1
|
Finzi A, Ottoboni S, Cellini M, Corcioni B, Gaudiano C, Fontana L. Color Doppler Imaging, Endothelin-1, Corneal Biomechanics and Scleral Rigidity in Asymmetric Age-Related Macular Degeneration. Clin Ophthalmol 2024; 18:2583-2591. [PMID: 39281979 PMCID: PMC11401527 DOI: 10.2147/opth.s479225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose Age-related macular degeneration (AMD) presents a multifaceted etiopathogenesis involving ischemic, inflammatory, and genetic components. This study investigates the correlation between ocular hemodynamics, scleral rigidity (SR), and plasma endothelin-1 (ET1) levels in treatment-naive patients with asymmetrical AMD. Patients and Methods This study included 20 treatment-naive patients (12 females and 8 males) with an average age of 76.4 ± 3.7 years, who presented with AMD with neovascular membrane formation (nAMD) in one eye, and intermediate grade 2 AMD (iAMD) in the other eye. The control group consisted of 20 healthy subjects (13 females and 7 males) with a mean age of 74.7 ± 3.9 years. All patients and healthy controls underwent color Doppler imaging (i) of the ophthalmic artery (OA), short posterior ciliary arteries (SPCAs), and central retinal artery (CRA); Plasma ET-1 levels were measured for all patients and healthy subjects. Corneal biomechanics were assessed using an Ocular Response Analyzer and two indices were obtained: corneal hysteresis (CH) and corneal resistance factor (CRF). Results Results showed reduced blood flow velocities and increased resistance indices in AMD eyes, particularly affecting the short posterior ciliary arteries. According to mechanical theory, ARMD eyes exhibited elevated scleral rigidity and corneal resistance factor compared to controls, with a notable rise in SR in neovascular AMD (nAMD) eyes. As per the chronic subacute inflammation theory, plasma ET-1 levels were significantly higher in AMD patients, correlating with abnormal SPCAs blood flow and increased resistance indices. Conclusion Findings suggest a multifactorial etiology of AMD involving an increase of ET-1 plasma levels with biomechanic damages of corneal and scleral tissue in nAMD.
Collapse
Affiliation(s)
- Alessandro Finzi
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Simone Ottoboni
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Mauro Cellini
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Beniamino Corcioni
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Caterina Gaudiano
- Department of Radiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luigi Fontana
- Ophthalmology Unit, Department of Experimental, Diagnostic and Specialty Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
2
|
Williams AM, Liu W, Ehrhardt MJ, Salehabadi SM, Panoskaltsis-Mortari A, Phillips NS, Mulrooney DA, Flerlage JE, Yasui Y, Srivastava D, Robison LL, Hudson MM, Ness KK, Sabin ND, Krull KR. Systemic Biological Mechanisms of Neurocognitive Dysfunction in Long-Term Survivors of Childhood Hodgkin Lymphoma. Clin Cancer Res 2024; 30:1822-1832. [PMID: 38381440 PMCID: PMC11062814 DOI: 10.1158/1078-0432.ccr-23-3709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/31/2024] [Accepted: 02/19/2024] [Indexed: 02/22/2024]
Abstract
PURPOSE Hodgkin lymphoma (HL) survivors experience neurocognitive impairment despite receiving no central nervous system-directed therapy, though little is known about the underlying mechanisms. EXPERIMENTAL DESIGN HL survivors (n = 197) and age-, sex- and race/ethnicity frequency-matched community controls (n = 199) underwent standardized neurocognitive testing, and serum collection. Luminex multiplex or ELISA assays measured markers of inflammation and oxidative stress. Linear regression models compared biomarker concentrations between survivors and controls and with neurocognitive outcomes, adjusting for age, sex, race, body mass index, anti-inflammatory medication, and recent infections. RESULTS HL survivors [mean (SD) current age 36 (8) years, 22 (8) years after diagnosis] demonstrated higher concentrations of interleukin-6 (IL6), high-sensitivity c-reactive protein (hs-CRP), oxidized low-density lipoprotein, and glutathione peroxidase (GPx), compared with controls (P's < 0.001). Among survivors, higher concentrations of IL6 were associated with worse visuomotor processing speed (P = 0.046). hs-CRP ≥3 mg/L was associated with worse attention, processing speed, memory, and executive function (P's < 0.05). Higher concentrations of malondialdehyde were associated with worse focused attention and visual processing speed (P's < 0.05). Homocysteine was associated with worse short-term recall (P = 0.008). None of these associations were statistically significant among controls. Among survivors, hs-CRP partially mediated associations between cardiovascular or endocrine conditions and visual processing speed, whereas IL6 partially mediated associations between pulmonary conditions and visuomotor processing speed. CONCLUSIONS Neurocognitive function in long-term survivors of HL appears to be associated with inflammation and oxidative stress, both representing potential targets for future intervention trials.
Collapse
Affiliation(s)
- AnnaLynn M. Williams
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Wei Liu
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Matthew J. Ehrhardt
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | | | - Nicholas S. Phillips
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Daniel A. Mulrooney
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jamie E. Flerlage
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yutaka Yasui
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Deokumar Srivastava
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN
| | - Leslie L. Robison
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Melissa M. Hudson
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
- Departments of Pediatrics and Medicine, University of Minnesota, Minneapolis, MN
| | - Kirsten K. Ness
- Departments of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Noah D. Sabin
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kevin R. Krull
- Department of Psychology and Biobehavioral Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
3
|
Madison AA, Andridge R, Kantaras AH, Renna ME, Bennett JM, Alfano CM, Povoski SP, Agnese DM, Lustberg M, Wesolowski R, Carson WE, Williams NO, Reinbolt RE, Sardesai SD, Noonan AM, Stover DG, Cherian MA, Malarkey WB, Kiecolt-Glaser JK. Depression, Inflammation, and Intestinal Permeability: Associations with Subjective and Objective Cognitive Functioning throughout Breast Cancer Survivorship. Cancers (Basel) 2023; 15:4414. [PMID: 37686689 PMCID: PMC10487080 DOI: 10.3390/cancers15174414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
About one-in-three breast cancer survivors have lingering cognitive complaints and objective cognitive impairment. Chronic inflammation and intestinal permeability (i.e., leaky gut), two risk factors for cognitive decline, can also fuel depression-another vulnerability for cognitive decline. The current study tested whether depression accompanied by high levels of inflammation or intestinal permeability predicted lower subjective and objective cognitive function in breast cancer survivors. We combined data from four breast cancer survivor studies (n = 613); some had repeated measurements for a total of 1015 study visits. All participants had a blood draw to obtain baseline measures of lipopolysaccharide binding protein-a measure of intestinal permeability, as well as three inflammatory markers that were incorporated into an inflammatory index: C-reactive protein, interleukin-6, and tumor necrosis factor-α. They reported depressive symptoms on the Center for Epidemiological Studies depression scale (CES-D), and a binary variable indicated clinically significant depressive symptoms (CES-D ≥ 16). The Kohli (749 observations) and the Breast Cancer Prevention Trial (591 observations) scales assessed subjective cognitive function. Objective cognitive function tests included the trail-making test, Hopkins verbal learning test, Conners continuous performance test, n-back test, FAS test, and animal-naming test (239-246 observations). Adjusting for education, age, BMI, cancer treatment type, time since treatment, study visit, and fatigue, women who had clinically elevated depressive symptoms accompanied by heightened inflammation or intestinal permeability reported poorer focus and marginally poorer memory. However, poorer performance across objective cognitive measures was not specific to inflammation-associated depression. Rather, there was some evidence of lower verbal fluency; poorer attention, verbal learning and memory, and working memory; and difficulties with visuospatial search among depressed survivors, regardless of inflammation. By themselves, inflammation and intestinal permeability less consistently predicted subjective or objective cognitive function. Breast cancer survivors with clinically significant depressive symptoms accompanied by either elevated inflammation or intestinal permeability may perceive greater cognitive difficulty, even though depression-related objective cognitive deficits may not be specific to inflammation- or leaky-gut-associated depression.
Collapse
Affiliation(s)
- Annelise A Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca Andridge
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Biostatistics, The Ohio State University, Columbus, OH 43210, USA
| | - Anthony H Kantaras
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Megan E Renna
- School of Psychology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Jeanette M Bennett
- Department of Psychological Science, University of North Carolina at Charlotte, Charlotte, NC 28213, USA
| | | | - Stephen P Povoski
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Doreen M Agnese
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Maryam Lustberg
- Center for Breast Cancer, Yale Cancer Center, Yale University, New Haven, CT 06519, USA
| | - Robert Wesolowski
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - William E Carson
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nicole O Williams
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Raquel E Reinbolt
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sagar D Sardesai
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anne M Noonan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel G Stover
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mathew A Cherian
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - William B Malarkey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Janice K Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
4
|
Suthahar N, Wang D, Aboumsallem JP, Shi C, de Wit S, Liu EE, Lau ES, Bakker SJL, Gansevoort RT, van der Vegt B, Jovani M, Kreger BE, Lee Splansky G, Benjamin EJ, Vasan RS, Larson MG, Levy D, Ho JE, de Boer RA. Association of Initial and Longitudinal Changes in C-reactive Protein With the Risk of Cardiovascular Disease, Cancer, and Mortality. Mayo Clin Proc 2023; 98:549-558. [PMID: 37019514 PMCID: PMC10698556 DOI: 10.1016/j.mayocp.2022.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 04/07/2023]
Abstract
OBJECTIVE To evaluate the value of serial C-reactive protein (CRP) measurements in predicting the risk of cardiovascular disease (CVD), cancer, and mortality. METHODS The analysis was performed using data from two prospective, population-based observational cohorts: the Prevention of Renal and Vascular End-Stage Disease (PREVEND) study and the Framingham Heart Study (FHS). A total of 9253 participants had CRP measurements available at two examinations (PREVEND: 1997-1998 and 2001-2002; FHS Offspring cohort: 1995-1998 and 1998-2001). All CRP measurements were natural log-transformed before analyses. Cardiovascular disease included fatal and nonfatal cardiovascular, cerebrovascular and peripheral vascular events, and heart failure. Cancer included all malignancies except nonmelanoma skin cancers. RESULTS The mean age of the study population at baseline was 52.4±12.1 years and 51.2% (n=4733) were women. Advanced age, female sex, smoking, body mass index, and total cholesterol were associated with greater increases in CRP levels over time (Pall<.001 in the multivariable model). Baseline CRP, as well as increase in CRP over time (ΔCRP), were associated with incident CVD (hazard ratio [HR]: 1.29 per 1-SD increase; 95% confidence interval [CI]: 1.29 to 1.47, and HR per 1-SD increase: 1.19; 95% CI: 1.09 to 1.29 respectively). Similar findings were observed for incident cancer (baseline CRP, HR: 1.17; 95% CI: 1.09 to 1.26; ΔCRP, HR: 1.08; 95% CI: 1.01 to 1.15) and mortality (baseline CRP, HR: 1.29; 95% CI: 1.21 to 1.37; ΔCRP, HR: 1.10; 95% CI: 1.05 to 1.16). CONCLUSION Initial as well as subsequent increases in CRP levels predict future CVD, cancer, and mortality in the general population.
Collapse
Affiliation(s)
- Navin Suthahar
- Department of Cardiology, University of Groningen, Groningen, the Netherlands; Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| | - Dongyu Wang
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Biostatistics, Boston University, Boston, MA, USA
| | | | - Canxia Shi
- Department of Cardiology, University of Groningen, Groningen, the Netherlands
| | - Sanne de Wit
- Department of Cardiology, University of Groningen, Groningen, the Netherlands
| | - Elizabeth E Liu
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily S Lau
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, the Netherlands
| | - Ron T Gansevoort
- Division of Nephrology, Department of Internal Medicine, University of Groningen, Groningen, the Netherlands
| | - Bert van der Vegt
- Department of Pathology, University of Groningen, Groningen, the Netherlands
| | - Manol Jovani
- Digestive Diseases and Nutrition, University of Kentucky Albert B. Chandler Hospital, Lexington, KY, USA
| | - Bernard E Kreger
- Department of Medicine, School of Medicine, Boston University, Boston, MA, USA; The Framingham Heart Study, Framingham, MA, USA
| | | | - Emelia J Benjamin
- Department of Biostatistics, Boston University, Boston, MA, USA; Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA; Department of Medicine, School of Medicine, Boston University, Boston, MA, USA; The Framingham Heart Study, Framingham, MA, USA
| | - Ramachandran S Vasan
- Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA; Department of Medicine, School of Medicine, Boston University, Boston, MA, USA; The Framingham Heart Study, Framingham, MA, USA
| | - Martin G Larson
- Department of Biostatistics, Boston University, Boston, MA, USA; The Framingham Heart Study, Framingham, MA, USA
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer E Ho
- Cardiovascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, Groningen, the Netherlands; Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
5
|
Walsh CP, Lindsay EK, Grosse P, Natale BN, Fairlie S, Bwint A, Schaffer L, McMahon K, Del Duke C, Forse J, Lamonja-Vicente N, Marsland AL. A systematic review and meta-analysis of the stability of peripheral immune markers in healthy adults. Brain Behav Immun 2023; 107:32-46. [PMID: 36152782 PMCID: PMC9729419 DOI: 10.1016/j.bbi.2022.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/09/2022] [Accepted: 09/16/2022] [Indexed: 01/03/2023] Open
Abstract
Peripheral immune markers are widely used to predict risk for inflammatory disease. However, whether single assessments of inflammatory biomarkers represent stable individual differences remains unclear. We reviewed 50 studies (N = 48,674; 57 % male; mean age 54 (range 13-79) years) that assessed markers of inflammation on >1 occasion, with time between measures ranging from 24 h to 7+ years. Separate random effects meta-analyses were conducted for each inflammatory marker and time interval. Markers that had broad coverage across most time intervals included C-reactive protein (CRP; k = 37), interleukin (IL)-6 (k = 22), TNF-α (k = 10), and fibrinogen (Fg; k = 9). For CRP, IL-6, and TNF-α, stability estimates generally decreased with time, with strong to moderate stability over intervals <6 months (r's = 0.80-0.61), modest to moderate stability over 6 months - 3 years (r's = 0.60-0.51), and low stability for >3 years (r's = 0.39-0.30). Estimates were less reliable for Fg for time intervals ≤ 3 years although they generally followed the same pattern; more reliable findings suggested greater stability for Fg than other markers for intervals >3 years (r = 0.53). These findings suggest that single measures of inflammatory biomarkers may be an adequate index of stable individual differences in the short term (<6 months), with repeated measures of inflammatory biomarkers recommended over intervals ≥ 6 months to 3 years, and absolutely necessary over intervals >3 years to reliably identify stable individual differences in health risk. These findings are consistent with stability estimates and clinical recommendations for repeated measurement of other cardiovascular measures of risk (e.g., blood lipids, blood pressure).
Collapse
Affiliation(s)
- Catherine P Walsh
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Emily K Lindsay
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Philip Grosse
- Clinical and Translational Science Institute, Forbes Tower, Suite 7057, Pittsburgh, PA 15213, USA.
| | - Brianna N Natale
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Samantha Fairlie
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Amanda Bwint
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Luke Schaffer
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Katie McMahon
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Colin Del Duke
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Jenny Forse
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| | - Noemi Lamonja-Vicente
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA; Department of Clinical Psychology and Psychobiology, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Anna L Marsland
- Department of Psychology, University of Pittsburgh, 210 Bouquet St., Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Kornej J, Qadan MA, Alotaibi M, Van Wagoner DR, Watrous JD, Trinquart L, Preis SR, Ko D, Jain M, Benjamin EJ, Cheng S, Lin H. The association between eicosanoids and incident atrial fibrillation in the Framingham Heart Study. Sci Rep 2022; 12:20218. [PMID: 36418854 PMCID: PMC9684401 DOI: 10.1038/s41598-022-21786-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 10/04/2022] [Indexed: 11/26/2022] Open
Abstract
Chronic inflammation is a continuous low-grade activation of the systemic immune response. Whereas downstream inflammatory markers are associated with atrial fibrillation (AF), upstream inflammatory effectors including eicosanoids are less studied. To examine the association between eicosanoids and incident AF. We used a liquid chromatography-mass spectrometry for the non-targeted measurement of 161 eicosanoids and eicosanoid-related metabolites in the Framingham Heart Study. The association of each eicosanoid and incident AF was assessed using Cox proportional hazards models and adjusted for AF risk factors, including age, sex, height, weight, systolic/diastolic blood pressure, current smoking, antihypertensive medication, diabetes, history of myocardial infarction and heart failure. False discovery rate (FDR) was used to adjust for multiple testing. Eicosanoids with FDR < 0.05 were considered significant. In total, 2676 AF-free individuals (mean age 66 ± 9 years, 56% females) were followed for mean 10.8 ± 3.4 years; 351 participants developed incident AF. Six eicosanoids were associated with incident AF after adjusting for multiple testing (FDR < 0.05). A joint score was built from the top eicosanoids weighted by their effect sizes, which was associated with incident AF (HR = 2.72, CI = 1.71-4.31, P = 2.1 × 10-5). In conclusion, six eicosanoids were associated with incident AF after adjusting for clinical risk factors for AF.
Collapse
Affiliation(s)
- Jelena Kornej
- National Heart, Lung, and Blood Institute, Boston University's Framingham Heart Study, Framingham, MA, USA. .,Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.
| | - Maha A. Qadan
- grid.239578.20000 0001 0675 4725Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH USA
| | - Mona Alotaibi
- grid.266100.30000 0001 2107 4242Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, La Jolla, CA USA
| | - David R. Van Wagoner
- grid.239578.20000 0001 0675 4725Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH USA
| | - Jeramie D. Watrous
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California, La Jolla, San Diego, CA USA
| | - Ludovic Trinquart
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Sarah R. Preis
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Darae Ko
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA USA
| | - Mohit Jain
- grid.266100.30000 0001 2107 4242Department of Medicine, University of California, La Jolla, San Diego, CA USA
| | - Emelia J. Benjamin
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.189504.10000 0004 1936 7558Section of Cardiovascular Medicine, Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA USA ,grid.189504.10000 0004 1936 7558Department of Epidemiology, Boston University School of Public Health, Boston, MA USA
| | - Susan Cheng
- grid.512369.aDepartment of Cardiology, Cedars-Sinai Medical Center, Smidt Heart Institute, Los Angeles, CA USA
| | - Honghuang Lin
- grid.510954.c0000 0004 0444 3861National Heart, Lung, and Blood Institute, Boston University’s Framingham Heart Study, Framingham, MA USA ,grid.168645.80000 0001 0742 0364Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA USA
| |
Collapse
|
7
|
Quantitative Comparison of Statistical Methods for Analyzing Human Metabolomics Data. Metabolites 2022; 12:metabo12060519. [PMID: 35736452 PMCID: PMC9227835 DOI: 10.3390/metabo12060519] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 01/26/2023] Open
Abstract
Emerging technologies now allow for mass spectrometry-based profiling of thousands of small molecule metabolites ('metabolomics') in an increasing number of biosamples. While offering great promise for insight into the pathogenesis of human disease, standard approaches have not yet been established for statistically analyzing increasingly complex, high-dimensional human metabolomics data in relation to clinical phenotypes, including disease outcomes. To determine optimal approaches for analysis, we formally compare traditional and newer statistical learning methods across a range of metabolomics dataset types. In simulated and experimental metabolomics data derived from large population-based human cohorts, we observe that with an increasing number of study subjects, univariate compared to multivariate methods result in an apparently higher false discovery rate as represented by substantial correlation between metabolites directly associated with the outcome and metabolites not associated with the outcome. Although the higher frequency of such associations would not be considered false in the strict statistical sense, it may be considered biologically less informative. In scenarios wherein the number of assayed metabolites increases, as in measures of nontargeted versus targeted metabolomics, multivariate methods performed especially favorably across a range of statistical operating characteristics. In nontargeted metabolomics datasets that included thousands of metabolite measures, sparse multivariate models demonstrated greater selectivity and lower potential for spurious relationships. When the number of metabolites was similar to or exceeded the number of study subjects, as is common with nontargeted metabolomics analysis of relatively small cohorts, sparse multivariate models exhibited the most-robust statistical power with more consistent results. These findings have important implications for metabolomics analysis in human disease.
Collapse
|
8
|
Rade JJ, Barton BA, Vasan RS, Kronsberg SS, Xanthakis V, Keaney JF, Hamburg NM, Kakouros N, Kickler TA. Association of Thromboxane Generation With Survival in Aspirin Users and Nonusers. J Am Coll Cardiol 2022; 80:233-250. [PMID: 35660296 DOI: 10.1016/j.jacc.2022.04.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Persistent systemic thromboxane generation, predominantly from nonplatelet sources, in aspirin (ASA) users with cardiovascular disease (CVD) is a mortality risk factor. OBJECTIVES This study sought to determine the mortality risk associated with systemic thromboxane generation in an unselected population irrespective of ASA use. METHODS Stable thromboxane B2 metabolites (TXB2-M) were measured by enzyme-linked immunosorbent assay in banked urine from 3,044 participants (mean age 66 ± 9 years, 53.8% women) in the Framingham Heart Study. The association of TXB2-M to survival over a median observation period of 11.9 years (IQR: 10.6-12.7 years) was determined by multivariable modeling. RESULTS In 1,363 (44.8%) participants taking ASA at the index examination, median TXB2-M were lower than in ASA nonusers (1,147 pg/mg creatinine vs 4,179 pg/mg creatinine; P < 0.0001). TXB2-M were significantly associated with all-cause and cardiovascular mortality irrespective of ASA use (HR: 1.96 and 2.41, respectively; P < 0.0001 for both) for TXB2-M in the highest quartile based on ASA use compared with lower quartiles, and remained significant after adjustment for mortality risk factors for similarly aged individuals (HR: 1.49 and 1.82, respectively; P ≤ 0.005 for both). In 2,353 participants without CVD, TXB2-M were associated with cardiovascular mortality in ASA nonusers (adjusted HR: 3.04; 95% CI: 1.29-7.16) but not in ASA users, while ASA use was associated with all-cause mortality in those with low (adjusted HR: 1.46; 95% CI: 1.14-1.87) but not elevated TXB2-M. CONCLUSIONS Systemic thromboxane generation is an independent risk factor for all-cause and cardiovascular mortality irrespective of ASA use, and its measurement may be useful for therapy modification, particularly in those without CVD.
Collapse
Affiliation(s)
- Jeffrey J Rade
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| | - Bruce A Barton
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - Shari S Kronsberg
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | - John F Keaney
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA; Boston University School of Medicine, Boston, Massachusetts, USA
| | - Naomi M Hamburg
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Nikolaos Kakouros
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
9
|
Millar CL, Costa E, Jacques PF, Dufour AB, Kiel DP, Hannan MT, Sahni S. Adherence to the Mediterranean-style diet and high intake of total carotenoids reduces the odds of frailty over 11 years in older adults: Results from the Framingham Offspring Study. Am J Clin Nutr 2022; 116:630-639. [PMID: 35551593 PMCID: PMC9437990 DOI: 10.1093/ajcn/nqac130] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/31/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The benefit of a Mediterranean-style diet in reducing frailty is not well established in older Americans. OBJECTIVES We sought to determine associations of a Mediterranean-style dietary pattern and related antioxidants with frailty onset and worsening of the Fried phenotype in adults. METHODS This prospective study included 2384 nonfrail adults from the Framingham Offspring Study with a Mediterranean-style dietary pattern score (MSDPS) and data on antioxidant intakes (vitamin C, E, and total carotenoids) estimated from an FFQ at the index examination (1998-2001) and 1 prior examination (if available), as well as a frailty assessment at the index examination and at least 1 follow-up. Frailty onset was defined as ≥3 of 5 Fried frailty phenotype criteria at follow-up and the worsening of the Fried frailty phenotype was defined as an increased number of frailty criteria over follow-up (yes or no). Logistic regression with generalized estimating equations estimated ORs and 95% CIs, adjusting for confounders. Analyses were stratified by age (<60 and ≥60 years) for significant interactions. RESULTS The mean ± SD age was 60 ± 9 years (range, 33-86 years) and 55% were female. In adjusted models, a 1-unit higher MSDPS reduced the odds of frailty by 3% (OR, 0.97; 95% CI: 0.96-0.99). Each 10-mg higher total carotenoid and vitamin E intake reduced the odds of frailty by 16% (OR, 0.84; 95% CI: 0.73-0.98) and 1% (OR, 0.99; 95% CI: 0.98-1.00), respectively. No association with vitamin C (P = 0.36) was observed. The associations among participants aged <60 years of age were stronger for each 1-unit higher MSDPS (OR, 0.93; 95% CI: 0.89-0.96) and total carotenoid intake (OR, 0.59; 95% CI: 0.41-0.82) than those observed in older individuals [ORs, 0.98 (95% CI: 0.97-1.00) and 0.92 (95% CI: 0.79-1.08), respectively]. CONCLUSIONS Our findings suggest that adherence to a Mediterranean-style diet and higher total carotenoid intake are associated with frailty prevention over time, particularly in adults <60 years.
Collapse
Affiliation(s)
- Courtney L Millar
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Elise Costa
- The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Paul F Jacques
- Nutritional Epidemiology Program, Jean Mayer, USDA Human Nutrition Research Center on Aging, Tufts University, Friedman School of Nutrition, Boston, MA, USA
| | - Alyssa B Dufour
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Douglas P Kiel
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marian T Hannan
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
10
|
Adiposity and Smoking Mediate the Relationship Between Depression History and Inflammation Among Young Adults. Int J Behav Med 2022; 29:787-795. [PMID: 35141821 DOI: 10.1007/s12529-022-10060-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Depression is associated with inflammation, but the mechanisms underlying this association are unclear. We examined adiposity and smoking as potential pathways through which childhood depression may lead to an elevated inflammatory status among young adults. METHODS The sample included 294 subjects with histories of depression (probands), 270 never-depressed siblings of probands (high-risk siblings), and 169 controls. C-reactive protein (CRP), interleukin-6 (IL-6), and soluble intercellular adhesion molecule-1 (sICAM-1) were assessed in serum samples. An adiposity score was computed from body mass index and waist circumference. Smoking behavior was evaluated during an interview. Mixed-effects models were used to test whether adiposity and smoking mediate the relationship between depression and inflammation. RESULTS Probands (p = .004), but not siblings (p = .071), had higher levels of sICAM-1 compared to controls. However, depression history and risk status had no direct effects on CRP (ps > .13) or IL-6 (ps > .16). Importantly, adiposity indirectly mediated the effect of group (probands vs. controls; siblings vs. controls) on all three inflammatory markers. Smoking indirectly mediated the effect of group (probands vs. controls; siblings vs. controls) on sICAM-1 only. CONCLUSIONS Among young adults, the adverse inflammatory consequences of depression history are significant for sICAM-1. Adiposity and smoking are pathways through which depression can indirectly impact several inflammatory markers, suggesting possible preventive interventions to improve the immunologic and cardiovascular health of depression-prone individuals.
Collapse
|
11
|
Turek-Jakubowska A, Dębski J, Jakubowski M, Szahidewicz-Krupska E, Gawryś J, Gawryś K, Janus A, Trocha M, Doroszko A. New Candidates for Biomarkers and Drug Targets of Ischemic Stroke-A First Dynamic LC-MS Human Serum Proteomic Study. J Clin Med 2022; 11:jcm11020339. [PMID: 35054033 PMCID: PMC8780942 DOI: 10.3390/jcm11020339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 01/27/2023] Open
Abstract
(1) Background: The aim of this dynamic-LC/MS-human-serum-proteomic-study was to identify potential proteins-candidates for biomarkers of acute ischemic stroke, their changes during acute phase of stroke and to define potential novel drug-targets. (2) Methods: A total of 32 patients (29–80 years) with acute ischemic stroke were enrolled to the study. The control group constituted 29 demographically-matched volunteers. Subjects with stroke presented clinical symptoms lasting no longer than 24 h, confirmed by neurological-examination and/or new cerebral ischemia visualized in the CT scans (computed tomography). The analysis of plasma proteome was performed using LC-MS (liquid chromatography–mass spectrometry). (3) Results: Ten proteins with significantly different serum concentrations between groups volunteers were: complement-factor-B, apolipoprotein-A-I, fibronectin, alpha-2-HS-glycoprotein, alpha-1B-glycoprotein, heat-shock-cognate-71kDa protein/heat-shock-related-70kDa-protein-2, thymidine phosphorylase-2, cytoplasmic-tryptophan-tRNA-ligase, ficolin-2, beta-Ala-His-dipeptidase. (4) Conclusions: This is the first dynamic LC-MS study performed on a clinical model which differentiates serum proteome of patients in acute phase of ischemic stroke in time series and compares to control group. Listed proteins should be considered as risk factors, markers of ischemic stroke or potential therapeutic targets. Further clinical validation might define their exact role in differential diagnostics, monitoring the course of the ischemic stroke or specifying them as novel drug targets.
Collapse
Affiliation(s)
| | - Janusz Dębski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warszawa, Poland;
| | - Maciej Jakubowski
- Lower Silesian Centre for Lung Diseases, Grabiszyńska 105, 53-439 Wroclaw, Poland;
| | - Ewa Szahidewicz-Krupska
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (E.S.-K.); (J.G.); (A.J.)
| | - Jakub Gawryś
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (E.S.-K.); (J.G.); (A.J.)
| | - Karolina Gawryś
- Department of Neurology, 4th Military Hospital, Weigla 5, 50-556 Wroclaw, Poland; (A.T.-J.); (K.G.)
| | - Agnieszka Janus
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (E.S.-K.); (J.G.); (A.J.)
| | - Małgorzata Trocha
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicz-Radecki 2, 50-349 Wroclaw, Poland;
| | - Adrian Doroszko
- Department of Internal Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (E.S.-K.); (J.G.); (A.J.)
- Correspondence: ; Tel.: +48-71-736-4000
| |
Collapse
|
12
|
Millar CL, Dufour AB, Shivappa N, Habtemariam D, Murabito JM, Benjamin EJ, Hebert JR, Kiel DP, Hannan MT, Sahni S. A proinflammatory diet is associated with increased odds of frailty after 12-year follow-up in a cohort of adults. Am J Clin Nutr 2021; 115:334-343. [PMID: 34558613 PMCID: PMC8827080 DOI: 10.1093/ajcn/nqab317] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/17/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Frailty occurs in 10-15% of community-living older adults and inflammation is a key determinant of frailty. Though diet is a modulator of inflammation, there are few prospective studies elucidating the role of diet-associated inflammation on frailty onset. OBJECTIVES We sought to determine whether a proinflammatory diet was associated with increased odds of frailty in adults from the Framingham Heart Study (FHS). DESIGN AND METHODS This study was nested in a prospective cohort that included individuals without frailty. Diet was assessed in 1998-2001 using a valid FFQ, and frailty was measured in 2011-2014. FFQ-derived energy-adjusted dietary inflammatory index (E-DII®) scores were computed, with higher E-DII scores indicating a more proinflammatory diet. Frailty was defined as fulfilling ≥3 of 5 Fried Phenotype criteria. Information on potential mediators, serum IL-6 and C-reactive protein was obtained in 1998-2001. Logistic regression estimated ORs and 95% CIs for E-DII (as continuous and in quartiles) and frailty onset adjusting for relevant confounders. RESULTS Of 1701 individuals without frailty at baseline (mean ± SD age: 58 ± 8 y; range: 33-81 y; 55% female), 224 developed frailty (13% incidence) over ∼12 y. The mean ± SD E-DII score was -1.95 ± 2.20; range: -6.71 to +5.40. After adjusting for relevant confounders, a 1-unit higher E-DII score was associated with 16% increased odds of developing frailty (95% CI: 1.07, 1.25). In categorical analyses, participants in the highest (proinflammatory) compared with lowest quartile of E-DII had >2-fold increased odds of frailty (ORquartile4vs.1: 2.22; 95% CI: 1.37, 3.60; P-trend < 0.01). IL-6 and C-reactive protein were not major contributors in the pathway. CONCLUSIONS In this cohort of middle-aged and older adults, a proinflammatory diet was associated with increased odds of frailty over ∼12 y of follow-up. Trials designed to increase consumption of anti-inflammatory foods for frailty prevention are warranted.
Collapse
Affiliation(s)
- Courtney L Millar
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alyssa B Dufour
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nitin Shivappa
- Department of Epidemiology and Biostatistics and the Cancer Prevention and Control Program, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA,Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC, USA
| | - Daniel Habtemariam
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
| | - Joanne M Murabito
- Boston University School of Medicine and Public Health, Boston, MA, USA,NHLBI's Framingham Heart Study, Framingham, MA, USA
| | - Emelia J Benjamin
- Boston University School of Medicine and Public Health, Boston, MA, USA,NHLBI's Framingham Heart Study, Framingham, MA, USA,Cardiovascular Section, Department of Medicine, Boston University School of Medicine, Boston, MA, USA,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - James R Hebert
- Department of Epidemiology and Biostatistics and the Cancer Prevention and Control Program, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA,Department of Nutrition, Connecting Health Innovations LLC, Columbia, SC, USA
| | - Douglas P Kiel
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Marian T Hannan
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
13
|
Kobak KA, Franczuk P, Schubert J, Dzięgała M, Kasztura M, Tkaczyszyn M, Drozd M, Kosiorek A, Kiczak L, Bania J, Ponikowski P, Jankowska EA. Primary Human Cardiomyocytes and Cardiofibroblasts Treated with Sera from Myocarditis Patients Exhibit an Increased Iron Demand and Complex Changes in the Gene Expression. Cells 2021; 10:cells10040818. [PMID: 33917391 PMCID: PMC8067399 DOI: 10.3390/cells10040818] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/28/2021] [Accepted: 04/01/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiac fibroblasts and cardiomyocytes are the main cells involved in the pathophysiology of myocarditis (MCD). These cells are especially sensitive to changes in iron homeostasis, which is extremely important for the optimal maintenance of crucial cellular processes. However, the exact role of iron status in the pathophysiology of MCD remains unknown. We cultured primary human cardiomyocytes (hCM) and cardiofibroblasts (hCF) with sera from acute MCD patients and healthy controls to mimic the effects of systemic inflammation on these cells. Next, we performed an initial small-scale (n = 3 per group) RNA sequencing experiment to investigate the global cellular response to the exposure on sera. In both cell lines, transcriptomic data analysis revealed many alterations in gene expression, which are related to disturbed canonical pathways and the progression of cardiac diseases. Moreover, hCM exhibited changes in the iron homeostasis pathway. To further investigate these alterations in sera-treated cells, we performed a larger-scale (n = 10 for controls, n = 18 for MCD) follow-up study and evaluated the expression of genes involved in iron metabolism. In both cell lines, we demonstrated an increased expression of transferrin receptor 1 (TFR1) and ferritin in MCD serum-treated cells as compared to controls, suggesting increased iron demand. Furthermore, we related TFR1 expression with the clinical profile of patients and showed that greater iron demand in sera-treated cells was associated with higher inflammation score (interleukin 6 (IL-6), C-reactive protein (CRP)) and advanced neurohormonal activation (NT-proBNP) in patients. Collectively, our data suggest that the malfunctioning of cardiomyocytes and cardiofibroblasts in the course of MCD might be related to alterations in the iron homeostasis.
Collapse
Affiliation(s)
- Kamil A. Kobak
- Laboratory for Applied Research on Cardiovascular System, Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.A.K.); (P.F.); (M.D.); (M.T.); (M.D.)
| | - Paweł Franczuk
- Laboratory for Applied Research on Cardiovascular System, Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.A.K.); (P.F.); (M.D.); (M.T.); (M.D.)
- Centre for Heart Diseases, University Hospital, 50-556 Wroclaw, Poland; (A.K.); (P.P.)
| | - Justyna Schubert
- Department of Food Hygiene and Consumer Health Protection, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (J.S.); (M.K.); (J.B.)
| | - Magdalena Dzięgała
- Laboratory for Applied Research on Cardiovascular System, Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.A.K.); (P.F.); (M.D.); (M.T.); (M.D.)
| | - Monika Kasztura
- Department of Food Hygiene and Consumer Health Protection, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (J.S.); (M.K.); (J.B.)
| | - Michał Tkaczyszyn
- Laboratory for Applied Research on Cardiovascular System, Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.A.K.); (P.F.); (M.D.); (M.T.); (M.D.)
- Centre for Heart Diseases, University Hospital, 50-556 Wroclaw, Poland; (A.K.); (P.P.)
| | - Marcin Drozd
- Laboratory for Applied Research on Cardiovascular System, Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.A.K.); (P.F.); (M.D.); (M.T.); (M.D.)
- Centre for Heart Diseases, University Hospital, 50-556 Wroclaw, Poland; (A.K.); (P.P.)
| | - Aneta Kosiorek
- Centre for Heart Diseases, University Hospital, 50-556 Wroclaw, Poland; (A.K.); (P.P.)
- Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Liliana Kiczak
- Department of Biochemistry and Molecular Biology, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| | - Jacek Bania
- Department of Food Hygiene and Consumer Health Protection, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (J.S.); (M.K.); (J.B.)
| | - Piotr Ponikowski
- Centre for Heart Diseases, University Hospital, 50-556 Wroclaw, Poland; (A.K.); (P.P.)
- Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Ewa A. Jankowska
- Laboratory for Applied Research on Cardiovascular System, Department of Heart Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.A.K.); (P.F.); (M.D.); (M.T.); (M.D.)
- Centre for Heart Diseases, University Hospital, 50-556 Wroclaw, Poland; (A.K.); (P.P.)
- Correspondence:
| |
Collapse
|
14
|
Jaafari J, Naddafi K, Yunesian M, Nabizadeh R, Hassanvand MS, Shamsipour M, Ghanbari Ghozikali M, Nazmara S, Shamsollahi HR, Yaghmaeian K. Associations between short term exposure to ambient particulate matter from dust storm and anthropogenic sources and inflammatory biomarkers in healthy young adults. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 761:144503. [PMID: 33352344 DOI: 10.1016/j.scitotenv.2020.144503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/18/2020] [Accepted: 12/10/2020] [Indexed: 05/13/2023]
Abstract
This study examined the association between particulate matter from anthropogenic and natural sources and inflammatory biomarkers, including hs-CRP, IL-6, sTNF-RII, and WBCs, in two groups of healthy young subjects. We followed up subjects of two panels (16 to 22 years old), including 22 subjects selected from the urban area (Tehran city) with high-level pollution background and 22 subjects selected from the rural area (Ahmadabad) with low-level pollution background. In each group, we collected 4 times blood samples in various air pollution conditions, In the subjects of the urban group, there was a substantial difference (p < 0.05) between inversion days and cold season control days, and between dust storm days and warm season control days for concentrations of hs-CRP, IL-6, and WBCs biomarkers. In the subjects of the rural group, a significant difference could be detected in the concentration of hs-CRP, IL-6, and WBCs biomarkers (p < 0.05) between inversion days and cold season control days, and between dust storm and warm control days. We found that the difference in concentrations of hs-CRP, IL-6, and WBCs biomarkers between dust storm days and warm control conditions in the rural group were higher than the difference in inversion and cold control conditions, which can be attributed to low background air pollution in the rural area. In the urban area, the health effect of anthropogenic sources of PM is higher than the dust storm condition, which can be attributed to the stronger effect of anthropogenic pollution effect.
Collapse
Affiliation(s)
- Jalil Jaafari
- Research Center of Health and Environment, School of Health, Guilan University of Medical Sciences, Rasht, Iran; Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran.
| | - Kazem Naddafi
- Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran; Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masud Yunesian
- Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran; Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Research Methodology and Data Analysis, Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran.
| | - Ramin Nabizadeh
- Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran; Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadegh Hassanvand
- Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran.
| | - Mansour Shamsipour
- Department of Research Methodology and Data Analysis, Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Shahrokh Nazmara
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Shamsollahi
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamyar Yaghmaeian
- Center for Air Pollution Research (CAPR), Institute for Environmental Research (IER), Tehran University of Medical Sciences, Tehran, Iran; Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Walker ME, Xanthakis V, Peterson LR, Duncan MS, Lee J, Ma J, Bigornia S, Moore LL, Quatromoni PA, Vasan RS, Jacques PF. Dietary Patterns, Ceramide Ratios, and Risk of All-Cause and Cause-Specific Mortality: The Framingham Offspring Study. J Nutr 2020; 150:2994-3004. [PMID: 32939554 PMCID: PMC7675031 DOI: 10.1093/jn/nxaa269] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/12/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Prior evidence suggests that diet modifies the association of blood ceramides with the risk of incident cardiovascular disease (CVD). It remains unknown if diet quality modifies the association of very long-chain-to-long-chain ceramide ratios with mortality in the community. OBJECTIVES Our objectives were to determine how healthy dietary patterns associate with blood ceramide concentrations and to examine if healthy dietary patterns modify associations of ceramide ratios (C22:0/C16:0 and C24:0/C16:0) with all-cause and cause-specific mortality. METHODS We examined 2157 participants of the Framingham Offspring Study (mean age = 66 y, 55% women). Blood ceramides were quantified using a validated assay. We evaluated prospective associations of the Dietary Guidelines Adherence Index (DGAI) and Mediterranean-style Diet Score (MDS) with incidence of all-cause and cause-specific mortality using Cox proportional hazards models. Cross-sectional associations of the DGAI and MDS with ceramides were evaluated using multivariable linear regression models. RESULTS The C22:0/C16:0 and C24:0/C16:0 ceramide ratios were inversely associated with all-cause, CVD, and cancer mortality; multivariable-adjusted HRs (95% CIs) were 0.73 (0.67, 0.80) and 0.70 (0.63, 0.77) for all-cause mortality, 0.74 (0.60, 0.90) and 0.69 (0.55, 0.86) for CVD mortality, and 0.75 (0.65, 0.87) and 0.75 (0.64, 0.88) for cancer mortality, respectively. Inverse associations of the C22:0/C16:0 and C24:0/C16:0 ceramide ratios with cancer mortality were attenuated among individuals with a higher diet quality (DGAI or MDS above the median, all P-interaction ≤0.1). The DGAI and MDS had distinct associations with ceramide ratios (DGAI: lower C22:0/C16:0 across quartiles; MDS: higher C24:0/C16:0 across quartiles; all P-trend ≤0.01). CONCLUSION In our community-based sample, ceramide ratios (C22:0/C16:0 and C24:0/C16:0) were associated with a lower risk of all-cause and cause-specific mortality. Further, we observed that a higher overall diet quality attenuates the association between blood ceramide ratios and cancer mortality and that dietary patterns have distinct relations with ceramide ratios.
Collapse
Affiliation(s)
- Maura E Walker
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Vanessa Xanthakis
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Linda R Peterson
- Division of Cardiovascular Medicine, Washington University, St Louis, MO, USA
| | - Meredith S Duncan
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Epidemiology, Vanderbilt University, Nashville, TN, USA
| | - Joowon Lee
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Jiantao Ma
- Framingham Heart Study, Framingham, MA, USA
- Division of Nutrition Data Science, Tufts University Friedman School of Nutrition Science and Policy, Boston, MA, USA
| | - Sherman Bigornia
- Department of Agriculture, Nutrition, and Food Systems, University of New Hampshire, Durham, NH, USA
| | - Lynn L Moore
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Paula A Quatromoni
- Department of Health Sciences, Sargent College of Health & Rehabilitation Sciences, Boston University, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Paul F Jacques
- Division of Nutrition Data Science, Tufts University Friedman School of Nutrition Science and Policy, Boston, MA, USA
- Nutrition Epidemiology, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
16
|
Abstract
Atherosclerosis, the pathology underlying heart attacks, strokes and peripheral artery disease, is a chronic inflammatory disease of the artery wall initiated by elevated low-density lipoprotein (LDL) cholesterol levels. LDL accumulates in the artery wall, where it can become oxidized to oxLDL. T cell responses to ApoB, a core protein found in LDL and other lipoproteins, are detectable in healthy mice and people. Most of the ApoB-specific CD4T cells are FoxP3+ regulatory T cells (Treg). In the course of atherosclerosis development, the number of ApoB-reactive T cells expands. At the same time, their phenotype changes, showing cell surface markers, transcription factors and transcriptomes resembling other T-helper lineages like Th17, Th1 and follicular helper (TFH) cells. TFH cells enter germinal centers and provide T cell help to B cells, enabling antibody isotype switch from IgM to IgG and supporting affinity maturation. In people and mice with atherosclerosis, IgG and IgM antibodies to oxLDL are detectable. Higher IgM antibody titers to oxLDL are associated with less, IgG antibodies with more atherosclerosis. Thus, both T and B cells play critical roles in atherosclerosis. Modifying the adaptive immune response to ApoB holds promise for preventing atherosclerosis and reducing disease burden.
Collapse
Affiliation(s)
- Klaus Ley
- Center for Autoimmunity and Inflammation, Inflammation Biology Laboratory, La Jolla Institute for Immunology, 9420 Athena Circle Drive, La Jolla, CA 92037, U.S.A
| |
Collapse
|
17
|
Ehinger E, Ghosheh Y, Pramod AB, Lin J, Hanna DB, Mueller K, Durant CP, Baas L, Qi Q, Wang T, Buscher K, Anastos K, Lazar JM, Mack WJ, Tien PC, Cohen MH, Ofotokun I, Gange S, Heath SL, Hodis HN, Tracy RP, Landay AL, Kaplan RC, Ley K. Classical monocyte transcriptomes reveal significant anti-inflammatory statin effect in women with chronic HIV. Cardiovasc Res 2020; 117:1166-1177. [PMID: 32658258 DOI: 10.1093/cvr/cvaa188] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/04/2020] [Accepted: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS During virally suppressed chronic HIV infection, persistent inflammation contributes to the development of cardiovascular disease (CVD), a major comorbidity in people living with HIV (LWH). Classical blood monocytes (CMs) remain activated during antiretroviral therapy and are a major source of pro-inflammatory and pro-thrombotic factors that contribute to atherosclerotic plaque development and instability. METHODS AND RESULTS Here, we identify transcriptomic changes in circulating CMs in peripheral blood mononuclear cell samples from participants of the Women's Interagency HIV Study, selected by HIV and subclinical CVD (sCVD) status. We flow-sorted CM from participants of the Women's Interagency HIV Study and deep-sequenced their mRNA (n = 92). CMs of HIV+ participants showed elevated interleukin (IL)-6, IL-1β, and IL-12β, overlapping with many transcripts identified in sCVD+ participants. In sCVD+ participants LWH, those reporting statin use showed reduced pro-inflammatory gene expression to a level comparable with healthy (HIV-sCVD-) participants. Statin non-users maintained an elevated inflammatory profile and increased cytokine production. CONCLUSION Statin therapy has been associated with a lower risk of cardiac events, such as myocardial infarction in the general population, but not in those LWH. Our data suggest that women LWH may benefit from statin therapy even in the absence of overt CVD.
Collapse
Affiliation(s)
- Erik Ehinger
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Yanal Ghosheh
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Akula Bala Pramod
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Juan Lin
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David B Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Karin Mueller
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Christopher P Durant
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Livia Baas
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Konrad Buscher
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Kathryn Anastos
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jason M Lazar
- Department of Medicine, State University of New York, Downstate Medical Center, Bronx, NY, USA.,Department of Epidemiology & Population Health, State University of New York, Downstate Medical Center, Bronx, NY, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Phyllis C Tien
- Department of Medicine and Medical Service, University of California, San Francisco, San Francisco, CA, USA.,Department of Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | - Mardge H Cohen
- Department of Medicine, John Stroger Hospital and Rush University, Chicago, IL, USA
| | - Igho Ofotokun
- Department of Medicine, Infectious Disease Division and Grady Health Care System, Emory University School of Medicine, Atlanta, GA, USA
| | - Stephen Gange
- Division of Cardiovascular Medicine, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sonya L Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Howard N Hodis
- Departments of Medicine and Preventative Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Russell P Tracy
- Department of Pathology & Laboratory Medicine and Biochemistry, University of Vermont Larner College of Medicine, Colchester, VT, USA
| | - Alan L Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA.,Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle WA 98109, USA
| | - Klaus Ley
- Laboratory of Inflammation Biology, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.,Department of Bioengineering, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
18
|
Sex-Specific Epidemiology of Heart Failure Risk and Mortality in Europe: Results From the BiomarCaRE Consortium. JACC-HEART FAILURE 2020; 7:204-213. [PMID: 30819375 DOI: 10.1016/j.jchf.2018.08.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/13/2018] [Accepted: 08/28/2018] [Indexed: 11/21/2022]
Abstract
OBJECTIVES This study investigates differences between women and men in heart failure (HF) risk and mortality. BACKGROUND Sex differences in HF epidemiology are insufficiently understood. METHODS In 78,657 individuals (median 49.5 years of age; age range 24.1 to 98.7 years; 51.7% women) from community-based European studies (FINRISK, DanMONICA, Moli-sani, Northern Sweden) of the BiomarCaRE (Biomarker for Cardiovascular Risk Assessment in Europe) consortium, the association between incident HF and mortality, the relationship of cardiovascular risk factors, prevalent cardiovascular diseases, biomarkers (C-reactive protein [CRP]; N-terminal pro-B-type natriuretic peptide [NT-proBNP]) with incident HF, and their attributable risks were tested in women vs. men. RESULTS Over a median follow-up of 12.7 years, fewer HF cases were observed in women (n = 2,399 [5.9%]) than in men (n = 2,771 [7.3%]). HF incidence increased markedly after 60 years of age, initially with a more rapid increase in men, whereas incidence in women exceeded that of men after 85 years of age. HF onset substantially increased mortality risk in both sexes. Multivariable-adjusted Cox models showed the following sex differences for the association with incident HF: systolic blood pressure hazard ratio (HR) according to SD in women of 1.09 (95% confidence interval [CI]: 1.05 to 1.14) versus HR of 1.19 (95% CI: 1.14 to 1.24) in men; heart rate HR of 0.98 (95% CI: 0.93 to 1.03) in women versus HR of 1.09 (95% CI: 1.04 to 1.13) in men; CRP HR of 1.10 (95% CI: 1.00 to 1.20) in women versus HR of 1.32 (95% CI: 1.24 to 1.41) in men; and NT-proBNP HR of 1.54 (95% CI: 1.37 to 1.74) in women versus HR of 1.89 (95% CI: 1.75 to 2.05) in men. Population-attributable risk of all risk factors combined was 59.0% in women and 62.9% in men. CONCLUSIONS Women had a lower risk for HF than men. Sex differences were seen for systolic blood pressure, heart rate, CRP, and NT-proBNP, with a lower HF risk in women.
Collapse
|
19
|
Christen T, Trompet S, Rensen PCN, Willems van Dijk K, Lamb HJ, Jukema JW, Rosendaal FR, le Cessie S, de Mutsert R. The role of inflammation in the association between overall and visceral adiposity and subclinical atherosclerosis. Nutr Metab Cardiovasc Dis 2019; 29:728-735. [PMID: 31138500 DOI: 10.1016/j.numecd.2019.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/23/2019] [Accepted: 03/17/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND AND AIMS Inflammation may underlie the association between obesity, atherosclerosis and cardiovascular disease. We investigated to what extent markers of inflammation mediate associations between overall and visceral body fat and subclinical atherosclerosis. METHODS AND RESULTS In this cross-sectional analysis of the Netherlands Epidemiology of Obesity study we estimated total body fat (TBF) by bio-impedance analysis, carotid artery intima media thickness (cIMT) by ultrasound, C-reactive protein (hs-CRP) and glycoprotein acetyls (GlycA) concentrations in fasting blood samples (n = 5627), and visceral adipose tissue (VAT) by magnetic resonance imaging (n = 2247). We examined associations between TBF and VAT, and cIMT using linear regression, adjusted for potential confounding factors, and for mediators: cardiometabolic risk factors (blood pressure, glucose and low-density lipoprotein cholesterol), and inflammation using CRP and GlycA as proxies. Mean (SD) cIMT was 615 (90) μm. Per SD of TBF (8%), cIMT was 19 μm larger (95% confidence interval, CI: 10, 28). This association was 17 μm (95% CI: 8, 27) after adjustment for cardiometabolic risk factors, and did not change after adjustment for markers of inflammation. Per SD (56 cm2) VAT, cIMT was 9 μm larger (95% CI: 2, 16) which changed to 5 μm (95% CI: -3, 12) after adjustment for cardiometabolic risk factors, and did not change after adjustment for inflammatory markers. CONCLUSION Our results suggest that associations between measures of overall and visceral body fat and subclinical atherosclerosis are not mediated by inflammation as measured by CRP and GlycA. Obesity may exert cardiovascular risk via other markers of systemic inflammation.
Collapse
Affiliation(s)
- T Christen
- Department of Clinical Epidemiology, Leiden University Medical Center (LUMC), PO Box 9600, 2300, RC, Leiden, the Netherlands.
| | - S Trompet
- Department of Medicine, Division of Gerontology and Geriatrics, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - P C N Rensen
- Department of Medicine, Division of Endocrinology, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - K Willems van Dijk
- Department of Medicine, Division of Endocrinology, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands; Department of Human Genetics, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - H J Lamb
- Department of Radiology, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - J W Jukema
- Department of Cardiology, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - F R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center (LUMC), PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - S le Cessie
- Department of Clinical Epidemiology, Leiden University Medical Center (LUMC), PO Box 9600, 2300, RC, Leiden, the Netherlands; Department of Biomedical Data Sciences, LUMC, PO Box 9600, 2300, RC, Leiden, the Netherlands
| | - R de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center (LUMC), PO Box 9600, 2300, RC, Leiden, the Netherlands
| |
Collapse
|
20
|
Murabito JM, Zhao Q, Larson MG, Rong J, Lin H, Benjamin EJ, Levy D, Lunetta KL. Measures of Biologic Age in a Community Sample Predict Mortality and Age-Related Disease: The Framingham Offspring Study. J Gerontol A Biol Sci Med Sci 2019; 73:757-762. [PMID: 28977464 DOI: 10.1093/gerona/glx144] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/19/2017] [Indexed: 01/19/2023] Open
Abstract
Background We tested the association of biologic age (BA) measures constructed from different types of biomarkers with mortality and disease in a community-based sample. Methods In Framingham Offspring participants at Exams 7 (1998-2001, mean age 62 ± 10) and 8 (2005-2008, mean age 67 ± 9), we used the Klemera-Doubal method to estimate clinical BA and inflammatory BA and computed the difference (∆age) between BA and CA. Clinical ∆age was computed at Exam 2 (1979-1983, mean age 45 ± 10). At Exam 8, we computed measures of intrinsic and extrinsic epigenetic age. Participants were followed through 2014 for outcomes. Cox proportional hazards models tested the association of each BA estimate with each outcome adjusting for covariates. Results Sample sizes ranged from 2532 to 3417 participants. In multivariable models, each 1-year increase in clinical ∆age at Exam 2 (hazard ratio [HR] = 1.04-1.06, p < 2 × 10-16) and clinical ∆age and inflammatory ∆age at Exam 7 significantly increased the hazards of mortality and incident cardiovascular disease (HR = 1.01-1.05, p < 2 × 10-7), whereas inflammatory ∆age increased the hazards of cancer (HR = 1.01, p < .05). At Exam 8, increased clinical ∆age, inflammatory ∆age, and extrinsic epigenetic age all significantly increased the hazard of mortality (HR = 1.03-1.05, all p < .05); clinical ∆age and inflammatory ∆age increased cardiovascular disease risk (HR = 1.04-1.05, all p < .01); and clinical ∆age increased cancer risk (HR = 1.03, p < .01) when all three BA measures were included in the model. Intrinsic epigenetic age was not significantly associated with any outcome. Conclusions Our findings suggest BA measures may be complementary in predicting risk for mortality and age-related disease.
Collapse
Affiliation(s)
- Joanne M Murabito
- Framingham Heart Study, Massachusetts
- Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Massachusetts
| | - Qiang Zhao
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | - Martin G Larson
- Framingham Heart Study, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | - Jian Rong
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| | - Honghuang Lin
- Framingham Heart Study, Massachusetts
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Massachusetts
| | - Emelia J Benjamin
- Framingham Heart Study, Massachusetts
- Department of Medicine, Section of Cardiovascular Medicine and Preventive Medicine, Boston University School of Medicine, Massachusetts
- Department of Epidemiology, Boston University School of Public Health, Massachusetts
| | - Daniel Levy
- Framingham Heart Study, Massachusetts
- The Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Kathryn L Lunetta
- Framingham Heart Study, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Massachusetts
| |
Collapse
|
21
|
Li W, Nyhan MM, Wilker EH, Vieira CLZ, Lin H, Schwartz JD, Gold DR, Coull BA, Aba AM, Benjamin EJ, Vasan RS, Koutrakis P, Mittleman MA. Recent exposure to particle radioactivity and biomarkers of oxidative stress and inflammation: The Framingham Heart Study. ENVIRONMENT INTERNATIONAL 2018; 121:1210-1216. [PMID: 30376999 PMCID: PMC6279550 DOI: 10.1016/j.envint.2018.10.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 05/21/2023]
Abstract
BACKGROUND Decay products of radioactive materials may attach to ambient fine particles and form radioactive aerosol. Internal ionizing radiation source from inhaled radioactive aerosol may contribute to the fine particulate matter (PM2.5)-inflammation pathway. However, few studies in humans have examined the associations. OBJECTIVES To examine the associations between particle radioactivity and biomarkers of oxidative stress and inflammation among participants from the Framingham Offspring and Third Generation cohorts. METHODS We included 3996 participants who were not current smokers and lived within 50 km from our central air pollution monitoring station. We estimated regional mean gross beta radioactivity from monitors in the northeastern U.S. as a surrogate for ambient radioactive particles, and calculated the 1- to 28-day moving averages. We used linear regression models for fibrinogen, tumor necrosis factor α, interleukin-6, and myeloperoxidase which were measured once, and linear mixed effect models for 8-epi-prostaglandin F2α, C-reactive protein, intercellular adhesion molecule-1 (ICAM-1), monocyte chemoattractant protein-1 (MCP-1), P-selectin, and tumor necrosis factor receptor-2 that were measured up to twice, adjusting for demographics, individual- and area-level socioeconomic positions, time, meteorology, and PM2.5. We also examined whether the associations differed by median age, sex, diabetes status, PM2.5 levels, and black carbon levels. RESULTS The mean age was 54 years and 54% were women. An interquartile range (3 × 10-3 pCi/m3) higher beta radioactivity level at the 7-day moving average was associated with 5.09% (95% CI: 0.92, 9.43), 2.65% (1.10, 4.22), and 4.71% (95% CI: 3.01, 6.44) higher levels of interleukin-6, MCP-1, and P-selectin, but with 7.01% (95% CI: -11.64, -2.15) and 2.70% (95% CI: -3.97, -1.42) lower levels of 8-epi-prostaglandin F2α and ICAM-1, respectively. CONCLUSIONS Regional mean particle radioactivity was positively associated with interleukin-6, MCP-1, and P-selectin, but negatively with ICAM-1 and 8-epi-prostaglandin F2α among our study participants.
Collapse
Affiliation(s)
- Wenyuan Li
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Cardiovascular Epidemiology Research Unit, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Marguerite M Nyhan
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Elissa H Wilker
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Cardiovascular Epidemiology Research Unit, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Carolina L Z Vieira
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Honghuang Lin
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Joel D Schwartz
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Diane R Gold
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | | | - Emelia J Benjamin
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States; Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Ramachandran S Vasan
- Department of Medicine, Boston University School of Medicine, Boston, MA, United States; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, United States; Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Murray A Mittleman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Cardiovascular Epidemiology Research Unit, Division of Cardiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
22
|
|
23
|
Krzanowski M, Krzanowska K, Dumnicka P, Gajda M, Woziwodzka K, Fedak D, Grodzicki T, Litwin JA, Sułowicz W. Elevated Circulating Osteoprotegerin Levels in the Plasma of Hemodialyzed Patients With Severe Artery Calcification. Ther Apher Dial 2018; 22:519-529. [PMID: 29974642 DOI: 10.1111/1744-9987.12681] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/05/2018] [Accepted: 02/15/2018] [Indexed: 12/30/2022]
Abstract
We studied the correlations between circulating osteoprotegerin (OPG) level and radial artery calcification (RAC) assessed histologically and carotid artery intima-media thickness (CCA-IMT). Moreover, we studied the relationship between OPG levels and all-cause and cardiovascular (CV) mortality during a 5-year observation period. The study comprised 59 CKD patients (36 hemodialyzed (HD), 23 predialysis). The biochemical parameters included: creatinine, calcium, phosphate, intact parathormone, C-reactive protein, interleukin-6, tumor necrosis factor receptor II (TNFRII), transforming growth factor-β, hepatocyte growth factor, fibroblast growth factor 23, osteonectin (ON), osteopontin, osteoprotegerin, and osteocalcin. CCA-IMT and the presence of atherosclerotic plaques was assessed by ultrasound. Fragments of radial artery obtained during creation of HD access were prepared for microscopy and stained for calcifications with alizarin red. RAC was detected in 34 patients (58%). In multiple regression adjusted for dialysis status, TNFRII, ON and Framingham risk score (FRS) were identified as the independent predictors of OPG. Serum OPG above the median value of 7.55 pmol/L significantly predicted the presence of RAC in simple logistic regression (OR 5.33; 95%CI 1.39-20.4; P = 0.012) and in multiple logistic regression adjusted for FRS, dialysis status and CCA-IMT values (OR 6.56; 95%CI 1.06-40.6; P = 0.036). OPG levels above the median were associated with higher CCA-IMT values (1.02 ± 0.10 vs. 0.86 ± 0.13; P < 0.001) and predicted the presence of atherosclerotic plaques in carotid artery (OR 14.4; 95%CI 2.84-72.9; P < 0.001), independently of FRS, dialysis status and RAC. In this study, elevated serum OPG levels correlated with higher CCA-IMT, the presence of atherosclerotic plaques and the severity of the RAC independently of each other. During follow-up, 25 patients (42%) died, including 21 due to CV causes. In multiple Cox regression, OPG above the median predicted overall survival independently of dialysis status, Framingham risk score, CCA-IMT above the median value, and the presence of atherosclerotic plaques in CCA, but not independently of RAC. We postulate that circulating OPG may play a dual role as a marker for both medial arterial calcification and atherosclerosis, hence it seems to be a valuable tool for assessing CV risk in patients with CKD. OPG might be an early indicator of all-cause mortality in CKD patients with advanced medial arterial calcification.
Collapse
Affiliation(s)
- Marcin Krzanowski
- Department of Nephrology, Jagiellonian University Medical College, Krakow, Poland
| | - Katarzyna Krzanowska
- Department of Nephrology, Jagiellonian University Medical College, Krakow, Poland
| | - Paulina Dumnicka
- Department of Medical Diagnostics, Jagiellonian University Medical College, Krakow, Poland
| | - Mariusz Gajda
- Department of Histology, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Woziwodzka
- Department of Nephrology, Jagiellonian University Medical College, Krakow, Poland
| | - Danuta Fedak
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz Grodzicki
- Department of Internal Medicine and Gerontology, Jagiellonian University Medical College, Krakow, Poland
| | - Jan A Litwin
- Department of Histology, Jagiellonian University Medical College, Krakow, Poland
| | - Władysław Sułowicz
- Department of Nephrology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
24
|
Shimanoe C, Hara M, Nishida Y, Nanri H, Otsuka Y, Horita M, Yasukata J, Miyoshi N, Yamada Y, Higaki Y, Tanaka K. Coping strategy and social support modify the association between perceived stress and C-reactive protein: a longitudinal study of healthy men and women. Stress 2018; 21:237-246. [PMID: 29402173 DOI: 10.1080/10253890.2018.1435638] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Inconsistent associations have been reported between perceived stress and C-reactive protein (CRP), a marker of systemic inflammation. We previously observed a male-specific inverse relationship between perceived stress and CRP in a cross-sectional study. In the present study, we examined the longitudinal association between changes in perceived stress and CRP, and further analyzed whether changes in coping strategies and social support modify this association. This study included 8454 participants in both a baseline survey and a follow-up survey 5 years later. Psychosocial measures (i.e. perceived stress, coping strategies, and social support) and CRP concentrations were measured by identical means in both surveys. Consistent with our previous findings, increased perceived stress was significantly associated with lower CRP in men (ptrend = .037), but not in women. Increased "emotional expression," a coping strategy, was also associated with lower CRP in women (ptrend = .024). Furthermore, interactions between perceived stress and a coping strategy (positive reappraisal) or social support on CRP were found in men (pinteraction = .007 and .038, respectively); the above inverse association between stress and CRP was not detected for participants with diminished positive reappraisal or social support. In conclusion, increases in perceived stress during a 5-year period were associated with decreases in CRP among healthy men, and the observed association was possibly modified by coping strategy or social support.
Collapse
Affiliation(s)
- Chisato Shimanoe
- a Department of Preventive Medicine, Faculty of Medicine , Saga University , Saga , Japan
| | - Megumi Hara
- a Department of Preventive Medicine, Faculty of Medicine , Saga University , Saga , Japan
| | - Yuichiro Nishida
- a Department of Preventive Medicine, Faculty of Medicine , Saga University , Saga , Japan
| | - Hinako Nanri
- b Department of Nutritional Science , National Institute of Health and Nutrition , Tokyo , Japan
| | - Yasuko Otsuka
- a Department of Preventive Medicine, Faculty of Medicine , Saga University , Saga , Japan
| | - Mikako Horita
- a Department of Preventive Medicine, Faculty of Medicine , Saga University , Saga , Japan
| | - Jun Yasukata
- c Laboratory of Exercise Physiology, Faculty of Sports and Health Science , Fukuoka University , Fukuoka , Japan
| | - Nobuyuki Miyoshi
- d Department of Childhood Care Education , Seika Women's Junior College , Fukuoka , Japan
| | - Yosuke Yamada
- b Department of Nutritional Science , National Institute of Health and Nutrition , Tokyo , Japan
| | - Yasuki Higaki
- c Laboratory of Exercise Physiology, Faculty of Sports and Health Science , Fukuoka University , Fukuoka , Japan
| | - Keitaro Tanaka
- a Department of Preventive Medicine, Faculty of Medicine , Saga University , Saga , Japan
| |
Collapse
|
25
|
Sumner JA, Chen Q, Roberts AL, Winning A, Rimm EB, Gilsanz P, Glymour MM, Tworoger SS, Koenen KC, Kubzansky LD. Cross-Sectional and Longitudinal Associations of Chronic Posttraumatic Stress Disorder With Inflammatory and Endothelial Function Markers in Women. Biol Psychiatry 2017; 82:875-884. [PMID: 28778657 PMCID: PMC5683901 DOI: 10.1016/j.biopsych.2017.06.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/24/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD) may contribute to heightened cardiovascular disease risk by promoting a proinflammatory state and impaired endothelial function. Previous research has demonstrated associations of PTSD with inflammatory and endothelial function biomarkers, but most work has been cross-sectional and does not separate the effects of trauma exposure from those of PTSD. METHODS We investigated associations of trauma exposure and chronic PTSD with biomarkers of inflammation (C-reactive protein and tumor necrosis factor alpha receptor II) and endothelial function (intercellular adhesion molecule-1 and vascular cell adhesion molecule-1) in 524 middle-aged women in the Nurses' Health Study II. Using linear mixed models, we examined associations of trauma/PTSD status with biomarkers measured twice, 10 to 16 years apart, in cardiovascular disease-free women, considering either average levels over time (cross-sectional) or change in levels over time (longitudinal). Biomarker levels were log-transformed. Trauma/PTSD status (based on structured diagnostic interviews) was defined as no trauma at either blood draw (n = 175), trauma at blood draw 1 but no PTSD at either draw (n = 175), and PTSD that persisted beyond blood draw 1 (chronic PTSD; n = 174). The reference group was women without trauma. RESULTS In models adjusted for known potential confounders, women with chronic PTSD had higher average C-reactive protein (B = 0.27, p < .05), tumor necrosis factor alpha receptor II (B = 0.07, p < .01), and intercellular adhesion molecule-1 (B = 0.04, p < .05) levels. Women with trauma but without PTSD had higher average tumor necrosis factor alpha receptor II levels (B = 0.05, p < .05). In addition, women with chronic PTSD had a greater increase in vascular cell adhesion molecule-1 over time (B = 0.003, p < .05). CONCLUSIONS Increased inflammation and impaired endothelial function may be pathways by which chronic PTSD increases cardiovascular disease risk.
Collapse
Affiliation(s)
- Jennifer A. Sumner
- Center for Behavioral Cardiovascular Health, Columbia University Medical Center, New York, NY,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Correspondence to: Jennifer Sumner, Center for Behavioral Cardiovascular Health, Columbia University Medical Center, 622 W. 168th St, PH 9-315, New York, NY 10032. Tel: 212-342-3133; Fax: 212-342-3431;
| | - Qixuan Chen
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY
| | - Andrea L. Roberts
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Ashley Winning
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Eric B. Rimm
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Paola Gilsanz
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA
| | - M. Maria Glymour
- Department of Epidemiology and Biostatistics, University of California San Francisco School of Medicine, San Francisco, CA
| | - Shelley S. Tworoger
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Karestan C. Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA,Psychiatric and Neurodevelopmental Genetics Unit and Department of Psychiatry, Massachusetts General Hospital, Boston, MA
| | - Laura D. Kubzansky
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
26
|
Lin H, Lunetta KL, Zhao Q, Rong J, Benjamin EJ, Mendelson MM, Joehanes R, Levy D, Larson MG, Murabito JM. Transcriptome-wide association study of inflammatory biologic age. Aging (Albany NY) 2017; 9:2288-2301. [PMID: 29135455 PMCID: PMC5723687 DOI: 10.18632/aging.101321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/02/2017] [Indexed: 05/16/2023]
Abstract
Chronic low grade inflammation is a fundamental mechanism of aging. We estimated biologic age using nine biomarkers from diverse inflammatory pathways and we hypothesized that genes associated with inflammatory biological age would provide insights into human aging. In Framingham Offspring Study participants at examination 8 (2005 to 2008), we used the Klemera-Doubal method to estimate inflammatory biologic age and we computed the difference (∆Age) between biologic age and chronologic age. Gene expression in whole blood was measured using the Affymetrix Human Exon 1.0 ST Array. We used linear mixed effect models to test associations between inflammatory ∆Age and gene expression (dependent variable) adjusting for age, sex, imputed cell counts, and technical covariates. Our study sample included 2386 participants (mean age 67A±9 years, 55% women). There were 448 genes significantly were associated with inflammatory ∆Age (P<2.8x10-6), 302 genes were positively associated and 146 genes were negatively associated. Pathway analysis among the identified genes highlighted the NOD-like receptor signaling and ubiquitin mediated proteolysis pathways. In summary, we identified 448 genes that were significantly associated with inflammatory biologic age. Future functional characterization may identify molecular interventions to delay aging and prolong healthspan in older adults.
Collapse
Affiliation(s)
- Honghuang Lin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kathryn L. Lunetta
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Qiang Zhao
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Jian Rong
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - 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
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, USA
| | - Michael M. Mendelson
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roby Joehanes
- Hebrew SeniorLife, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Levy
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Martin G. Larson
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Joanne M. Murabito
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA
- Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
27
|
Patel MS, Miranda-Nieves D, Chen J, Haller CA, Chaikof EL. Targeting P-selectin glycoprotein ligand-1/P-selectin interactions as a novel therapy for metabolic syndrome. Transl Res 2017; 183:1-13. [PMID: 28034759 PMCID: PMC5393932 DOI: 10.1016/j.trsl.2016.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 11/13/2016] [Indexed: 12/22/2022]
Abstract
Obesity-induced insulin resistance and metabolic syndrome continue to pose an important public health challenge worldwide as they significantly increase the risk of type 2 diabetes and atherosclerotic cardiovascular disease. Advances in the pathophysiologic understanding of this process has identified that chronic inflammation plays a pivotal role. In this regard, given that both animal models and human studies have demonstrated that the interaction of P-selectin glycoprotein ligand-1 (PSGL-1) with P-selectin is not only critical for normal immune response but also is upregulated in the setting of metabolic syndrome, PSGL-1/P-selectin interactions provide a novel target for preventing and treating resultant disease. Current approaches of interfering with PSGL-1/P-selectin interactions include targeted antibodies, recombinant immunoglobulins that competitively bind P-selectin, and synthetic molecular therapies. Experimental models as well as clinical trials assessing the role of these modalities in a variety of diseases have continued to contribute to the understanding of PSGL-1/P-selectin interactions and have demonstrated the difficulty in creating clinically relevant therapeutics. Most recently, however, computational simulations have further enhanced our understanding of the structural features of PSGL-1 and related glycomimetics, which are responsible for high-affinity selectin interactions. Leveraging these insights for the design of next generation agents has thus led to development of a promising synthetic method for generating PSGL-1 glycosulfopeptide mimetics for the treatment of metabolic syndrome.
Collapse
Affiliation(s)
- Madhukar S Patel
- Department of Surgery, Massachusetts General Hospital, Boston, Mass; Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass
| | - David Miranda-Nieves
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass; Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Mass
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Carolyn A Haller
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
28
|
Spracklen CN, Smith CJ, Saftlas AF, Triche EW, Bjonnes A, Keating BJ, Saxena R, Breheny PJ, Dewan AT, Robinson JG, Hoh J, Ryckman KK. Genetic predisposition to elevated levels of C-reactive protein is associated with a decreased risk for preeclampsia. Hypertens Pregnancy 2017; 36:30-35. [PMID: 27657194 PMCID: PMC5538572 DOI: 10.1080/10641955.2016.1223303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 08/03/2016] [Accepted: 08/08/2016] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To examine the association between genetic predisposition to elevated C-reactive protein (CRP)and risk for preeclampsia using validated genetic loci for C-reactive protein. METHODS Preeclampsia cases (n = 177) and normotensive controls (n = 116) were selected from live birth certificates to nulliparous Iowa women during the period August 2002-May 2005. Disease status was verified by the medical chart review. Genetic predisposition to CRP was estimated by a genetic risk score on the basis of established loci for CRP levels. Logistic regression analyses were used to evaluate the relationships between the genotype score and preeclampsia. Replication analyses were performed in an independent, US population of preeclampsia cases (n = 516) and controls (n = 1,097) of European ancestry. RESULTS The genetic risk score (GRS) related to higher levels of CRP demonstrated a significantly decreased risk of preeclampsia (OR 0.89, 95% CI 0.82-0.96). When the GRS was analyzed by quartile, an inverse linear trend was observed (p = 0.0006). The results were similar after adjustments for the body mass index (BMI), smoking, and leisure-time physical activity. In the independent replication population, the association with the CRP GRS was also marginally significant (OR 0.97, 95% CI 0.92, 1.02). Meta-analysis of the two studies was statistically significant (OR 0.95, 95% CI 0.90, 0.99). CONCLUSION Our data suggest an inverse, counterintuitive association between the genetic predisposition to elevated levels of CRP and a decreased risk of preeclampsia. This suggests that the blood CRP level is a marker of preeclampsia, but it does not appear to be a factor on the causal pathway.
Collapse
Affiliation(s)
- Cassandra N. Spracklen
- Present address: Department of Genetics, University of North Carolina-Chapel Hill, 5100 Genetic Medicine Building, CB #7264, 120 Mason Farm Road, Chapel Hill, NC 27599 (work was performed at Department of Epidemiology, University of Iowa College of Public Health, 145 Riverside Drive, S471 CPHB, Iowa City, IA 52242)
| | - Caitlin J. Smith
- Department of Epidemiology, University of Iowa College of Public Health, 145 Riverside Drive, S471 CPHB, Iowa City, IA 52242
| | - Audrey F. Saftlas
- Department of Epidemiology, University of Iowa College of Public Health, 145 Riverside Drive, S427 CPHB, Iowa City, IA 52242
| | - Elizabeth W. Triche
- Department of Epidemiology, Division of Biology and Medicine, Brown University, 121 S. Main St., 2 floor, Box G-S121-2, Providence, Rhode Island
| | - Andrew Bjonnes
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5, Boston, MA 02114 and Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge MA 02142
| | - Brendan J. Keating
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, Division of Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Richa Saxena
- Center for Human Genetic Research and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 5, Boston, MA 02114 and Program in Medical and Population Genetics, Broad Institute, 7 Cambridge Center, Cambridge MA 02142
| | - Patrick J. Breheny
- Department of Biostatistics, University of Iowa College of Public Health, 145Riverside Drive, N336 CPHB, Iowa City, IA 52242
| | - Andrew T. Dewan
- Division of Chronic Disease Epidemiology, Yale School of Public Health, 60 College Street, Room 403, New Haven, CT, 06520
| | - Jennifer G. Robinson
- Department of Epidemiology, University of Iowa College of Public Health, 145 Riverside Drive, S455 CPHB, Iowa City, IA 52242
| | - Josephine Hoh
- Department of Environmental Health Sciences, Yale School of Public Health, 60 College Street, New Haven, CT, 06520
| | - Kelli K. Ryckman
- Department of Epidemiology, University of Iowa College of Public Health, 145 Riverside Drive, S414 CPHB, Iowa City, IA 52242
| |
Collapse
|
29
|
Wu Z, Huang Z, Jin W, Rimm EB, Lichtenstein AH, Kris-Etherton PM, Wu S, Gao X. Peripheral Inflammatory Biomarkers for Myocardial Infarction Risk: A Prospective Community-Based Study. Clin Chem 2016; 63:663-672. [PMID: 28031418 DOI: 10.1373/clinchem.2016.260828] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 10/18/2016] [Indexed: 11/06/2022]
Abstract
BACKGROUND Most previous studies regarding chronic inflammation and risk of myocardial infarction (MI) have lacked repeated measures of high-sensitivity C-reactive protein (hs-CRP) and/or white blood cell (WBC) count over time. We examined whether cumulative average and longitudinal changes in these biomarkers were associated with subsequent MI risk. METHODS In this prospective, community-based study, we included 82544 Chinese participants [66796 men and 15748 women; mean (SD) age 55.1 (9.86) y] without prior cardiovascular diseases or cancer at baseline (2006-2007). hs-CRP, WBC and other clinical covariates were assessed at baseline and every 2 years during follow-up. RESULTS During 6 years of follow-up (2006-2012), we documented 714 incident MI cases. Higher baseline and cumulative average concentrations of hs-CRP and/or WBC were consistently associated with increased risk of MI (Ptrend <0.001 for both). Longitudinal increase in hs-CRP (Ptrend <0.001), but not WBC, was also associated with a higher future risk of MI, after adjustment for their baseline values and other covariates. Each 1-mg/L increment per year in hs-CRP was associated with a 9.3% increase in risk for future MI [hazard ratio (HR) = 1.09, 95% CI, 1.03; 1.17]. Participants with high-grade inflammatory status (hs-CRP ≥10 mg/L and WBC ≥10 × 109/L) had a higher risk of MI occurring <3 months after hs-CRP/WBC assessments vs those with hs-CRP <0.5 mg/L and WBC <5 × 109/L (HR = 6.64; 95% CI, 1.49-29.6), as compared with MI occurring ≥4 years (HR = 2.95; 95% CI, 0.90, 9.65). CONCLUSIONS Plasma hs-CRP concentration and WBC predicted MI risk. Longitudinal increase in hs-CRP was also associated with a higher risk of MI.
Collapse
Affiliation(s)
- Zhijun Wu
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Zhe Huang
- Department of Cardiology, Kailuan Hospital, Tangshan, People's Republic of China
| | - Wei Jin
- Department of Cardiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Eric B Rimm
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA
| | - Shouling Wu
- Department of Cardiology, Kailuan Hospital, Tangshan, People's Republic of China;
| | - Xiang Gao
- Department of Nutritional Sciences, Pennsylvania State University, State College, PA.
| |
Collapse
|
30
|
Werler MM, Parker SE, Hedman K, Gissler M, Ritvanen A, Surcel HM. Maternal Antibodies to Herpes Virus Antigens and Risk of Gastroschisis in Offspring. Am J Epidemiol 2016; 184:902-912. [PMID: 27856447 DOI: 10.1093/aje/kww114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/08/2016] [Indexed: 12/16/2022] Open
Abstract
Gastroschisis risk is highest in offspring of young women and is increasing in prevalence, suggesting that exposures that are increasingly common among younger females may be causal. Some infections by viruses in the herpes family are more common in the earlier childbearing years and have been increasing in prevalence over time. Data from the Finnish Maternity Cohort were linked to Finnish malformation and birth registers (1987-2012) for this study, a nested case-control study of mothers of offspring with gastroschisis and age-matched controls. Maternal antibody responses in early pregnancy (mean gestational age = 11.1 weeks) to Epstein Barr virus (EBV), herpes simplex virus types 1 and 2 (HSV-1 and HSV-2), and cytomegalovirus were measured. Conditional logistic regression models were used to estimate odds ratios (and 95% confidence intervals) for high immunoglobulin reactivity. Odds ratios for high immunoglobulin M (IgM) reactivity to EBV-viral capsid antigen and HSV-1 or HSV-2 (as indicators of recent infection) were 2.16 (95% confidence interval (CI): 0.97, 4.79) and 1.94 (95% CI: 0.74, 5.12), respectively. For higher immunoglobulin G (IgG) reactivity to EBV-viral capsid antigen and HSV-2 IgG, odds ratios were 2.16 (95% CI: 0.82, 5.70) and 2.48 (95% CI: 1.50, 4.10), respectively. Reactivities to HSV-1 IgG, cytomegalovirus IgM, or cytomegalovirus IgG did not appear to increase gastroschisis risk. Primary EBV infection was not associated with gastroschisis, but observed associations with both IgM and IgG reactivities to EBV and HSV suggest that reactivations may be risk factors for it.
Collapse
|
31
|
Malik A, Saleem S, Basit Ashraf MA, Qazi MH. 1, 25-dihydroxyvitamin D 3, a potential role player in the development of thyroid disorders in schizophrenics. Pak J Med Sci 2016; 32:1370-1374. [PMID: 28083028 PMCID: PMC5216284 DOI: 10.12669/pjms.326.11157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective: The present study was designed to assess the role of vitamin-D, in the development of autoimmune thyroid dysfunction in newly diagnosed schizophrenics. Methods: For the present study 100 patients and 100 controls were screened out and studied for their thyroid antibodies, GSH, homocysteine, NOS and vitamin D levels by appropriate protocols to assess the underlying mechanism involved in the schizophrenics susceptible to autoimmune thyroid diseases. Results: The results of the present study depicted that in schizophrenics, levels of cytokines like IL-6 (7.98±0.67 pg/ml), TNF-α, (40.76±6.98 pg/ml), homocysteine (16.98±1.09 µmol/L), Tg-Ab (30.93±3.87 IU/L), TPO-Ab (10.33±1.78 IU/L) and TSHr-Ab (3.76±0.055 IU/L) increased whereas, those of Vit-D (12.76±0.99 pmol/L), NOS (5.99±0.87 IU/L), GSH (4.48±.965 µg/dl) and NO (16.87±3.98 ng/ml) were decreased in the patients as compared to healthy control subjects. Conclusion: Vitamin-D in schizophrenia is involved in augmentation of hyperhomocysteinemia, inflammation, oxidative stress and thyroid antibodies, thereby playing a significant role not only in induction of schizophrenic symptoms but may also result in autoimmune thyroid diseases. Thus, earlier detection and rectification of its levels are helpful to limit the miseries of schizophrenia.
Collapse
Affiliation(s)
- Arif Malik
- Dr. Arif Malik, PhD, Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Shamaila Saleem
- Dr. Shamaila Saleem, MBBS, M.Phil, Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Muhammad Abdul Basit Ashraf
- Dr. Muhammad Abdul Basit Ashraf, MBBS, Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Mahmood Husain Qazi
- Dr. Mahmood Husain Qazi, PhD. Centre for Research in Molecular Medicine (CRiMM), The University of Lahore, Lahore, Pakistan
| |
Collapse
|
32
|
Potential Diagnostic and Prognostic Biomarkers of Epigenetic Drift within the Cardiovascular Compartment. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2465763. [PMID: 26942189 PMCID: PMC4749768 DOI: 10.1155/2016/2465763] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/02/2015] [Accepted: 11/24/2015] [Indexed: 12/15/2022]
Abstract
Biomarkers encompass a wide range of different measurable indicators, representing a tangible link to physiological changes occurring within the body. Accessibility, sensitivity, and specificity are significant factors in biomarker suitability. New biomarkers continue to be discovered, and questions over appropriate selection and assessment of their usefulness remain. If traditional markers of inflammation are not sufficiently robust in their specificity, then perhaps alternative means of detection may provide more information. Epigenetic drift (epigenetic modifications as they occur as a direct function with age), and its ancillary elements, including platelets, secreted microvesicles (MVs), and microRNA (miRNA), may hold enormous predictive potential. The majority of epigenetic drift observed in blood is independent of variations in blood cell composition, addressing concerns affecting traditional blood-based biomarker efficacy. MVs are found in plasma and other biological fluids in healthy individuals. Altered MV/miRNA profiles may also be found in individuals with various diseases. Platelets are also highly reflective of physiological and lifestyle changes, making them extremely sensitive biomarkers of human health. Platelets release increased levels of MVs in response to various stimuli and under a plethora of disease states, which demonstrate a functional effect on other cell types.
Collapse
|
33
|
Xie X, Song X, Yuan S, Cai H, Chen Y, Chang X, Liang B, Huang D. Histone acetylation regulates orphan nuclear receptor NR4A1 expression in hypercholesterolaemia. Clin Sci (Lond) 2015; 129:1151-61. [PMID: 26396259 DOI: 10.1042/cs20150346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/21/2015] [Indexed: 02/04/2023]
Abstract
Hypercholesterolaemia and inflammation are correlated with atherogenesis. Orphan nuclear receptor NR4A1, as a key regulator of inflammation, is closely associated with lipid levels in vivo. However, the mechanism by which lipids regulate NR4A1 expression remains unknown. We aimed to elucidate the underlying mechanism of NR4A1 expression in monocytes during hypercholesterolaemia, and reveal the potential role of NR4A1 in hypercholesterolaemia-induced circulating inflammation. Circulating leucocytes were collected from blood samples of 139 patients with hypercholesterolaemia and 139 sex- and age-matched healthy subjects. We found that there was a low-grade inflammatory state and higher expression of NR4A1 in patients. Both total cholesterol and low-density lipoprotein cholesterol levels in plasma were positively correlated with NR4A1 mRNA level. ChIP revealed that acetylation of histone H3 was enriched in the NR4A1 promoter region in patients. Human mononuclear cell lines THP-1 and U937 were treated with cholesterol. Supporting our clinical observations, cholesterol enhanced p300 acetyltransferase and decreased HDAC7 (histone deacetylase 7) recruitment to the NR4A1 promoter region, resulting in histone H3 hyperacetylation and further contributing to NR4A1 up-regulation in monocytes. Moreover, cytosporone B, an NR4A1 agonist, completely reversed cholesterol-induced IL-6 (interleukin 6) and MCP-1 (monocyte chemoattractant protein 1) expression to below basal levels, and knockdown of NR4A1 expression by siRNA not only mimicked, but also exaggerated the effects of cholesterol on inflammatory biomarker up-regulation. Thus we conclude that histone acetylation contributes to the regulation of NR4A1 expression in hypercholesterolaemia, and that NR4A1 expression reduces hypercholesterolaemia-induced inflammation.
Collapse
MESH Headings
- Acetylation
- Adult
- Aged
- Binding Sites
- Case-Control Studies
- Chemokine CCL2/metabolism
- Cholesterol/metabolism
- Female
- Gene Expression Regulation
- Histone Deacetylases/metabolism
- Histones/metabolism
- Humans
- Hypercholesterolemia/blood
- Hypercholesterolemia/genetics
- Hypercholesterolemia/metabolism
- Inflammation/blood
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/prevention & control
- Inflammation Mediators/blood
- Inflammation Mediators/metabolism
- Interleukin-6/metabolism
- Male
- Middle Aged
- Monocytes/drug effects
- Monocytes/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/blood
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Phenylacetates/pharmacology
- Promoter Regions, Genetic
- Protein Processing, Post-Translational
- RNA Interference
- RNA, Messenger/metabolism
- Transfection
- U937 Cells
- p300-CBP Transcription Factors/metabolism
Collapse
Affiliation(s)
- Xina Xie
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Xuhong Song
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Song Yuan
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Haitao Cai
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Yequn Chen
- Department of Community Surveillance, The First Affiliated Hospital of Shantou, University Medical College, Shantou, 515041, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Bin Liang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Key Laboratory of Molecular Biology in High Cancer Incidence Coastal Chaoshan Area of Guangdong Higher Education Institutes, Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
34
|
Pilling LC, Joehanes R, Melzer D, Harries LW, Henley W, Dupuis J, Lin H, Mitchell M, Hernandez D, Ying SX, Lunetta KL, Benjamin EJ, Singleton A, Levy D, Munson P, Murabito JM, Ferrucci L. Gene expression markers of age-related inflammation in two human cohorts. Exp Gerontol 2015; 70:37-45. [PMID: 26087330 PMCID: PMC4600657 DOI: 10.1016/j.exger.2015.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/13/2015] [Accepted: 05/19/2015] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Chronically elevated circulating inflammatory markers are common in older persons but mechanisms are unclear. Many blood transcripts (>800 genes) are associated with interleukin-6 protein levels (IL6) independent of age. We aimed to identify gene transcripts statistically mediating, as drivers or responders, the increasing levels of IL6 protein in blood at older ages. METHODS Blood derived in-vivo RNA from the Framingham Heart Study (FHS, n=2422, ages 40-92 yrs) and InCHIANTI study (n=694, ages 30-104 yrs), with Affymetrix and Illumina expression arrays respectively (>17,000 genes tested), were tested for statistical mediation of the age-IL6 association using resampling techniques, adjusted for confounders and multiple testing. RESULTS In FHS, IL6 expression was not associated with IL6 protein levels in blood. 102 genes (0.6% of 17,324 expressed) statistically mediated the age-IL6 association of which 25 replicated in InCHIANTI (including 5 of the 10 largest effect genes). The largest effect gene (SLC4A10, coding for NCBE, a sodium bicarbonate transporter) mediated 19% (adjusted CI 8.9 to 34.1%) and replicated by PCR in InCHIANTI (n=194, 35.6% mediated, p=0.01). Other replicated mediators included PRF1 (perforin, a cytolytic protein in cytotoxic T lymphocytes and NK cells) and IL1B (Interleukin 1 beta): few other cytokines were significant mediators. CONCLUSIONS This transcriptome-wide study on human blood identified a small distinct set of genes that statistically mediate the age-IL6 association. Findings are robust across two cohorts and different expression technologies. Raised IL6 levels may not derive from circulating white cells in age related inflammation.
Collapse
Affiliation(s)
- Luke C Pilling
- Epidemiology and Public Health, Medical School, University of Exeter, RILD, Exeter EX2 5DW, UK
| | - Roby Joehanes
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA
| | - David Melzer
- Epidemiology and Public Health, Medical School, University of Exeter, RILD, Exeter EX2 5DW, UK
| | - Lorna W Harries
- Institute of Biomedical and Clinical Sciences, Medical School, University of Exeter, RILD, Exeter EX2 5DW, UK
| | - William Henley
- Institute for Health Services Research, University of Exeter Medical School, Exeter EX1 2LU, UK
| | - Josée Dupuis
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Honghuang Lin
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Marcus Mitchell
- Institute of Biomedical and Clinical Sciences, Medical School, University of Exeter, RILD, Exeter EX2 5DW, UK
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sai-Xia Ying
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA
| | - Kathryn L Lunetta
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Emelia J Benjamin
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Section of Cardiovascular Medicine and Preventive Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Levy
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; The Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Peter Munson
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA
| | - Joanne M Murabito
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
35
|
Engelberger RP, Limacher A, Kucher N, Baumann F, Silbernagel G, Benghozi R, Do DD, Willenberg T, Baumgartner I. Biological variation of established and novel biomarkers for atherosclerosis: Results from a prospective, parallel-group cohort study. Clin Chim Acta 2015; 447:16-22. [DOI: 10.1016/j.cca.2015.05.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 01/26/2023]
|
36
|
Fontes JD, Rahman F, Lacey S, Larson MG, Vasan RS, Benjamin EJ, Harris WS, Robins SJ. Red blood cell fatty acids and biomarkers of inflammation: a cross-sectional study in a community-based cohort. Atherosclerosis 2015; 240:431-6. [PMID: 25897795 PMCID: PMC4511480 DOI: 10.1016/j.atherosclerosis.2015.03.043] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 03/13/2015] [Accepted: 03/31/2015] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Inflammation and inflammatory biomarkers have emerged as integral components and predictors of incident cardiovascular (CV) disease. Omega-3 fatty acids, particularly eicosapentaenoic and docosahexaenoic acids (EPA and DHA) have anti-inflammatory properties, and have been variably associated with lower blood pressure, favorable blood lipid changes, and reduced CV events. METHODS AND RESULTS We examined the cross-sectional association of red blood cell (RBC) fatty acids, representative of body membrane fatty acid composition, with 10 biomarkers active in multiple inflammatory pathways in 2724 participants (mean age 66 ± 9 years, 54% women, 8% minorities) from the Framingham Offspring and minority Omni Cohorts. After multivariable adjustment, the RBC EPA and DHA content was inversely correlated (all P ≤ 0.001) with 8 biomarkers: urinary isoprostanes (r = -0.16); and soluble interleukin-6 (r = -0.10); C-reactive protein (r = -0.08); tumor necrosis factor receptor 2 (r = -0.08); intercellular adhesion molecule-1 (r = -0.08); P-selectin (r = -0.06); lipoprotein-associated phospholipase-A2 mass (r = -0.11) and activity (r = -0.08). The correlations for monocyte chemoattractant protein-1 was -0.05, P = 0.006 and osteoprotegerin (r = -0.06, P = 0.002) were only nominally significant. CONCLUSION In our large community-based study, we observed modest inverse associations between several types of inflammatory biomarkers with RBC omega-3 fatty acid levels. Our findings are consistent with the hypothesis that omega-3 fatty acids have anti-inflammatory properties.
Collapse
Affiliation(s)
- João D Fontes
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Cardiology and Preventive Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Faisal Rahman
- Boston University Medical Center, Boston, MA 02118, USA
| | - Sean Lacey
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
| | - Martin G Larson
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Mathematics and Statistics, Boston University, Boston, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ramachandran S Vasan
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Cardiology and Preventive Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Epidemiology Department, Boston University School of Public Health, Boston, MA, USA
| | - Emelia J Benjamin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Cardiology and Preventive Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Epidemiology Department, Boston University School of Public Health, Boston, MA, USA
| | - William S Harris
- Department of Medicine, Sanford School of Medicine, University of South Dakota, and OmegaQuant Analytics, LLC, 5009 W. 12th St, Ste 8, Sioux Falls, SD 57106, USA.
| | - Sander J Robins
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Cardiology and Preventive Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
37
|
Alehagen U, Lindahl TL. sP-selectin is a useful biomarker for cardiovascular risk. Cardiovasc Endocrinol 2015. [DOI: 10.1097/xce.0000000000000042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
38
|
Davenport C, Ashley DT, O'Sullivan EP, McHenry CM, Agha A, Thompson CJ, O'Gorman DJ, Smith D. The Effects of Atorvastatin on Arterial Stiffness in Male Patients with Type 2 Diabetes. J Diabetes Res 2015; 2015:846807. [PMID: 26064990 PMCID: PMC4430667 DOI: 10.1155/2015/846807] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 04/01/2015] [Accepted: 04/09/2015] [Indexed: 12/02/2022] Open
Abstract
Statin therapy improves lipid profiles and reduces vascular inflammation, but its effects on central arterial stiffness in type 2 diabetes are unclear. The aim of this study was to determine whether statin therapy reduces central arterial stiffness, in a dose-dependent manner, in male patients with type 2 diabetes. Fifty-one patients ceased statin therapy for 6 weeks, followed by randomisation to either 10 or 80 mg of atorvastatin. At randomization, 3 and 12 months, central arterial stiffness was measured via carotid-femoral pulse wave velocity (PWV), along with serum markers of vascular inflammation including high-sensitivity c-reactive protein (hsCRP) and osteoprotegerin (OPG). PWV decreased from 10.37 ± 1.30 to 9.68 ± 1.19 m/sec (p < 0.01 from baseline) at 3 months and 9.10 ± 1.17 m/sec (p < 0.001 from baseline) at 12 months. hsCRP and OPG decreased significantly at 3 and 12 months. Reductions in PWV did not differ significantly between the groups. Baseline PWV and OPG values correlated strongly (r = 0.48, p < 0.01), as did their response to atorvastatin over 12 months (r = 0.36 delta-OPG and delta-PWV, p < 0.01). Atorvastatin therapy appeared to reduce central arterial stiffness in male type 2 diabetes, with no dose-dependent effect observed. The correlation observed between reductions in PWV and OPG suggests that atorvastatin reduces PWV via direct anti-inflammatory effects on the vasculature.
Collapse
Affiliation(s)
- Colin Davenport
- Department of Academic Endocrinology, Beaumont Hospital, Dublin, Ireland
- *Colin Davenport:
| | - David T. Ashley
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Eoin P. O'Sullivan
- Department of Academic Endocrinology, Beaumont Hospital, Dublin, Ireland
| | - Claire M. McHenry
- Department of Academic Endocrinology, Beaumont Hospital, Dublin, Ireland
| | - Amar Agha
- Department of Academic Endocrinology, Beaumont Hospital, Dublin, Ireland
| | | | - Donal J. O'Gorman
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Diarmuid Smith
- Department of Academic Endocrinology, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
39
|
Lin H, Joehanes R, Pilling LC, Dupuis J, Lunetta KL, Ying SX, Benjamin EJ, Hernandez D, Singleton A, Melzer D, Munson PJ, Levy D, Ferrucci L, Murabito JM. Whole blood gene expression and interleukin-6 levels. Genomics 2014; 104:490-5. [PMID: 25311648 PMCID: PMC4262595 DOI: 10.1016/j.ygeno.2014.10.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/03/2014] [Accepted: 10/03/2014] [Indexed: 12/29/2022]
Abstract
BACKGROUND Circulating interleukin-6 levels increase with advancing age and are a risk factor for various diseases and mortality. The characterization of gene expression profiles associated with interleukin-6 levels might suggest important molecular events underlying its regulation. METHODS AND RESULTS We studied the association of transcriptional profiles with interleukin-6 levels in 2422 participants from the Framingham Heart Study Offspring Cohort using Affymetrix Human Exon 1.0 ST Array. We identified 4139 genes that were significantly associated with interleukin-6 levels (FDR<0.05) after adjusting for age, sex and blood cell components. We then replicated 807 genes in the InCHIANTI study with 694 participants. Many of the top genes are involved in inflammation-related pathways or erythrocyte function, including JAK/Stat signaling pathway and interleukin-10 signaling pathway. CONCLUSION We identified and replicated 807 genes that were associated with circulating interleukin-6 levels. Future characterization of interleukin-6 regulation networks may facilitate the identification of additional potential targets for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Honghuang Lin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA; National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA.
| | - Roby Joehanes
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA; Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Luke C Pilling
- Epidemiology and Public Health, Medical School, University of Exeter, Exeter EX1 2 LU, UK
| | - Josée Dupuis
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Kathryn L Lunetta
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Sai-Xia Ying
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA
| | - Emelia J Benjamin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Section of Cardiovascular Medicine and Preventive Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - David Melzer
- Epidemiology and Public Health, Medical School, University of Exeter, Exeter EX1 2 LU, UK
| | - Peter J Munson
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA; Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Daniel Levy
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Population Sciences Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Luigi Ferrucci
- Clinical Research Branch, National Institute on Aging, Baltimore, MD, USA
| | - Joanne M Murabito
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA; Section of General Internal Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
40
|
Benjamin I, Brown N, Burke G, Correa A, Houser SR, Jones DW, Loscalzo J, Vasan RS, Whitman GR. American Heart Association Cardiovascular Genome-Phenome Study: foundational basis and program. Circulation 2014; 131:100-12. [PMID: 25411155 PMCID: PMC4286232 DOI: 10.1161/circulationaha.114.014190] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Ivor Benjamin
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Nancy Brown
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Gregory Burke
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Adolfo Correa
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Steven R Houser
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Daniel W Jones
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Joseph Loscalzo
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.).
| | - Ramachandran S Vasan
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| | - Gayle R Whitman
- From the Medical College of Wisconsin, Milwaukee (I.B.); American Heart Association, Dallas, TX (N.B., G.W.); Wake Forest University School of Medicine, Winston-Salem, NC (G.B.); Mississippi Medical Center, Jackson Heart Study, Jackson (A.C.); Temple University School of Medicine, Philadelphia, PA (S.R.H.); University of Mississippi, Oxford (D.J.); Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.); and Boston University School of Medicine and Framingham Heart Study, Boston, MA (R.S.V.)
| |
Collapse
|
41
|
Systematic review and metaanalysis on nonclassic cardiovascular biomarkers after hypertensive pregnancy disorders. Am J Obstet Gynecol 2014; 211:373.e1-9. [PMID: 24637129 DOI: 10.1016/j.ajog.2014.03.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 02/09/2014] [Accepted: 03/12/2014] [Indexed: 12/26/2022]
Abstract
OBJECTIVE The aim of this study was to investigate which nonclassic cardiovascular biomarkers are associated with persistent endothelial dysfunction after pregnancy in women with a history of hypertensive pregnancy disorders compared with women with uncomplicated pregnancies. STUDY DESIGN This was a systematic review and metaanalysis of observational studies. A search was performed in PubMed, Embase, Cochrane, and Cinahl including articles from inception to Feb. 27, 2013. Included were cohort studies and case-control studies. Cases were women with a history of hypertension in pregnancy, control subjects were women with a history of uncomplicated pregnancies. Of the 3136 found, 21 studies on 16 nonclassic cardiovascular biomarkers are described in this review; 12 studies on 5 biomarkers were included in the metaanalysis. RESULTS Women with a history of hypertensive pregnancy disorders had a higher homocysteine level compared with women with a history of uncomplicated pregnancies (5 studies; pooled mean difference, 0.77 ng/mL; 95% confidence interval, 0.27-1.26; P < .01). For the other nonclassic cardiovascular biomarkers including markers in areas of inflammation, thrombosis, and angiogenesis, we found no significant differences. CONCLUSION This review and metaanalysis showed that women with a history of hypertensive pregnancy disorders have higher homocysteine levels compared with women with a history of uncomplicated pregnancies. These data suggest persistent endothelial alteration after pregnancies complicated by hypertensive disorders.
Collapse
|
42
|
Kitahara CM, Trabert B, Katki HA, Chaturvedi AK, Kemp TJ, Pinto LA, Moore SC, Purdue MP, Wentzensen N, Hildesheim A, Shiels MS. Body mass index, physical activity, and serum markers of inflammation, immunity, and insulin resistance. Cancer Epidemiol Biomarkers Prev 2014; 23:2840-9. [PMID: 25249326 DOI: 10.1158/1055-9965.epi-14-0699-t] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Epidemiologic studies examining circulating levels of inflammatory markers in relation to obesity and physical inactivity may aid in our understanding of the role of inflammation in obesity-related cancers. However, previous studies on this topic have focused on a limited set of markers. METHODS We evaluated associations between body mass index (BMI) and vigorous physical activity level, based on self-report, and serum levels of 78 inflammation-related markers. Markers were measured using a bead-based multiplex method among 1,703 men and women, ages 55-74 years, and with no prior history of cancer at blood draw, and selected for case-control studies nested within the Prostate, Lung, Ovarian, and Colorectal Cancer Screening Trial. Analyses were adjusted for age, sex, smoking, case-control study, physical activity, and BMI. RESULTS Twelve markers were positively associated with BMI after FDR correction. ORs and 95% confidence interval (CI) for highest versus lowest levels of CCL2/MCP-1, CXCL5/ENA-78, sTNFRII, CXCL10/IP-10, CXCL6/GCP2, CCL13/MCP-4, amylin, CRP, C-peptide, CCL19/MIP-3b, insulin, and leptin were: 1.50 (1.14-1.98), 1.52 (1.12-2.05), 1.61 (1.17-2.20), 1.69 (1.25-2.28), 1.74 (1.24-2.44), 1.75 (1.22-2.50), 1.91 (1.31-2.78), 2.41 (1.36-4.25), 2.78 (1.83-4.24), 3.30 (2.28-4.78), 4.05 (2.51-6.55), and 50.03 (19.87-125.99) per 5 kg/m(2), respectively. Only CXCL12/SDF-1a was associated with physical activity (≥3 vs. <1 h/wk; OR, 3.28; 95% CI, 1.55-6.94) after FDR correction. CONCLUSIONS BMI was associated with a wide range of circulating markers involved in the inflammatory response. IMPACT This cross-sectional analysis identified serum markers could be considered in future studies aimed at understanding the underlying mechanisms linking inflammation with obesity and obesity-related cancers.
Collapse
Affiliation(s)
- Cari M Kitahara
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland.
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Hormuzd A Katki
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Anil K Chaturvedi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Troy J Kemp
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ligia A Pinto
- HPV Immunology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Steven C Moore
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Mark P Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
43
|
Klein R, Myers CE, Cruickshanks KJ, Gangnon RE, Danforth LG, Sivakumaran TA, Iyengar SK, Tsai MY, Klein BEK. Markers of inflammation, oxidative stress, and endothelial dysfunction and the 20-year cumulative incidence of early age-related macular degeneration: the Beaver Dam Eye Study. JAMA Ophthalmol 2014; 132:446-55. [PMID: 24481424 PMCID: PMC4076038 DOI: 10.1001/jamaophthalmol.2013.7671] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
IMPORTANCE Modifying levels of factors associated with age-related macular degeneration (AMD) may decrease the risk for visual impairment in older persons. OBJECTIVE To examine the relationships of markers of inflammation, oxidative stress, and endothelial dysfunction to the 20-year cumulative incidence of early AMD. DESIGN, SETTING, AND PARTICIPANTS This longitudinal population-based cohort study involved a random sample of 975 persons in the Beaver Dam Eye Study without signs of AMD who participated in the baseline examination in 1988-1990 and up to 4 follow-up examinations in 1993-1995, 1998-2000, 2003-2005, and 2008-2010. EXPOSURES Serum markers of inflammation (high-sensitivity C-reactive protein, tumor necrosis factor-α receptor 2, interleukin-6, and white blood cell count), oxidative stress (8-isoprostane and total carbonyl content), and endothelial dysfunction (soluble vascular cell adhesion molecule-1 and soluble intercellular adhesion molecule-1) were measured. Interactions with complement factor H (rs1061170), age-related maculopathy susceptibility 2 (rs10490924), complement component 3 (rs2230199), and complement component 2/complement factor B (rs4151667) were examined using multiplicative models. Age-related macular degeneration was assessed from fundus photographs. MAIN OUTCOMES AND MEASURES Early AMD defined by the presence of any size drusen and the presence of pigmentary abnormalities or by the presence of large-sized drusen (≥125-μm diameter) in the absence of late AMD. RESULTS The 20-year cumulative incidence of early AMD was 23.0%. Adjusting for age, sex, and other risk factors, high-sensitivity C-reactive protein (odds ratio comparing fourth with first quartile, 2.18; P = .005), tumor necrosis factor-α receptor 2 (odds ratio, 1.78; P = .04), and interleukin-6 (odds ratio, 1.78; P = .03) were associated with the incidence of early AMD. Increased incidence of early AMD was associated with soluble vascular cell adhesion molecule-1 (odds ratio per SD on the logarithmic scale, 1.21; P = .04). CONCLUSIONS AND RELEVANCE We found modest evidence of relationships of serum high-sensitivity C-reactive protein, tumor necrosis factor-α receptor 2, interleukin-6, and soluble vascular cell adhesion molecule-1 to the 20-year cumulative incidence of early AMD independent of age, smoking status, and other factors. It is not known whether these associations represent a cause and effect relationship or whether other unknown confounders accounted for the findings. Even if inflammatory processes are a cause of early AMD, it is not known whether interventions that reduce systemic inflammatory processes will reduce the incidence of early AMD.
Collapse
Affiliation(s)
- Ronald Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison2Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison
| | - Chelsea E Myers
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison
| | - Karen J Cruickshanks
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison3Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison
| | - Ronald E Gangnon
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison3Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison
| | - Lorraine G Danforth
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison
| | - Theru A Sivakumaran
- Departments of Epidemiology and Biostatistics, and Genetics and Ophthalmology, Case Western Reserve University, Cleveland, Ohio5Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sudha K Iyengar
- Departments of Epidemiology and Biostatistics, and Genetics and Ophthalmology, Case Western Reserve University, Cleveland, Ohio
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis
| | - Barbara E K Klein
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison
| |
Collapse
|