will emphasize on key discoveries and paradigm shifts reflecting the complex spatio-temporal nature of compartmentalized cAMP signalling networks in health and disease. As airway fibroblasts and airway smooth muscle cells are recognized as central players in the development and progression of asthma and COPD, we will focus on the role of cAMP signalling in their function in relation to airway function and plasticity. We will recapture on the recent identification of cAMP-sensing multi-protein complexes maintained by cAMP effectors, including A-kinase anchoring proteins (AKAPs), proteins kinase A (PKA), exchange protein directly activated by cAMP (Epac), cAMP-elevating seven-transmembrane (7TM) receptors and phosphodiesterases (PDEs) and we will report on findings indicating that the pertubation of compartmentalized cAMP signalling correlates with the pathopysiology of obstructive lung diseases. Future challenges include studies on cAMP dynamics and compartmentalization in the lung and the development of novel drugs targeting these systems for therapeutic interventions in chronic obstructive inflammatory diseases.
Collapse
Affiliation(s)
- Bart G J Dekkers
- Department of Molecular Pharmacology, University Center of Pharmacy, University of Groningen, The Netherlands.
| | | | | |
Collapse
|
43
|
Mohan S, Ahmad AS, Glushakov AV, Chambers C, Doré S. Putative role of prostaglandin receptor in intracerebral hemorrhage. Front Neurol 2012; 3:145. [PMID: 23097645 PMCID: PMC3477820 DOI: 10.3389/fneur.2012.00145] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Accepted: 09/30/2012] [Indexed: 01/21/2023] Open
Abstract
Each year, approximately 795,000 people experience a new or recurrent stroke. Of all strokes, 84% are ischemic, 13% are intracerebral hemorrhage (ICH) strokes, and 3% are subarachnoid hemorrhage strokes. Despite the decreased incidence of ischemic stroke, there has been no change in the incidence of hemorrhagic stroke in the last decade. ICH is a devastating disease 37–38% of patients between the ages of 45 and 64 die within 30 days. In an effort to prevent ischemic and hemorrhagic strokes we and others have been studying the role of prostaglandins and their receptors. Prostaglandins are bioactive lipids derived from the metabolism of arachidonic acid. They sustain homeostatic functions and mediate pathogenic mechanisms, including the inflammatory response. Most prostaglandins are produced from specific enzymes and act upon cells via distinct G-protein coupled receptors. The presence of multiple prostaglandin receptors cross-reactivity and coupling to different signal transduction pathways allow differentiated cells to respond to prostaglandins in a unique manner. Due to the number of prostaglandin receptors, prostaglandin-dependent signaling can function either to promote neuronal survival or injury following acute excitotoxicity, hypoxia, and stress induced by ICH. To better understand the mechanisms of neuronal survival and neurotoxicity mediated by prostaglandin receptors, it is essential to understand downstream signaling. Several groups including ours have discovered unique roles for prostaglandin receptors in rodent models of ischemic stroke, excitotoxicity, and Alzheimer disease, highlighting the emerging role of prostaglandin receptor signaling in hemorrhagic stroke with a focus on cyclic-adenosine monophosphate and calcium (Ca2+) signaling. We review current ICH data and discuss future directions notably on prostaglandin receptors, which may lead to the development of unique therapeutic targets against hemorrhagic stroke and brain injuries alike.
Collapse
Affiliation(s)
- Shekher Mohan
- Department of Anesthesiology, College of Medicine, University of Florida Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
44
|
Munshi A, Roy S, Thangaraj K, Kaul S, Babu MS, Jyothy A. Association of SNP41, SNP56 and a novel SNP in PDE4D gene with stroke and its subtypes. Gene 2012; 506:31-5. [PMID: 22771915 DOI: 10.1016/j.gene.2012.06.079] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 06/21/2012] [Accepted: 06/25/2012] [Indexed: 01/08/2023]
Abstract
An association between phosphodiesterase 4D (PDE4D) gene and risk of stroke has been suggested by deCODE group in an Icelandic population. In the present case-control study we investigated the association of SNP41 (rs12153798) and SNP56 (rs702553) with ischemic stroke and stroke subtypes. Five hundred and sixteen ischemic stroke patients and 513 healthy age and sex matched controls were included in the study. The genotypes were determined by subjecting the PCR products to sequencing. Both the SNPs 56 and 41 associated significantly with stroke [adjusted OR=1.97; 95% CI (1.262-3.082); p=0.003: adjusted OR=5.42; 95% CI (3.45-8.5); p<0.001 respectively]. In addition to this, a novel SNP at position 59736747 T>G was found while sequencing the PCR products including SNP56. This novel SNP was found in patients as well as controls but did not show a significant association with the disease. We found significant association of SNPs 56 and 41 with large artery atherosclerosis, lacunar and cardioembolic stroke. In conclusion PDE4D gene plays a key part in the pathogenesis of ischemic stroke in the South Indian population from Andhra Pradesh.
Collapse
Affiliation(s)
- Anjana Munshi
- Institute of Genetics and Hospital for Genetic Diseases, Osmania University, Begumpet, Hyderabad-500016, India.
| | | | | | | | | | | |
Collapse
|
45
|
Zimmerman NP, Kumar SN, Turner JR, Dwinell MB. Cyclic AMP dysregulates intestinal epithelial cell restitution through PKA and RhoA. Inflamm Bowel Dis 2012; 18:1081-91. [PMID: 21993975 PMCID: PMC3258471 DOI: 10.1002/ibd.21898] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 08/22/2011] [Indexed: 01/17/2023]
Abstract
BACKGROUND Mucosal homeostasis is dependent on the establishment and maintenance of the cell-cell contacts that comprise the physiological barrier. Breaks in the barrier are linked to multiple diseases such as inflammatory bowel disease. While increased cyclic adenosine monophosphate (cAMP) levels limit inflammation by decreasing leukocyte infiltration, the effects of elevated cAMP on intestinal epithelial repair are unknown. METHODS Restitution in animals administered rolipram was monitored by microscopic examination after laser wounding of the intestinal epithelium or in mice treated with dextran sodium sulfate (DSS). In vitro analysis was conducted using IEC6 and T84 cells to determine the role of elevated cAMP in altering Rho-dependent cellular migration signaling pathways. RESULTS We show that treatment with rolipram, forskolin, and cAMP analogs decrease intestinal epithelial cell migration in vitro. In vivo cell imaging revealed that increased cAMP resulted in a decreased cellular migration rate, with cells at the edge displaying the highest activity. As expected, elevated cAMP elicited increased protein kinase A (PKA) activity, in turn resulting in the inactivation and sequestration of RhoA and decreased actin reorganization. The ablation of restitution by cAMP was not restricted to cell culture, as forskolin and rolipram treatment significantly decreased epithelial microwound closure induced by the two photon confocal injury model. CONCLUSIONS Together, these data suggest that administration of cAMP-elevating agents paradoxically decrease infiltration of damage-causing leukocytes while also preventing epithelial repair and barrier maintenance. We propose that treatment with cAMP-elevating agents severely limits mucosal reepithelialization and should be contraindicated for use in chronic inflammatory bowel disorders.
Collapse
Affiliation(s)
- Noah P. Zimmerman
- Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226
| | - Suresh N. Kumar
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226
| | | | - Michael B. Dwinell
- Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226
| |
Collapse
|
46
|
Shirshev SV. Role of Epac proteins in mechanisms of cAMP-dependent immunoregulation. BIOCHEMISTRY (MOSCOW) 2012; 76:981-98. [PMID: 22082266 DOI: 10.1134/s000629791109001x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review presents observations on the role of Epac proteins (exchange protein directly activated by cAMP) in immunoregulation mechanisms. Signaling pathways that involve Epac proteins and their domain organization and functions are considered. The role of Epac1 protein expressed in the immune system cells is especially emphasized. Molecular mechanisms of the cAMP-dependent signal via Epac1 are analyzed in monocytes/macrophages, T-cells, and B-lymphocytes. The role of Epac1 is shown in the regulation of adhesion, leukocyte chemotaxis, as well as in phagocytosis and bacterial killing. The molecular cascade initiated by Epac1 is examined under conditions of antigen activation of T-cells and immature B-lymphocytes.
Collapse
Affiliation(s)
- S V Shirshev
- Institute of Ecology and Genetics of Microorganisms, Ural Branch of the Russian Academy of Sciences, Perm, Russia.
| |
Collapse
|
47
|
Models and mechanisms of acute lung injury caused by direct insults. Eur J Cell Biol 2012; 91:590-601. [PMID: 22284832 DOI: 10.1016/j.ejcb.2011.11.004] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 11/18/2011] [Accepted: 11/30/2011] [Indexed: 11/22/2022] Open
Abstract
Acute lung injury (ALI) and its more severe form acute respiratory distress syndrome (ARDS) are life-threatening diseases that are characterized by acute onset, pulmonary inflammation, oedema due to increased vascular permeability and severe hypoxemia. Clinically, ARDS can be divided into ARDS due to direct causes such as pneumonia, aspiration or injurious ventilation, and due to extrapulmonary indirect causes such as sepsis, severe burns or pancreatitis. In order to identify potential therapeutic targets, we asked here whether common molecular mechanisms can be identified that are relevant in different models of the direct form of ALI/ARDS. To this end, we reviewed three widely used models: (a) one based on a biological insult, i.e. instillation of bacterial endotoxins; (b) one based on a chemical insult, i.e. instillation of acid; and (c) one based on a mechanical insult, i.e. injurious ventilation. Studies were included only if the mediator or mechanism of interest was studied in at least two of the three animal models listed above. As endpoints, we selected neutrophil sequestration, permeability, hypoxemia (physiological dysfunction) and survival. Our analysis showed that most studies have focused on mechanisms of pulmonary neutrophil sequestration and models with moderate forms of oedema. The underlying mechanisms that involve canonical inflammatory pathways such as MAP kinases, CXCR2 chemokines, PAF, leukotrienes, adhesions molecules (CD18, ICAM-1) and elastase have been defined relatively well. Further mechanisms including TNF, DARC, HMGB1, PARP, GADD45 and collagenase are under investigation. Such mechanisms that are shared between the three ALI models may represent viable therapeutic targets. However, only few studies have linked these pathways to hypoxemia, the most important clinical aspect of ALI/ARDS. Since moderate oedema does not necessarily lead to hypoxemia, we suggest that the clinical relevance of experimental studies can be further improved by putting greater emphasis on gas exchange.
Collapse
|
48
|
Ninković J, Roy S. Morphine decreases bacterial phagocytosis by inhibiting actin polymerization through cAMP-, Rac-1-, and p38 MAPK-dependent mechanisms. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1068-1079. [PMID: 22248582 DOI: 10.1016/j.ajpath.2011.11.034] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 11/08/2011] [Accepted: 11/14/2011] [Indexed: 11/18/2022]
Abstract
Morphine increases the susceptibility to opportunistic infection by attenuating bacterial clearance through inhibition of Fcγ receptor (FcgR)-mediated phagocytosis. Mechanisms by which morphine inhibits this process remain to be investigated. Actin polymerization is essential for FcgR-mediated internalization; therefore, disruption of the signaling mechanisms involved in this process is detrimental to the phagocytic ability of macrophages. To our knowledge, this study is the first to propose the modulation of actin polymerization and upstream signaling effectors [cAMP, Rac1-GTP, and p38 mitogen-activated protein kinase (MAPK)] as key mechanisms by which morphine leads to inhibition of pathogen clearance. Our results indicate that long-term morphine treatment in vitro and in vivo, through activation of the μ-opioid receptor, leads to an increase in intracellular cAMP, activation of protein kinase A, and inhibition of Rac1-GTPase and p38 MAPK, thereby attenuating actin polymerization and reducing membrane ruffling. Furthermore, because of long-term morphine treatment, FcgR-mediated internalization of opsonized dextran beads is also reduced. Morphine's inhibition of Rac1-GTPase activation is abolished in J774 macrophages transfected with constitutively active pcDNA3-EGFP-Rac1-Q61L plasmid. Dibutyryl-cAMP inhibits, whereas H89 restores, activation of Rac-GTPase and abolishes morphine's inhibitory effect, implicating cAMP as the key effector in morphine's modulation of actin polymerization. These findings indicate that long-term morphine treatment, by increasing intracellular cAMP and activating protein kinase A, leads to inhibition of Rac1-GTPase and p38 MAPK, causing attenuation of actin polymerization, FcgR-mediated phagocytosis, and decreased bacterial clearance.
Collapse
Affiliation(s)
- Jana Ninković
- Department of Pharmacology, University of Minnesota, School of Medicine, Minneapolis, Minnesota
| | - Sabita Roy
- Department of Pharmacology, University of Minnesota, School of Medicine, Minneapolis, Minnesota; Department of Surgery, University of Minnesota, School of Medicine, Minneapolis, Minnesota.
| |
Collapse
|
49
|
Nguyen C, Feng C, Zhan M, Cross AS, Goldblum SE. Bacillus anthracis-derived edema toxin (ET) counter-regulates movement of neutrophils and macromolecules through the endothelial paracellular pathway. BMC Microbiol 2012; 12:2. [PMID: 22230035 PMCID: PMC3277462 DOI: 10.1186/1471-2180-12-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2011] [Accepted: 01/09/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A common finding amongst patients with inhalational anthrax is a paucity of polymorphonuclear leukocytes (PMNs) in infected tissues in the face of abundant circulating PMNs. A major virulence determinant of anthrax is edema toxin (ET), which is formed by the combination of two proteins produced by the organism, edema factor (EF), which is an adenyl cyclase, and protective antigen (PA). Since cAMP, a product of adenyl cyclase, is known to enhance endothelial barrier integrity, we asked whether ET might decrease extravasation of PMNs into tissues through closure of the paracellular pathway through which PMNs traverse. RESULTS Pretreatment of human microvascular endothelial cell(EC)s of the lung (HMVEC-L) with ET decreased interleukin (IL)-8-driven transendothelial migration (TEM) of PMNs with a maximal reduction of nearly 60%. This effect required the presence of both EF and PA. Conversely, ET did not diminish PMN chemotaxis in an EC-free system. Pretreatment of subconfluent HMVEC-Ls decreased transendothelial 14 C-albumin flux by ~ 50% compared to medium controls. Coadministration of ET with either tumor necrosis factor-α or bacterial lipopolysaccharide, each at 100 ng/mL, attenuated the increase of transendothelial 14 C-albumin flux caused by either agent alone. The inhibitory effect of ET on TEM paralleled increases in protein kinase A (PKA) activity, but could not be blocked by inhibition of PKA with either H-89 or KT-5720. Finally, we were unable to replicate the ET effect with either forskolin or 3-isobutyl-1-methylxanthine, two agents known to increase cAMP. CONCLUSIONS We conclude that ET decreases IL-8-driven TEM of PMNs across HMVEC-L monolayers independent of cAMP/PKA activity.
Collapse
Affiliation(s)
- Chinh Nguyen
- Southern Arizona Veterans Affairs Health Care Systems, Tucson, AZ 85723, USA.
| | | | | | | | | |
Collapse
|
50
|
Jongsma M, Matas-Rico E, Rzadkowski A, Jalink K, Moolenaar WH. LPA is a chemorepellent for B16 melanoma cells: action through the cAMP-elevating LPA5 receptor. PLoS One 2011; 6:e29260. [PMID: 22195035 PMCID: PMC3237609 DOI: 10.1371/journal.pone.0029260] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 11/23/2011] [Indexed: 01/08/2023] Open
Abstract
Lysophosphatidic acid (LPA), a lipid mediator enriched in serum, stimulates cell migration, proliferation and other functions in many cell types. LPA acts on six known G protein-coupled receptors, termed LPA1–6, showing both overlapping and distinct signaling properties. Here we show that, unexpectedly, LPA and serum almost completely inhibit the transwell migration of B16 melanoma cells, with alkyl-LPA(18∶1) being 10-fold more potent than acyl-LPA(18∶1). The anti-migratory response to LPA is highly polarized and dependent on protein kinase A (PKA) but not Rho kinase activity; it is associated with a rapid increase in intracellular cAMP levels and PIP3 depletion from the plasma membrane. B16 cells express LPA2, LPA5 and LPA6 receptors. We show that LPA-induced chemorepulsion is mediated specifically by the alkyl-LPA-preferring LPA5 receptor (GPR92), which raises intracellular cAMP via a noncanonical pathway. Our results define LPA5 as an anti-migratory receptor and they implicate the cAMP-PKA pathway, along with reduced PIP3 signaling, as an effector of chemorepulsion in B16 melanoma cells.
Collapse
Affiliation(s)
- Maikel Jongsma
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elisa Matas-Rico
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Adrian Rzadkowski
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees Jalink
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wouter H. Moolenaar
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|