51
|
Schubert R, Geoffroy E, Gregga I, Mulford AJ, Aguet F, Ardlie K, Gerszten R, Clish C, Van Den Berg D, Taylor KD, Durda P, Johnson WC, Cornell E, Guo X, Liu Y, Tracy R, Conomos M, Blackwell T, Papanicolaou G, Lappalainen T, Mikhaylova AV, Thornton TA, Cho MH, Gignoux CR, Lange L, Lange E, Rich SS, Rotter JI, Manichaikul A, Im HK, Wheeler HE. Protein prediction for trait mapping in diverse populations. PLoS One 2022; 17:e0264341. [PMID: 35202437 PMCID: PMC8870552 DOI: 10.1371/journal.pone.0264341] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Genetically regulated gene expression has helped elucidate the biological mechanisms underlying complex traits. Improved high-throughput technology allows similar interrogation of the genetically regulated proteome for understanding complex trait mechanisms. Here, we used the Trans-omics for Precision Medicine (TOPMed) Multi-omics pilot study, which comprises data from Multi-Ethnic Study of Atherosclerosis (MESA), to optimize genetic predictors of the plasma proteome for genetically regulated proteome-wide association studies (PWAS) in diverse populations. We built predictive models for protein abundances using data collected in TOPMed MESA, for which we have measured 1,305 proteins by a SOMAscan assay. We compared predictive models built via elastic net regression to models integrating posterior inclusion probabilities estimated by fine-mapping SNPs prior to elastic net. In order to investigate the transferability of predictive models across ancestries, we built protein prediction models in all four of the TOPMed MESA populations, African American (n = 183), Chinese (n = 71), European (n = 416), and Hispanic/Latino (n = 301), as well as in all populations combined. As expected, fine-mapping produced more significant protein prediction models, especially in African ancestries populations, potentially increasing opportunity for discovery. When we tested our TOPMed MESA models in the independent European INTERVAL study, fine-mapping improved cross-ancestries prediction for some proteins. Using GWAS summary statistics from the Population Architecture using Genomics and Epidemiology (PAGE) study, which comprises ∼50,000 Hispanic/Latinos, African Americans, Asians, Native Hawaiians, and Native Americans, we applied S-PrediXcan to perform PWAS for 28 complex traits. The most protein-trait associations were discovered, colocalized, and replicated in large independent GWAS using proteome prediction model training populations with similar ancestries to PAGE. At current training population sample sizes, performance between baseline and fine-mapped protein prediction models in PWAS was similar, highlighting the utility of elastic net. Our predictive models in diverse populations are publicly available for use in proteome mapping methods at https://doi.org/10.5281/zenodo.4837327.
Collapse
Affiliation(s)
- Ryan Schubert
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL, United States of America
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| | - Elyse Geoffroy
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| | - Isabelle Gregga
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
| | - Ashley J. Mulford
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| | - Francois Aguet
- Broad Institute, Cambridge, MA, United States of America
| | - Kristin Ardlie
- Broad Institute, Cambridge, MA, United States of America
| | - Robert Gerszten
- Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Clary Clish
- Broad Institute, Cambridge, MA, United States of America
| | - David Van Den Berg
- University of Southern California, Los Angeles, CA, United States of America
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Peter Durda
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, United States of America
| | - W. Craig Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, United States of America
| | - Elaine Cornell
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, United States of America
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States of America
| | - Russell Tracy
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, United States of America
| | - Matthew Conomos
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Tom Blackwell
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States of America
| | - George Papanicolaou
- Epidemiology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States of America
| | - Tuuli Lappalainen
- New York Genome Center and Department of Systems Biology, Columbia University, New York, NY United States of America
| | - Anna V. Mikhaylova
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Christopher R. Gignoux
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Leslie Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Ethan Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States of America
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States of America
| | | | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States of America
| | - Hae Kyung Im
- Section of Genetic Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Heather E. Wheeler
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| |
Collapse
|
52
|
Kim KY, Kim JO, Kim YS, Choi JE, Park JM, Han K, Park DH, Park YC, Kim BT, Hong KW. Genome-wide association of individual vulnerability with alcohol-associated liver disease: A Korean genome and epidemiology study. Hepatology 2022; 75:391-402. [PMID: 34387878 PMCID: PMC9300105 DOI: 10.1002/hep.32115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND AIMS The quantity of alcohol leading to alcohol-associated liver disease (ALD) varies individually. Genetic backgrounds contributing to the divergence in individual susceptibility to alcohol-induced liver damage have not been elucidated in detail. APPROACH AND RESULTS Based on the Korean Genome and Epidemiology Study Health Examination (KoGES_HEXA) cohort data, 21,919 participants (40-79 years old) were included and divided into cases and controls based on the ALD diagnostic criteria proposed by the American College of Gastroenterology. Data generated by a genome wide-association study were analyzed using logistic regression to assess the risk of ALD development in nondrinkers, light drinkers, and heavy drinkers. We detected three loci, gamma-glutamyltransferase 1 (GGT1), zinc protein finger 827 (ZNF827) and HNF1 homeobox A (HNF1A), which were significantly associated with ALD risk. The GGT1 rs2006227 minor allele was strongly associated with all groups. Among the minor alleles of single nucleotide polymorphisms (SNPs) in HNF1A, rs1183910 had the strongest association with a protective effect from ALD in light drinkers. However, this association was not observed in heavy drinkers. Five SNPs on chromosome 11 showed suggestive significance in protective effects against ALD. CONCLUSIONS SNPs, including HNF1A rs1183910 minor allele, are the most promising genetic candidates for protection against ALD. The expression of genes contributing to ALD development may be altered by the amount of alcohol consumed.
Collapse
Affiliation(s)
- Kwang Yoon Kim
- Department of Family Practice & Community HealthAjou University School of MedicineSuwonRepublic of Korea
| | - Jung Oh Kim
- Healthcare R&D DivisionTheragen Bio Co. Ltd.SuwonRepublic of Korea
| | - Young-Sang Kim
- Department of Family MedicineCHA Bundang Medical CenterCHA UniversitySeongnamRepublic of Korea
| | - Ja-Eun Choi
- Healthcare R&D DivisionTheragen Bio Co. Ltd.SuwonRepublic of Korea
| | - Jae-Min Park
- Department of Family MedicineGangnam Severance HospitalYonsei University College of MedicineSeoulRepublic of Korea
| | - Kunhee Han
- Department of Family MedicineSeonam HospitalSeoulRepublic of Korea
| | - Da-Hyun Park
- Healthcare R&D DivisionTheragen Bio Co. Ltd.SuwonRepublic of Korea
| | - Yon Chul Park
- Department of Family MedicineWonju Severance Christian HospitalWonjuRepublic of Korea.,Department of Medical EducationYonsei University Wonju College of MedicineWonjuRepublic of Korea
| | - Bom Taeck Kim
- Department of Family Practice & Community HealthAjou University School of MedicineSuwonRepublic of Korea
| | - Kyung-Won Hong
- Healthcare R&D DivisionTheragen Bio Co. Ltd.SuwonRepublic of Korea
| |
Collapse
|
53
|
Liu Y, Zhang C, Jiang L, Xu L, Tian J, Zhao X, Feng X, Wang D, Zhang Y, Sun K, Xu J, Liu R, Xu B, Zhao W, Hui R, Gao R, Gao Z, Song L, Yuan J. Relationship Between High-Sensitivity C-Reactive Protein and Long-Term Outcomes in Elderly Patients With 3-Vessel Disease. Angiology 2022; 73:60-67. [PMID: 34109809 DOI: 10.1177/00033197211021195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The prognostic value of high-sensitivity C-reactive protein (hsCRP) in complex coronary artery disease has not been fully established. We aimed to determine the association between hsCRP and long-term outcomes in elderly patients with 3-vessel disease (TVD). From April 2004 to February 2011, 3069 patients aged ≥65 years with TVD were consecutively enrolled and received medical treatment alone, percutaneous coronary intervention, or coronary artery bypass grafting. The patients were divided into 2 groups according to their hsCRP levels: <3.00 mg/L (62.1%) and ≥3.00 mg/L (37.9%). The mean age was 71 ± 4 years. The high hsCRP group had more risk factors and more frequently received conservative treatment than the low hsCRP group. During a median follow-up period of 6.2 years, elevated hsCRP was significantly associated with increased all-cause death (19.5% vs 29.6%, P < .001), cardiac death (9.4% vs 15.2%, P = .001), and major adverse cardiovascular and cerebrovascular events (34.1% vs 42.5%, P = .001). Multivariable Cox regression analyses revealed that hsCRP was an independent predictor for all of these events. Combining hsCRP with Synergy between PCI with TAXUS and Cardiac Surgery score II further improved the predictive power of the score. The relationship between hsCRP and mortality was relatively consistent across subgroups. Overall, hsCRP could prove useful for risk prediction in elderly patients.
Collapse
Affiliation(s)
- Yue Liu
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Ce Zhang
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Lin Jiang
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Lianjun Xu
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Jian Tian
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Xueyan Zhao
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Xinxing Feng
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Dong Wang
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Yin Zhang
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Kai Sun
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Jingjing Xu
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Ru Liu
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Bo Xu
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Wei Zhao
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Rutai Hui
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Runlin Gao
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhan Gao
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Lei Song
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Jinqing Yuan
- 34736Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
54
|
Anghel D, Sîrbu CA, Hoinoiu EM, Petrache OG, Pleșa CF, Negru MM, Ioniţă-Radu F. Influence of anti-TNF therapy and homocysteine level on carotid intima-media thickness in rheumatoid arthritis patients. Exp Ther Med 2021; 23:59. [PMID: 34917185 DOI: 10.3892/etm.2021.10981] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/07/2021] [Indexed: 01/18/2023] Open
Abstract
It is a well-known fact that disruptions in the immune system and systemic inflammation are associated with accelerated atherosclerosis in rheumatoid arthritis (RA) patients. Elevated levels of tumor necrosis factor α (TNF-α), a major pro-inflammatory cytokine, are involved in endothelial cell activation of medium and large arteries, leading to increased endothelial permeability, generation of superoxide anion radical and hydrogen peroxide, and decreased availability of nitric oxide (NO). The present study aims to determine the influence of anti-TNF therapy and homocysteine (Hcy) levels on the carotid intima-media thickness (IMT) in patients with RA. Assessments were performed on 115 patients diagnosed with RA on biological treatment to determine the evolution of IMT and Hcy levels. Carotid ultrasonography was used to assess the IMT, as a fast and easy tool for the prediction of cardiovascular events in patients with RA. The first measurement of IMT was noted as IMT1, followed by a second measurement after 1 year, noted as IMT2. The group of patients was divided into approximately three equal groups, each being treated with a different biological product, respectively, etanercept, adalimumab, and infliximab. In the 3 groups, after 1 year of anti-TNF-α therapy, IMT2 progression was significantly reduced compared to baseline. No significant differences were found among the three groups of treatment. A strong association was observed between IMT1-IMT2 in the etanercept group (P<0.001, r=0.758), in the adalimumab group (P<0.001, r=0.761) and in the infliximab group (P<0.001, r=0.829). The low level of Hcy2 after 12 months of anti-TNF-α therapy was significantly correlated with a decrease in IMT2 (P<0.001) in patients who had a high level of Hcy and IMT >0.9 mm at baseline. The results from the present study showed that biological treatment and the low level of homocysteinemia reduced the cardiovascular risk in RA, regardless of the treatment chosen (infliximab, adalimumab, or etanercept).
Collapse
Affiliation(s)
- Daniela Anghel
- Department of Internal Medicine, Central Military Emergency University Hospital, 010242 Bucharest, Romania.,Department of Medico-Surgical and Prophylactic Disciplines, Faculty of Medicine, 'Titu Maiorescu' University, 031593 Bucharest, Romania
| | - Carmen Adella Sîrbu
- Department of Medico-Surgical and Prophylactic Disciplines, Faculty of Medicine, 'Titu Maiorescu' University, 031593 Bucharest, Romania.,Department of Neurology, Central Military Emergency University Hospital, 010242 Bucharest, Romania
| | - Elena-Mădălina Hoinoiu
- Department of Internal Medicine, Central Military Emergency University Hospital, 010242 Bucharest, Romania
| | - Oana-Georgiana Petrache
- Department of Internal Medicine, Central Military Emergency University Hospital, 010242 Bucharest, Romania
| | - Cristina-Florentina Pleșa
- Department of Neurology, Central Military Emergency University Hospital, 010242 Bucharest, Romania.,Department of Preclinical Disciplines, Faculty of Medicine, 'Titu Maiorescu' University, 031593 Bucharest, Romania
| | - Maria Magdalena Negru
- Department of Internal Medicine and Rheumatology, 'Sf. Maria' Clinical Hospital, 011172 Bucharest, Romania.,Department of Internal Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Florentina Ioniţă-Radu
- Department of Internal Medicine, 'Carol Davila' University of Medicine and Pharmacy, 020022 Bucharest, Romania.,Department of Gastroenterology, Central Military Emergency University Hospital, 010242 Bucharest, Romania
| |
Collapse
|
55
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:785-796. [PMID: 34857118 DOI: 10.1016/j.repce.2021.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
56
|
Monomeric C reactive protein (mCRP) regulates inflammatory responses in human and mouse chondrocytes. J Transl Med 2021; 101:1550-1560. [PMID: 33767361 DOI: 10.1038/s41374-021-00584-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
C-reactive protein (CRP) is an acute-phase protein that is used as an established biomarker to follow disease severity and progression in a plethora of inflammatory diseases. However, its pathophysiologic mechanisms of action are still poorly defined and remain elusive. CRP, in its pentameric form, exhibits weak anti-inflammatory activity. On the contrary, the monomeric isoform (mCRP) exhibits potent pro-inflammatory properties in endothelial cells, leukocytes, and platelets. So far, no data exists regarding mCRP effects in human or mouse chondrocytes. This work aimed to verify the pathophysiological relevance of mCRP in the etiology and/or progression of osteoarthritis (OA). We investigated the effects of mCRP in cultured human primary chondrocytes and in the chondrogenic ATDC5 mouse cell line. We determined mRNA and protein levels of relevant factors involved in inflammatory responses and the modulation of nitric oxide synthase type II (NOS2), an early inflammatory molecular target. We demonstrate, for the first time, that monomeric C reactive protein increases NOS2, COX2, MMP13, VCAM1, IL-6, IL-8, and LCN2 expression in human and murine chondrocytes. We also demonstrated that NF-kB is a key factor in the intracellular signaling of mCRP-driven induction of pro-inflammatory and catabolic mediators in chondrocytes. We concluded that mCRP exerts a sustained catabolic effect on human and murine chondrocytes, increasing the expression of inflammatory mediators and proteolytic enzymes, which can promote extracellular matrix (ECM) breakdown in healthy and OA cartilage. In addition, our results implicate the NF-kB signaling pathway in catabolic effects mediated by mCRP.
Collapse
|
57
|
Wang IC, Sugai J, Majzoub J, Johnston J, Giannobile WV, Wang HL. Pro-inflammatory Profiles in Cardiovascular Disease Patients with Peri-implantitis. J Periodontol 2021; 93:824-836. [PMID: 34807456 DOI: 10.1002/jper.21-0419] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/28/2021] [Accepted: 11/16/2021] [Indexed: 11/06/2022]
Abstract
AIM To investigate the pro-inflammatory cytokine profiles in patients with or without cardiovascular disease (CVD) and with or without peri-implantitis. METHODS Serum, peri-implant crevicular fluid (PICF), and gingival crevicular fluid (GCF) were collected from patients with (n = 82) or without CVD (n = 46) at the most severe peri-implantitis site including sites with periodontitis. A panel of proinflammatory molecules including high-sensitivity C-reactive protein (hsCRP), This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- I-Ching Wang
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Currently, Department of Periodontics, College of Dentistry, University of Iowa, Iowa City, Iowa, USA
| | - James Sugai
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Jad Majzoub
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Jeffery Johnston
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Vice President, Chief Science Officer, and Director of the Research and Data Institute at Delta Dental of Michigan
| | - William V Giannobile
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Currently, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Hom-Lay Wang
- Department of Periodontics & Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
58
|
Khafagy R, Dash S. Obesity and Cardiovascular Disease: The Emerging Role of Inflammation. Front Cardiovasc Med 2021; 8:768119. [PMID: 34760952 PMCID: PMC8573144 DOI: 10.3389/fcvm.2021.768119] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/27/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity is a growing public health challenge across the globe. It is associated with increased morbidity and mortality. Cardiovascular disease (CVD) is the leading cause of mortality for people with obesity. Current strategies to reduce CVD are largely focused on addressing traditional risk factors such as dyslipidemia, type 2 diabetes (T2D) and hypertension. Although this approach is proven to reduce CVD, substantial residual risk remains for people with obesity. This necessitates a better understanding of the etiology of CVD in people with obesity and alternate therapeutic approaches. Reducing inflammation may be one such strategy. A wealth of animal and human data indicates that obesity is associated with adipose tissue and systemic inflammation. Inflammation is a known contributor to CVD in humans and can be successfully targeted to reduce CVD. Here we will review the etiology and pathogenesis of inflammation in obesity associated metabolic disease as well as CVD. We will review to what extent these associations are causal based on human genetic studies and pharmacological studies. The available data suggests that anti-inflammatory treatments can be used to reduce CVD, but off-target effects such as increased infection have precluded its broad therapeutic application to date. The role of anti-inflammatory therapies in improving glycaemia and metabolic parameters is less established. A number of clinical trials are currently ongoing which are evaluating anti-inflammatory agents to lower CVD. These studies will further clarify whether anti-inflammatory agents can safely reduce CVD.
Collapse
Affiliation(s)
- Rana Khafagy
- Department of Pharmacy, The Hospital for Sick Children, Toronto, ON, Canada.,Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University Health Network, Toronto, ON, Canada
| | - Satya Dash
- Banting & Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University Health Network, Toronto, ON, Canada
| |
Collapse
|
59
|
Association between Biomarkers of Oxidative Stress and Inflammation with Cardiac Necrosis and Heart Failure in Non-ST Segment Elevation Myocardial Infarction Patients and Various Degrees of Kidney Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3090120. [PMID: 34760045 PMCID: PMC8575633 DOI: 10.1155/2021/3090120] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/09/2021] [Accepted: 10/22/2021] [Indexed: 01/29/2023]
Abstract
The aim of this study was to explore the possible association between markers of inflammation and oxidative stress (OS) and markers of cardiac function and necrosis in 100 NSTEMI (non-ST-elevation myocardial infarction) patients with various degrees of kidney dysfunction. At admission, ejection fraction (EF), brain natriuretic peptide (BNP), troponin (TnI), creatinine phosphokinase (CPK), alanine transaminase (ALT), aspartate transaminase (AST), high-sensitive C-reactive protein (hs-CRP), interleukins 6 and 10 (IL-6, IL10), myeloperoxidase (MPO), transforming growth factor beta (TGF-β1), glomerular filtration rate (GFR), and albuminuria were assessed. Study participants were divided into 2 subgroups based on the median level of EF. Compared to the high, patients in the low EF group had higher GFR, BNP, CPK, hs-CRP, IL-10, IL-6, and MPO values and lower albuminuria levels. The levels of EF decreased in parallel with the progression of CKD, whereas the levels of BNP, IL-6, and TGF-β were significantly higher in late stages of CKD. Spearman's rho correlation analysis showed that EF was inversely correlated with MPO (r = -0.20, p = 0.05) BNP (r = -0.30, p = 0.002), hs-CRP (r = -0.38, p < 0.0001), IL-10 (r = -0.30, p = 0.003), and IL-6 (r = -0.24, p = 0.02) and positively with GFR (r = 0.27, p = 0.008). TnI was correlated with CPK (r = 0.44, p < 0.0001), CPK-MB (r = 0.31, p = 0.002), ALT (r = 0.50, p < 0.0001), AST (r = 0.29, p = 0.004), IL-10 (r = 0.22, p = 0.03), and MPO (r = -0.28, p = 0.006). In multivariate regression analysis, only BNP (β = -0.011, p = 0.004), hs-CRP (β = -0.11, p = 0.001), and GFR (β = 0.12, p = 0.0029) were independent determinants of EF. Similarly, MPO (β = -1.69, p = 0.02), IL-10 (β = 0.15, p = 0.006), and AST (β = 0.04, p = 0.001) were the 3 major determinants of TnI. Based on these associations, we built a predictive model including markers of inflammation and OS (MPO, IL-10, and hs-CRP) to identify patients with the most severe cardiac injury (combined EF below median and troponin above median values). Receiver-operator characteristic (ROC) analysis showed that the area under the ROC curve of this model to detect patients with low EF and high TnI was 0.67 (p = 0.015, 95%confidence interval = 0.53-0.81).
Collapse
|
60
|
Abstract
During the past decade, genome-wide association studies (GWAS) have transformed our understanding of many heritable traits. Three recent large-scale GWAS meta-analyses now further markedly expand the knowledge on coronary artery disease (CAD) genetics in doubling the number of loci with genome-wide significant signals. Here, we review the unprecedented discoveries of CAD GWAS on low-frequency variants, underrepresented populations, sex differences and integrated polygenic risk. We present the milestones of CAD GWAS and post-GWAS studies from 2007 to 2021, and the trend in identification of variants with smaller odds ratio by year due to the increasing sample size. We compile the 321 CAD loci discovered thus far and classify candidate genes as well as distinct functional pathways on the road to indepth biological investigation and identification of novel treatment targets. We draw attention to systems genetics in integrating these loci into gene regulatory networks within and across tissues. We review the traits, biomarkers and diseases scrutinized by Mendelian randomization studies for CAD. Finally, we discuss the potentials and concerns of polygenic scores in predicting CAD risk in patient care as well as future directions of GWAS and post-GWAS studies in the field of precision medicine.
Collapse
Affiliation(s)
- Zhifen Chen
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Munich Heart Alliance, Munich, Germany
| |
Collapse
|
61
|
El-Far M, Hanna DB, Durand M, Larouche-Anctil E, Sylla M, Chartrand-Lefebvre C, Cloutier G, Goulet JP, Kassaye S, Karim R, Kizer JR, French AL, Gange SJ, Lazar JM, Hodis HN, Routy JP, Ancuta P, Chomont N, Landay AL, Kaplan RC, Tremblay CL. Brief Report: Subclinical Carotid Artery Atherosclerosis Is Associated With Increased Expression of Peripheral Blood IL-32 Isoforms Among Women Living With HIV. J Acquir Immune Defic Syndr 2021; 88:186-191. [PMID: 34138771 PMCID: PMC8434945 DOI: 10.1097/qai.0000000000002746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Persistent inflammation in HIV infection is associated with elevated cardiovascular disease (CVD) risk, even with viral suppression. Identification of novel surrogate biomarkers can enhance CVD risk stratification and suggest novel therapies. We investigated the potential of interleukin 32 (IL-32), a proinflammatory multi-isoform cytokine, as a biomarker for subclinical carotid artery atherosclerosis in virologically suppressed women living with HIV (WLWH). METHODS AND RESULTS Nested within the Women's Interagency HIV Study, we conducted a cross-sectional comparison of IL-32 between 399 WLWH and 100 women without HIV, followed by a case-control study of 72 WLWH (36 carotid artery plaque cases vs. 36 age-matched controls without plaque). Plasma IL-32 protein was measured by ELISA, and mRNA of IL-32 isoforms (IL-32α, β, γ, D, ε, and θ) was quantified by reverse transcription polymerase chain reaction from peripheral blood mononuclear cells. Plasma IL-32 protein levels were higher in WLWH compared with women without HIV (P = 0.02). Among WLWH, although plasma IL-32 levels did not differ significantly between plaque cases and controls, expression of IL-32 isoforms α, β, and ε mRNA was significantly higher in peripheral blood mononuclear cells from cases (P = 0.01, P = 0.005, and P = 0.018, respectively). Upregulation of IL-32β and IL-32ε among WLWH with carotid artery plaque persisted after adjustment for age, race/ethnicity, smoking, systolic blood pressure, body mass index, and history of hepatitis C virus (P = 0.04 and P = 0.045); the adjusted association for IL-32α was marginally significant (P = 0.07). CONCLUSIONS IL-32 isoforms should be studied further as potential CVD biomarkers. This is of particular interest in WLWH by virtue of altered IL-32 levels in this population.
Collapse
Affiliation(s)
| | | | - Madeleine Durand
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | | | | | - Carl Chartrand-Lefebvre
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Radiologie, Radio-oncologie et Médecine Nucléaire, Faculté de Médecine, Université de Montréal
| | - Guy Cloutier
- CHUM-Research Centre, Montréal, QC, Canada
- Département de radiologie et Institut de génie biomedical, Université de Montréal, Montréal, QC, Canada
| | | | - Seble Kassaye
- Department of medicine, Georgetown University, Washington, DC, USA
| | - Roksana Karim
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jorge R. Kizer
- Cardiology Section, San Francisco Veterans Affairs Health Care System, and Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Audrey L. French
- Division of Infectious Diseases, Stroger Hospital of Cook County, Chicago IL, USA
| | - Stephen J. Gange
- Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jason M. Lazar
- Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Howard N. Hodis
- Atherosclerosis Research Unit, University of Southern California, Los Angeles, CA, USA
| | - Jean-Pierre Routy
- Research Institute of McGill University Health Centre, Montréal, QC, Canada
| | - Petronela Ancuta
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | - Nicolas Chomont
- CHUM-Research Centre, Montréal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, QC, Canada
| | | | - Robert C. Kaplan
- Albert Einstein College of Medicine, Bronx, NY, USA
- Divsion of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle WA, USA
| | - Cécile L. Tremblay
- CHUM-Research Centre, Montréal, QC, Canada
- Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
62
|
Febbraio M, Roy CB, Levin L. Is There a Causal Link Between Periodontitis and Cardiovascular Disease? A Concise Review of Recent Findings. Int Dent J 2021; 72:37-51. [PMID: 34565546 PMCID: PMC9275186 DOI: 10.1016/j.identj.2021.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/19/2021] [Accepted: 07/23/2021] [Indexed: 01/08/2023] Open
Abstract
There is substantial evidence in support of an association between periodontitis and cardiovascular disease. The most important open question related to this association is causality. This article revisits the question of causality by reviewing intervention studies and systematic reviews and meta analyses published in the last 3 years. Where are we now in answering this question? Whilst systematic reviews and epidemiological studies continue to support an association between the diseases, intervention studies fall short in determining causality. There is a dearth of good-quality, blinded randomised control trials with cardiovascular disease outcomes. Most studies use surrogate markers/biomarkers for endpoints, and this is problematic as they may not be reflective of cardiovascular disease status. This review further highlights another issue with surrogate markers/biomarkers: the potential for collider bias. Ethical considerations surrounding nontreatment have led to calls for a well-annotated database containing in-depth dental health data. Finally, a relatively new and important risk factor for cardiovascular disease, clonal haematopoiesis of indeterminate potential, is discussed. Clonal haematopoiesis of indeterminate potential increases cardiovascular risk by more than 40%, and inflammation is a contributing factor. The impact of periodontal disease on this emerging risk factor has yet to be explored. Although the question of causality in the association between periodontal disease and cardiovascular disease remains unanswered, the importance of good oral health in maintaining good heart health is reiterated.
Collapse
Affiliation(s)
- Maria Febbraio
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | | | - Liran Levin
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
63
|
Roy S, Sleiman MB, Jha P, Ingels JF, Chapman CJ, McCarty MS, Ziebarth JD, Hook M, Sun A, Zhao W, Huang J, Neuner SM, Wilmott LA, Shapaker TM, Centeno AG, Ashbrook DG, Mulligan MK, Kaczorowski CC, Makowski L, Cui Y, Read RW, Miller RA, Mozhui K, Williams EG, Sen S, Lu L, Auwerx J, Williams RW. Gene-by-environment modulation of lifespan and weight gain in the murine BXD family. Nat Metab 2021; 3:1217-1227. [PMID: 34552269 PMCID: PMC8478125 DOI: 10.1038/s42255-021-00449-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/06/2021] [Indexed: 02/07/2023]
Abstract
How lifespan and body weight vary as a function of diet and genetic differences is not well understood. Here we quantify the impact of differences in diet on lifespan in a genetically diverse family of female mice, split into matched isogenic cohorts fed a low-fat chow diet (CD, n = 663) or a high-fat diet (HFD, n = 685). We further generate key metabolic data in a parallel cohort euthanized at four time points. HFD feeding shortens lifespan by 12%: equivalent to a decade in humans. Initial body weight and early weight gains account for longevity differences of roughly 4-6 days per gram. At 500 days, animals on a HFD typically gain four times as much weight as control, but variation in weight gain does not correlate with lifespan. Classic serum metabolites, often regarded as health biomarkers, are not necessarily strong predictors of longevity. Our data indicate that responses to a HFD are substantially modulated by gene-by-environment interactions, highlighting the importance of genetic variation in making accurate individualized dietary recommendations.
Collapse
Affiliation(s)
- Suheeta Roy
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pooja Jha
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jesse F Ingels
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Casey J Chapman
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Melinda S McCarty
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Jesse D Ziebarth
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Michael Hook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Anna Sun
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Wenyuan Zhao
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Jinsong Huang
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Sarah M Neuner
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Lynda A Wilmott
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Thomas M Shapaker
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Arthur G Centeno
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - David G Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | | | - Liza Makowski
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yan Cui
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Robert W Read
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Richard A Miller
- Department of Pathology, University of Michigan Geriatrics Center, Ann Arbor, MI, USA
| | - Khyobeni Mozhui
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Evan G Williams
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Saunak Sen
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Interfaculty Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center (UTHSC), Memphis, TN, USA.
| |
Collapse
|
64
|
Dao Bui Quy Q, Pham Ngoc Huy T, Nguyen Minh T, Nguyen Duc L, Nguyen Minh T, Nguyen Trung K, Tran Viet T, Do Q, Le Viet T. High Serum Uric Acid and High-Sensitivity C Reactive Protein Concentrations Predict Three-Year Cardiovascular Mortality in Patients Treated With Continuous Ambulatory Peritoneal Dialysis. Cureus 2021; 13:e17900. [PMID: 34532198 PMCID: PMC8435087 DOI: 10.7759/cureus.17900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2021] [Indexed: 12/03/2022] Open
Abstract
AIMS This study aims to access the predicting value of serum uric acid (UA) and high-sensitivity C reactive protein (hs-CRP) concentration on three-year cardiovascular-related mortality in patients performing continuous ambulatory peritoneal dialysis (CAPD). METHODS A total of267 CAPD patients [150 male (56.2%); mean age 48.93 ± 13.58 years] were included in our study. All patients had measured serum UA and hs-CRP concentration. A high-sensitivity particle-enhanced immunoturbidimetric assay determined serum hs-CRP; serum UA levels were determined using an enzymatic colorimetric assay. All patients were followed for three years to detect cardiovascular-related mortality by cardiologists and stroke specialists. RESULTS Mean serum UA level was 415.16 ± 84.28 µmol/L, 58.4% of patients had increased serum UA level. Median serum hs-CRP level was 2 (1-4) mg/L, 12.4% of patients had increased serum hs-CRP level. During 36 months of follow-up, 41 patients (15.4%) had cardiovascular-related mortality. The results of Cox proportional hazards regression showed that hypertension, diabetes, high serum UA and hs-CRP were risk factors that related to cardiovascular-related mortality (p<0.05). The receiver operating characteristic (ROC) curve and Kaplan-Meier analysis results showed that UA and hs-CRP level had predictive value for three-year cardiovascular-related mortality in CAPD patients [uric acid: area under the curve (AUC)=0.822; hs-CRP: AUC=0.834, p < 0.001]. CONCLUSION High serum UA and hs-CRP levels were predictive factors of cardiovascular-related mortality in CAPD patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Quyet Do
- Director, Vietnam Military Medical University, Hanoi, VNM
| | - Thang Le Viet
- Nephrology and Hemodialysis, Military Hospital 103, Hanoi, VNM
| |
Collapse
|
65
|
Luan YY, Yin CH, Yao YM. Update Advances on C-Reactive Protein in COVID-19 and Other Viral Infections. Front Immunol 2021; 12:720363. [PMID: 34447386 PMCID: PMC8382792 DOI: 10.3389/fimmu.2021.720363] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/21/2021] [Indexed: 01/08/2023] Open
Abstract
Severe coronavirus disease 2019 (COVID-19) can manifest as a viral-induced hyperinflammation with multiorgan dysfunction. It has been documented that severe COVID-19 is associated with higher levels of inflammatory mediators than a mild disease, and tracking these markers may allow early identification or even prediction of disease progression. It is well known that C-reactive protein (CRP) is the acute-phase protein and the active regulator of host innate immunity, which is highly predictive of the need for mechanical ventilation and may guide escalation of treatment of COVID-19-related uncontrolled inflammation. There are numerous causes of an elevated CRP, including acute and chronic responses, and these can be infectious or non-infectious in etiology. CRP are normally lacking in viral infections, while adaptive immunity appears to be essential for COVID-19 virus clearance, and the macrophage activation syndrome may explain the high serum CRP contents and contribute to the disease progression. Nevertheless, for the assessment of host inflammatory status and identification of viral infection in other pathologies, such as bacterial sepsis, the acute-phase proteins, including CRP and procalcitonin, can provide more important information for guiding clinical diagnosis and antibiotic therapy. This review is aimed to highlight the current and most recent studies with regard to the clinical significance of CRP in severe COVID-19 and other viral associated illnesses, including update advances on the implication of CRP and its form specifically on the pathogenesis of these diseases. The progressive understanding in these areas may be translated into promising measures to prevent severe outcomes and mitigate appropriate treatment modalities in critical COVID-19 and other viral infections.
Collapse
Affiliation(s)
- Ying-Yi Luan
- Translational Medicine Research Center, Medical Innovation Research Division and the Fourth Medical Center of PLA General Hospital, Beijing, China.,Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Cheng-Hong Yin
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and the Fourth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
66
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:S0870-2551(21)00321-8. [PMID: 34456098 DOI: 10.1016/j.repc.2021.02.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 10/20/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
67
|
Montarello NJ, Singh K, Sinhal A, Wong DTL, Alcock R, Rajendran S, Dautov R, Barlis P, Patel S, Nidorf SM, Thompson PL, Salagaras T, Butters J, Nerlekar N, Di Giovanni G, Ottaway JL, Nicholls SJ, Psaltis PJ. Assessing the Impact of Colchicine on Coronary Plaque Phenotype After Myocardial Infarction with Optical Coherence Tomography: Rationale and Design of the COCOMO-ACS Study. Cardiovasc Drugs Ther 2021; 36:1175-1186. [PMID: 34432196 PMCID: PMC8384919 DOI: 10.1007/s10557-021-07240-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 12/31/2022]
Abstract
Introduction Recurrent event rates after myocardial infarction (MI) remain unacceptably high, in part because of the continued growth and destabilization of residual coronary atherosclerotic plaques, which may occur despite lipid-lowering therapy. Inflammation is an important contributor to this ongoing risk. Recent studies have shown that the broad-acting anti-inflammatory agent, colchicine, may reduce adverse cardiovascular events in patients post-MI, although the mechanistic basis for this remains unclear. Advances in endovascular arterial wall imaging have allowed detailed characterization of the burden and compositional phenotype of coronary plaque, along with its natural history and responsiveness to treatment. One such example has been the use of optical coherence tomography (OCT) to demonstrate the plaque-stabilizing effects of statins on both fibrous cap thickness and the size of lipid pools within plaque. Methods The Phase 2, multi-centre, double-blind colchicine for coronary plaque modification in acute coronary syndrome (COCOMO-ACS) study will evaluate the effect of colchicine 0.5 mg daily on coronary plaque features using serial OCT imaging in patients following MI. Recruitment for the trial has been completed with 64 participants with non-ST elevation MI randomized 1:1 to colchicine or placebo in addition to guideline recommended therapies, including high-intensity statins. The primary endpoint is the effect of colchicine on the minimal fibrous cap thickness of non-culprit plaque over an 18-month period. Summary The COCOMO-ACS study will determine whether addition of colchicine 0.5 mg daily to standard post-MI treatment has incremental benefits on high-risk features of coronary artery plaques. If confirmed, this will provide new mechanistic insights into how colchicine may confer clinical benefits in patients with atherosclerotic cardiovascular disease. Trial Registration ANZCTR trial registration number: ACTRN12618000809235. Date of trial registration: 11th of May 2018.
Collapse
Affiliation(s)
- Nicholas J Montarello
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia
| | - Kuljit Singh
- Department of Cardiology, Gold Coast University Hospital, Gold Coast, Australia
| | - Ajay Sinhal
- Flinders Medical Centre, Flinders University, Adelaide, Australia
| | - Dennis T L Wong
- Victorian Heart Institute, Monash University, Clayton, Australia
| | | | | | | | | | | | - Stefan M Nidorf
- GenesisCare Western Australia, Perth, Australia.,Heart and Vascular Research Institute of Western Australia, Perth, Australia
| | - Peter L Thompson
- GenesisCare Western Australia, Perth, Australia.,Heart and Vascular Research Institute of Western Australia, Perth, Australia.,Sir Charles Gairdner Hospital, Perth, Australia
| | - Thalia Salagaras
- South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, SA, 5001, Australia
| | - Julie Butters
- Victorian Heart Institute, Monash University, Clayton, Australia.,South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, SA, 5001, Australia
| | - Nitesh Nerlekar
- Victorian Heart Institute, Monash University, Clayton, Australia.,Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Giuseppe Di Giovanni
- South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, SA, 5001, Australia
| | - Juanita L Ottaway
- South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, SA, 5001, Australia
| | | | - Peter J Psaltis
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide, Australia. .,South Australian Health and Medical Research Institute, PO Box 11060, Adelaide, SA, 5001, Australia. .,Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
68
|
Si S, Li J, Tewara MA, Xue F. Genetically Determined Chronic Low-Grade Inflammation and Hundreds of Health Outcomes in the UK Biobank and the FinnGen Population: A Phenome-Wide Mendelian Randomization Study. Front Immunol 2021; 12:720876. [PMID: 34386016 PMCID: PMC8353321 DOI: 10.3389/fimmu.2021.720876] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022] Open
Abstract
Background C-reactive protein (CRP) has been used as a biomarker of chronic low-grade inflammation in observational studies. We aimed to determine whether genetically determined CRP was associated with hundreds of human phenotypes to guide anti-inflammatory interventions. Methods We used individual data from the UK Biobank to perform a phenome-wide two-stage least squares (2SLS) Mendelian randomization (MR) analysis for CRP with 879 diseases. Summary-level data from the FinnGen consortium were utilized to perform phenome-wide two-sample MR analysis on 821 phenotypes. Systematic two-sample MR methods included MR-IVW, MR-WME, MR-Mod, and MR-PRESSO as sensitivity analyses combined with multivariable MR to identify robust associations. Genetic correlation analysis was applied to identify shared genetic risks. Results We found genetically determined CRP was robustly associated with 15 diseases in the UK Biobank and 11 diseases in the FinnGen population (P < 0.05 for all MR analyses). CRP was positively associated with tongue cancer, bronchitis, hydronephrosis, and acute pancreatitis and negatively associated with colorectal cancer, colon cancer, cerebral ischemia, electrolyte imbalance, Parkinson's disease, epilepsy, anemia of chronic disease, encephalitis, psychophysical visual disturbances, and aseptic necrosis of bone in the UK Biobank. There were positive associations with impetigo, vascular dementia, bipolar disorders, hypercholesterolemia, vertigo, and neurological diseases, and negative correlations with degenerative macular diseases, metatarsalgia, interstitial lung disease, and idiopathic pulmonary fibrosis, and others. in the FinnGen population. The electrolyte imbalance and anemia of chronic disease in UK Biobank and hypercholesterolemia and neurological diseases in FinnGen pass the FDR corrections. Neurological diseases and bipolar disorders also presented positive genetic correlations with CRP. We found no overlapping causal associations between the populations. Previous causal evidence also failed to support these associations (except for bipolar disorders). Conclusions Genetically determined CRP was robustly associated with several diseases in the UK Biobank and the FinnGen population, but could not be replicated, suggesting heterogeneous and non-repeatable effects of CRP across populations. This implies that interventions at CRP are unlikely to result in decreased risk for most human diseases in the general population but may benefit specific high-risk populations. The limited causal evidence and potential double-sided effects remind us to be cautious about CRP interventions.
Collapse
Affiliation(s)
- Shucheng Si
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Shandong University, Jinan, China
- National Institute of Health Data Science of China, Shandong University, Jinan, China
| | - Jiqing Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Shandong University, Jinan, China
- National Institute of Health Data Science of China, Shandong University, Jinan, China
| | - Marlvin Anemey Tewara
- Center for Health Promotion and Research (Former Tuberculosis Reference Laboratory), Bamenda, Cameroon
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Shandong University, Jinan, China
- National Institute of Health Data Science of China, Shandong University, Jinan, China
| |
Collapse
|
69
|
Libby P. Inflammation in Atherosclerosis-No Longer a Theory. Clin Chem 2021; 67:131-142. [PMID: 33393629 DOI: 10.1093/clinchem/hvaa275] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/23/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Inflammation links to atherosclerosis and its complications in various experimental investigations. Animal studies have implicated numerous inflammatory mediators in the initiation and complication of atherosclerosis. Numerous studies in humans have shown associations of biomarkers of inflammation with cardiovascular events provoked by atheromata. Inflammatory status, determined by the biomarker C-reactive protein, can guide the allocation of statin therapy to individuals without elevated low-density lipoprotein (LDL) concentrations to prevent first ever adverse cardiovascular events. CONTENT Until recently, no direct evidence has shown that an intervention that selectively limits inflammation can improve outcomes in patients with atherosclerosis. A recent study, based on decades of preclinical investigation, treated patients who had sustained a myocardial infarction and whose LDL was well-controlled on statin treatment with an antibody that neutralizes interleukin-1 beta. This trial, conducted in over 10 000 individuals, showed a reduction in major adverse cardiac events, establishing for the first time the clinical efficacy of an anti-inflammatory intervention in atherosclerosis. Two large subsequent studies have shown that colchicine treatment can also prevent recurrent events in patients recovering from an acute coronary syndrome or in the stable phase of coronary artery disease. These clinical trials have transformed inflammation in atherosclerosis from theory to practice. SUMMARY Much work remains to optimize further anti-inflammatory interventions, minimize unwanted actions, and refine patient selection. This long road from discovery in the laboratory to successful clinical trials represents a victory for medical science, and opens a new avenue to reducing the risk that remains despite current treatments for atherosclerosis.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
70
|
Niu PP, Wang X, Xu YM. Association of Interleukin-6 Signaling and C-Reactive Protein With Intracranial Aneurysm: A Mendelian Randomization and Genetic Correlation Study. Front Genet 2021; 12:679363. [PMID: 34168680 PMCID: PMC8219052 DOI: 10.3389/fgene.2021.679363] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Background and objective Evidence suggests that interleukin-6 (IL6) signaling is causally associated with aortic aneurysm independently of the effect of C-reactive protein (CRP). We aimed to explore the genetic overlap and associations between inflammation (IL6 signaling and CRP) and intracranial aneurysm (IA) risk. Methods Two-sample Mendelian randomization (MR) methods were used to assess the causal effects of soluble IL6 receptor (sIL6R) (n = 21,758) and CRP (n = 204,402) levels on IA (7,495 cases and 71,934 controls) risk using genome-wide association study summary data of European individuals. Cross-trait linkage disequilibrium score regression was used to estimate the genetic correlations of CRP (n = 400,094) with IA. Results MR analyses showed that circulating sIL6R and CRP levels were not associated with the risk of IA. The odds ratios based on the inverse variance-weighted method were 0.986 (0.950-1.023, p = 0.45) and 0.957 (0.846-1.084, p = 0.49) for sIL6R and CRP, respectively. MR analyses using data of ruptured and unruptured IA each showed no association. Linkage disequilibrium score regression showed that the genetic correlation between CRP and IA was 0.16 (SE = 0.04, p = 0.0003). The genetic correlation diminished after conditioning IA on blood pressure (0.07 ± 0.05, p = 0.16), smoking (0.02 ± 0.05, p = 0.65), or blood pressure plus smoking (-0.03 ± 0.05, p = 0.53). Conclusion Using associated genetic variants as instrument variables, two-sample MR analyses showed no evidence that circulating sIL6R and CRP levels were associated with IA risk. Although a positive genetic correlation was found between CRP levels and IA risk, it was mainly driven by the shared genetic background of blood pressure and smoking with both CRP and IA.
Collapse
Affiliation(s)
- Peng-Peng Niu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
71
|
Wang J, Zhao Q, Bowden J, Hemani G, Davey Smith G, Small DS, Zhang NR. Causal inference for heritable phenotypic risk factors using heterogeneous genetic instruments. PLoS Genet 2021; 17:e1009575. [PMID: 34157017 PMCID: PMC8301661 DOI: 10.1371/journal.pgen.1009575] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/23/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Over a decade of genome-wide association studies (GWAS) have led to the finding of extreme polygenicity of complex traits. The phenomenon that "all genes affect every complex trait" complicates Mendelian Randomization (MR) studies, where natural genetic variations are used as instruments to infer the causal effect of heritable risk factors. We reexamine the assumptions of existing MR methods and show how they need to be clarified to allow for pervasive horizontal pleiotropy and heterogeneous effect sizes. We propose a comprehensive framework GRAPPLE to analyze the causal effect of target risk factors with heterogeneous genetic instruments and identify possible pleiotropic patterns from data. By using GWAS summary statistics, GRAPPLE can efficiently use both strong and weak genetic instruments, detect the existence of multiple pleiotropic pathways, determine the causal direction and perform multivariable MR to adjust for confounding risk factors. With GRAPPLE, we analyze the effect of blood lipids, body mass index, and systolic blood pressure on 25 disease outcomes, gaining new information on their causal relationships and potential pleiotropic pathways involved.
Collapse
Affiliation(s)
- Jingshu Wang
- Department of Statistics, University of Chicago, Chicago, Illinois, United States of America
| | - Qingyuan Zhao
- Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, Cambridge, United Kingdom
| | - Jack Bowden
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Dylan S. Small
- Department of Statistics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nancy R. Zhang
- Department of Statistics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
72
|
Ahluwalia TS, Prins BP, Abdollahi M, Armstrong NJ, Aslibekyan S, Bain L, Jefferis B, Baumert J, Beekman M, Ben-Shlomo Y, Bis JC, Mitchell BD, de Geus E, Delgado GE, Marek D, Eriksson J, Kajantie E, Kanoni S, Kemp JP, Lu C, Marioni RE, McLachlan S, Milaneschi Y, Nolte IM, Petrelis AM, Porcu E, Sabater-Lleal M, Naderi E, Seppälä I, Shah T, Singhal G, Standl M, Teumer A, Thalamuthu A, Thiering E, Trompet S, Ballantyne CM, Benjamin EJ, Casas JP, Toben C, Dedoussis G, Deelen J, Durda P, Engmann J, Feitosa MF, Grallert H, Hammarstedt A, Harris SE, Homuth G, Hottenga JJ, Jalkanen S, Jamshidi Y, Jawahar MC, Jess T, Kivimaki M, Kleber ME, Lahti J, Liu Y, Marques-Vidal P, Mellström D, Mooijaart SP, Müller-Nurasyid M, Penninx B, Revez JA, Rossing P, Räikkönen K, Sattar N, Scharnagl H, Sennblad B, Silveira A, Pourcain BS, Timpson NJ, Trollor J, van Dongen J, Van Heemst D, Visvikis-Siest S, Vollenweider P, Völker U, Waldenberger M, Willemsen G, Zabaneh D, Morris RW, Arnett DK, Baune BT, Boomsma DI, Chang YPC, Deary IJ, Deloukas P, Eriksson JG, Evans DM, Ferreira MA, Gaunt T, Gudnason V, Hamsten A, Heinrich J, Hingorani A, Humphries SE, Jukema JW, Koenig W, Kumari M, et alAhluwalia TS, Prins BP, Abdollahi M, Armstrong NJ, Aslibekyan S, Bain L, Jefferis B, Baumert J, Beekman M, Ben-Shlomo Y, Bis JC, Mitchell BD, de Geus E, Delgado GE, Marek D, Eriksson J, Kajantie E, Kanoni S, Kemp JP, Lu C, Marioni RE, McLachlan S, Milaneschi Y, Nolte IM, Petrelis AM, Porcu E, Sabater-Lleal M, Naderi E, Seppälä I, Shah T, Singhal G, Standl M, Teumer A, Thalamuthu A, Thiering E, Trompet S, Ballantyne CM, Benjamin EJ, Casas JP, Toben C, Dedoussis G, Deelen J, Durda P, Engmann J, Feitosa MF, Grallert H, Hammarstedt A, Harris SE, Homuth G, Hottenga JJ, Jalkanen S, Jamshidi Y, Jawahar MC, Jess T, Kivimaki M, Kleber ME, Lahti J, Liu Y, Marques-Vidal P, Mellström D, Mooijaart SP, Müller-Nurasyid M, Penninx B, Revez JA, Rossing P, Räikkönen K, Sattar N, Scharnagl H, Sennblad B, Silveira A, Pourcain BS, Timpson NJ, Trollor J, van Dongen J, Van Heemst D, Visvikis-Siest S, Vollenweider P, Völker U, Waldenberger M, Willemsen G, Zabaneh D, Morris RW, Arnett DK, Baune BT, Boomsma DI, Chang YPC, Deary IJ, Deloukas P, Eriksson JG, Evans DM, Ferreira MA, Gaunt T, Gudnason V, Hamsten A, Heinrich J, Hingorani A, Humphries SE, Jukema JW, Koenig W, Kumari M, Kutalik Z, Lawlor DA, Lehtimäki T, März W, Mather KA, Naitza S, Nauck M, Ohlsson C, Price JF, Raitakari O, Rice K, Sachdev PS, Slagboom E, Sørensen TIA, Spector T, Stacey D, Stathopoulou MG, Tanaka T, Wannamethee SG, Whincup P, Rotter JI, Dehghan A, Boerwinkle E, Psaty BM, Snieder H, Alizadeh BZ. Genome-wide association study of circulating interleukin 6 levels identifies novel loci. Hum Mol Genet 2021; 30:393-409. [PMID: 33517400 PMCID: PMC8098112 DOI: 10.1093/hmg/ddab023] [Show More Authors] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/02/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Interleukin 6 (IL-6) is a multifunctional cytokine with both pro- and anti-inflammatory properties with a heritability estimate of up to 61%. The circulating levels of IL-6 in blood have been associated with an increased risk of complex disease pathogenesis. We conducted a two-staged, discovery and replication meta genome-wide association study (GWAS) of circulating serum IL-6 levels comprising up to 67 428 (ndiscovery = 52 654 and nreplication = 14 774) individuals of European ancestry. The inverse variance fixed effects based discovery meta-analysis, followed by replication led to the identification of two independent loci, IL1F10/IL1RN rs6734238 on chromosome (Chr) 2q14, (Pcombined = 1.8 × 10-11), HLA-DRB1/DRB5 rs660895 on Chr6p21 (Pcombined = 1.5 × 10-10) in the combined meta-analyses of all samples. We also replicated the IL6R rs4537545 locus on Chr1q21 (Pcombined = 1.2 × 10-122). Our study identifies novel loci for circulating IL-6 levels uncovering new immunological and inflammatory pathways that may influence IL-6 pathobiology.
Collapse
Affiliation(s)
- Tarunveer S Ahluwalia
- Steno Diabetes Center Copenhagen, Gentofte DK2820, Denmark.,Department of Biology, The Bioinformatics Center, University of Copenhagen, Copenhagen DK2200, Denmark
| | - Bram P Prins
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Mohammadreza Abdollahi
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | | | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama 35233, USA
| | - Lisa Bain
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Barbara Jefferis
- Department of Primary Care & Population Health, UCL Institute of Epidemiology & Health Care, University College London, London NW3 2PF, UK
| | - Jens Baumert
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol BS8 2PS, UK
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| | - Eco de Geus
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Diana Marek
- SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Joel Eriksson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Centre for Bone and Arthritis Research (CBAR), University of Gothenburg, Gothenburg 41345, Sweden
| | - Eero Kajantie
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, PO Box 30, Helsinki 00271, Finland.,Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki 00014, Finland
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts & the London Medical School, Queen Mary University of London, London EC1M 6BQ, UK
| | - John P Kemp
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Chen Lu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Stela McLachlan
- Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam 1081 HJ, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | | | - Eleonora Porcu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato (CA) 09042, Italy
| | - Maria Sabater-Lleal
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden.,Unit of Genomics of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Barcelona 08041, Spain
| | - Elnaz Naderi
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Tina Shah
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Gaurav Singhal
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia
| | - Elisabeth Thiering
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany.,Division of Metabolic Diseases and Nutritional Medicine, Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Munich 80337, Germany
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands.,Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | | | - Emelia J Benjamin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA.,Section of Cardiovascular Medicine and Preventive Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Juan P Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA 02130, USA
| | - Catherine Toben
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - George Dedoussis
- 44Department of Nutrition-Dietetics, Harokopio University, Athens 17671, Greece
| | - Joris Deelen
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands.,Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jorgen Engmann
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany.,German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg SE-41345, Sweden
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17475, Germany
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku 20520, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku 20520, Finland
| | - Yalda Jamshidi
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George's University of London, London SW17 0RE, UK
| | - Magdalene C Jawahar
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - Tine Jess
- 55Department of Epidemiology Research, Statens Serum Institute, Copenhagen DK2300, Denmark
| | - Mika Kivimaki
- Department of Epidemiology & Public Health, UCL Institute of Epidemiology & Health Care, University College London, London WC1E 7HB, UK
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Jari Lahti
- Turku Institute for Advanced Studies, University of Turku, Turku 20014, Finland.,Department of Psychology and Logopedics, University of Helsinki, Helsinki 00014, Finland
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University Hospital (CHUV), Lausanne 1011, Switzerland.,University of Lausanne, Lausanne 1011, Switzerland
| | - Dan Mellström
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Centre for Bone and Arthritis Research (CBAR), University of Gothenburg, Gothenburg 41345, Sweden
| | - Simon P Mooijaart
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Martina Müller-Nurasyid
- IBE, Faculty of Medicine, Ludwig Maximilians University (LMU) Munich, Munich 81377, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johhanes Gutenberg University, Mainz 55101, Germany
| | - Brenda Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam 1081 HJ, The Netherlands
| | - Joana A Revez
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte DK2820, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen DK2200, Denmark
| | - Katri Räikkönen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki 00014, Finland
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow G12 8TA, UK
| | - Hubert Scharnagl
- 66Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria
| | - Bengt Sennblad
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden.,Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala 75124, Sweden
| | - Angela Silveira
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK.,Max Planck Institute for Psycholinguistics, Nijmegen XD 6525, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, The Netherlands
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Julian Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia.,Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | | | - Jenny van Dongen
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | | | | | - Peter Vollenweider
- Department of Internal Medicine, Lausanne University Hospital (CHUV), Lausanne 1011, Switzerland.,University of Lausanne, Lausanne 1011, Switzerland
| | - Uwe Völker
- MediCity Research Laboratory, University of Turku, Turku 20520, Finland
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Gonneke Willemsen
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Delilah Zabaneh
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London WC1E 6BT, UK
| | - Richard W Morris
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1UD, UK
| | - Donna K Arnett
- Dean's Office, College of Public Health, University of Kentucky, Lexington, KY 40536, USA
| | - Bernhard T Baune
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville 3000, Australia.,Department of Psychiatry and Psychotherapy, University of Muenster, Muenster 48149, Germany.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville 3000, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Yen-Pei C Chang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts & the London Medical School, Queen Mary University of London, London EC1M 6BQ, UK.,77Centre for Genomic Health, Queen Mary University of London, London EC1M 6BQ, UK
| | - Johan G Eriksson
- National Institute for Health and Welfare, University of Helsinki, Helsinki 00014, Finland.,Department of General Practice and Primary Health Care, University of Helsinki, Helsinki 00014, Finland
| | - David M Evans
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | | | - Tom Gaunt
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS6 2BN, UK.,Population Health Science, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur 201, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Anders Hamsten
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Joachim Heinrich
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany.,Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich 81377, Germany.,Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne 3010, Australia
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Steve E Humphries
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - J Wouter Jukema
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands.,Durrer Center for Cardiogenetic Research, Amsterdam 1105 AZ, The Netherlands
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich 80636, Germany.,88DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich 80336, Germany.,Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm 89081, Germany
| | - Meena Kumari
- Department of Epidemiology & Public Health, UCL Institute of Epidemiology & Health Care, University College London, London WC1E 7HB, UK.,Institute for Social and Economic Research, University of Essex, Colchester CO4 3SQ, Germany
| | - Zoltan Kutalik
- SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,University Center for Primary Care and Public Health, University of Lausanne, Lausanne 1010, Switzerland
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS6 2BN, UK.,Population Health Science, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany.,66Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria.,SYNLAB Academy, SYNALB Holding Deutschland GmbH, Mannheim 68163, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia.,Neuroscience Research Australia, Sydney 2031, Australia
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato (CA) 09042, Italy
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald 17475, Germany.,DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald 17475, Germany
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Centre for Bone and Arthritis Research (CBAR), University of Gothenburg, Gothenburg 41345, Sweden
| | - Jackie F Price
- Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku 20520, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20014, Finland
| | - Ken Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney 2031, Australia
| | - Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands.,Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center For Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK2200, Denmark.,Department of Public Health, Section on Epidemiology, University of Copenhagen, Copenhagen DK1014, Denmark
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - David Stacey
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | | | - Toshiko Tanaka
- Longitudinal Study Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | - S Goya Wannamethee
- Department of Primary Care & Population Health, UCL Institute of Epidemiology & Health Care, University College London, London NW3 2PF, UK
| | - Peter Whincup
- Population Health Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus MC, Rotterdam 3000 CA, The Netherlands
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA.,Departments of Epidemiology and Health Services, University of Washington, Seattle, WA 98101, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Behrooz Z Alizadeh
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| |
Collapse
|
73
|
Sun W, Han Y, Yang S, Zhuang H, Zhang J, Cheng L, Fu L. The Assessment of Interleukin-18 on the Risk of Coronary Heart Disease. Med Chem 2021; 16:626-634. [PMID: 31584380 DOI: 10.2174/1573406415666191004115128] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/13/2019] [Accepted: 08/23/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Observational studies support the inflammation hypothesis in coronary heart disease (CHD). As a pleiotropic proinflammatory cytokine, Interleukin-18 (IL-18), has also been found to be associated with the risk of CHD. However, to our knowledge, the method of Mendelian Randomization has not been used to explore the causal effect of IL-18 on CHD. OBJECTIVE To assess the causal effect of IL-18 on the risk of CHD. METHODS AND RESULTS Genetic variant instruments for IL-18 were obtained from information of the CHS and InCHIANTI cohort, and consisted of the per-allele difference in mean IL-18 for 16 independent variants that reached genome-wide significance. The per-allele difference in log-odds of CHD for each of these variants was estimated from CARDIoGRAMplusC4D, a two-stage meta -analysis. Two-sample Mendelian Randomization (MR) was then performed. Various MR analyses were used, including weighted inverse-variance, MR-Egger regression, robust regression, and penalized regression. The OR of elevated IL-18 associated with CHD was only 0.005 (95%CI -0.105~0.095; P-value=0.927). Similar results were obtained with the use of MR-Egger regression, suggesting that directional pleiotropy was unlikely biasing these results (intercept -0.050, P-value=0.220). Moreover, results from the robust regression and penalized regression analyses also revealed essentially similar findings. CONCLUSION Our findings indicate that, by itself, IL-18 is unlikely to represent even a modest causal factor for CHD risk.
Collapse
Affiliation(s)
- Weiju Sun
- Cardiovascular Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Han
- Cardiovascular Department, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - He Zhuang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jingwen Zhang
- Department of Physiology and Biology, University of Mississippi Medical Center, United States
| | - Liang Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lu Fu
- Cardiovascular Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
74
|
Best LG, Azure C, Martell K, Tsosie KS, Voels B. Unactivated leukocyte expression of C-reactive protein is minimal and not dependent on rs1205 genotype. Sci Rep 2021; 11:5691. [PMID: 33707594 PMCID: PMC7952394 DOI: 10.1038/s41598-021-85272-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/22/2021] [Indexed: 11/12/2022] Open
Abstract
C-reactive protein (CRP), a prominent component of the innate immune system, is implicated in the pathophysiology of many conditions. CRP production primarily occurs in the liver; but contributions from other tissues is unclear. The Genotype-Tissue Expression Portal shows essentially no expression in whole blood and reports in the literature are conflicting. Multiple genomic variants influence serum levels of CRP. We measured CRP mRNA expression in leukocytes and sought to determine if rs1205 genotype influences leukocyte expression. Leukocytes were obtained from 20 women differing by genotype. Quantitative, real-time PCR (RT-qPCR) detected CRP and reference gene (GAPDH) mRNA. Leukocyte expression was calculated by the 2ΔCT method, and against a standard curve. Digital drop PCR was also used to calculate expression ratios. Student's t test and linear regression methods examined possible differences between genotypes. During 32 runs (10 replicates each), the RT-qPCR mean (SD) CRP/GAPDH ratio was 3.39 × 10–4 (SD 1.73 × 10–4) and 3.15 × 10–4 (SD 1.64 × 10–4) for TT and CC genotypes respectively, p = 0.76; and digital drop PCR results were similar. Serum CRP was not significantly different between genotypes, nor correlated with leukocyte expression. CRP is minimally expressed in unactivated leukocytes and this expression is not likely influenced by rs1205 genotype.
Collapse
Affiliation(s)
- L G Best
- University of North Dakota, Grand Forks, ND, USA. .,Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA. .,, 1935 118th Ave NW, Watford City, ND, 58854, USA.
| | - C Azure
- Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA
| | - K Martell
- Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA
| | - K S Tsosie
- Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA
| | - B Voels
- Science, Cankdeska Cikana Community College, Fort Totten, ND, USA
| |
Collapse
|
75
|
Gardner RM, Lee BK, Brynge M, Sjöqvist H, Dalman C, Karlsson H. Neonatal Levels of Acute Phase Proteins and Risk of Autism Spectrum Disorder. Biol Psychiatry 2021; 89:463-475. [PMID: 33187600 DOI: 10.1016/j.biopsych.2020.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Immune signaling pathways influence neurodevelopment and are hypothesized to contribute to the etiology of autism spectrum disorder (ASD). We aimed to assess risk of ASD in relation to levels of neonatal acute phase proteins (APPs), key components of innate immune function, measured in neonatal dried blood spots. METHODS We included 924 ASD cases, 1092 unaffected population-based controls, and 203 unaffected siblings of ASD cases in this case-control study nested within the register-based Stockholm Youth Cohort. Concentrations of 9 different APPs were measured in eluates from neonatal dried blood spots from cases, controls, and siblings using a bead-based multiplex assay. RESULTS Neonatal C-reactive protein was consistently associated with odds of ASD in case-control comparisons, with higher odds associated with the highest quintile compared with the middle quintile (odds ratio [OR] = 1.50, 95% confidence interval [CI] = 1.10-2.04) in adjusted analyses. In contrast, the lowest quintiles of α-2-macroglobulin (OR = 3.71, CI = 1.21-11.33), ferritin (OR = 4.20, CI = 1.40-12.65), and serum amyloid P (OR = 3.05, CI = 1.16-8.01) were associated with odds of ASD in the matched sibling comparison. Neonatal APPs varied with perinatal environmental factors and maternal/fetal phenotypes. Significant interactions in terms of risk for ASD were observed between neonatal APPs and maternal infection during late pregnancy, maternal anemia, and maternal psychiatric history. CONCLUSIONS Indicators of the neonatal innate immune response are associated with risk of ASD, although the nature of these associations varies considerably with factors in the perinatal environment and the genetic background of the comparison group.
Collapse
Affiliation(s)
- Renee M Gardner
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| | - Brian K Lee
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology and Biostatistics, Drexel University School of Public Health, Philadelphia, Pennsylvania; A.J. Drexel Autism Institute, Philadelphia, Pennsylvania
| | - Martin Brynge
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Hugo Sjöqvist
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Christina Dalman
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden; Centre for Epidemiology and Community Medicine, Stockholm County Council, Stockholm, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
76
|
Sherman BT, Hu X, Singh K, Haine L, Rupert AW, Neaton JD, Lundgren JD, Imamichi T, Chang W, Lane HC. Genome-wide association study of high-sensitivity C-reactive protein, D-dimer, and interleukin-6 levels in multiethnic HIV+ cohorts. AIDS 2021; 35:193-204. [PMID: 33095540 PMCID: PMC7789909 DOI: 10.1097/qad.0000000000002738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/28/2020] [Accepted: 10/12/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Elevated levels of interleukin-6 (IL-6), D-dimer, and C-reactive protein (hsCRP) are associated with increased incidence of comorbid disease and mortality among people living with HIV (PLWH). Prior studies suggest a genetic basis for these biomarker elevations in the general population. The study objectives are to identify the genetic basis for these biomarkers among PLWH. METHODS Baseline levels of hsCRP, D-dimer, and IL-6, and single nucleotide polymorphisms (SNPs) were determined for 7768 participants in three HIV treatment trials. Single variant analysis was performed for each biomarker on samples from each of three ethnic groups [African (AFR), Admixed American (AMR), European (EUR)] within each trial including covariates relevant to biomarker levels. For each ethnic group, the results were pooled across trials, then further pooled across ethnicities. RESULTS The transethnic analysis identified three, two, and one known loci associated with hsCRP, D-dimer, and IL-6 levels, respectively, and two novel loci, FGB and GCNT1, associated with D-dimer levels. Lead SNPs exhibited similar effects across ethnicities. Additionally, three novel, ethnic-specific loci were identified: CATSPERG associated with D-dimer in AFR and PROX1-AS1 and TRAPPC9 associated with IL-6 in AFR and AMR, respectively. CONCLUSION Eleven loci associated with three biomarker levels were identified in PLWH from the three studies including six loci known in the general population and five novel loci associated with D-dimer and IL-6 levels. These findings support the hypothesis that host genetics may partially contribute to chronic inflammation in PLWH and help to identify potential targets for intervention of serious non-AIDS complications.
Collapse
Affiliation(s)
- Brad T. Sherman
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick
| | - Xiaojun Hu
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick
| | - Kanal Singh
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Lillian Haine
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Adam W. Rupert
- AIDS Monitoring Laboratory, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - James D. Neaton
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Jens D. Lundgren
- Centre of Excellence for Health, Immunity and Infections, Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Denmark
| | - Tomozumi Imamichi
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick
| | - Weizhong Chang
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick
| | - H. Clifford Lane
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| |
Collapse
|
77
|
Abstract
Atherosclerotic cardiovascular disease (ASCVD) proceeds through a series of stages: initiation, progression (or regression), and complications. By integrating known biology regarding molecular signatures of each stage with recent advances in high-dimensional molecular data acquisition platforms (to assay the genome, epigenome, transcriptome, proteome, metabolome, and gut microbiome), snapshots of each phase of atherosclerotic cardiovascular disease development can be captured. In this review, we will summarize emerging approaches for assessment of atherosclerotic cardiovascular disease risk in humans using peripheral blood molecular signatures and molecular imaging approaches. We will then discuss the potential (and challenges) for these snapshots to be integrated into a personalized movie providing dynamic readouts of an individual's atherosclerotic cardiovascular disease risk status throughout the life course.
Collapse
Affiliation(s)
- Matthew Nayor
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Kemar J. Brown
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Ramachandran S. Vasan
- Sections of Preventive Medicine & Epidemiology, and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA; Department of Epidemiology, Boston University School of Public Health; Boston University Center for Computing and Data Sciences
| |
Collapse
|
78
|
Stein JH, Kime N, Korcarz CE, Ribaudo H, Currier JS, Delaney JC. Effects of HIV Infection on Arterial Endothelial Function: Results From a Large Pooled Cohort Analysis. Arterioscler Thromb Vasc Biol 2021; 41:512-522. [PMID: 33327750 PMCID: PMC7770018 DOI: 10.1161/atvbaha.120.315435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/26/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To determine the effects of HIV serostatus and disease severity on endothelial function in a large pooled cohort study of people living with HIV infection and HIV- controls. Approach and Results: We used participant-level data from 9 studies: 7 included people living with HIV (2 treatment-naïve) and 4 had HIV- controls. Brachial artery flow-mediated dilation (FMD) was measured using a standardized ultrasound imaging protocol with central reading. After data harmonization, multiple linear regression was used to examine the effects of HIV- serostatus, HIV disease severity measures, and cardiovascular disease risk factors on FMD. Of 2533 participants, 986 were people living with HIV (mean 44.4 [SD 11.8] years old) and 1547 were HIV- controls (42.9 [12.2] years old). The strongest and most consistent associates of FMD were brachial artery diameter, age, sex, and body mass index. The effect of HIV+ serostatus on FMD was strongly influenced by kidney function. In the highest tertile of creatinine (1.0 mg/dL), the effect of HIV+ serostatus was strong (β=-1.59% [95% CI, -2.58% to -0.60%], P=0.002), even after covariate adjustment (β=-1.36% [95% CI, -2.46% to -0.47%], P=0.003). In the lowest tertile (0.8 mg/dL), the effect of HIV+ serostatus was strong (β=-1.90% [95% CI, -2.58% to -1.21%], P<0.001), but disappeared after covariate adjustment. HIV RNA viremia, CD4+ T-cell count, and use of antiretroviral therapy were not meaningfully associated with FMD. CONCLUSIONS The significant effect of HIV+ serostatus on FMD suggests that people living with HIV are at increased cardiovascular disease risk, especially if they have kidney disease.
Collapse
Affiliation(s)
- James H. Stein
- University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Noah Kime
- University of Washington Collaborative Health Studies Coordinating Center, Seattle, WA
| | - Claudia E. Korcarz
- University of Wisconsin School of Medicine and Public Health; Madison, WI
| | | | - Judith S. Currier
- David Geffen School of Medicine at University of California -Los Angeles; Los Angeles, CA
| | - Joseph C. Delaney
- University of Washington Collaborative Health Studies Coordinating Center, Seattle, WA
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba; Winnipeg, MB
| |
Collapse
|
79
|
Markozannes G, Koutsioumpa C, Cividini S, Monori G, Tsilidis KK, Kretsavos N, Theodoratou E, Gill D, Ioannidis JP, Tzoulaki I. Global assessment of C-reactive protein and health-related outcomes: an umbrella review of evidence from observational studies and Mendelian randomization studies. Eur J Epidemiol 2021; 36:11-36. [PMID: 32978716 PMCID: PMC7847446 DOI: 10.1007/s10654-020-00681-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
C-reactive protein (CRP) has been studied extensively for association with a large number of non-infectious diseases and outcomes. We aimed to evaluate the breadth and validity of associations between CRP and non-infectious, chronic health outcomes and biomarkers. We conducted an umbrella review of systematic reviews and meta-analyses and a systematic review of Mendelian randomization (MR) studies. PubMed, Scopus, and Cochrane Database of Systematic Reviews were systematically searched from inception up to March 2019. Meta-analyses of observational studies and MR studies examining associations between CRP and health outcomes were identified, excluding studies on the diagnostic value of CRP for infections. We found 113 meta-analytic comparisons of observational studies and 196 MR analyses, covering a wide range of outcomes. The overwhelming majority of the meta-analyses of observational studies reported a nominally statistically significant result (95/113, 84.1%); however, the majority of the meta-analyses displayed substantial heterogeneity (47.8%), small study effects (39.8%) or excess significance (41.6%). Only two outcomes, cardiovascular mortality and venous thromboembolism, showed convincing evidence of association with CRP levels. When examining the MR literature, we found MR studies for 53/113 outcomes examined in the observational study meta-analyses but substantial support for a causal association with CRP was not observed for any phenotype. Despite the striking amount of research on CRP, convincing evidence for associations and causal effects is remarkably limited.
Collapse
Affiliation(s)
- Georgios Markozannes
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110, Ioannina, Greece
| | - Charalampia Koutsioumpa
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110, Ioannina, Greece
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- BBS Program, Harvard Medical School, 220 Longwood Avenue, Boston, MA, 02115, USA
| | - Sofia Cividini
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Grace Monori
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Nikolaos Kretsavos
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110, Ioannina, Greece
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - John Pa Ioannidis
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Biomedical Data Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, CA, 94305, USA
- Meta-Research Innovation Center at Stanford (METRICS), Stanford, CA, 94305, USA
| | - Ioanna Tzoulaki
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110, Ioannina, Greece.
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
80
|
Reynoso-Villalpando GL, Casillas-Muñoz FA, Padilla-Gutiérrez JR, Sevillano-Collantes C, Moreno-Ruiz I, Del Cañizo-Gómez FJ, Valdez-Haro A, Martínez-Fernández DE, Valle Y. The Genetic Variants -717T>C (rs2794521), 1444G>A (rs1130864), and 1846 C > T (rs1205) of CRP Gene, Their Haplotypes, and Their Association with Serum CRP Levels, Acute Coronary Syndrome, and Diabetes in Patients from Western Mexico. Metab Syndr Relat Disord 2020; 19:127-136. [PMID: 33296257 DOI: 10.1089/met.2020.0080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background: C-reactive protein (CRP) is involved in inflammatory pathways that are associated with the onset and progression of type 2 diabetes mellitus (T2DM) as well as an increased risk of an acute coronary syndrome (ACS). This research aimed to evaluate the potential association of the genetic variants -717T>C, 1444G>A, and 1846 C > T of CRP gene on CRP levels, ACS, and T2DM in participants from Western Mexico. Methods: Six hundred three participants were studied: (1) control group (CG); (2) ACS participants classified as unstable angina (UA), myocardial infarction without ST-segment elevation (NSTEMI), and myocardial infarction with ST-segment elevation (STEMI); (3) T2DM Participants; and (4) ACS plus T2DM participants (ACS+T2DM). Genetic variants were genotyped using allelic discrimination with TaqMan® probes, and high-sensitivity CRP (hs-CRP) was measured by Turbidimetry. Results: TAC haplotype frequency was significantly higher in ACS+T2DM versus CG and versus ACS participants (odds ratio [OR] = 2.774, P = 0.017 and OR = 3.479, P = 0.020, respectively). hs-CRP levels were especially higher for ACS and for ACS+T2DM participants with respect to CG and T2DM (with P < 0.0001). We observed higher hs-CRP levels in NSTEMI and STEMI versus UA in ACS scenario (P = 0.001, P = 0.027, respectively) and for ACS+T2DM scenario (P = 0.0001, P = 0.002, respectively). Conclusion: hs-CRP level fluctuations are related to the presence of T2DM and the presence and severity of ACS. Very high levels (>10 mg/L) are a risk marker of cardiovascular complications. Our results demonstrate a possible relationship between TAC haplotype and an increased risk for T2DM and ACS.
Collapse
Affiliation(s)
- Gabriela Lizet Reynoso-Villalpando
- Departamento de Clínicas Médicas, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Colonia Independencia, Guadalajara, México.,Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Guadalajara, México
| | - Fidel Antonio Casillas-Muñoz
- Departamento de Clínicas Médicas, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Colonia Independencia, Guadalajara, México.,Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Guadalajara, México
| | - Jorge Ramón Padilla-Gutiérrez
- Departamento de Clínicas Médicas, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Colonia Independencia, Guadalajara, México
| | - Cristina Sevillano-Collantes
- Sección de Endocrinología y Nutrición, Hospital Universitario Infanta Leonor, Facultad de Medicina, Universidad Complutense, Madrid, España
| | - Inmaculada Moreno-Ruiz
- Sección de Endocrinología y Nutrición, Hospital Universitario Infanta Leonor, Facultad de Medicina, Universidad Complutense, Madrid, España
| | - Francisco Javier Del Cañizo-Gómez
- Sección de Endocrinología y Nutrición, Hospital Universitario Infanta Leonor, Facultad de Medicina, Universidad Complutense, Madrid, España
| | - Angélica Valdez-Haro
- Hospital Infantil del Estado de Sonora, Departamento de Enseñanza y Calidad, Hermosillo, Sonora, Mexico
| | | | - Yeminia Valle
- Departamento de Clínicas Médicas, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Colonia Independencia, Guadalajara, México
| |
Collapse
|
81
|
Shattuck-Heidorn H, Eick GN, Kramer KL, Sugiyama LS, Snodgrass JJ, Ellison PT. Variability of C-reactive protein in first-generation Ecuadorian immigrants living in the United States. Am J Hum Biol 2020; 33:e23547. [PMID: 33289200 DOI: 10.1002/ajhb.23547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES Establish the variability of C-reactive protein (CRP) within a population of first-generation immigrants living in the United States. Prior work has theorized that individuals with high levels of childhood pathogen exposure may have lower CRP levels in adulthood, and therefore that for these individuals, CRP may not be as accurate an index of chronic disease risk related to low-level inflammation as is presumed based on data from wealthy populations. This potentially has major implications for the interpretation of CRP as a biomarker of chronic inflammation. METHODS This longitudinal study collected a total of 125 dried blood spot (DBS) samples from 31 participants (median 4 samples each) and CRP levels in these DBS were assayed by enzyme-linked immunosorbant assay. Surveys were administered to characterize childhood pathogen exposure, and current illness. Variance was estimated using mixed effects regression models. RESULTS On average, participants were adults (mean = 41.9 years old) who had immigrated to the United States nearly 20 years prior to the study and had nearly universally experienced childhood helminth infection and other major pathogen exposures. Median serum-equivalent CRP was 0.77 mg/L. Individuals reliably differed in subacute CRP levels, and, depending on whether untransformed or log-transformed CRP was the outcome variable, 45% or 62% of variance in CRP was attributable to between-individual differences. CONCLUSIONS The variability of CRP levels in individuals with relatively high childhood pathogen exposure is comparable to previously reported studies in North America and Europe. However, CRP values are relatively low. CRP is an appropriate measure of subacute inflammation in this sample.
Collapse
Affiliation(s)
- Heather Shattuck-Heidorn
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
- Women and Gender Studies, University of Southern Maine, Portland, Maine, USA
| | - Geeta N Eick
- Department of Anthropology, University of Oregon, Eugene, Oregon, USA
| | - Karen L Kramer
- Department of Anthropology, University of Utah, Salt Lake City, Utah, USA
| | | | | | - Peter T Ellison
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
82
|
Dekker M, Waissi F, Timmerman N, Silvis MJM, Timmers L, de Kleijn DPV. Extracellular Vesicles in Diagnosing Chronic Coronary Syndromes the Bumpy Road to Clinical Implementation. Int J Mol Sci 2020; 21:E9128. [PMID: 33266227 PMCID: PMC7729611 DOI: 10.3390/ijms21239128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 12/15/2022] Open
Abstract
Coronary artery disease (CAD), comprising both acute coronary syndromes (ACS) and chronic coronary syndromes (CCS), remains one of the most important killers throughout the entire world. ACS is often quickly diagnosed by either deviation on an electrocardiogram or elevated levels of troponin, but CCS appears to be more complicated. The most used noninvasive strategies to diagnose CCS are coronary computed tomography and perfusion imaging. Although both show reasonable accuracy (80-90%), these modalities are becoming more and more subject of debate due to costs, radiation and increasing inappropriate use in low-risk patients. A reliable, blood-based biomarker is not available for CCS but would be of great clinical importance. Extracellular vesicles (EVs) are lipid-bilayer membrane vesicles containing bioactive contents e.g., proteins, lipids and nucleic acids. EVs are often referred to as the "liquid biopsy" since their contents reflect changes in the condition of the cell they originate from. Although EVs are studied extensively for their role as biomarkers in the cardiovascular field during the last decade, they are still not incorporated into clinical practice in this field. This review provides an overview on EV biomarkers in CCS and discusses the clinical and technological aspects important for successful clinical application of EVs.
Collapse
Affiliation(s)
- Mirthe Dekker
- Department of Vascular Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.D.); (F.W.); (N.T.)
- Department of Cardiology, Amsterdam University Medical Centre, Mijbergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Farahnaz Waissi
- Department of Vascular Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.D.); (F.W.); (N.T.)
- Department of Cardiology, Amsterdam University Medical Centre, Mijbergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Nathalie Timmerman
- Department of Vascular Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.D.); (F.W.); (N.T.)
| | - Max J. M. Silvis
- Department of Cardiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Leo Timmers
- Department of Cardiology, St. Antonius Hospital Nieuwegein, 3435 CM Nieuwegein, The Netherlands;
| | - Dominique P. V. de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (M.D.); (F.W.); (N.T.)
- Netherlands Heart Institute, 3511 EP Utrecht, The Netherlands
| |
Collapse
|
83
|
Abstract
Statins, ezetimibe, and PCSK9 inhibitors are currently the standard of care for the prevention and treatment of coronary artery disease. Despite their widespread use, coronary artery disease remains the leading cause of death worldwide, a fact that pleads for the development of new protective therapies. In no small part due to advances in the field of human genetics, many new therapies targeting various lipid traits or inflammation have recently received approval from regulatory agencies such as the US Food and Drug Administration or fared favorably in clinical trials. This wave of new therapies promises to transform the care of patients at risk for life-threatening coronary events.
Collapse
Affiliation(s)
- Kiran Musunuru
- Cardiovascular Institute, Department of Medicine, Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
84
|
Alfaddagh A, Martin SS, Leucker TM, Michos ED, Blaha MJ, Lowenstein CJ, Jones SR, Toth PP. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am J Prev Cardiol 2020; 4:100130. [PMID: 34327481 PMCID: PMC8315628 DOI: 10.1016/j.ajpc.2020.100130] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation constitutes a complex, highly conserved cascade of molecular and cellular events. Inflammation has been labeled as “the fire within,” is highly regulated, and is critical to host defense and tissue repair. In general, inflammation is beneficial and has evolved to promote survival. However, inflammation can also be maladaptive when chronically activated and sustained, leading to progressive tissue injury and reduced survival. Examples of a maladaptive response include rheumatologic disease and atherosclerosis. Despite evidence gathered by Virchow over 100 years ago showing that inflammatory white cells play a role in atherogenesis, atherosclerosis was until recently viewed as a disease of passive cholesterol accumulation in the subendothelial space. This view has been supplanted by considerable basic scientific and clinical evidence demonstrating that every step of atherogenesis, from the development of endothelial cell dysfunction to foam cell formation, plaque formation and progression, and ultimately plaque rupture stemming from architectural instability, is driven by the cytokines, interleukins, and cellular constituents of the inflammatory response. Herein we provide an overview of the role of inflammation in atherosclerotic cardiovascular disease, discuss the predictive value of various biomarkers involved in inflammation, and summarize recent clinical trials that evaluated the capacity of various pharmacologic interventions to attenuate the intensity of inflammation and impact risk for acute cardiovascular events.
Collapse
Affiliation(s)
- Abdulhamied Alfaddagh
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth S Martin
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten M Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erin D Michos
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles J Lowenstein
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven R Jones
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter P Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
85
|
Shakour N, Ruscica M, Hadizadeh F, Cirtori C, Banach M, Jamialahmadi T, Sahebkar A. Statins and C-reactive protein: in silico evidence on direct interaction. Arch Med Sci 2020; 16:1432-1439. [PMID: 33224343 PMCID: PMC7667423 DOI: 10.5114/aoms.2020.100304] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Statins are known to lower CRP, and this reduction has been suggested to contribute to the established efficacy of these drugs in reducing cardiovascular events and outcomes. However, the exact mechanism underlying the CRP-lowering effect of statins remains elusive. METHODS In order to test the possibility of direct interaction, we performed an in silico study by testing the orientation of the respective ligands (statins) and phosphorylcholine (the standard ligand of CRP) in the CRP active site using Molecular Operating Environment (MOE) software. RESULTS Docking experiments showed that all statins could directly interact with CRP. Among statins, rosuvastatin had the strongest interaction with CRP (pKi = 16.14), followed by fluvastatin (pKi = 15.58), pitavastatin (pKi = 15.26), atorvastatin (pKi = 14.68), pravastatin (pKi = 13.95), simvastatin (pKi = 7.98) and lovastatin (pKi = 7.10). According to the above-mentioned results, rosuvastatin, fluvastatin, pitavastatin and atorvastatin were found to have stronger binding to CRP compared with the standard ligand phosphocholine (pKi = 14.55). CONCLUSIONS This finding suggests a new mechanism of interaction between statins and CRP that could be independent of the putative cholesterol-lowering activity of statins.
Collapse
Affiliation(s)
- Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Farzin Hadizadeh
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Cesare Cirtori
- Centro Dislipidemie, A.S.S.T. Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital, Medical University of Lodz, Lodz, Poland
- Polish Mother’s Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Halal Research Center of IRI, FDA, Tehran, Iran
| |
Collapse
|
86
|
Napoli R, Ruvolo A, Triggianese P, Prevete N, Schiattarella GG, Nigro C, Miele C, Magliulo F, Grassi S, Pecoraro A, Cittadini A, Esposito G, de Paulis A, Spadaro G. Immunoglobulins G modulate endothelial function and affect insulin sensitivity in humans. Nutr Metab Cardiovasc Dis 2020; 30:2085-2092. [PMID: 32807637 DOI: 10.1016/j.numecd.2020.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/12/2020] [Accepted: 07/01/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND AIMS Data from animals suggest that immunoglobulins G (IgG) play a mechanistic role in atherosclerosis and diabetes through endothelial dysfunction and insulin resistance. Patients with common variable immunodeficiency (CVID), who have low circulating levels of IgG and are treated with intravenous polyclonal IgG (IVIgG), may provide an ideal model to clarify whether circulating IgG modulate endothelial function and affect insulin sensitivity in humans. METHODS AND RESULTS We studied 24 patients with CVID and 17 matched healthy controls (HC). Endothelial function was evaluated as flow mediated dilation (FMD) of the brachial artery at baseline and 1, 7, 14, and 21 days after IVIgG infusion in the CVID patients. We measured also plasma glucose, insulin, and calculated the HOMA-IR index. We also investigated the role of human IgG on the production of Nitric Oxide (NO) in vitro in Human Coronary Artery Endothelial Cells (HCAEC). Compared to HC, FMD of CVID patients was significantly impaired at baseline (9.4 ± 0.9 and 7.6 ± 0.6% respectively, p < 0.05) but rose above normal levels 1 and 7 days after IVIgG infusion to return at baseline at 14 and 21 days. Serum insulin concentration and HOMA-IR index dropped by 50% in CVID patients after IVIgG (p < 0.002 vs. baseline). In vitro IgG stimulated NO production in HCAEC. CONCLUSIONS Reduced IgG levels are associated with endothelial dysfunction and IVIgG stimulates endothelial function directly while improving insulin sensitivity. The current findings may suggest an anti-atherogenic role of human IgG.
Collapse
Affiliation(s)
- Raffaele Napoli
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy.
| | - Antonio Ruvolo
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Paola Triggianese
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Nella Prevete
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Gabriele G Schiattarella
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Cecilia Nigro
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Claudia Miele
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Fabio Magliulo
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Simona Grassi
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Antonio Cittadini
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, Federico II University School of Medicine, Naples, Italy
| |
Collapse
|
87
|
Li C, Wang Y, Zhang Q, Wang L, Li K, Yang X. Incorporating the erythrocyte sedimentation rate for enhanced accuracy of the global registry of acute coronary event score in patients with ST-segment elevated myocardial infarction: A retrospective cohort study. Medicine (Baltimore) 2020; 99:e22523. [PMID: 33031294 PMCID: PMC7544386 DOI: 10.1097/md.0000000000022523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
There is scarce evidence that the erythrocyte sedimentation rate (ESR) could efficiently improve the prediction accuracy of the Global Registry of Acute Coronary Events (GRACE) risk score in cases of ST-elevation myocardial infarction (STEMI).A cohort of 1094 STEMI patients undergoing primary percutaneous coronary intervention was retrospectively recruited. Patients were categorized based on the ESR values. Final endpoints included cardiovascular death and major adverse cardiovascular event (MACE) occurrence. The predictive value of combined models with the GRACE score and ESR was assessed by receiver operating characteristic (ROC) analysis, net reclassification improvement (NRI), and integrated discrimination improvement.During the mean follow-up of 23 months, 34 patients died and 190 experienced MACEs, of which 23 patients died in the first year; both endpoints were more frequent in the higher group. The ESR and high-sensitivity C-reactive protein (hs-CRP) were independent risk factors of 1-year cardiovascular death, together with the GRACE score (ESR: hazard ratio = 1.03, P = .006 hs-CRP: hazard ratio = 1.00, P = .001; GRACE: 1.03, P = .012). Although no statistical improvement in the area under the ROC curve was observed in either the GRACE/ESR or the GRACE/hs-CRP model (GRACE/ESR models: 0. 8073 vs GRACE: 0.7714, P = .22; GRACE/ESR models: 0. 7815 vs GRACE: 0.7714, P = .61), the GRACE score and ESR together significantly improved the NRI (0.633; P< .001) compared with the GRACE alone. Regarding the mid-term mortality, adding the ESR to the GRACE score not only improved the NRI (0.8433; P < .001), but also increased the integrated discrimination improvement (0.0509; P = .04).The ESR is an independent risk factor of cardiovascular death and MACE in STEMI patients receiving primary percutaneous coronary intervention. The ESR comparatively enhanced the predictive values of the prognostic model, including the GRACE risk score.
Collapse
|
88
|
Liu S, Zhao W, Liu X, Cheng L. Metagenomic analysis of the gut microbiome in atherosclerosis patients identify cross-cohort microbial signatures and potential therapeutic target. FASEB J 2020; 34:14166-14181. [PMID: 32939880 DOI: 10.1096/fj.202000622r] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022]
Abstract
The gut microbiota is associated with cardiovascular diseases, including atherosclerosis. However, the composition, functional capacity, and metabolites of the gut microbiome about atherosclerosis have not been comprehensively studied. Here, we reanalyzed 25 metagenomic stool samples from Sweden and 385 metagenomic stool samples from China using HUMAnN2, PanPhlAn, and MelonnPan to obtain more sufficient information. We found that the samples from atherosclerotic patients in both cohorts were depleted in Bacteroides xylanisolvens, Odoribacter splanchnicus, Eubacterium eligens, Roseburia inulinivorans, and Roseburia intestinalis. At the functional level, healthy metagenomes were both enriched in pathways of starch degradation V, glycolysis III (from glucose), CDP-diacylglycerol biosynthesis, and folate transformations. R inulinivorans and R intestinalis are major contributors to starch degradation V, while E eligens greatly contribute to the pathway CDP-diacylglycerol biosynthesis, and B xylanisolvens and B uniformis contribute to folate transformations II. The 11 marker species selected from the Chinese cohort distinguish patients from controls with an area under the receiver operating characteristics curve (AUC) of 0.86. Strain-level microbial analysis revealed a geographically associated adaptation of the strains from E eligens, B uniformis, and E coli. Two gut microbial metabolites, nicotinic acid and hydrocinnamic acid, had significantly higher predicted abundance in the control samples compared to the patients in the Chinese cohort, and interestinglynicotinic acid is already an effective lipid-lowering drug to reducing cardiovascular risk. Our results indicate intestinal bacteria such as B xylanisolvens, E eligens, and R inulinivorans could be promising probiotics and potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Sheng Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Wenjing Zhao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xueyan Liu
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Lixin Cheng
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, China
| |
Collapse
|
89
|
Rajab IM, Hart PC, Potempa LA. How C-Reactive Protein Structural Isoforms With Distinctive Bioactivities Affect Disease Progression. Front Immunol 2020; 11:2126. [PMID: 33013897 PMCID: PMC7511658 DOI: 10.3389/fimmu.2020.02126] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/05/2020] [Indexed: 12/22/2022] Open
Abstract
C-reactive protein (CRP) is a widely known, hepatically synthesized protein whose blood levels change rapidly and pronouncedly in response to any tissue damaging event associated with an inflammatory response. The synthesis and secretion of CRP is stimulated by interleukin-6, an early pleiotropic cytokine released by macrophages, endothelial, and other cells that are activated when localized normal tissue structures are compromised by trauma or disease. Serum CRP levels can change rapidly and robustly from 10-100-fold within 6–72 h of any tissue damaging event. Elevated blood levels correlate with the onset and extent of both activated inflammation and the acute phase biochemical response to the tissue insult. Because its functional bioactivity as the prototypic acute phase reactant has eluded clear definition for decades, diagnosticians of various conditions and diseases use CRP blood levels as a simple index for ongoing inflammation. In many pathologies, which involves many different tissues, stages of disease, treatments, and responses to treatments, its interpretive diagnostic value requires a deeper understanding of the localized tissue processes and events that contribute signals which regulate protective or pathological host defense bioactivities. This report presents concepts that describe how local tissue activation events can lead to a non-proteolytic, conformational rearrangement of CRP into a unique isoform with distinctive solubility, antigenicity, binding reactivities and bioactivities from that protein widely known and measured in serum. By describing factors that control the expression, tissue localization, half-life and pro-inflammatory amplification activity of this CRP isoform, a unifying explanation for the diagnostic significance of CRP measurement in disease is advanced.
Collapse
Affiliation(s)
- Ibraheem M Rajab
- Roosevelt University College of Pharmacy, Schaumburg, IL, United States
| | - Peter C Hart
- Roosevelt University College of Pharmacy, Schaumburg, IL, United States
| | | |
Collapse
|
90
|
Determinants of placental leptin receptor gene expression and association with measures at birth. Placenta 2020; 100:89-95. [PMID: 32891006 DOI: 10.1016/j.placenta.2020.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/04/2020] [Accepted: 08/10/2020] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The leptin signalling pathway is important in metabolic health during pregnancy. However, few studies have investigated the determinants and extent of leptin receptor gene (LEPR) expression in the placenta, nor the relationship with infant health in early life. Here, we investigate the genetic and maternal in utero determinants of placental LEPR expression, and whether this expression is linked to anthropometric and inflammatory measures at birth in healthy newborns in the Barwon Infant Study. METHODS Placental LEPR expression was measured using RT-qPCR (n = 854 placentae). Associations between genetic variation in LEPR, maternal in utero factors, measures at birth and placental LEPR expression were assessed using multivariable linear regression modelling. RESULTS We found that the genotype at two intronic SNPs, rs9436301 and rs9436746, was independently associated with placental LEPR expression. Maternal pre-pregnancy body mass index, gestational diabetes mellitus, weight gain and smoking in pregnancy were not associated with LEPR expression. Placental LEPR expression was negatively associated with high sensitivity C-Reactive Protein in umbilical cord blood, which persisted after adjustment for potential confounders. DISCUSSION Overall, our findings suggest that genetic variation in LEPR plays a key role in regulating placental LEPR expression, which is in turn is associated with inflammatory markers in cord blood at birth. Further studies encompassing other aspects of leptin signalling are warranted to understand if these relationships are causal and have health implications.
Collapse
|
91
|
Orlandi M, Graziani F, D'Aiuto F. Periodontal therapy and cardiovascular risk. Periodontol 2000 2020; 83:107-124. [PMID: 32385887 DOI: 10.1111/prd.12299] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiovascular diseases are the worldwide leading cause of mortality. Cardiovascular diseases are noncommunicable conditions with a complex pathogenesis, and their clinical manifestations include major cardiovascular events such as myocardial infarction and stroke. Epidemiologic evidence suggests a consistent association between periodontitis and increased risk of cardiovascular diseases. Some evidence supports a beneficial effect of the treatment of periodontitis on both surrogate and hard cardiovascular outcomes. This narrative review has been conducted as an update of the most recent evidence on the effects of periodontitis treatment on cardiovascular outcomes since the last commissioned review of the European Federation of Periodontology-American Academy of Periodontology World Workshop in 2012. Newer evidence originating from published randomized controlled trials confirms a positive effect of periodontal treatment on surrogate measures of cardiovascular diseases, whereas there have been no randomized controlled trials investigating the effect of periodontal treatment on the incidence of cardiovascular disease events such as myocardial infarction and stroke. In conclusion, there is sufficient evidence from observational and experimental studies on surrogate cardiovascular measures to justify the design and conduct of appropriately powered randomized controlled trials investigating the effect of effective periodontal interventions on cardiovascular disease outcomes (ie, myocardial infarction and stroke) with adequate control of traditional cardiovascular risk factors.
Collapse
Affiliation(s)
- Marco Orlandi
- Periodontology Unit, UCL Eastman Dental Institute, London, UK
| | - Filippo Graziani
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy.,Sub-Unit of Periodontology, Halitosis and Periodontal Medicine, University Hospital of Pisa, Pisa, Italy
| | | |
Collapse
|
92
|
Khandaker GM, Zuber V, Rees JMB, Carvalho L, Mason AM, Foley CN, Gkatzionis A, Jones PB, Burgess S. Shared mechanisms between coronary heart disease and depression: findings from a large UK general population-based cohort. Mol Psychiatry 2020; 25:1477-1486. [PMID: 30886334 PMCID: PMC7303009 DOI: 10.1038/s41380-019-0395-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/29/2019] [Accepted: 02/14/2019] [Indexed: 12/15/2022]
Abstract
While comorbidity between coronary heart disease (CHD) and depression is evident, it is unclear whether the two diseases have shared underlying mechanisms. We performed a range of analyses in 367,703 unrelated middle-aged participants of European ancestry from UK Biobank, a population-based cohort study, to assess whether comorbidity is primarily due to genetic or environmental factors, and to test whether cardiovascular risk factors and CHD are likely to be causally related to depression using Mendelian randomization. We showed family history of heart disease was associated with a 20% increase in depression risk (95% confidence interval [CI] 16-24%, p < 0.0001), but a genetic risk score that is strongly associated with CHD risk was not associated with depression. An increase of 1 standard deviation in the CHD genetic risk score was associated with 71% higher CHD risk, but 1% higher depression risk (95% CI 0-3%; p = 0.11). Mendelian randomization analyses suggested that triglycerides, interleukin-6 (IL-6), and C-reactive protein (CRP) are likely causal risk factors for depression. The odds ratio for depression per standard deviation increase in genetically-predicted triglycerides was 1.18 (95% CI 1.09-1.27; p = 2 × 10-5); per unit increase in genetically-predicted log-transformed IL-6 was 0.74 (95% CI 0.62-0.89; p = 0.0012); and per unit increase in genetically-predicted log-transformed CRP was 1.18 (95% CI 1.07-1.29; p = 0.0009). Our analyses suggest that comorbidity between depression and CHD arises largely from shared environmental factors. IL-6, CRP and triglycerides are likely to be causally linked with depression, so could be targets for treatment and prevention of depression.
Collapse
Affiliation(s)
- Golam M Khandaker
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Verena Zuber
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Jessica M B Rees
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Livia Carvalho
- Department of Clinical Pharmacology, Queen Mary University of London, London, UK
| | - Amy M Mason
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | | | - Peter B Jones
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK.
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|
93
|
Ruscica M, Corsini A, Ferri N, Banach M, Sirtori CR. Clinical approach to the inflammatory etiology of cardiovascular diseases. Pharmacol Res 2020; 159:104916. [PMID: 32445957 PMCID: PMC7238995 DOI: 10.1016/j.phrs.2020.104916] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023]
Abstract
Inflammation is an obligatory marker of arterial disease, both stemming from the inflammatory activity of cholesterol itself and from well-established molecular mechanisms. Raised progenitor cell recruitment after major events and clonal hematopoiesis related mechanisms have provided an improved understanding of factors regulating inflammatory phenomena. Trials with inflammation antagonists have led to an extensive evaluation of biomarkers such as the high sensitivity C reactive protein (hsCRP), not exerting a causative role, but frequently indicative of the individual cardiovascular (CV) risk. Aim of this review is to provide indication on the anti-inflammatory profile of agents of general use in CV prevention, i.e. affecting lipids, blood pressure, diabetes as well nutraceuticals such as n-3 fatty acids. A crucial issue in the evaluation of the benefit of the anti-inflammatory activity is the frequent discordance between a beneficial activity on a major risk factor and associated changes of hsCRP, as in the case of statins vs PCSK9 antagonists. In hypertension, angiotensin converting enzyme inhibitors exert an optimal anti-inflammatory activity, vs the case of sartans. The remarkable preventive activity of SLGT-2 inhibitors in heart failure is not associated with a clear anti-inflammatory mechanism. Finally, icosapent ethyl has been shown to reduce the CV risk in hypertriglyceridemia, with a 27 % reduction of hsCRP. The inflammation-based approach to arterial disease has considerably gained from an improved understanding of the clinical diagnostic strategy and from a better knowledge on the mode of action of numerous agents, including nutraceuticals.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy; Multimedica IRCCS, Milano, Italy
| | - Nicola Ferri
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padua, Italy
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland.
| | - Cesare R Sirtori
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
94
|
Gan X, Gong T, Zheng Y, Gopinath SCB, Zhao K. Electroimmunodetection of cardiac C-reactive protein for determining myocardial Injury. Biotechnol Appl Biochem 2020; 68:272-278. [PMID: 32275089 DOI: 10.1002/bab.1921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/03/2020] [Indexed: 11/07/2022]
Abstract
C-reactive protein (CRP) is an acute phase reactant to be a marker of inflammation and has been correlated with the cardiac injury. An immunoassay was performed using anti-human CRP antibody on an InterDigitated electrode (IDE) sensor to determine and specify CRP concentration for diagnosing the condition of myocardial inflammation. To promote the detection, gold nanoparticle (GNP) was seeded on the aminated-IDE surface. Anti-CRP was hitched on the GNP-seeded surface and identified the abundance of CRP. The limit of quantification was found as 100 fM, and the higher current response was noticed by increasing CRP concentrations with the sensitivity at 1 pM. Furthermore, CRP-spiked human serum did not interfere the determination of CRP and increased the current response, indicating suitability for a real-life sample. Similarly, the control experiments with nonimmune antibody Troponin I are not showing the definite current responses, proving the selective identification of CRP. This method of diagnosing is needful to determine the cardiovascular injury at the right time.
Collapse
Affiliation(s)
- Xiaoya Gan
- Department of Cardiology, Shandong Provincial Taishan Hospital, Taian, Shandong Province, China
| | - Tao Gong
- Department of Critical Medicine, Pingyi County Hospital of Traditional Chinese Medicine, Pingyi County, Linyi, Shandong Province, China
| | - Yin Zheng
- Department of Cardiology, Hainan Cancer Hospital, Xiuying District, Haikou, Hainan, China.,Hainan Chengmei International Health Management Center, Xiuying District, Haikou, Hainan, China
| | - Subash C B Gopinath
- Universiti Malaysia Perlis, Institute of Nano Electronic Engineering, Kangar, Perlis, Malaysia.,School of Bioprocess Engineering, Universiti Malaysia Perlis, Arau, Perlis, Malaysia
| | - Ketong Zhao
- Department of Cardiology, Hainan Cancer Hospital, Xiuying District, Haikou, Hainan, China.,Hainan Chengmei International Health Management Center, Xiuying District, Haikou, Hainan, China
| |
Collapse
|
95
|
Hadrup N, Zhernovkov V, Jacobsen NR, Voss C, Strunz M, Ansari M, Schiller HB, Halappanavar S, Poulsen SS, Kholodenko B, Stoeger T, Saber AT, Vogel U. Acute Phase Response as a Biological Mechanism-of-Action of (Nano)particle-Induced Cardiovascular Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907476. [PMID: 32227434 DOI: 10.1002/smll.201907476] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 05/15/2023]
Abstract
Inhaled nanoparticles constitute a potential health hazard due to their size-dependent lung deposition and large surface to mass ratio. Exposure to high levels contributes to the risk of developing respiratory and cardiovascular diseases, as well as of lung cancer. Particle-induced acute phase response may be an important mechanism of action of particle-induced cardiovascular disease. Here, the authors review new important scientific evidence showing causal relationships between inhalation of particle and nanomaterials, induction of acute phase response, and risk of cardiovascular disease. Particle-induced acute phase response provides a means for risk assessment of particle-induced cardiovascular disease and underscores cardiovascular disease as an occupational disease.
Collapse
Affiliation(s)
- Niels Hadrup
- National Research Centre for the Working Environment, Copenhagen, DK-2100, Denmark
| | - Vadim Zhernovkov
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | | | - Carola Voss
- Comprehensive Pneumology Center (CPC)/Institute of Lung Biology and Disease (ILBD), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, D-85764, Germany
| | - Maximilian Strunz
- Comprehensive Pneumology Center (CPC)/Institute of Lung Biology and Disease (ILBD), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, D-85764, Germany
| | - Meshal Ansari
- Comprehensive Pneumology Center (CPC)/Institute of Lung Biology and Disease (ILBD), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, D-85764, Germany
| | - Herbert B Schiller
- Comprehensive Pneumology Center (CPC)/Institute of Lung Biology and Disease (ILBD), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, D-85764, Germany
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Sarah S Poulsen
- National Research Centre for the Working Environment, Copenhagen, DK-2100, Denmark
| | - Boris Kholodenko
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| | - Tobias Stoeger
- Comprehensive Pneumology Center (CPC)/Institute of Lung Biology and Disease (ILBD), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, D-85764, Germany
| | - Anne Thoustrup Saber
- National Research Centre for the Working Environment, Copenhagen, DK-2100, Denmark
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, DK-2100, Denmark
- DTU Health, Technical University of Denmark, Kgs. Lyngby, DK-2800, Denmark
| |
Collapse
|
96
|
Scarale MG, Copetti M, Garofolo M, Fontana A, Salvemini L, De Cosmo S, Lamacchia O, Penno G, Trischitta V, Menzaghi C. The Synergic Association of hs-CRP and Serum Amyloid P Component in Predicting All-Cause Mortality in Patients With Type 2 Diabetes. Diabetes Care 2020; 43:1025-1032. [PMID: 32144164 DOI: 10.2337/dc19-2489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/12/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 2 diabetes is characterized by increased death rate. In order to tackle this dramatic event, it becomes essential to discover novel biomarkers capable of identifying high-risk patients to be exposed to more aggressive preventive and treatment strategies. hs-CRP and serum amyloid P component (SAP) are two acute-phase inflammation proteins, which interact physically and share structural and functional features. We investigated their combined role in associating with and improving prediction of mortality in type 2 diabetes. RESEARCH DESIGN AND METHODS Four cohorts comprising 2,499 patients with diabetes (643 all-cause deaths) were analyzed. The improvement of mortality prediction was addressed using two well-established prediction models, namely, EstimatioN oF mORtality risk in type 2 diabetiC patiEnts (ENFORCE) and Risk Equations for Complications of Type 2 Diabetes (RECODe). RESULTS Both hs-CRP and SAP were independently associated with all-cause mortality (hazard ratios [HRs] [95% CIs]: 1.46 [1.34-1.58] [P < 0.001] and 0.82 [0.76-0.89] [P < 0.001], respectively). Patients with SAP ≤33 mg/L were at increased risk of death versus those with SAP >33 mg/L only if hs-CRP was relatively high (>2 mg/L) (HR 1.96 [95% CI 1.52-2.54] [P < 0.001] and 1.20 [0.91-1.57] [P = 0.20] in hs-CRP >2 and ≤2 mg/L subgroups, respectively; hs-CRP-by-SAP strata interaction P < 0.001). The addition of hs-CRP and SAP significantly (all P < 0.05) improved several discrimination and reclassification measures of both ENFORCE and RECODe all-cause mortality prediction models. CONCLUSIONS In type 2 diabetes, hs-CRP and SAP show opposite and synergic associations with all-cause mortality. The use of both markers, possibly in combination with others yet to be unraveled, might improve the ability to predict the risk of death in the real-life setting.
Collapse
Affiliation(s)
- Maria Giovanna Scarale
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics, Fondazione IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy
| | - Monia Garofolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Fontana
- Unit of Biostatistics, Fondazione IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy
| | - Lucia Salvemini
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy
| | - Salvatore De Cosmo
- Department of Clinical Sciences, Fondazione IRCCS "Casa Sollievo Della Sofferenza," San Giovanni Rotondo, Italy
| | - Olga Lamacchia
- Unit of Endocrinology and Diabetology, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Penno
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Trischitta
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy .,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Menzaghi
- Research Unit of Diabetes and Endocrine Diseases, Fondazione IRCCS "Casa Sollievo della Sofferenza," San Giovanni Rotondo, Italy
| |
Collapse
|
97
|
Çolak Y, Afzal S, Lange P, Nordestgaard BG. Smoking, Systemic Inflammation, and Airflow Limitation: A Mendelian Randomization Analysis of 98 085 Individuals From the General Population. Nicotine Tob Res 2020; 21:1036-1044. [PMID: 29688528 DOI: 10.1093/ntr/nty077] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 04/20/2018] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Smoking is associated with systemic and local inflammation in the lungs. Furthermore, in chronic obstructive pulmonary disease, which is often caused by smoking, there is often systemic inflammation that is linked to lung function impairment. However, the causal pathways linking smoking, systemic inflammation, and airflow limitation are still unknown. We tested whether higher tobacco consumption is associated with higher systemic inflammation, observationally and genetically and whether genetically higher systemic inflammation is associated with airflow limitation. METHODS We included 98 085 individuals aged 20-100 years from the Copenhagen General Population Study; 36589 were former smokers and 16172 were current smokers. CHRNA3 rs1051730 genotype was used as a proxy for higher tobacco consumption and the IL6R rs2228145 genotype was used for higher systemic inflammation. Airflow limitation was defined as forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) <70%. RESULTS Difference in plasma level of C-reactive protein was 4.8% (95% CI = 4.4% to 5.2%) per 10 pack-year increase and 1.6% (95% CI = 0.4% to 2.8%) per T allele. Corresponding differences were 1.2% (95% CI = 1.1% to 1.3%) and 0.5% (95% CI = 0.3% to 0.8%) for fibrinogen, 1.2% (95% CI = 1.2% to 1.3%) and 0.7% (95% CI = 0.5% to 1.0%) for α1-antitrypsin, 2.0% (95% CI = 1.8% to 2.1%) and 0.7% (95% CI = 0.4% to 1.1%) for leukocytes, 1.9% (95% CI = 1.8% to 2.1%) and 0.8% (95% CI = 0.4% to 1.2%) for neutrophils, and 0.8% (95% CI = 0.7% to 1.0%) and 0.4% (95% CI = 0.1% to 0.7%) for thrombocytes. The differences in these levels were lower for former smokers compared with current smokers. The IL6R rs2228145 genotype was associated with higher plasma acute-phase reactants but not with airflow limitation. Compared with the C/C genotype, the odds ratio for airflow limitation was 0.95 (95% CI = 0.89 to 1.02) for A/C genotype and 0.94 (95% CI = 0.87 to 1.01) for A/A genotype. CONCLUSIONS Higher tobacco consumption is associated with higher systemic inflammation both genetically and observationally, whereas systemic inflammation was not associated with airflow limitation genetically. IMPLICATIONS The association between higher tobacco consumption and higher systemic inflammation may be causal, and the association is stronger among current smokers compared to former smokers, indicating that smoking cessation may reduce the effects of smoking on systemic inflammation. Systemic inflammation does not seem to be a causal driver in development of airflow limitation. These findings can help to understand the pathogenic effects of smoking and the interplay between smoking, systemic inflammation, and airflow limitation and hence development and progression of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Yunus Çolak
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Shoaib Afzal
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Peter Lange
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Internal Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Department of Public Health, Section of Social Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
98
|
Systemic inflammation is associated with incident stroke and heart disease in East Asians. Sci Rep 2020; 10:5605. [PMID: 32221345 PMCID: PMC7101367 DOI: 10.1038/s41598-020-62391-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Systemic inflammation, reflected by increased plasma concentrations of C-reactive protein (CRP) and fibrinogen, is associated with increased risk of coronary heart disease, but its relevance for stroke types remains unclear. Moreover, evidence is limited in non-European populations. We investigated associations of CRP and fibrinogen with risks of incident major coronary events (MCE), ischemic stroke (IS) and intracerebral hemorrhage (ICH) in a cohort of Chinese adults. A nested case-control study within the prospective China Kadoorie Biobank included 1,508 incident MCE cases, 5,418 IS cases, 4,476 ICH cases, and 5,285 common controls, aged 30–79 years. High-sensitivity CRP and low-density lipoprotein cholesterol (LDL-C) were measured in baseline plasma samples from all participants, and fibrinogen in a subset (n = 9,380). Logistic regression yielded adjusted odds ratios (ORs) per SD higher usual levels of log-transformed CRP and fibrinogen. The overall mean (SD) baseline LDL-C was 91.6 mg/dL (24.0) and geometric mean (95% CI) CRP and fibrinogen were 0.90 mg/L (0.87–0.93) and 3.01 g/L (2.98–3.03), respectively. There were approximately log-linear positive associations of CRP with each outcome, which persisted after adjustment for LDL-C and other risk factors, with adjusted ORs (95% CI) per SD higher CRP of 1.67 (1.44–1.94) for MCE and 1.22 (1.10–1.36) for both IS and ICH. No associations of fibrinogen with MCE, IS, or ICH were identified. Adding CRP to prediction models based on established risk factors improved model fit for each of MCE, IS, and ICH, with small improvements in C-statistic and correct reclassification of controls to lower risk groups. Among Chinese adults, who have low mean LDL-C, CRP, but not fibrinogen, was independently associated with increased risks of MCE and stroke.
Collapse
|
99
|
Haybar H, Maleki Behzad M, Shahrabi S, Ansari N, Saki N. Expression of Blood Cells Associated CD Markers and Cardiovascular Diseases: Clinical Applications in Prognosis. Lab Med 2020; 51:122-142. [PMID: 31340048 DOI: 10.1093/labmed/lmz049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) are a major cause of mortality worldwide. The results of various studies have shown that abnormality in the frequency and function of blood cells can be involved in CVD complications. In this review, we have focused on abnormalities in the expression of the CD (cluster of differentiation) markers of blood cells to assess the association of these abnormalities with CVD prognosis. METHODS We identified the relevant literature through a PubMed search (1990-2018) of English-language articles using the terms "Cardiovascular diseases", "CD markers", "leukocytes", "platelets", and "endothelial cells". RESULTS There is a variety of mechanisms for the effect of CD-marker expressions on CVDs prognosis, ranging from proinflammatory processes to dysfunctional effects in blood cells. CONCLUSION Considering the possible effects of CD-marker expression on CVDs prognosis, particularly prognosis of acute myocardial infarction and atherosclerosis, long-term studies in large cohorts are required to identify the prognostic value of CD markers and to target them with appropriate therapeutic agents.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masumeh Maleki Behzad
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Ansari
- Isfahan Bone Metabolic Disorders Research Center, Department of Internal Medicine, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
100
|
Leite FRM, Nascimento GG, Peres KG, Demarco FF, Horta BL, Peres MA. Collider bias in the association of periodontitis and carotid intima‐media thickness. Community Dent Oral Epidemiol 2020; 48:264-270. [DOI: 10.1111/cdoe.12525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Fábio R. M. Leite
- Section of Periodontology Department of Dentistry and Oral Health Aarhus University Aarhus Denmark
| | - Gustavo G. Nascimento
- Section of Periodontology Department of Dentistry and Oral Health Aarhus University Aarhus Denmark
| | - Karen G. Peres
- School of Dentistry and Oral Health Griffith University, Gold Coast Campus Southport QLD Australia
| | - Flávio F. Demarco
- Graduate Program in Dentistry Federal University of Pelotas Pelotas Brazil
- Graduate Program in Epidemiology Federal University of Pelotas Pelotas Brazil
| | - Bernardo L. Horta
- Graduate Program in Epidemiology Federal University of Pelotas Pelotas Brazil
| | - Marco A. Peres
- School of Dentistry and Oral Health Griffith University, Gold Coast Campus Southport QLD Australia
- Menzies Health Institute Queensland Griffith University, Gold Coast Campus Southport QLD Australia
| |
Collapse
|