1
|
Chu S, Shen F, Liu W, Zhang J, Wang X, Jiang M, Bai G. Sinapine targeting PLCβ3 EF hands disrupts Gαq-PLCβ3 interaction and ameliorates cardiovascular diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155200. [PMID: 38387273 DOI: 10.1016/j.phymed.2023.155200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 02/24/2024]
Abstract
BACKGROUND The renin-angiotensin-aldosterone system (RAAS) over-activation is highly involved in cardiovascular diseases (CVDs), with the Gαq-PLCβ3 axis acting as a core node of RAAS. PLCβ3 is a potential target of CVDs, and the lack of inhibitors has limited its drug development. PURPOSE Sinapine (SP) is a potential leading compound for treating CVDs. Thus, we aimed to elucidate the regulation of SP towards the Gαq-PLCβ3 axis and its molecular mechanism. STUDY DESIGN Aldosteronism and hypertension animal models were employed to investigate SP's inhibitory effect on the abnormal activation of the RAAS through the Gαq-PLCβ3 axis. We used chemical biology methods to identify potential targets and elucidate the underlying molecular mechanisms. METHODS The effects of SP on aldosteronism and hypertension were evaluated using an established animal model in our laboratory. Target identification and underlying molecular mechanism research were performed using activity-based protein profiling with a bio-orthogonal click chemistry reaction and other biochemical methods. RESULTS SP alleviated aldosteronism and hypertension in animal models by targeting PLCβ3. The underlying mechanism for blocking the Gαq-PLCβ3 interaction involves targeting the EF hands through the Asn-260 amino acid residue. SP regulated the Gαq-PLCβ3 axis more precisely than the Gαq-GEFT or Gαq-PKCζ axis in the cardiovascular system. CONCLUSION SP alleviated RAAS over-activation via Gαq-PLCβ3 interaction blockade by targeting the PLCβ3 EF hands domain, which provided a novel PLC inhibitor for treating CVDs. Unlike selective Gαq inhibitors, SP reduced the risk of side effects compared to Gαq inhibitors in treating CVDs.
Collapse
Affiliation(s)
- Simeng Chu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Fukui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Wenjuan Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Jin Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China
| | - Xiaoying Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China.
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin, 300353, China.
| |
Collapse
|
2
|
Alavi MS, Soheili V, Roohbakhsh A. The role of transient receptor potential (TRP) channels in phagocytosis: A comprehensive review. Eur J Pharmacol 2024; 964:176302. [PMID: 38154767 DOI: 10.1016/j.ejphar.2023.176302] [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: 08/24/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
When host cells are exposed to foreign particles, dead cells, or cell hazards, a sophisticated process called phagocytosis begins. During this process, macrophages, dendritic cells, and neutrophils engulf the target by expanding their membranes. Phagocytosis of apoptotic cells is called efferocytosis. This process is of significant importance as billions of cells are eliminated daily without provoking inflammation. Both phagocytosis and efferocytosis depend on Ca2+ signaling. A big family of Ca2+ permeable channels is transient receptor potentials (TRPs) divided into nine subfamilies. We aimed to review their roles in phagocytosis. The present review article shows that various TRP channels such as TRPV1, 2, 3, 4, TRPM2, 4, 7, 8, TRPML1, TRPA1, TRPC1, 3, 5, 6 have roles at various stages of phagocytosis. They are involved in the phagocytosis of amyloid β, α-synuclein, myelin debris, bacteria, and apoptotic cells. In particular, TRPC3 and TRPM7 contribute to efferocytosis. These effects are mediated by changing Ca2+ signaling or targeting intracellular enzymes such as Akt. In addition, they contribute to the chemotaxis of phagocytic cells towards targets. Although a limited number of studies have assessed the role of TRP channels in phagocytosis and efferocytosis, their findings indicate that they have critical roles in these processes. In some cases, their ablation completely abolished the phagocytic function of the cells. As a result, TRP channels are potential targets for developing new therapeutics that modulate phagocytosis.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Soheili
- Pharmaceutical Control Department, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Sun B, Lin S, Zheng M, Zheng B, Mao L, Gu Y, Cai J, Dai Y, Zheng M, Lou Y. Phosphatidylinositol-specific phospholipase C can decrease Müller cell viability and suppress its phagocytic activity by modulating PI3K/AKT signaling pathway. Can J Microbiol 2023; 69:501-511. [PMID: 37672795 DOI: 10.1139/cjm-2023-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Bacillus cereus endophthalmitis is a devastating eye infection that causes rapid blindness through the release of extracellular tissue-destructive exotoxins. The phagocytic and antibacterial functions of ocular cells are the keys to limiting ocular bacterial infections. In a previous study, we identified a new virulence gene, plcA-2 (different from the original plcA-1 gene), that was strongly associated with the plcA gene of Listeria monocytogenes. This plcA gene had been confirmed to play an important role in phagocytosis. However, how the Bc-phosphatidylinositol-specific phospholipase C (PI-PLC) proteins encoded by the plcA-1/2 genes affect phagocytes remains unclear in B. cereus endophthalmitis. Here, we found that the enzymatic activity of Bc-PI-PLC-A2 was approximately twofold higher than that of Bc-PI-PLC-A1, and both proteins inhibited the viability of Müller cells. In addition, PI-PLC proteins reduced phagocytosis of Müller cells by decreasing the phosphorylation levels of key proteins in the PI3K/AKT signaling pathway. In conclusion, we showed that PI-PLC proteins contribute to inhibit the viability of and suppress the phagocytosis of Müller cells, providing new insights into the pathogenic mechanism of B. cereus endophthalmitis.
Collapse
Affiliation(s)
- Bianjin Sun
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Shudan Lin
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Clinical Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou, China
| | - Mengmeng Zheng
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Beijia Zheng
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Liping Mao
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yunfeng Gu
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiabei Cai
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yiran Dai
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Meiqin Zheng
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yongliang Lou
- Zhejiang Provincial Key Laboratory for Technology and Application of Model Organisms, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Kanemaru K, Nakamura Y. Activation Mechanisms and Diverse Functions of Mammalian Phospholipase C. Biomolecules 2023; 13:915. [PMID: 37371495 DOI: 10.3390/biom13060915] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Phospholipase C (PLC) plays pivotal roles in regulating various cellular functions by metabolizing phosphatidylinositol 4,5-bisphosphate in the plasma membrane. This process generates two second messengers, inositol 1,4,5-trisphosphate and diacylglycerol, which respectively regulate the intracellular Ca2+ levels and protein kinase C activation. In mammals, six classes of typical PLC have been identified and classified based on their structure and activation mechanisms. They all share X and Y domains, which are responsible for enzymatic activity, as well as subtype-specific domains. Furthermore, in addition to typical PLC, atypical PLC with unique structures solely harboring an X domain has been recently discovered. Collectively, seven classes and 16 isozymes of mammalian PLC are known to date. Dysregulation of PLC activity has been implicated in several pathophysiological conditions, including cancer, cardiovascular diseases, and neurological disorders. Therefore, identification of new drug targets that can selectively modulate PLC activity is important. The present review focuses on the structures, activation mechanisms, and physiological functions of mammalian PLC.
Collapse
Affiliation(s)
- Kaori Kanemaru
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| | - Yoshikazu Nakamura
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba 278-8510, Japan
| |
Collapse
|
5
|
Kök GF, Türsen Ü. The Immunogenetics of Granulomatous Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:349-368. [DOI: 10.1007/978-3-030-92616-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Wang W, Wu J, Liu P, Tang X, Pang H, Xie T, Xu F, Shao J, Chen Y, Liu B, Zheng Y. Urinary Proteomics Identifying Novel Biomarkers for the Diagnosis and Phenotyping of Carotid Artery Stenosis. Front Mol Biosci 2021; 8:714706. [PMID: 34447787 PMCID: PMC8383446 DOI: 10.3389/fmolb.2021.714706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/26/2021] [Indexed: 01/12/2023] Open
Abstract
Background: Carotid artery stenosis (CAS) is caused by the formation of atherosclerotic plaques inside the arterial wall and accounts for 20–30% of all strokes. The development of an early, noninvasive diagnostic method and the identification of high-risk patients for ischemic stroke is essential to the management of CAS in clinical practice. Methods: We used the data-independent acquisition (DIA) technique to conduct a urinary proteomic study in patients with CAS and healthy controls. We identified the potential diagnosis and risk stratification biomarkers of CAS. And Ingenuity pathway analysis was used for functional annotation of differentially expressed proteins (DEPs). Furthermore, receiver operating characteristic (ROC) analysis was performed to evaluate the diagnostic values of DEPs. Results: A total of 194 DEPs were identified between CAS patients and healthy controls by DIA quantification. The bioinformatics analysis showed that these DEPs were correlated with the pathogenesis of CAS. We further identified 32 DEPs in symptomatic CAS compared to asymptomatic CAS, and biological function analysis revealed that these proteins are mainly related to immune/inflammatory pathways. Finally, a biomarker panel of six proteins (ACP2, PLD3, HLA-C, GGH, CALML3, and IL2RB) exhibited potential diagnostic value in CAS and good discriminative power for differentiating symptomatic and asymptomatic CAS with high sensitivity and specificity. Conclusions: Our study identified novel potential urinary biomarkers for noninvasive early screening and risk stratification of CAS.
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianqiang Wu
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyue Tang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyu Pang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Xie
- State Key Laboratory of Complex Severe and Rare Diseases, Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Xu
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang Shao
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuexin Chen
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bao Liu
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuehong Zheng
- State Key Laboratory of Complex Severe and Rare Disease, Department of Vascular Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
7
|
Surendran P, Feofanova EV, Lahrouchi N, Ntalla I, Karthikeyan S, Cook J, Chen L, Mifsud B, Yao C, Kraja AT, Cartwright JH, Hellwege JN, Giri A, Tragante V, Thorleifsson G, Liu DJ, Prins BP, Stewart ID, Cabrera CP, Eales JM, Akbarov A, Auer PL, Bielak LF, Bis JC, Braithwaite VS, Brody JA, Daw EW, Warren HR, Drenos F, Nielsen SF, Faul JD, Fauman EB, Fava C, Ferreira T, Foley CN, Franceschini N, Gao H, Giannakopoulou O, Giulianini F, Gudbjartsson DF, Guo X, Harris SE, Havulinna AS, Helgadottir A, Huffman JE, Hwang SJ, Kanoni S, Kontto J, Larson MG, Li-Gao R, Lindström J, Lotta LA, Lu Y, Luan J, Mahajan A, Malerba G, Masca NGD, Mei H, Menni C, Mook-Kanamori DO, Mosen-Ansorena D, Müller-Nurasyid M, Paré G, Paul DS, Perola M, Poveda A, Rauramaa R, Richard M, Richardson TG, Sepúlveda N, Sim X, Smith AV, Smith JA, Staley JR, Stanáková A, Sulem P, Thériault S, Thorsteinsdottir U, Trompet S, Varga TV, Velez Edwards DR, Veronesi G, Weiss S, Willems SM, Yao J, Young R, Yu B, Zhang W, Zhao JH, Zhao W, Zhao W, Evangelou E, Aeschbacher S, Asllanaj E, Blankenberg S, Bonnycastle LL, Bork-Jensen J, Brandslund I, Braund PS, Burgess S, Cho K, Christensen C, Connell J, Mutsert RD, Dominiczak AF, Dörr M, Eiriksdottir G, Farmaki AE, Gaziano JM, Grarup N, Grove ML, Hallmans G, Hansen T, Have CT, Heiss G, Jørgensen ME, Jousilahti P, Kajantie E, Kamat M, Käräjämäki A, Karpe F, Koistinen HA, Kovesdy CP, Kuulasmaa K, Laatikainen T, Lannfelt L, Lee IT, Lee WJ, Linneberg A, Martin LW, Moitry M, Nadkarni G, Neville MJ, Palmer CNA, Papanicolaou GJ, Pedersen O, Peters J, Poulter N, Rasheed A, Rasmussen KL, Rayner NW, Mägi R, Renström F, Rettig R, Rossouw J, Schreiner PJ, Sever PS, Sigurdsson EL, Skaaby T, Sun YV, Sundstrom J, Thorgeirsson G, Esko T, Trabetti E, Tsao PS, Tuomi T, Turner ST, Tzoulaki I, Vaartjes I, Vergnaud AC, Willer CJ, Wilson PWF, Witte DR, Yonova-Doing E, Zhang H, Aliya N, Almgren P, Amouyel P, Asselbergs FW, Barnes MR, Blakemore AI, Boehnke M, Bots ML, Bottinger EP, Buring JE, Chambers JC, Chen YDI, Chowdhury R, Conen D, Correa A, Davey Smith G, Boer RAD, Deary IJ, Dedoussis G, Deloukas P, Di Angelantonio E, Elliott P, Felix SB, Ferrières J, Ford I, Fornage M, Franks PW, Franks S, Frossard P, Gambaro G, Gaunt TR, Groop L, Gudnason V, Harris TB, Hayward C, Hennig BJ, Herzig KH, Ingelsson E, Tuomilehto J, Järvelin MR, Jukema JW, Kardia SLR, Kee F, Kooner JS, Kooperberg C, Launer LJ, Lind L, Loos RJF, Majumder AAS, Laakso M, McCarthy MI, Melander O, Mohlke KL, Murray AD, Nordestgaard BG, Orho-Melander M, Packard CJ, Padmanabhan S, Palmas W, Polasek O, Porteous DJ, Prentice AM, Province MA, Relton CL, Rice K, Ridker PM, Rolandsson O, Rosendaal FR, Rotter JI, Rudan I, Salomaa V, Samani NJ, Sattar N, Sheu WHH, Smith BH, Soranzo N, Spector TD, Starr JM, Sebert S, Taylor KD, Lakka TA, Timpson NJ, Tobin MD, van der Harst P, van der Meer P, Ramachandran VS, Verweij N, Virtamo J, Völker U, Weir DR, Zeggini E, Charchar FJ, Wareham NJ, Langenberg C, Tomaszewski M, Butterworth AS, Caulfield MJ, Danesh J, Edwards TL, Holm H, Hung AM, Lindgren CM, Liu C, Manning AK, Morris AP, Morrison AC, O'Donnell CJ, Psaty BM, Saleheen D, Stefansson K, Boerwinkle E, Chasman DI, Levy D, Newton-Cheh C, Munroe PB, Howson JMM. Discovery of rare variants associated with blood pressure regulation through meta-analysis of 1.3 million individuals. Nat Genet 2020; 52:1314-1332. [PMID: 33230300 PMCID: PMC7610439 DOI: 10.1038/s41588-020-00713-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/08/2020] [Indexed: 01/14/2023]
Abstract
Genetic studies of blood pressure (BP) to date have mainly analyzed common variants (minor allele frequency > 0.05). In a meta-analysis of up to ~1.3 million participants, we discovered 106 new BP-associated genomic regions and 87 rare (minor allele frequency ≤ 0.01) variant BP associations (P < 5 × 10-8), of which 32 were in new BP-associated loci and 55 were independent BP-associated single-nucleotide variants within known BP-associated regions. Average effects of rare variants (44% coding) were ~8 times larger than common variant effects and indicate potential candidate causal genes at new and known loci (for example, GATA5 and PLCB3). BP-associated variants (including rare and common) were enriched in regions of active chromatin in fetal tissues, potentially linking fetal development with BP regulation in later life. Multivariable Mendelian randomization suggested possible inverse effects of elevated systolic and diastolic BP on large artery stroke. Our study demonstrates the utility of rare-variant analyses for identifying candidate genes and the results highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Elena V Feofanova
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Najim Lahrouchi
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences Amsterdam, Amsterdam, the Netherlands
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - James Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Lingyan Chen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Borbala Mifsud
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Chen Yao
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aldi T Kraja
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - James H Cartwright
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jacklyn N Hellwege
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Ayush Giri
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Vinicius Tragante
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
| | | | - Dajiang J Liu
- Institute of Personalized Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Bram P Prins
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Isobel D Stewart
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Claudia P Cabrera
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Artur Akbarov
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Paul L Auer
- Joseph J Zilber School of Public Health, University of Wisconsin, Milwaukee, WI, USA
| | - Lawrence F Bielak
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Vickie S Braithwaite
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Nutrition and Bone Health Group, University of Cambridge, Cambridge, UK
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, Gambia
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - E Warwick Daw
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Fotios Drenos
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
| | - Sune Fallgaard Nielsen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Cristiano Fava
- Department of Medicine, University of Verona, Verona, Italy
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Teresa Ferreira
- The Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Christopher N Foley
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - He Gao
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
| | - Olga Giannakopoulou
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Genomic Health, Queen Mary University of London, London, UK
- Division of Psychiatry, University College of London, London, UK
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Aki S Havulinna
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | | | - Jennifer E Huffman
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, UK
| | - Shih-Jen Hwang
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Genomic Health, Life Sciences, Queen Mary University of London, London, UK
| | - Jukka Kontto
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Martin G Larson
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Biostatistics Department, Boston University School of Public Health, Boston, MA, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jaana Lindström
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt Epidemiology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Giovanni Malerba
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Nicholas G D Masca
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Hao Mei
- Department of Data Science, School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - David Mosen-Ansorena
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Medicine I, Ludwig-Maximilians-University Munich, Munich, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU, Munich, Germany
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dirk S Paul
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Markus Perola
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Clinical and Molecular Metabolism Research Program (CAMM), Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alaitz Poveda
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Melissa Richard
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nuno Sepúlveda
- Department of Infection Biology, Faculty of Tropical and Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Centre of Statistics and Applications of University of Lisbon, Lisbon, Portugal
| | - Xueling Sim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Saw Swee Hock School of Public Health, National University of, Singapore, Singapore
| | - Albert V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - James R Staley
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Alena Stanáková
- University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | | | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Stella Trompet
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tibor V Varga
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Tennessee Valley Health Systems VA, Nashville, TN, USA
| | - Giovanni Veronesi
- Research Center in Epidemiology and Preventive Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and University of Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Robin Young
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Bing Yu
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
| | - Jing-Hua Zhao
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Wei Zhao
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | | | - Eralda Asllanaj
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Stefan Blankenberg
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
- University Medical Center Hamburg Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lori L Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ivan Brandslund
- Department of Clinical Biochemistry, Lillebaelt Hospital, Vejle, Denmark
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - John Connell
- University of Dundee, Ninewells Hospital & Medical School, Dundee, UK
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anna F Dominiczak
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | | | - Aliki-Eleni Farmaki
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
- Department of Population Science and Experimental Medicine, Institute of Cardiovascular Science, University College London, London, UK
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian T Have
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | - Pekka Jousilahti
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Eero Kajantie
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- PEDEGO Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Mihir Kamat
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
| | - AnneMari Käräjämäki
- Department of Primary Health Care, Vaasa Central Hospital, Vaasa, Finland
- Diabetes Center, Vaasa Health Care Center, Vaasa, Finland
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| | - Heikki A Koistinen
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Csaba P Kovesdy
- Nephrology Section, Memphis VA Medical Center, Memphis, TN, USA
| | - Kari Kuulasmaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Tiina Laatikainen
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, , Chung Shan Medical University, Taichung, Taiwan
- College of Science, Tunghai University, Taichung, Taiwan
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Social Work, Tunghai University, Taichung, Taiwan
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisa W Martin
- George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Marie Moitry
- Department of Public health, Strasbourg University Hospital, University of Strasbourg, Strasbourg, France
| | - Girish Nadkarni
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| | - Colin N A Palmer
- Medical Research Institute, University of Dundee, Ninewells Hospital and Medical School, Dundee, Scotland, UK
| | | | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - James Peters
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Neil Poulter
- International Centre for Circulatory Health, Imperial College London, London, UK
| | - Asif Rasheed
- Centre for Non-Communicable Diseases, Karachi, Pakistan
| | - Katrine L Rasmussen
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - N William Rayner
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Reedik Mägi
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Frida Renström
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Rainer Rettig
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Jacques Rossouw
- Division of Cardiovascular Sciences, NHLBI, Bethesda, MD, USA
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Peter S Sever
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Emil L Sigurdsson
- Department of Family Medicine, University of Iceland, Reykjavik, Iceland
- Development Centre for Primary Health Care in Iceland, Reykjavik, Iceland
| | - Tea Skaaby
- Center for Clinical Research and Disease Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Johan Sundstrom
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gudmundur Thorgeirsson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Internal Medicine, Division of Cardiology, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Elisabetta Trabetti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Philip S Tsao
- VA Palo Alto Health Care System, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Tiinamaija Tuomi
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Endocrinology, Helsinki University Central Hospital, Helsinki, Finland
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden Institute for Molecular Medicine Helsinki (FIMM), Helsinki University, Helsinki, Finland
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Ilonca Vaartjes
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, University of Utrecht, Utrecht, the Netherlands
- Center for Circulatory Health, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Anne-Claire Vergnaud
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Peter W F Wilson
- Atlanta VAMC and Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Diabetes Academy, Odense, Denmark
- Steno Diabetes Center Aarhus, Aarhus, Denmark
| | - Ekaterina Yonova-Doing
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - He Zhang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Naheed Aliya
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Peter Almgren
- Department of Medicine, Lund University, Malmö, Sweden
| | - Philippe Amouyel
- Univ Lille, U1167 - RID-AGE - Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, France
- INSERM, U1167, Lille, France
- CHU Lille, U1167, Lille, France
- Institut Pasteur de Lille, U1167, Lille, France
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Institute of Cardiovascular Science, Faculty of Population Health Sciences, University College London, London, UK
- Health Data Research UK, Institute of Health Informatics, University College London, London, UK
| | - Michael R Barnes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Alexandra I Blakemore
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
- Section of Investigative Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Michiel L Bots
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University of Utrecht, University of Utrecht, Utrecht, the Netherlands
- Center for Circulatory Health, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie E Buring
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Imperial College Healthcare NHS Trust, London, UK
| | - Yii-Der Ida Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Rajiv Chowdhury
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Non-communicable Disease Research (CNCR), Dhaka, Bangladesh
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Cardiovascular Research Institute Basel, Basel, Switzerland
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Rudolf A de Boer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
- Centre for Genomic Health, Life Sciences, Queen Mary University of London, London, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Health Data Research UK-London at Imperial College London, London, UK
- UKDRI, Dementia Research Institute at Imperial College London, London, UK
- British Heart Foundation (BHF) Centre of Research Excellence, Imperial College London, London, UK
| | - Stephan B Felix
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Jean Ferrières
- Department of Cardiology and Department of Epidemiology, INSERM UMR 1027, Toulouse University Hospital, Toulouse, France
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul W Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, Skåne University Hospital Malmö, Malmö, Sweden
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
- Oxford Center for Diabetes, Endocrinology & Metabolism, Radcliff Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen Franks
- Institute of Reproductive & Developmental Biology, Imperial College London, London, UK
| | | | - Giovanni Gambaro
- Division of Nephrology, Department of Medicine, University of Verona, Verona, Italy
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Helsinki (FIMM), Helsinki University, Helsinki, Finland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute of Aging, Bethesda, MD, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, IGMM, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, UK
| | - Branwen J Hennig
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, Gambia
- Wellcome Trust, London, UK
| | - Karl-Heinz Herzig
- Institute of Biomedicine, Medical Research Center (MRC), University of Oulu, and University Hospital Oulu, Oulu, Finland
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Jaakko Tuomilehto
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
- National Institute of Public Health, Madrid, Spain
| | - Marjo-Riitta Järvelin
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
- Department of Epidemiology and Biostatistics, MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Unit of Primary Care, Oulu University Hospital, Kajaanintie, Oulu, Finland
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Frank Kee
- Centre for Public Health, Queens University Belfast, Belfast, UK
| | - Jaspal S Kooner
- National Institute for Health Research (NIHR) Imperial Biomedical Research Centre, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, Middlesex, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute of Aging, Bethesda, MD, USA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Genentech, South San Francisco, San Francisco, CA, USA
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Alison D Murray
- The Institute of Medical Sciences, Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | | | | | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Ozren Polasek
- Department of Public Health, University of Split School of Medicine, Split, Croatia
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Andrew M Prentice
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, Gambia
- MRC International Nutrition Group at London School of Hygiene and Tropical Medicine, Keppel St, London, UK
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics and Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St Louis, MO, USA
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Olov Rolandsson
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, UK
| | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
- Institute of Medical Technology, National Chung-Hsing University, Taichung, Taiwan
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Nicole Soranzo
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Research Centre, University of Edinburgh, Edinburgh, UK
| | - Sylvain Sebert
- Center for Life-Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Timo A Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio, Finland
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Martin D Tobin
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, the Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, the Netherlands
| | - Peter van der Meer
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Vasan S Ramachandran
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Niek Verweij
- University Medical Center Groningen, Groningen, the Netherlands
| | - Jarmo Virtamo
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and University of Greifswald, Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Eleftheria Zeggini
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Institute of Translational Genomics, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Fadi J Charchar
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Health Innovation and Transformation Center, Federation University Australia, Ballarat, Victoria, Australia
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
- Division of Medicine, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit (BTRU) in Donor Health and Genomics at the University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Hilma Holm
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
| | - Adriana M Hung
- VA Tennessee Valley Healthcare System, Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- The Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Chunyu Liu
- Boston University School of Public Health, Boston, MA, USA
| | - Alisa K Manning
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christopher J O'Donnell
- VA Boston Healthcare, Section of Cardiology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Danish Saleheen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Kari Stefansson
- deCODE genetics/Amgen, Inc, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Daniel Levy
- Boston University and National Heart, Lung and Blood Institute Framingham Heart Study, Framingham, MA, USA
- Population Sciences, Branch, National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, MD, USA
| | - Christopher Newton-Cheh
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK.
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK.
- Novo Nordisk Research Centre Oxford, Novo Nordisk Ltd, Oxford, UK.
| |
Collapse
|
8
|
Katan M, Cockcroft S. Phospholipase C families: Common themes and versatility in physiology and pathology. Prog Lipid Res 2020; 80:101065. [PMID: 32966869 DOI: 10.1016/j.plipres.2020.101065] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/14/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
Phosphoinositide-specific phospholipase Cs (PLCs) are expressed in all mammalian cells and play critical roles in signal transduction. To obtain a comprehensive understanding of these enzymes in physiology and pathology, a detailed structural, biochemical, cell biological and genetic information is required. In this review, we cover all these aspects to summarize current knowledge of the entire superfamily. The families of PLCs have expanded from 13 enzymes to 16 with the identification of the atypical PLCs in the human genome. Recent structural insights highlight the common themes that cover not only the substrate catalysis but also the mechanisms of activation. This involves the release of autoinhibitory interactions that, in the absence of stimulation, maintain classical PLC enzymes in their inactive forms. Studies of individual PLCs provide a rich repertoire of PLC function in different physiologies. Furthermore, the genetic studies discovered numerous mutated and rare variants of PLC enzymes and their link to human disease development, greatly expanding our understanding of their roles in diverse pathologies. Notably, substantial evidence now supports involvement of different PLC isoforms in the development of specific cancer types, immune disorders and neurodegeneration. These advances will stimulate the generation of new drugs that target PLC enzymes, and will therefore open new possibilities for treatment of a number of diseases where current therapies remain ineffective.
Collapse
Affiliation(s)
- Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Shamshad Cockcroft
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, 21 University Street, London WC1E 6JJ, UK.
| |
Collapse
|
9
|
Interface of Phospholipase Activity, Immune Cell Function, and Atherosclerosis. Biomolecules 2020; 10:biom10101449. [PMID: 33076403 PMCID: PMC7602611 DOI: 10.3390/biom10101449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022] Open
Abstract
Phospholipases are a family of lipid-altering enzymes that can either reduce or increase bioactive lipid levels. Bioactive lipids elicit signaling responses, activate transcription factors, promote G-coupled-protein activity, and modulate membrane fluidity, which mediates cellular function. Phospholipases and the bioactive lipids they produce are important regulators of immune cell activity, dictating both pro-inflammatory and pro-resolving activity. During atherosclerosis, pro-inflammatory and pro-resolving activities govern atherosclerosis progression and regression, respectively. This review will look at the interface of phospholipase activity, immune cell function, and atherosclerosis.
Collapse
|
10
|
Ren C, Yuan Q, Jian X, Randazzo PA, Tang W, Wu D. Small GTPase ARF6 Is a Coincidence-Detection Code for RPH3A Polarization in Neutrophil Polarization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1012-1021. [PMID: 31924649 PMCID: PMC6994837 DOI: 10.4049/jimmunol.1901080] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/10/2019] [Indexed: 12/21/2022]
Abstract
Cell polarization is a key step for leukocytes adhesion and transmigration during leukocytes' inflammatory infiltration. Polarized localization of plasma membrane (PM) phosphatidylinositol-4-phosphate (PtdIns4P) directs the polarization of RPH3A, which contains a PtdIns4P binding site. Consequently, RPH3A mediates the RAB21 and PIP5K1C90 polarization, which is important for neutrophil adhesion to endothelia during inflammation. However, the mechanism by which RPH3A is recruited only to PM PtdIns4P rather than Golgi PtdIns4P remains unclear. By using ADP-ribosylation factor 6 (ARF6) small interfering RNA, ARF6 dominant-negative mutant ARF6(T27N), and ARF6 activation inhibitor SecinH3, we demonstrate that ARF6 plays an important role in the polarization of RPH3A, RAB21, and PIP5K1C90 in murine neutrophils. PM ARF6 is polarized and colocalized with RPH3A, RAB21, PIP5K1C90, and PM PtdIns4P in mouse and human neutrophils upon integrin stimulation. Additionally, ARF6 binds to RPH3A and enhances the interaction between the PM PtdIns4P and RPH3A. Consistent with functional roles of polarization of RPH3A, Rab21, and PIP5K1C90, ARF6 is also required for neutrophil adhesion on the inflamed endothelial layer. Our study reveals a previously unknown role of ARF6 in neutrophil polarization as being the coincidence-detection code with PM PtdIns4P. Cooperation of ARF6 and PM PtdIns4P direct RPH3A polarization, which is important for neutrophil firm adhesion to endothelia.
Collapse
Affiliation(s)
- Chunguang Ren
- Department of Pharmacology, Vascular Biology and Therapeutic Program, School of Medicine, Yale University, New Haven, CT 06520; and
| | - Qianying Yuan
- Department of Pharmacology, Vascular Biology and Therapeutic Program, School of Medicine, Yale University, New Haven, CT 06520; and
| | - Xiaoying Jian
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Paul A Randazzo
- Laboratory of Cellular and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Wenwen Tang
- Department of Pharmacology, Vascular Biology and Therapeutic Program, School of Medicine, Yale University, New Haven, CT 06520; and
| | - Dianqing Wu
- Department of Pharmacology, Vascular Biology and Therapeutic Program, School of Medicine, Yale University, New Haven, CT 06520; and
| |
Collapse
|
11
|
Liu F, Dai S, Feng D, Peng X, Qin Z, Kearns AC, Huang W, Chen Y, Ergün S, Wang H, Rappaport J, Bryda EC, Chandrasekhar A, Aktas B, Hu H, Chang SL, Gao B, Qin X. Versatile cell ablation tools and their applications to study loss of cell functions. Cell Mol Life Sci 2019; 76:4725-4743. [PMID: 31359086 PMCID: PMC6858955 DOI: 10.1007/s00018-019-03243-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 12/22/2022]
Abstract
Targeted cell ablation is a powerful approach for studying the role of specific cell populations in a variety of organotypic functions, including cell differentiation, and organ generation and regeneration. Emerging tools for permanently or conditionally ablating targeted cell populations and transiently inhibiting neuronal activities exhibit a diversity of application and utility. Each tool has distinct features, and none can be universally applied to study different cell types in various tissue compartments. Although these tools have been developed for over 30 years, they require additional improvement. Currently, there is no consensus on how to select the tools to answer the specific scientific questions of interest. Selecting the appropriate cell ablation technique to study the function of a targeted cell population is less straightforward than selecting the method to study a gene's functions. In this review, we discuss the features of the various tools for targeted cell ablation and provide recommendations for optimal application of specific approaches.
Collapse
Affiliation(s)
- Fengming Liu
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Shen Dai
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiao Peng
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Zhongnan Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Alison C Kearns
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Wenfei Huang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Yong Chen
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
- Key Lab for Immunology in Universities of Shandong Province, School of Clinical Medicine, Weifang Medical University, 261053, Weifang, People's Republic of China
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximillan University, 97070, Wurzburg, Germany
| | - Hong Wang
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA
| | - Jay Rappaport
- Division of Pathology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, 4011 Discovery Drive, Columbia, MO, 65201, USA
| | - Anand Chandrasekhar
- Division of Biological Sciences, 340D Life Sciences Center, University of Missouri, 1201 Rollins St, Columbia, MO, USA
| | - Bertal Aktas
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sulie L Chang
- Institute of NeuroImmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuebin Qin
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, 3500 N Broad Street, Philadelphia, PA, 19140, USA.
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA.
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
12
|
Ren C, Yuan Q, Braun M, Zhang X, Petri B, Zhang J, Kim D, Guez-Haddad J, Xue W, Pan W, Fan R, Kubes P, Sun Z, Opatowsky Y, Polleux F, Karatekin E, Tang W, Wu D. Leukocyte Cytoskeleton Polarization Is Initiated by Plasma Membrane Curvature from Cell Attachment. Dev Cell 2019; 49:206-219.e7. [PMID: 30930167 DOI: 10.1016/j.devcel.2019.02.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 01/15/2019] [Accepted: 02/25/2019] [Indexed: 12/30/2022]
Abstract
Cell polarization is important for various biological processes. However, its regulation, particularly initiation, is incompletely understood. Here, we investigated mechanisms by which neutrophils break their symmetry and initiate their cytoskeleton polarization from an apolar state in circulation for their extravasation during inflammation. We show here that a local increase in plasma membrane (PM) curvature resulting from cell contact to a surface triggers the initial breakage of the symmetry of an apolar neutrophil and is required for subsequent polarization events induced by chemical stimulation. This local increase in PM curvature recruits SRGAP2 via its F-BAR domain, which in turn activates PI4KA and results in PM PtdIns4P polarization. Polarized PM PtdIns4P is targeted by RPH3A, which directs PIP5K1C90 and subsequent phosphorylated myosin light chain polarization, and this polarization signaling axis regulates neutrophil firm attachment to endothelium. Thus, this study reveals a mechanism for the initiation of cell cytoskeleton polarization.
Collapse
Affiliation(s)
- Chunguang Ren
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale University, New Haven, CT 06520, USA
| | - Qianying Yuan
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale University, New Haven, CT 06520, USA
| | - Martha Braun
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA; Nanobiology Institute, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Xia Zhang
- Department of Geriatrics, the First affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Björn Petri
- Snyder Institute for Chronic Diseases Mouse Phenomics Resource Laboratory, University of Calgary, Calgary AB T2N 4N1, Canada; Department of Microbiology, Immunology, and Infectious Diseases, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Jiasheng Zhang
- Department of Internal Medicine, Yale University, New Haven, CT 06520, USA
| | - Dongjoo Kim
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Julia Guez-Haddad
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Wenzhi Xue
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Weijun Pan
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences (CAS), Shanghai, China
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Paul Kubes
- Snyder Institute for Chronic Diseases Mouse Phenomics Resource Laboratory, University of Calgary, Calgary AB T2N 4N1, Canada; Department of Physiology and Pharmacology, Cumming School of Medicine, and Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary AB T2N 4N1, Canada
| | - Zhaoxia Sun
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Franck Polleux
- Department of Neuroscience, Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10025, USA
| | - Erdem Karatekin
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520, USA; Nanobiology Institute, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, Yale University, New Haven, CT 06520, USA; Centre National de la Recherche Scientifique (CNRS), Paris, France.
| | - Wenwen Tang
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale University, New Haven, CT 06520, USA.
| | - Dianqing Wu
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
13
|
Chan EC, Ren C, Xie Z, Jude J, Barker T, Koziol-White CA, Ma M, Panettieri RA, Wu D, Rosenberg HF, Druey KM. Regulator of G protein signaling 5 restricts neutrophil chemotaxis and trafficking. J Biol Chem 2018; 293:12690-12702. [PMID: 29929985 DOI: 10.1074/jbc.ra118.002404] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/14/2018] [Indexed: 01/09/2023] Open
Abstract
Neutrophils are white blood cells that are mobilized to damaged tissues and to sites of pathogen invasion, providing the first line of host defense. Chemokines displayed on the surface of blood vessels promote migration of neutrophils to these sites, and tissue- and pathogen-derived chemoattractant signals, including N-formylmethionylleucylphenylalanine (fMLP), elicit further migration to sites of infection. Although nearly all chemoattractant receptors use heterotrimeric G proteins to transmit signals, many of the mechanisms lying downstream of chemoattractant receptors that either promote or limit neutrophil motility are incompletely defined. Here, we show that regulator of G protein signaling 5 (RGS5), a protein that modulates G protein activity, is expressed in both human and murine neutrophils. We detected significantly more neutrophils in the airways of Rgs5-/- mice than WT counterparts following acute respiratory virus infection and in the peritoneum in response to injection of thioglycollate, a biochemical proinflammatory stimulus. RGS5-deficient neutrophils responded with increased chemotaxis elicited by the chemokines CXC motif chemokine ligand 1 (CXCL1), CXCL2, and CXCL12 but not fMLP. Moreover, adhesion of these cells was increased in the presence of both CXCL2 and fMLP. In summary, our results indicate that RGS5 deficiency increases chemotaxis and adhesion, leading to more efficient neutrophil mobilization to inflamed tissues in mice. These findings suggest that RGS5 expression and activity in neutrophils determine their migrational patterns in the complex microenvironments characteristic of inflamed tissues.
Collapse
Affiliation(s)
- Eunice C Chan
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Chunguang Ren
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Zhihui Xie
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Joseph Jude
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Tolga Barker
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Cynthia A Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Michelle Ma
- Inflammation Immunobiology Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute of New Jersey, Rutgers New Jersey School of Medicine, Rutgers, New Jersey 07103
| | - Dianqing Wu
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06510
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Kirk M Druey
- Molecular Signal Transduction Section, NIAID, National Institutes of Health, Bethesda, Maryland 20892.
| |
Collapse
|
14
|
Huang W, Wang X, Endo-Streeter S, Barrett M, Waybright J, Wohlfeld C, Hajicek N, Harden TK, Sondek J, Zhang Q. A membrane-associated, fluorogenic reporter for mammalian phospholipase C isozymes. J Biol Chem 2017; 293:1728-1735. [PMID: 29263090 DOI: 10.1074/jbc.ra117.000926] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 12/05/2017] [Indexed: 11/06/2022] Open
Abstract
A diverse group of cell-surface receptors, including many G protein-coupled receptors and receptor tyrosine kinases, activate phospholipase C (PLC) isozymes to hydrolyze phosphatidylinositol 4,5-bisphosphate into the second messengers diacylglycerol and 1,4,5-inositol trisphosphate. Consequently, PLCs control various cellular processes, and their aberrant regulation contributes to many diseases, including cancer, atherosclerosis, and rheumatoid arthritis. Despite the widespread importance of PLCs in human biology and disease, it has been impossible to directly monitor the real-time activation of these enzymes at membranes. To overcome this limitation, here we describe XY-69, a fluorogenic reporter that preferentially partitions into membranes and provides a selective tool for measuring the real-time activity of PLCs as either purified enzymes or in cellular lysates. Indeed, XY-69 faithfully reported the membrane-dependent activation of PLC-β3 by Gαq Therefore, XY-69 can replace radioactive phosphatidylinositol 4,5-bisphosphate used in conventional PLC assays and will enable high-throughput screens to identify both orthosteric and allosteric PLC inhibitors. In the future, cell-permeable variants of XY-69 represent promising candidates for reporting the activation of PLCs in live cells with high spatiotemporal resolution.
Collapse
Affiliation(s)
- Weigang Huang
- From the Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy
| | - Xiaoyang Wang
- From the Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy
| | | | | | - Jarod Waybright
- From the Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy
| | - Christian Wohlfeld
- From the Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy
| | | | | | - John Sondek
- Departments of Pharmacology and.,Biochemistry and Biophysics, School of Medicine, and.,the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Qisheng Zhang
- From the Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, .,Departments of Pharmacology and.,the Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
15
|
Xiong W, Wang H, Lu L, Xi R, Wang F, Gu G, Tao R. The macrophage C-type lectin receptor CLEC5A (MDL-1) expression is associated with early plaque progression and promotes macrophage survival. J Transl Med 2017; 15:234. [PMID: 29126450 PMCID: PMC5681784 DOI: 10.1186/s12967-017-1336-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/30/2017] [Indexed: 12/11/2022] Open
Abstract
Background Biomarkers of early plaque progression are still elusive. Myeloid DAP12-associating lectin-1 (MDL-1), also called CLEC5A, is a C-type lectin receptor implicated in the progression of multiple acute and chronic inflammatory diseases. However, the relationship between its level and atherosclerosis is unknown. In this study, we aimed to investigate the correlation between macrophage MDL-1 expression and early atherosclerosis progression. Methods Immunofluorescence staining, real-time PCR and western blot were performed to analyze MDL-1 expression in aorta or mice macrophages. The role of MDL-1 in macrophage survival was further investigated by adenovirus infection and TUNEL assay. Results Significant MDL-1 expression was found in advanced human and apoE−/− mice atherosclerotic plaques, especially in lesional macrophages. In the model of atherosclerosis regression, we found MDL-1 expression was highly downregulated in lesional macrophages from ldlr−/− mouse regressive plaques, coincident with a reduction in lesional macrophage content and marker of M1 proinflammatory macrophages. Furthermore, we found MDL-1 was significantly expressed in inflammatory M1 subtype polarized bone marrow-derived macrophages. In vitro experiments, the level of MDL-1 was remarkably elevated in macrophages treated with pathophysiological drivers of plaque progression, such as oxidized low-density lipoprotein (ox-LDL) and hypoxia. Mechanistically, we demonstrated that MDL-1 overexpression notably promoted macrophage survival and decreased cleaved caspase-3 expression under ox-LDL stimulation, which suggested that it could maintain lesional macrophage survival and cause its accumulation. Conclusions This study firstly demonstrated that MDL-1 is mainly expressed in atherosclerotic lesional macrophages and increased macrophage MDL-1 expression is associated with early plaque progression and promotes macrophage survival. Electronic supplementary material The online version of this article (10.1186/s12967-017-1336-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Weixin Xiong
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China
| | - Haibo Wang
- Institute of Cardiovascular Disease, Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lin Lu
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China.,Institute of Cardiovascular Disease, Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui Xi
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China
| | - Fang Wang
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China
| | - Gang Gu
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China.
| | - Rong Tao
- Department of Cardiology, Ruijin Hospital, Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Huangpu District, Shanghai, 200025, China.
| |
Collapse
|
16
|
The Complement System Component C5a Produces Thermal Hyperalgesia via Macrophage-to-Nociceptor Signaling That Requires NGF and TRPV1. J Neurosci 2017; 36:5055-70. [PMID: 27147658 DOI: 10.1523/jneurosci.3249-15.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The complement cascade is a principal component of innate immunity. Recent studies have underscored the importance of C5a and other components of the complement system in inflammatory and neuropathic pain, although the underlying mechanisms are largely unknown. In particular, it is unclear how the complement system communicates with nociceptors and which ion channels and receptors are involved. Here we demonstrate that inflammatory thermal and mechanical hyperalgesia induced by complete Freund's adjuvant was accompanied by C5a upregulation and was markedly reduced by C5a receptor (C5aR1) knock-out or treatment with the C5aR1 antagonist PMX53. Direct administration of C5a into the mouse hindpaw produced strong thermal hyperalgesia, an effect that was absent in TRPV1 knock-out mice, and was blocked by the TRPV1 antagonist AMG9810. Immunohistochemistry of mouse plantar skin showed prominent expression of C5aR1 in macrophages. Additionally, C5a evoked strong Ca(2+) mobilization in macrophages. Macrophage depletion in transgenic macrophage Fas-induced apoptosis mice abolished C5a-dependent thermal hyperalgesia. Examination of inflammatory mediators following C5a injection revealed a rapid upregulation of NGF, a mediator known to sensitize TRPV1. Preinjection of an NGF-neutralizing antibody or Trk inhibitor GNF-5837 prevented C5a-induced thermal hyperalgesia. Notably, NGF-induced thermal hyperalgesia was unaffected by macrophage depletion. Collectively, these results suggest that complement fragment C5a induces thermal hyperalgesia by triggering macrophage-dependent signaling that involves mobilization of NGF and NGF-dependent sensitization of TRPV1. Our findings highlight the importance of macrophage-to-neuron signaling in pain processing and identify C5a, NGF, and TRPV1 as key players in this cross-cellular communication. SIGNIFICANCE STATEMENT This study provides mechanistic insight into how the complement system, a key component of innate immunity, regulates the development of pain hypersensitivity. We demonstrate a crucial role of the C5a receptor, C5aR1, in the development of inflammatory thermal and mechanical sensitization. By focusing on the mechanisms of C5a-induced thermal hyperalgesia, we show that this process requires recruitment of macrophages and initiation of macrophage-to-nociceptor signaling. At the molecular level, we demonstrate that this signaling depends on NGF and is mediated by the heat-sensitive nociceptive channel TRPV1. This deeper understanding of how immune cells and neurons interact to regulate pain processing is expected to facilitate mechanism-based approaches in the development of new analgesics.
Collapse
|
17
|
Macrophages and Phospholipases at the Intersection between Inflammation and the Pathogenesis of HIV-1 Infection. Int J Mol Sci 2017; 18:ijms18071390. [PMID: 28661459 PMCID: PMC5535883 DOI: 10.3390/ijms18071390] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/22/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022] Open
Abstract
Persistent low grade immune activation and chronic inflammation are nowadays considered main driving forces of the progressive immunologic failure in effective antiretroviral therapy treated HIV-1 infected individuals. Among the factors contributing to this phenomenon, microbial translocation has emerged as a key driver of persistent immune activation. Indeed, the rapid depletion of gastrointestinal CD4+ T lymphocytes occurring during the early phases of infection leads to a deterioration of the gut epithelium followed by the translocation of microbial products into the systemic circulation and the subsequent activation of innate immunity. In this context, monocytes/macrophages are increasingly recognized as an important source of inflammation, linked to HIV-1 disease progression and to non-AIDS complications, such as cardiovascular disease and neurocognitive decline, which are currently main challenges in treated patients. Lipid signaling plays a central role in modulating monocyte/macrophage activation, immune functions and inflammatory responses. Phospholipase-mediated phospholipid hydrolysis leads to the production of lipid mediators or second messengers that affect signal transduction, thus regulating a variety of physiologic and pathophysiologic processes. In this review, we discuss the contribution of phospholipases to monocyte/macrophage activation in the context of HIV-1 infection, focusing on their involvement in virus-associated chronic inflammation and co-morbidities.
Collapse
|
18
|
Yuan Q, Ren C, Xu W, Petri B, Zhang J, Zhang Y, Kubes P, Wu D, Tang W. PKN1 Directs Polarized RAB21 Vesicle Trafficking via RPH3A and Is Important for Neutrophil Adhesion and Ischemia-Reperfusion Injury. Cell Rep 2017; 19:2586-2597. [PMID: 28636945 PMCID: PMC5548392 DOI: 10.1016/j.celrep.2017.05.080] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/18/2017] [Accepted: 05/24/2017] [Indexed: 01/08/2023] Open
Abstract
Polarized vesicle transport plays an important role in cell polarization, but the mechanisms underlying this process and its role in innate immune responses are not well understood. Here, we describe a phosphorylation-regulated polarization mechanism that is important for neutrophil adhesion to endothelial cells during inflammatory responses. We show that the protein kinase PKN1 phosphorylates RPH3A, which enhances binding of RPH3A to guanosine triphosphate (GTP)-bound RAB21. These interactions are important for polarized localization of RAB21 and RPH3A in neutrophils, which leads to PIP5K1C90 polarization. Consistent with the roles of PIP5K1C90 polarization, the lack of PKN1 or RPH3A impairs neutrophil integrin activation, adhesion to endothelial cells, and infiltration in inflammatory models. Furthermore, myeloid-specific loss of PKN1 decreases tissue injury in a renal ischemia-reperfusion model. Thus, this study characterizes a mechanism for protein polarization in neutrophils and identifies a potential protein kinase target for therapeutic intervention in reperfusion-related tissue injury.
Collapse
Affiliation(s)
- Qianying Yuan
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Chunguang Ren
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Wenwen Xu
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Björn Petri
- Snyder Institute for Chronic Diseases Mouse Phenomics Resource Laboratory, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jiasheng Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yong Zhang
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Paul Kubes
- Snyder Institute for Chronic Diseases Mouse Phenomics Resource Laboratory, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Dianqing Wu
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Wenwen Tang
- Department of Pharmacology, Vascular Biology and Therapeutic Program, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
19
|
Affiliation(s)
- Trent D Evans
- From the Department of Medicine, Cardiovascular Division (T.D.E., I.S., X.Z., B.R.) and Department of Pathology & Immunology (B.R.), Washington University School of Medicine, St Louis, MO
| | - Ismail Sergin
- From the Department of Medicine, Cardiovascular Division (T.D.E., I.S., X.Z., B.R.) and Department of Pathology & Immunology (B.R.), Washington University School of Medicine, St Louis, MO
| | - Xiangyu Zhang
- From the Department of Medicine, Cardiovascular Division (T.D.E., I.S., X.Z., B.R.) and Department of Pathology & Immunology (B.R.), Washington University School of Medicine, St Louis, MO
| | - Babak Razani
- From the Department of Medicine, Cardiovascular Division (T.D.E., I.S., X.Z., B.R.) and Department of Pathology & Immunology (B.R.), Washington University School of Medicine, St Louis, MO.
| |
Collapse
|
20
|
Mao G, Jin J, Kunapuli SP, Rao AK. Nuclear factor-κB regulates expression of platelet phospholipase C-β2 (PLCB2). Thromb Haemost 2016; 116:931-940. [PMID: 27465150 PMCID: PMC6919569 DOI: 10.1160/th15-09-0749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 07/10/2016] [Indexed: 11/05/2022]
Abstract
Phospholipase C (PLC)-β2 (gene PLCB2) is a critical regulator of platelet responses upon activation. Mechanisms regulating of PLC-β2 expression in platelets/MKs are unknown. Our studies in a patient with platelet PLC-β2 deficiency revealed the PLCB2 coding sequence to be normal and decreased platelet PLC-β2 mRNA, suggesting a defect in transcriptional regulation. PLCB2 5'- upstream region of the patient revealed a heterozygous 13 bp deletion (-1645/-1633 bp) encompassing a consensus sequence for nuclear factor-κB (NF-κB). This was subsequently detected in three of 50 healthy subjects. To understand the mechanisms regulating PLC-β2, we studied the effect of this variation in the PLCB2. Gel-shift studies using nuclear extracts from human erythroleukaemia (HEL) cells or recombinant p65 showed NF-κB binding to oligonucleotide with NF-κB site; in luciferase reporter studies its deletion reduced PLCB2 promoter activity. PLCB2 expression was decreased by siRNA knockdown of NF-κB p65 subunit and increased by p65 overexpression. By immunoblotting platelet PLC-β2 in 17 healthy subjects correlated with p65 (r=0.76, p=0.0005). These studies provide the first evidence that NF-κB regulates MK/platelet PLC-β2 expression. This interaction is important because of the major role of PLC-β2 in platelet activation and of NF-κB in processes, including inflammation and atherosclerosis, where both are intimately involved.
Collapse
Affiliation(s)
| | | | | | - A Koneti Rao
- A. Koneti Rao, MD, Sol Sherry Thrombosis Research Center, Temple University School of Medicine, 3400 North Broad Street, MRB-204, Philadelphia, PA 19140, USA, Tel.: +1 215 707 4684, Fax: +1 215 707 2783, E-mail:
| |
Collapse
|
21
|
Zhong W, Pan G, Wang L, Li S, Ou J, Xu M, Li J, Zhu B, Cao X, Ma H, Li C, Xu J, Olkkonen VM, Staels B, Yan D. ORP4L Facilitates Macrophage Survival via G-Protein-Coupled Signaling: ORP4L-/- Mice Display a Reduction of Atherosclerosis. Circ Res 2016; 119:1296-1312. [PMID: 27729467 DOI: 10.1161/circresaha.116.309603] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/02/2016] [Accepted: 10/11/2016] [Indexed: 02/05/2023]
Abstract
RATIONALE Macrophage survival within the arterial wall is a central factor contributing to atherogenesis. Oxysterols, major components of oxidized low-density lipoprotein, exert cytotoxic effects on macrophages. OBJECTIVE To determine whether oxysterol-binding protein-related protein 4 L (ORP4L), an oxysterol-binding protein, affects macrophage survival and the pathogenesis of atherosclerosis. METHODS AND RESULTS By hiring cell biological approaches and ORP4L-/- mice, we show that ORP4L coexpresses with and forms a complex with Gαq/11 and phospholipase C (PLC)-β3 in macrophages. ORP4L facilitates G-protein-coupled ligand-induced PLCβ3 activation, IP3 production, and Ca2+ release from the endoplasmic reticulum. Through this mechanism, ORP4L sustains antiapoptotic Bcl-XL expression through Ca2+-mediated c-AMP responsive element binding protein transcriptional regulation and thus protects macrophages from apoptosis. Excessive stimulation with the oxysterol 25-hydroxycholesterol disassembles the ORP4L/Gαq/11/PLCβ3 complexes, resulting in reduced PLCβ3 activity, IP3 production, and Ca2+ release, as well as decreased Bcl-XL expression and increased apoptosis. Overexpression of ORP4L counteracts these oxysterol-induced defects. Mice lacking ORP4L exhibit increased apoptosis of macrophages in atherosclerotic lesions and a reduced lesion size. CONCLUSIONS ORP4L is crucial for macrophage survival. It counteracts the cytotoxicity of oxysterols/oxidized low-density lipoprotein to protect macrophage from apoptosis, thus playing an important role in the development of atherosclerosis.
Collapse
Affiliation(s)
- Wenbin Zhong
- From the Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China (W.Z., G.P., L.W., J.L., B.Z., X.C., H.M., C.L., D.Y.); Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland (S.L., V.M.O.); Division of Cardiac Surgery, the First Affiliated Hospital (J.O.) and Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (M.X., J.X.); and U1011 Inserm, EGID, Université Lille, CHU Lille, Institut Pasteur de Lille, France (B.S.)
| | - Guoping Pan
- From the Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China (W.Z., G.P., L.W., J.L., B.Z., X.C., H.M., C.L., D.Y.); Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland (S.L., V.M.O.); Division of Cardiac Surgery, the First Affiliated Hospital (J.O.) and Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (M.X., J.X.); and U1011 Inserm, EGID, Université Lille, CHU Lille, Institut Pasteur de Lille, France (B.S.)
| | - Lin Wang
- From the Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Department of Biotechnology, College of Life Science and Technology, Jinan University, Guangzhou, China (W.Z., G.P., L.W., J.L., B.Z., X.C., H.M., C.L., D.Y.); Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, Finland (S.L., V.M.O.); Division of Cardiac Surgery, the First Affiliated Hospital (J.O.) and Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China (M.X., J.X.); and U1011 Inserm, EGID, Université Lille, CHU Lille, Institut Pasteur de Lille, France (B.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rai S, Bhatnagar S. Hyperlipidemia, Disease Associations, and Top 10 Potential Drug Targets: A Network View. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:152-68. [DOI: 10.1089/omi.2015.0172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sneha Rai
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology, Dwarka, New Delhi, India
| |
Collapse
|
23
|
Cocco L, Follo MY, Manzoli L, Suh PG. Phosphoinositide-specific phospholipase C in health and disease. J Lipid Res 2015; 56:1853-60. [PMID: 25821234 DOI: 10.1194/jlr.r057984] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Indexed: 12/20/2022] Open
Abstract
Phospholipases are widely occurring and can be found in several different organisms, including bacteria, yeast, plants, animals, and viruses. Phospholipase C (PLC) is a class of phospholipases that cleaves phospholipids on the diacylglycerol (DAG) side of the phosphodiester bond producing DAGs and phosphomonoesters. Among PLCs, phosphoinositide-specific PLC (PI-PLC) constitutes an important step in the inositide signaling pathways. The structures of PI-PLC isozymes show conserved domains as well as regulatory specific domains. This is important, as most PI-PLCs share a common mechanism, but each of them has a peculiar role and can have a specific cell distribution that is linked to a specific function. More importantly, the regulation of PLC isozymes is fundamental in health and disease, as there are several PLC-dependent molecular mechanisms that are associated with the activation or inhibition of important physiopathological processes. Moreover, PI-PLC alternative splicing variants can play important roles in complex signaling networks, not only in cancer but also in other diseases. That is why PI-PLC isozymes are now considered as important molecules that are essential for better understanding the molecular mechanisms underlying both physiology and pathogenesis, and are also potential molecular targets useful for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Lucio Cocco
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Matilde Y Follo
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Lucia Manzoli
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Pann-Ghill Suh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 689-798, Korea
| |
Collapse
|
24
|
Borst O, Schaub M, Walker B, Schmid E, Münzer P, Voelkl J, Alesutan I, Rodríguez JM, Vogel S, Schoenberger T, Metzger K, Rath D, Umbach A, Kuhl D, Müller II, Seizer P, Geisler T, Gawaz M, Lang F. Pivotal Role of Serum- and Glucocorticoid-Inducible Kinase 1 in Vascular Inflammation and Atherogenesis. Arterioscler Thromb Vasc Biol 2015; 35:547-57. [DOI: 10.1161/atvbaha.114.304454] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective—
Atherosclerosis, an inflammatory disease of arterial vessel walls, requires migration and matrix metalloproteinase (MMP)-9–dependent invasion of monocytes/macrophages into the vascular wall. MMP-9 expression is stimulated by transcription factor nuclear factor-κB, which is regulated by inhibitor κB (IκB) and thus IκB kinase. Regulators of nuclear factor-κB include serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored involvement of SGK1 in vascular inflammation and atherogenesis.
Approach and Results—
Gene-targeted apolipoprotein E (ApoE)–deficient mice without (
apoe
−/−
sgk1
+/+
) or with (
apoe
−/−
sgk1
−/−
) additional SGK1 knockout received 16-week cholesterol-rich diet. According to immunohistochemistry atherosclerotic lesions in aorta and carotid artery, vascular CD45
+
leukocyte infiltration, Mac-3
+
macrophage infiltration, vascular smooth muscle cell content, MMP-2, and MMP-9 positive areas in atherosclerotic tissue were significantly less in
apoe
−/−
sgk1
−/−
mice than in
apoe
−/−
sgk1
+/+
mice. As determined by Boyden chamber, thioglycollate-induced peritonitis and air pouch model, migration of SGK1-deficient CD11b
+
F4/80
+
macrophages was significantly diminished in vitro and in vivo. Zymographic MMP-2 and MMP-9 production, MMP-9 activity and invasion through matrigel in vitro were significantly less in
sgk1
−/−
than in
sgk1
+/+
macrophages and in control plasmid–transfected or inactive
K127N
SGK1-transfected than in constitutively active
S422D
SGK1-transfected THP-1 cells. Confocal microscopy revealed reduced macrophage number and macrophage MMP-9 content in plaques of
apoe
−/−
sgk1
−/−
mice. In THP-1 cells, MMP-inhibitor GM6001 (25 μmol/L) abrogated
S422D
SGK1-induced MMP-9 production and invasion. According to reverse transcription polymerase chain reaction, MMP-9 transcript levels were significantly reduced in
sgk1
−/−
macrophages and strongly upregulated in
S422D
SGK1-transfected THP-1 cells compared with control plasmid–transfected or
K127N
SGK1-transfected THP-1 cells. According to immunoblotting and confocal microscopy, phosphorylation of IκB kinase and inhibitor κB and nuclear translocation of p50 were significantly lower in
sgk1
−/−
macrophages than in
sgk1
+/+
macrophages and significantly higher in
S422D
SGK1-transfected THP-1 cells than in control plasmid–transfected or
K127N
SGK1-transfected THP-1 cells. Treatment of
S422D
SGK1-transfected THP-1 cells with IκB kinase-inhibitor BMS-345541 (10 μmol/L) abolished
S422D
SGK1-induced increase of MMP-9 transcription and gelatinase activity.
Conclusions—
SGK1 plays a pivotal role in vascular inflammation during atherogenesis. SGK1 participates in the regulation of monocyte/macrophage migration and MMP-9 transcription via regulation of nuclear factor-κB.
Collapse
Affiliation(s)
- Oliver Borst
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Malte Schaub
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Britta Walker
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Evi Schmid
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Patrick Münzer
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Jakob Voelkl
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Ioana Alesutan
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - José M. Rodríguez
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Sebastian Vogel
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Tanja Schoenberger
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Katja Metzger
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Dominik Rath
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Anja Umbach
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Dietmar Kuhl
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Iris I. Müller
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Peter Seizer
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Tobias Geisler
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Meinrad Gawaz
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| | - Florian Lang
- From the Department of Cardiology and Cardiovascular Medicine (O.B., M.S., S.V., T.S., K.M., D.R., I.I.M., P.S., T.G., M.G.), Department of Physiology (O.B., B.W., E.S., P.M., J.V., I.A., A.U., F.L.), Department of Pediatric Surgery and Urology, University Children’s Hospital (E.S.), University of Tuebingen, Tuebingen, Germany; Laboratory of Biochemical Genetics and Metabolism, The Rockefeller University of New York (J.M.R.); and Center for Molecular Neurobiology (ZMNH), Institute for Molecular and
| |
Collapse
|
25
|
Gao K, Tang W, Li Y, Zhang P, Wang D, Yu L, Wang C, Wu D. Front-signal-dependent accumulation of the RHOA inhibitor FAM65B at leading edges polarizes neutrophils. J Cell Sci 2015; 128:992-1000. [PMID: 25588844 DOI: 10.1242/jcs.161497] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A hallmark of neutrophil polarization is the back localization of active RHOA and phosphorylated myosin light chain (pMLC, also known as MYL2). However, the mechanism for the polarization is not entirely clear. Here, we show that FAM65B, a newly identified RHOA inhibitor, is important for the polarization. When FAM65B is phosphorylated, it binds to 14-3-3 family proteins and becomes more stable. In neutrophils, chemoattractants stimulate FAM65B phosphorylation largely depending on the signals from the front of the cells that include those mediated by phospholipase Cβ (PLCβ) and phosphoinositide 3-kinase γ (PI3Kγ), leading to FAM65B accumulation at the leading edge. Concordantly, FAM65B deficiency in neutrophils resulted in an increase in RHOA activity and localization of pMLC to the front of cells, as well as defects in chemotaxis directionality and adhesion to endothelial cells under flow. These data together elucidate a mechanism for RHOA and pMLC polarization in stimulated neutrophils through direct inhibition of RHOA by FAM65B at the leading edge.
Collapse
Affiliation(s)
- Kun Gao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Institute of Genetics, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China Vascular Biology and Therapeutic Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06410, USA
| | - Wenwen Tang
- Vascular Biology and Therapeutic Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06410, USA
| | - Yuan Li
- Vascular Biology and Therapeutic Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06410, USA
| | - Pingzhao Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Institute of Genetics, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Dejie Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Institute of Genetics, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Institute of Genetics, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Chenji Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Institute of Genetics, School of Life Sciences, Fudan University, 220 Handan Road, Shanghai 200433, P.R. China
| | - Dianqing Wu
- Vascular Biology and Therapeutic Program, Department of Pharmacology, Yale University School of Medicine, 10 Amistad Street, New Haven, CT 06410, USA
| |
Collapse
|
26
|
Abstract
Rapid progress has recently been made regarding how phospholipase C (PLC)-β functions downstream of G protein-coupled receptors and how PLC-β functions in the nucleus. PLC-β has also been shown to interplay with tyrosine kinase-based signaling pathways, specifically to inhibit Stat5 activation by recruiting the protein-tyrosine phosphatase SHP-1. In this regard, a new multimolecular signaling platform, named SPS complex, has been identified. The SPS complex has important regulatory roles in tumorigenesis and immune cell activation. Furthermore, a growing body of work suggests that PLC-β also participates in the differentiation and activation of immune cells that control both the innate and adaptive immune systems.
Collapse
Affiliation(s)
- Wenbin Xiao
- Department of Pathology, University Hospital Case Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
27
|
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
|
28
|
Kawakami T, Xiao W. Phospholipase C-β in immune cells. Adv Biol Regul 2013; 53:249-57. [PMID: 23981313 DOI: 10.1016/j.jbior.2013.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 07/29/2013] [Accepted: 08/02/2013] [Indexed: 12/22/2022]
Abstract
Great progress has recently been made in structural and functional research of phospholipase C (PLC)-β. We now understand how PLC-β isoforms (β1-β4) are activated by GTP-bound Gαq downstream of G protein-coupled receptors. Numerous studies indicate that PLC-βs participate in the differentiation and activation of immune cells that control both the innate and adaptive immune systems. The PLC-β3 isoform also interplays with tyrosine kinase-based signaling pathways, to inhibit Stat5 activation by recruiting the protein-tyrosine phosphatase SHP-1, with which PLC-β3 and Stat5 form a multi-molecular signaling platform, named SPS complex. The SPS complex has important regulatory roles in tumorigenesis and immune cell activation.
Collapse
Affiliation(s)
- Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA; Laboratory of Allergic Disease, RIKEN Center for Integrative Medical Sciences (IMS-RCAI), Yokohama 230-0045, Japan.
| | | |
Collapse
|
29
|
Bohdanowicz M, Schlam D, Hermansson M, Rizzuti D, Fairn GD, Ueyama T, Somerharju P, Du G, Grinstein S. Phosphatidic acid is required for the constitutive ruffling and macropinocytosis of phagocytes. Mol Biol Cell 2013; 24:1700-12, S1-7. [PMID: 23576545 PMCID: PMC3667723 DOI: 10.1091/mbc.e12-11-0789] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Phagocytes spontaneously ruffle as they probe their environment and take up antigens. These cells are uniquely enriched in phosphatidic acid, which is necessary for ruffling and macropinocytosis. Macrophages and dendritic cells continuously survey their environment in search of foreign particles and soluble antigens. Such surveillance involves the ongoing extension of actin-rich protrusions and the consequent formation of phagosomes and macropinosomes. The signals inducing this constitutive cytoskeletal remodeling have not been defined. We report that, unlike nonphagocytic cells, macrophages and immature dendritic cells have elevated levels of phosphatidic acid (PA) in their plasma membrane. The plasmalemmal PA is synthesized by phosphorylation of diacylglycerol, which is in turn generated by a G protein–stimulated phospholipase C. Inhibition of diacylglycerol kinase activity results in the detachment of T-cell lymphoma invasion and metastasis–inducing protein 1 (TIAM1)—a Rac guanine exchange factor—from the plasma membrane, thereby depressing Rac activity and abolishing the constitutive ruffling and macropinocytosis that characterize macrophages and immature dendritic cells. Accumulation of PA and binding of TIAM1 to the membrane require the activity of phosphatidylinositol-4,5-bisphosphate 3-kinase. Thus a distinctive, constitutive pathway of PA biosynthesis promotes the actin remodeling required for immune surveillance.
Collapse
Affiliation(s)
- Michal Bohdanowicz
- Division of Cell Biology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Huang W, Barrett M, Hajicek N, Hicks S, Harden TK, Sondek J, Zhang Q. Small molecule inhibitors of phospholipase C from a novel high-throughput screen. J Biol Chem 2013; 288:5840-8. [PMID: 23297405 DOI: 10.1074/jbc.m112.422501] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Phospholipase C (PLC) isozymes are important signaling molecules, but few small molecule modulators are available to pharmacologically regulate their function. With the goal of developing a general approach for identification of novel PLC inhibitors, we developed a high-throughput assay based on the fluorogenic substrate reporter WH-15. The assay is highly sensitive and reproducible: screening a chemical library of 6280 compounds identified three novel PLC inhibitors that exhibited potent activities in two separate assay formats with purified PLC isozymes in vitro. Two of the three inhibitors also inhibited G protein-coupled receptor-stimulated PLC activity in intact cell systems. These results demonstrate the power of the high-throughput assay for screening large collections of small molecules to identify novel PLC modulators. Potent and selective modulators of PLCs will ultimately be useful for dissecting the roles of PLCs in cellular processes, as well as provide lead compounds for the development of drugs to treat diseases arising from aberrant phospholipase activity.
Collapse
Affiliation(s)
- Weigang Huang
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Ritsma L, Steller EJA, Beerling E, Loomans CJM, Zomer A, Gerlach C, Vrisekoop N, Seinstra D, van Gurp L, Schafer R, Raats DA, de Graaff A, Schumacher TN, de Koning EJP, Rinkes IHB, Kranenburg O, Rheenen JV. Intravital Microscopy Through an Abdominal Imaging Window Reveals a Pre-Micrometastasis Stage During Liver Metastasis. Sci Transl Med 2012; 4:158ra145. [DOI: 10.1126/scitranslmed.3004394] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
32
|
Fischer A, Schmid B, Ellinghaus D, Nothnagel M, Gaede KI, Schürmann M, Lipinski S, Rosenstiel P, Zissel G, Höhne K, Petrek M, Kolek V, Pabst S, Grohé C, Grunewald J, Ronninger M, Eklund A, Padyukov L, Gieger C, Wichmann HE, Nebel A, Franke A, Müller-Quernheim J, Hofmann S, Schreiber S. A novel sarcoidosis risk locus for Europeans on chromosome 11q13.1. Am J Respir Crit Care Med 2012; 186:877-85. [PMID: 22837380 DOI: 10.1164/rccm.201204-0708oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
RATIONALE Sarcoidosis is a complex inflammatory disease with a heterogeneous clinical picture. Among others, an acute and chronic clinical course can be distinguished, for which specific genetic risk factors are known. OBJECTIVES To identify additional risk loci for sarcoidosis and its acute and chronic subforms, we analyzed imputed data from a genome-wide association scan for these phenotypes. METHODS After quality control, the genome-wide association scan comprised nearly 1.3 million imputed single-nucleotide polymorphisms based on an Affymetrix 6.0 Gene Chip dataset of 564 German sarcoidosis cases, including 176 acute and 354 chronic cases and 1,575 control subjects. MEASUREMENTS AND MAIN RESULTS We identified chromosome 11q13.1 (rs479777) as a novel locus influencing susceptibility to sarcoidosis with genome-wide significance. The marker was significantly associated in three distinct German case-control populations and in an additional German family sample with odds ratios ranging from 0.67 to 0.77. This finding was further replicated in two independent European case-control populations from the Czech Republic (odds ratio, 0.75) and from Sweden (odds ratio, 0.79). In a meta-analysis of the included European case-control samples the marker yielded a P value of 2.68 × 10(-18). The locus was previously reported to be associated with Crohn disease, psoriasis, alopecia areata, and leprosy. For sarcoidosis, fine-mapping and expression analysis suggest KCNK4, PRDX5, PCLB3, and most promising CCDC88B as candidates for the underlying risk gene in the associated region. CONCLUSIONS This study provides striking evidence for association of chromosome 11q13.1 with sarcoidosis in Europeans, and thus identified a further genetic risk locus shared by sarcoidosis, Crohn disease and psoriasis.
Collapse
Affiliation(s)
- Annegret Fischer
- Christian-Albrechts University, Institute of Clinical Molecular Biology, Schittenhelmstrasse 12, Kiel, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Diverse roles of macrophages in atherosclerosis: from inflammatory biology to biomarker discovery. Mediators Inflamm 2012; 2012:693083. [PMID: 22577254 PMCID: PMC3337637 DOI: 10.1155/2012/693083] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Accepted: 01/11/2012] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease, a leading cause of mortality in developed countries, is mainly caused by atherosclerosis, a chronic inflammatory disease. Macrophages, which differentiate from monocytes that are recruited from the blood, account for the majority of leukocytes in atherosclerotic plaques. Apoptosis and the suppressed clearance of apoptotic macrophages (efferocytosis) are associated with vulnerable plaques that are prone to rupture, leading to thrombosis. Based on the central functions of macrophages in atherogenesis, cytokines, chemokines, enzymes, or microRNAs related to or produced by macrophages have become important clinical prognostic or diagnostic biomarkers. This paper discusses the impact of monocyte-derived macrophages in early atherogenesis and advanced disease. The role and possible future development of macrophage inflammatory biomarkers are also described.
Collapse
|
34
|
Bennett M, Yu H, Clarke M. Signalling from dead cells drives inflammation and vessel remodelling. Vascul Pharmacol 2012; 56:187-92. [PMID: 22306421 DOI: 10.1016/j.vph.2012.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/17/2012] [Accepted: 01/21/2012] [Indexed: 11/16/2022]
Abstract
Death of vascular smooth muscle cells (VSMCs) has been demonstrated in vessel development and in disease, most notably in atherosclerosis, but also after injury and remodelling. VSMC death promotes multiple features of vulnerable plaques, but also induces features of normal vessel ageing and cystic medial necrosis, including loss of VSMCs, elastin fragmentation and loss, increased glycosaminoglycans and speckled calcification. VSMC apoptosis in the absence of efficient phagocytosis also produces inflammation due to secondary necrosis; in contrast, VSMC apoptosis in normal vessels can be silent. We have investigated the consequences of VSMC apoptosis in both disease and during vessel remodelling. We find that VSMCs release specific cytokines dependent upon the mode of cell death; IL-1β predominates during apoptosis, whilst IL-1α predominates during necrosis. Both IL-1α and β promote release of further cytokines from adjacent live cells, in particular IL-6 and MCP-1. The balance of cytokines results in pathology with differing compositions, including inflammation or neointima formation/vascular repair, via direct promotion of VSMC proliferation and migration. Thus, VSMC death can promote either pathology or repair, depending upon the context and cytokine signalling.
Collapse
Affiliation(s)
- Martin Bennett
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, United Kingdom.
| | | | | |
Collapse
|
35
|
Zheng Y, Gardner SE, Clarke MCH. Cell death, damage-associated molecular patterns, and sterile inflammation in cardiovascular disease. Arterioscler Thromb Vasc Biol 2012; 31:2781-6. [PMID: 22096097 DOI: 10.1161/atvbaha.111.224907] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cell death and inflammation are ancient processes of fundamental biological importance in both normal physiology and pathology. This is evidenced by the profound conservation of mediators, with ancestral homologues identified from plants to humans, and the number of diseases driven by aberrant control of either process. Apoptosis is the most well-studied cell death, but many forms exist, including autophagy, necrosis, pyroptosis, paraptosis, and the obscure dark cell death. Cell death occurs throughout the cardiovascular system, from initial shaping of the heart and vasculature during development to involvement in pathologies, including atherosclerosis, aneurysm, cardiomyopathy, restenosis, and vascular graft rejection. However, determining whether cell death primarily drives pathology or is a secondary bystander effect is difficult. Inflammation, the primary response of innate immunity, is considered essential in initiating and driving vascular diseases. Cell death and inflammation are inextricably linked with their effectors modulating the other process. Indeed, an evolutionary link between cell death and inflammation occurs at caspase-1 (which activates interleukin-1β), which can induce death by pyroptosis, and is a member of the caspase family vital for apoptosis. This review examines cell death in vascular disease, how it can induce inflammation, and finally the emergence of inflammasomes in vascular pathology.
Collapse
Affiliation(s)
- Yue Zheng
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrooke's Hospital, Cambridge CB2 2QQ, United Kingdom
| | | | | |
Collapse
|
36
|
Novel inflammatory mechanisms of accelerated atherosclerosis in kidney disease. Kidney Int 2011; 80:453-63. [DOI: 10.1038/ki.2011.178] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
37
|
Donadeu FX, Esteves CL, Doyle LK, Walker CA, Schauer SN, Diaz CA. Phospholipase Cβ3 mediates LH-induced granulosa cell differentiation. Endocrinology 2011; 152:2857-69. [PMID: 21586561 DOI: 10.1210/en.2010-1298] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies showed that under certain conditions LH can stimulate not only adenylate cyclase (AC) but also phospholipase Cβ (PLCβ) signaling in target cells; however, the physiological involvement of PLCβ in LH-induced ovarian follicular cell differentiation has not been determined. To address this, ex vivo expression analyses and specific PLCβ targeting were performed in primary bovine granulosa cells. Expression analyses in cells from small (2.0-5.9 mm), medium (6.0-9.9 mm), and ovulatory-size (10.0-13.9 mm) follicles revealed an increase in mRNA and protein levels of heterotrimeric G protein subunits-αs, -αq, -α11, and -αi2 in ovulatory-size follicles, simultaneous with a substantial increase in LH receptor expression. Among the four known PLCβ isoforms, PLCβ3 (PLCB3) was specifically up-regulated in cells from ovulatory-size follicles, in association with a predominantly cytoplasmic location of PLCB3 in these cells and a significant inositol phosphate response to LH stimulation. Furthermore, RNA interference-mediated PLCB3 down-regulation reduced the ability of LH to induce hallmark differentiation responses of granulosa cells, namely transcriptional up-regulation of prostaglandin-endoperoxide synthase 2 and down-regulation of both aromatase expression and estradiol production. Responses to the AC agonist, forskolin, however, were not affected. In addition, PLCB3 down-regulation did not alter cAMP responses to LH in granulosa cells, ruling out a primary involvement of AC in mediating the effects of PLCB3. In summary, we provide evidence of a physiological involvement of PLCβ signaling in ovulatory-size follicles and specifically identify PLCB3 as a mediator of LH-induced differentiation responses of granulosa cells.
Collapse
Affiliation(s)
- Francesc X Donadeu
- Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, Scotland, United Kingdom.
| | | | | | | | | | | |
Collapse
|
38
|
Song S, Chew C, Dale BM, Traum D, Peacock J, Yamazaki T, Clynes R, Kurosaki T, Greenberg S. A requirement for the p85 PI3K adapter protein BCAP in the protection of macrophages from apoptosis induced by endoplasmic reticulum stress. THE JOURNAL OF IMMUNOLOGY 2011; 187:619-25. [PMID: 21685326 DOI: 10.4049/jimmunol.0903425] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Macrophages are innate immune cells that play key roles in regulation of the immune response and in tissue injury and repair. In response to specific innate immune stimuli, macrophages may exhibit signs of endoplasmic reticulum (ER) stress and progress to apoptosis. Factors that regulate macrophage survival under these conditions are poorly understood. In this study, we identified B cell adapter protein (BCAP), a p85 PI3K-binding adapter protein, in promoting survival in response to the combined challenge of LPS and ER stress. BCAP was unique among nine PI3K adapter proteins in being induced >10-fold in response to LPS. LPS-stimulated macrophages incubated with thapsigargin, a sarcoplasmic/endoplasmic reticulum calcium ATPase inhibitor that induces ER stress, underwent caspase-3 activation and apoptosis. Macrophages from BCAP(-/-) mice exhibited increased apoptosis in response to these stimuli. BCAP-deficient macrophages demonstrated decreased activation of Akt, but not ERK, and, unlike BCAP-deficient B cells, expressed normal amounts of the NF-κB subunits, c-Rel and RelA. Retroviral transduction of BCAP-deficient macrophages with wild-type BCAP, but not a Y4F BCAP mutant defective in binding the SH2 domain of p85 PI3K, reversed the proapoptotic phenotype observed in BCAP-deficient macrophages. We conclude that BCAP is a nonredundant PI3K adapter protein in macrophages that is required for maximal cell survival in response to ER stress. We suggest that as macrophages engage their pathogenic targets, innate immune receptors trigger increased expression of BCAP, which endows them with the capacity to withstand further challenges from ongoing cellular insults, such as ER stress.
Collapse
Affiliation(s)
- SungWon Song
- Program in Biotechnology, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Tano JY, Smedlund K, Lee R, Abramowitz J, Birnbaumer L, Vazquez G. Impairment of survival signaling and efferocytosis in TRPC3-deficient macrophages. Biochem Biophys Res Commun 2011; 410:643-7. [PMID: 21684255 DOI: 10.1016/j.bbrc.2011.06.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 06/06/2011] [Indexed: 10/18/2022]
Abstract
We have recently shown that in macrophages proper operation of the survival pathways phosphatidylinositol-3-kinase (PI3K)/AKT and nuclear factor kappa B (NFkB) has an obligatory requirement for constitutive, non-regulated Ca(2+) influx. In the present work we examined if Transient Receptor Potential Canonical 3 (TRPC3), a member of the TRPC family of Ca(2+)-permeable cation channels, contributes to the constitutive Ca(2+) influx that supports macrophage survival. We used bone marrow-derived macrophages obtained from TRPC3(-/-) mice to determine the activation status of survival signaling pathways, apoptosis and their efferocytic properties. Treatment of TRPC3(+/+) macrophages with the pro-apoptotic cytokine TNFα induced time-dependent phosphorylation of IκBα, AKT and BAD, and this was drastically reduced in TRPC3(-/-) macrophages. Compared to TRPC3(+/+) cells TRPC3(-/-) macrophages exhibited reduced constitutive cation influx, increased apoptosis and impaired efferocytosis. The present findings suggest that macrophage TRPC3, presumably through its constitutive function, contributes to survival signaling and efferocytic properties.
Collapse
Affiliation(s)
- Jean-Yves Tano
- Department of Physiology and Pharmacology and the Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Health Science Campus, 3000 Arlington Av, Toledo, OH 43614, USA
| | | | | | | | | | | |
Collapse
|
40
|
Bezzerri V, d'Adamo P, Rimessi A, Lanzara C, Crovella S, Nicolis E, Tamanini A, Athanasakis E, Tebon M, Bisoffi G, Drumm ML, Knowles MR, Pinton P, Gasparini P, Berton G, Cabrini G. Phospholipase C-β3 is a key modulator of IL-8 expression in cystic fibrosis bronchial epithelial cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:4946-58. [PMID: 21411730 DOI: 10.4049/jimmunol.1003535] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Respiratory insufficiency is the major cause of morbidity and mortality in patients affected by cystic fibrosis (CF). An excessive neutrophilic inflammation, mainly orchestrated by the release of IL-8 from bronchial epithelial cells and amplified by chronic bacterial infection with Pseudomonas aeruginosa, leads to progressive tissue destruction. The anti-inflammatory drugs presently used in CF patients have several limitations, indicating the need for identifying novel molecular targets. To address this issue, we preliminarily studied the association of 721 single nucleotide polymorphisms from 135 genes potentially involved in signal transduction implicated in neutrophil recruitment in a cohort of F508del homozygous CF patients with either severe or mild progression of lung disease. The top ranking association was found for a nonsynonymous polymorphism of the phospholipase C-β3 (PLCB3) gene. Studies in bronchial epithelial cells exposed to P. aeruginosa revealed that PLCB3 is implicated in extracellular nucleotide-dependent intracellular calcium signaling, leading to activation of the protein kinase Cα and Cβ and of the nuclear transcription factor NF-κB p65. The proinflammatory pathway regulated by PLCB3 acts by potentiating the Toll-like Receptors' signaling cascade and represents an interesting molecular target to attenuate the excessive recruitment of neutrophils without completely abolishing the inflammatory response.
Collapse
Affiliation(s)
- Valentino Bezzerri
- Laboratory of Molecular Pathology, University Hospital of Verona, 37126 Verona, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Babaev VR, Whitesell RR, Li L, Linton MF, Fazio S, May JM. Selective macrophage ascorbate deficiency suppresses early atherosclerosis. Free Radic Biol Med 2011; 50:27-36. [PMID: 20974251 PMCID: PMC3014415 DOI: 10.1016/j.freeradbiomed.2010.10.702] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/22/2010] [Accepted: 10/17/2010] [Indexed: 11/17/2022]
Abstract
To test whether severe ascorbic acid deficiency in macrophages affects progression of early atherosclerosis, we used fetal liver cell transplantation to generate atherosclerosis-prone apolipoprotein E-deficient (apoE(-/-)) mice that selectively lacked the ascorbate transporter (SVCT2) in hematopoietic cells, including macrophages. After 13 weeks of chow diet, apoE(-/-) mice lacking the SVCT2 in macrophages had surprisingly less aortic atherosclerosis, decreased lesion macrophage numbers, and increased macrophage apoptosis compared to control-transplanted mice. Serum lipid levels were similar in both groups. Peritoneal macrophages lacking the SVCT2 had undetectable ascorbate; increased susceptibility to H(2)O(2)-induced mitochondrial dysfunction and apoptosis; decreased expression of genes for COX-2, IL1β, and IL6; and decreased lipopolysaccharide-stimulated NF-κB and antiapoptotic gene expression. These changes were associated with decreased expression of both the receptor for advanced glycation end products and HIF-1α, either or both of which could have been the proximal cause of decreased macrophage activation and apoptosis in ascorbate-deficient macrophages.
Collapse
Affiliation(s)
- Vladimir R Babaev
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
42
|
Lammers B, Chandak PG, Aflaki E, Van Puijvelde GHM, Radovic B, Hildebrand RB, Meurs I, Out R, Kuiper J, Van Berkel TJC, Kolb D, Haemmerle G, Zechner R, Levak-Frank S, Van Eck M, Kratky D. Macrophage adipose triglyceride lipase deficiency attenuates atherosclerotic lesion development in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol 2010; 31:67-73. [PMID: 21030715 DOI: 10.1161/atvbaha.110.215814] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The consequences of macrophage triglyceride (TG) accumulation on atherosclerosis have not been studied in detail so far. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme for the initial step in TG hydrolysis. Because ATGL knockout (KO) mice exhibit massive TG accumulation in macrophages, we used ATGL KO mice to study the effects of macrophage TG accumulation on atherogenesis. METHODS AND RESULTS Low-density lipoprotein receptor (LDLr) KO mice were transplanted with bone marrow from ATGL KO (ATGL KO→LDLr KO) or wild-type (WT→LDLr KO) mice and challenged with a Western-type diet for 9 weeks. Despite TG accumulation in ATGL KO macrophages, atherosclerosis in ATGL KO→LDLr KO mice was 43% reduced associated with decreased plasma monocyte chemoattractant protein-1 (MCP-1) and macrophage interleukin-6 concentrations. This coincided with a reduced amount of macrophages, possibly because of a 39% increase in intraplaque apoptosis and a decreased migratory capacity of ATGL KO macrophages. The reduced number of white blood cells might be due to a 36% decreased Lin(-)Sca-1(+)cKit(+) hematopoietic stem cell population. CONCLUSIONS We conclude that the attenuation of atherogenesis in ATGL KO→LDLr KO mice is due to decreased infiltration of less inflammatory macrophages into the arterial wall and increased macrophage apoptosis.
Collapse
Affiliation(s)
- Bart Lammers
- Division of Biopharmaceutics, Gorlaeus Laboratories, Einsteinweg 55, 2333CC Leiden, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Xu W, Wang P, Petri B, Zhang Y, Tang W, Sun L, Kress H, Mann T, Shi Y, Kubes P, Wu D. Integrin-induced PIP5K1C kinase polarization regulates neutrophil polarization, directionality, and in vivo infiltration. Immunity 2010; 33:340-50. [PMID: 20850356 DOI: 10.1016/j.immuni.2010.08.015] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 07/19/2010] [Accepted: 08/11/2010] [Indexed: 11/15/2022]
Abstract
Neutrophils are important in innate immunity and acute inflammatory responses. However, the regulation of their recruitment to sites of inflammation has not been well characterized. Here, we investigated the kinase PIP5K1C and showed that PIP5K1C deficiency impaired neutrophil recruitment because of an adhesion defect. PIP5K1C regulated the adhesion through facilitating RhoA GTPase and integrin activation by chemoattractants. Integrins could induce polarization of an isoform of PIP5K1C, PIP5K1C-90, in neutrophils through intracellular vesicle transport independently of exogenous chemoattractant. PIP5K1C-90 polarization was required for polarized RhoA activation at uropods and provided an initial directional cue for neutrophil polarization on the endothelium. Importantly, the polarization was also required for circumventing the inhibition of lamellipodium formation by RhoA so that neutrophils could form leading edges required for transendothelial migration. Because integrins are not known to regulate neutrophil polarization, our study revealed a previously underappreciated role of integrin signaling in neutrophil regulation.
Collapse
Affiliation(s)
- Wenwen Xu
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Fukami K, Inanobe S, Kanemaru K, Nakamura Y. Phospholipase C is a key enzyme regulating intracellular calcium and modulating the phosphoinositide balance. Prog Lipid Res 2010; 49:429-37. [PMID: 20553968 DOI: 10.1016/j.plipres.2010.06.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Spatial and temporal activation of phosphoinositide turnover enables eukaryotic cells to perform various functions such as cell proliferation/differentiation, fertilization, neuronal functions, and cell motility. In this system, phospholipase C (PLC) is a key enzyme, which hydrolyzes phosphatidylinositol 4,5-bisphosphate (PI(4,5)P(2)) into two second messengers, inositol 1,4,5-trisphosphate (Ins(1,4,5)P(3)) and diacylglycerol (DAG). Ins(1,4,5)P(3) triggers the release of calcium from intracellular stores, and DAG mediates the activation of protein kinase C (PKC). In parallel, PI(4,5)P(2) also directly regulates a variety of cellular functions, including cytoskeletal remodeling, cytokinesis, phagocytosis, membrane dynamics, and channel activity, in addition to its role as a substrate for PLC and phosphatidylinositol 3-kinase (PI3K), which generates PI(3,4,5)P(3). An imbalance of these phosphoinositides contributes to the pathogeneses of various human diseases. Therefore, strict regulation of the levels of PI(4,5)P(2) and PI(3,4,5)P(3) by PLC or other interconverting enzymes is necessary for cellular functions. In this review, we focus on the roles of PLC as a calcium-regulating enzyme and as a modulator of the phosphoinositide balance.
Collapse
Affiliation(s)
- Kiyoko Fukami
- Laboratory of the Genome and Biosignals, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo, Japan.
| | | | | | | |
Collapse
|
45
|
Rebres RA, Moon C, Decamp D, Lin KM, Fraser ID, Milne SB, Roach TIA, Brown HA, Seaman WE. Clostridium difficile toxin B differentially affects GPCR-stimulated Ca2+ responses in macrophages: independent roles for Rho and PLA2. J Leukoc Biol 2010; 87:1041-57. [PMID: 20200401 DOI: 10.1189/jlb.1108708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Clostridium difficile toxins cause acute colitis by disrupting the enterocyte barrier and promoting inflammation. ToxB from C. difficile inactivates Rho family GTPases and causes release of cytokines and eicosanoids by macrophages. We studied the effects of ToxB on GPCR signaling in murine RAW264.7 macrophages and found that ToxB elevated Ca(2+) responses to Galphai-linked receptors, including the C5aR, but reduced responses to Galphaq-linked receptors, including the UDP receptors. Other Rho inhibitors also reduced UDP Ca(2+) responses, but they did not affect C5a responses, suggesting that ToxB inhibited UDP responses by inhibiting Rho but enhanced C5a responses by other mechanisms. By using PLCbeta isoform-deficient BMDM, we found that ToxB inhibited Ca(2+) signaling through PLCbeta4 but enhanced signaling through PLCbeta3. Effects of ToxB on GPCR Ca(2+) responses correlated with GPCR use of PLCbeta3 versus PLCbeta4. ToxB inhibited UDP Ca(2+) signaling without reducing InsP3 production or the sensitivity of cellular Ca(2+) stores to exogenous InsP3, suggesting that ToxB impairs UDP signaling at the level of InsP3/Ca(2+)coupling. In contrast, ToxB elevated InsP3 production by C5a, and the enhancement of Ca(2+) signaling by C5a was prevented by inhibition of PLA(2) or 5-LOX but not COX, implicating LTs but not prostanoids in the mechanism. In sum, ToxB has opposing, independently regulated effects on Ca(2+) signaling by different GPCR-linked PLCbeta isoforms in macrophages.
Collapse
Affiliation(s)
- Robert A Rebres
- Alliance for Cellular Signaling at Northern California Institute for Research and Education, VA Medical Center, San Francisco, California, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang H, Zhang W, Tang R, Zhu C, Bucher C, Blazar BR, Geng JG, Zhang C, Linden J, Wu C, Huo Y. Adenosine receptor A2A deficiency in leukocytes increases arterial neointima formation in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2010; 30:915-22. [PMID: 20167656 DOI: 10.1161/atvbaha.109.202572] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To use the mice deficient in both adenosine receptor A(2A)(A(2A)R(-/-)) and apolipoprotein E (apoE(-/-)) to investigate the role of A(2A)R in mediating the interactions of leukocytes with injured arterial walls and the formation of arterial neointima induced by a guide wire. METHODS AND RESULTS In apoE(-/-) mice, A(2A)R deficiency increased the size of the arterial neointima in injured carotid arteries by 83%. Arterial neointima formation was also enhanced in chimeric mice that underwent bone marrow transplantation (these mice lacked A(2A)R in their bone marrow-derived cells). Epifluorescence intravital microscopy showed that neutrophil rolling and adherence to the injured arterial area were enhanced by 80% and 110% in A(2A)R(-/-)/apoE(-/-) mice, respectively. This phenomenon occurred even though the protein levels of homing molecules on A(2A)R-deficient neutrophils were unchanged from those of wild-type neutrophils. A(2A)R-deficient neutrophils exhibited an increase in the phosphorylation of p38 mitogen-activated protein kinase, P-selectin glycoprotein ligand-1 (PSGL-1) clustering, and the affinity of b(2) integrins. The inhibition of p38 phosphorylation abrogated the increased PSGL-1 clustering and beta(2) integrin affinity, thus reversing the increased homing ability of A(2A)R-deficient leukocytes. CONCLUSION A(2A)R plays a complex role in inflammation and tissue injury. The deficiency of A(2A)R enhances the homing ability of leukocytes and increases the formation of the arterial neointima after injury. A(2A)R antagonists are being tested for the treatment of neurodegenerative and other chronic diseases. An evaluation of the effect of A(2A)R antagonists on arterial restenosis after arterial angioplasty should be conducted.
Collapse
Affiliation(s)
- Huan Wang
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Clarke MCH, Talib S, Figg NL, Bennett MR. Vascular smooth muscle cell apoptosis induces interleukin-1-directed inflammation: effects of hyperlipidemia-mediated inhibition of phagocytosis. Circ Res 2010; 106:363-72. [PMID: 19926874 DOI: 10.1161/circresaha.109.208389] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Atherosclerosis is characterized by lipid accumulation in the vessel wall, inflammation, and both macrophage and vascular smooth muscle cell (VSMC) apoptosis. However, whereas VSMC apoptosis in mice with established atherosclerotic plaques or hyperlipidemia increases serum levels of the proatherogenic cytokines monocyte chemotactic protein (MCP)-1, tumor necrosis factor alpha, and interleukin (IL)-6, the link between hyperlipidemia, apoptosis and inflammation, and the mechanisms by which apoptotic cells promote inflammation in atherosclerosis are unknown. OBJECTIVE To determine whether hyperlipidemia affects apoptotic cell clearance, and identify the molecular pathways downstream of VSMC apoptosis that may promote inflammation. METHODS AND RESULTS We find that human VSMCs are potent and efficient phagocytes of apoptotic human VSMCs, but phagocytosis is significantly reduced by oxidized low-density lipoprotein in vitro or hyperlipidemia in vivo. Necrotic human aortic VSMCs release IL-1alpha, which induces IL-6 and MCP-1 production from viable human VSMCs in vitro. In contrast, secondary necrotic VSMCs release both IL-1alpha and caspase-activated IL-1beta, augmenting IL-6 and MCP-1 production. Conditionally inducing VSMC apoptosis in situ in hyperlipidemic SM22alpha-hDTR/ApoE(-/-) mice to levels seen in human plaques increases serum MCP-1, tumor necrosis factor alpha, and IL-6, which is prevented by blocking IL-1. CONCLUSIONS We conclude that VSMC necrosis releases IL-1alpha, whereas secondary necrosis of apoptotic VSMCs releases both IL-1alpha and beta. IL-1 from necrotic VSMCs induces the surrounding viable VSMCs to produce proinflammatory cytokines. Thus, failed clearance of apoptotic VSMCs caused by hyperlipidemia in vivo may promote the increased serum cytokines and chronic inflammation associated with atherosclerosis.
Collapse
MESH Headings
- Animals
- Apolipoproteins E/genetics
- Apolipoproteins E/metabolism
- Apoptosis/physiology
- Blotting, Western
- Cells, Cultured
- Chemokine CCL2/blood
- Chemokine CCL2/metabolism
- Heparin-binding EGF-like Growth Factor
- Humans
- Hyperlipidemias/physiopathology
- Immunohistochemistry
- Inflammation/metabolism
- Inflammation/physiopathology
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Interleukin-1alpha/metabolism
- Interleukin-6/blood
- Interleukin-6/metabolism
- Mice
- Mice, Knockout
- Mice, Transgenic
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/parasitology
- Phagocytosis/physiology
Collapse
Affiliation(s)
- Murray C H Clarke
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK.
| | | | | | | |
Collapse
|
48
|
Grinberg S, Hasko G, Wu D, Leibovich SJ. Suppression of PLCbeta2 by endotoxin plays a role in the adenosine A(2A) receptor-mediated switch of macrophages from an inflammatory to an angiogenic phenotype. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:2439-53. [PMID: 19850892 DOI: 10.2353/ajpath.2009.090290] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Toll-like receptor (TLR) 2, 4, 7, and 9 agonists, together with adenosine A(2A) receptor (A(2A)R) agonists, switch macrophages from an inflammatory (M1) to an angiogenic (M2-like) phenotype. This switch involves induction of A(2A)Rs by TLR agonists, down-regulation of tumor necrosis factor alpha (TNFalpha) and interleukin-12, and up-regulation of vascular endothelial growth factor (VEGF) and interleukin-10 expression. We show here that the TLR4 agonist lipopolysaccharide (LPS) induces rapid and specific post-transcriptional down-regulation of phospholipase C(PLC)beta1 and beta2 expression in macrophages by de-stabilizing their mRNAs. The PLCbeta inhibitor U73122 down-regulates TNFalpha expression by macrophages, and in the presence of A(2A)R agonists, up-regulates VEGF, mimicking the synergistic action of LPS with A(2A)R agonists. Selective down-regulation of PLCbeta2, but not PLCbeta1, using small-interfering RNA resulted in increased VEGF expression in response to A(2A)R agonists, but did not suppress TNFalpha expression. Macrophages from PLCbeta2(-/-) mice also expressed increased VEGF in response to A(2A)R agonists. LPS-mediated suppression of PLCbeta1 and beta2 is MyD88-dependent. In a model of endotoxic shock, LPS (35 microg/mouse, i.p.) suppressed PLCbeta1 and beta2 expression in spleen, liver, and lung of wild-type but not MyD88(-/-) mice. These studies indicate that LPS suppresses PLCbeta1 and beta2 expression in macrophages in vitro and in several tissues in vivo. These results suggest that suppression of PLCbeta2 plays an important role in switching M1 macrophages into an M2-like state.
Collapse
Affiliation(s)
- Stan Grinberg
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, New Jersey 07103, USA
| | | | | | | |
Collapse
|
49
|
Litosch I, Pujari R, Lee SJ. Phosphatidic acid regulates signal output by G protein coupled receptors through direct interaction with phospholipase C-β1. Cell Signal 2009; 21:1379-84. [DOI: 10.1016/j.cellsig.2009.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Accepted: 04/27/2009] [Indexed: 12/25/2022]
|
50
|
Abstract
Atherosclerosis is an inflammatory disease of the wall of large- and medium-sized arteries that is precipitated by elevated levels of low-density lipoprotein (LDL) cholesterol in the blood. Although dendritic cells (DCs) and lymphocytes are found in the adventitia of normal arteries, their number is greatly expanded and their distribution changed in human and mouse atherosclerotic arteries. Macrophages, DCs, foam cells, lymphocytes, and other inflammatory cells are found in the intimal atherosclerotic lesions. Beneath these lesions, adventitial leukocytes organize in clusters that resemble tertiary lymphoid tissues. Experimental interventions can reduce the number of available blood monocytes, from which macrophages and most DCs and foam cells are derived, and reduce atherosclerotic lesion burden without altering blood lipids. Under proatherogenic conditions, nitric oxide production from endothelial cells is reduced and the burden of reactive oxygen species (ROS) and advanced glycation end products (AGE) is increased. Incapacitating ROS-generating NADPH oxidase or the receptor for AGE (RAGE) has beneficial effects. Targeting inflammatory adhesion molecules also reduces atherosclerosis. Conversely, removing or blocking IL-10 or TGF-beta accelerates atherosclerosis. Regulatory T cells and B1 cells secreting natural antibodies are atheroprotective. This review summarizes our current understanding of inflammatory and immune mechanisms in atherosclerosis.
Collapse
Affiliation(s)
- Elena Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507-1696, USA.
| | | |
Collapse
|