1
|
Liu Y, Shamaitijiang X, Skudder-Hill L, Kimita W, Sequeira-Bisson IR, Petrov MS. Relationship of high-density lipoprotein subfractions and apolipoprotein A-I with fat in the pancreas. Diabetes Obes Metab 2024. [PMID: 39377129 DOI: 10.1111/dom.15990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024]
Abstract
AIM To investigate the associations of high-density lipoprotein (HDL) subfractions and apolipoprotein A-I (apo A-I) with fat in the pancreas. METHODS A total of 170 individuals were studied. All participants underwent magnetic resonance imaging on a single 3.0-Tesla scanner to determine the presence/absence of fatty pancreas. HDL subfractions were measured using a commercially available lipoprotein subfractions testing system and classed as large, intermediate and small HDL. Both unadjusted and adjusted (accounting for demographics, anthropometrics, insulin resistance and other covariates) logistic regression models were built. RESULTS Individuals with fatty pancreas had significantly lower circulating levels of the large HDL class and apo A-I. Every unit decrease in the large HDL class was associated with a 93% increase in the likelihood of fatty pancreas in the most adjusted model (P < .001). Every unit decrease in apo A-I was associated with a 45% increase in the likelihood of fatty pancreas in the most adjusted model (P = .012). The intermediate and small HDL classes were not significantly associated with fatty pancreas. CONCLUSIONS Fat in the pancreas is inversely associated with the circulating levels of large HDL particles and apo A-I. Purposely designed studies are warranted to investigate the potential of fatty pancreas as an indicator of the risk of cardiovascular diseases.
Collapse
Affiliation(s)
- Yutong Liu
- School of Medicine, University of Auckland, Auckland, New Zealand
| | | | | | - Wandia Kimita
- School of Medicine, University of Auckland, Auckland, New Zealand
| | - Ivana R Sequeira-Bisson
- Human Nutrition Unit, School of Biological Sciences, University of Auckland, Auckland, New Zealand
- The Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Kempegowda SN, Sugur K, Thimmulappa RK. Dysfunctional HDL Diagnostic Metrics for Cardiovascular Disease Risk Stratification: Are we Ready to Implement in Clinics? J Cardiovasc Transl Res 2024:10.1007/s12265-024-10559-x. [PMID: 39298091 DOI: 10.1007/s12265-024-10559-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/09/2024] [Indexed: 09/21/2024]
Abstract
Epidemiological studies have revealed that patients with higher levels of high-density lipoprotein cholesterol (HDL-C) were more resistant to cardiovascular diseases (CVD), and yet targeting HDL for CVD prevention, risk assessment, and pharmacological management has not proven to be very effective. The mechanistic investigations have demonstrated that HDL exerts anti-atherogenic functions via mediating reverse cholesterol transport, antioxidant action, anti-inflammatory activity, and anti-thrombotic activity. Contrary to expectations, however, adverse cardiovascular events were reported in clinical trials of drugs that raised HDL levels. This has sparked a debate between HDL quantity and quality. Patients with atherosclerotic CVD are associated with dysfunctional HDL, and the degree of HDL dysfunction is correlated with the severity of the disease, independent of HDL-C levels. This growing body of evidence has underscored the need for integrating HDL functional assays in clinical practice for CVD risk management. Because HDL exerts diverse athero-protective functions, there is no single method for capturing HDL functionality. This review critically evaluates the various techniques currently being used for monitoring HDL functionality and discusses key structural changes in HDL indicative of dysfunctional HDL and the technical challenges that need to be addressed to enable the integration of HDL function-based metrics in clinical practice for CVD risk estimation and the development of newer therapies targeting HDL function.
Collapse
Affiliation(s)
- Swetha N Kempegowda
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Kavya Sugur
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Rajesh K Thimmulappa
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India.
| |
Collapse
|
3
|
Bhale AS, Meilhac O, d'Hellencourt CL, Vijayalakshmi MA, Venkataraman K. Cholesterol transport and beyond: Illuminating the versatile functions of HDL apolipoproteins through structural insights and functional implications. Biofactors 2024; 50:922-956. [PMID: 38661230 DOI: 10.1002/biof.2057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/02/2024] [Indexed: 04/26/2024]
Abstract
High-density lipoproteins (HDLs) play a vital role in lipid metabolism and cardiovascular health, as they are intricately involved in cholesterol transport and inflammation modulation. The proteome of HDL particles is indeed complex and distinct from other components in the bloodstream. Proteomics studies have identified nearly 285 different proteins associated with HDL; however, this review focuses more on the 15 or so traditionally named "apo" lipoproteins. Important lipid metabolizing enzymes closely working with the apolipoproteins are also discussed. Apolipoproteins stand out for their integral role in HDL stability, structure, function, and metabolism. The unique structure and functions of each apolipoprotein influence important processes such as inflammation regulation and lipid metabolism. These interactions also shape the stability and performance of HDL particles. HDLs apolipoproteins have multifaceted roles beyond cardiovascular diseases (CVDs) and are involved in various physiological processes and disease states. Therefore, a detailed exploration of these apolipoproteins can offer valuable insights into potential diagnostic markers and therapeutic targets. This comprehensive review article aims to provide an in-depth understanding of HDL apolipoproteins, highlighting their distinct structures, functions, and contributions to various physiological processes. Exploiting this knowledge holds great potential for improving HDL function, enhancing cholesterol efflux, and modulating inflammatory processes, ultimately benefiting individuals by limiting the risks associated with CVDs and other inflammation-based pathologies. Understanding the nature of all 15 apolipoproteins expands our knowledge of HDL metabolism, sheds light on their pathological implications, and paves the way for advancements in the diagnosis, prevention, and treatment of lipid and inflammatory-related disorders.
Collapse
Affiliation(s)
- Aishwarya Sudam Bhale
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Olivier Meilhac
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | - Christian Lefebvre d'Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, Saint-Pierre, France
| | | | - Krishnan Venkataraman
- Centre for Bio-Separation Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| |
Collapse
|
4
|
Abubakar M, Irfan U, Abdelkhalek A, Javed I, Khokhar MI, Shakil F, Raza S, Salim SS, Altaf MM, Habib R, Ahmed S, Ahmed F. Comprehensive Quality Analysis of Conventional and Novel Biomarkers in Diagnosing and Predicting Prognosis of Coronary Artery Disease, Acute Coronary Syndrome, and Heart Failure, a Comprehensive Literature Review. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10540-8. [PMID: 38995611 DOI: 10.1007/s12265-024-10540-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Coronary artery disease (CAD), acute coronary syndrome (ACS), and heart failure (HF) are major global health issues with high morbidity and mortality rates. Biomarkers like cardiac troponins (cTn) and natriuretic peptides (NPs) are crucial tools in cardiology, but numerous new biomarkers have emerged, proving increasingly valuable in CAD/ACS. These biomarkers are classified based on their mechanisms, such as fibrosis, metabolism, inflammation, and congestion. The integration of established and emerging biomarkers into clinical practice is an ongoing process, and recognizing their strengths and limitations is crucial for their accurate interpretation, incorporation into clinical settings, and improved management of CVD patients. We explored established biomarkers like cTn, NPs, and CRP, alongside newer biomarkers such as Apo-A1, IL-17E, IgA, Gal-3, sST2, GDF-15, MPO, H-FABP, Lp-PLA2, and ncRNAs; provided evidence of their utility in CAD/ACS diagnosis and prognosis; and empowered clinicians to confidently integrate these biomarkers into clinical practice based on solid evidence.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan.
| | - Umema Irfan
- Department of Internal Medicine, Deccan College of Medical Sciences, Hyderabad, India
| | - Ahmad Abdelkhalek
- Department of Internal Medicine, Zhejiang University, Zhejiang, China
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | | | - Fraz Shakil
- Department of Emergency Medicine, Mayo Hospital, Lahore, Pakistan
| | - Saud Raza
- Department of Anesthesia, Social Security Teaching Hospital, Lahore, Punjab, Pakistan
| | - Siffat Saima Salim
- Department of Surgery, Holy Family Red Crescent Medical College Hospital, Dhaka, Bangladesh
| | - Muhammad Mahran Altaf
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | - Rizwan Habib
- Department of Internal Medicine and Emergency, Indus Hospital, Lahore, Pakistan
| | - Simra Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| | - Farea Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| |
Collapse
|
5
|
Cheng W, Li T, Wang X, Xu T, Zhang Y, Chen J, Wei Z. The neutrophil-to-apolipoprotein A1 ratio is associated with adverse outcomes in patients with acute decompensated heart failure at different glucose metabolic states: a retrospective cohort study. Lipids Health Dis 2024; 23:118. [PMID: 38649986 PMCID: PMC11034163 DOI: 10.1186/s12944-024-02104-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/06/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The present study was performed to assess the association between the neutrophil-to-apolipoprotein A1 ratio (NAR) and outcomes in patients with acute decompensated heart failure (ADHF) at different glucose metabolism states. METHODS We recruited 1233 patients with ADHF who were admitted to Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University from December 2014 to October 2019. The endpoints were defined as composites of cardiovascular death, nonfatal myocardial infarction, nonfatal ischemic stroke and exacerbation of chronic heart failure. The restricted cubic spline was used to determine the best cutoff of NAR, and patients were divided into low and high NAR groups. Kaplan-Meier plots and multivariable Cox proportional hazard models were used to investigate the association between NAR and the risk of adverse outcomes. RESULTS During the five-year follow-up period, the composite outcome occurred in 692 participants (56.1%). After adjusting for potential confounding factors, a higher NAR was associated with a higher incidence of composite outcomes in the total cohort (Model 1: HR = 1.42, 95% CI = 1.22-1.65, P<0.001; Model 2: HR = 1.29, 95% CI = 1.10-1.51, P = 0.002; Model 3: HR = 1.20, 95% CI = 1.01-1.42, P = 0.036). At different glucose metabolic states, a high NAR was associated with a high risk of composite outcomes in patients with diabetes mellitus (DM) (Model 1: HR = 1.54, 95% CI = 1.25-1.90, P<0.001; Model 2: HR = 1.40, 95% CI = 1.13-1.74, P = 0.002; Model 3: HR = 1.31, 95% CI = 1.04-1.66, P = 0.022), and the above association was not found in patients with prediabetes mellitus (Pre-DM) or normal glucose regulation (NGR) (both P>0.05). CONCLUSIONS The NAR has predictive value for adverse outcomes of ADHF with DM, which implies that the NAR could be a potential indicator for the management of ADHF.
Collapse
Affiliation(s)
- Weimeng Cheng
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Tianyue Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiaohan Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Tingting Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Ying Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Jianzhou Chen
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
6
|
Li J, Ge X, Liu X, Fu C, Miao J, Zhao W, Miao L, Hang D. Serum apolipoproteins and mortality risk: evidence from observational and Mendelian randomization analyses. Am J Clin Nutr 2024; 119:981-989. [PMID: 38211689 DOI: 10.1016/j.ajcnut.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Apolipoproteins (APOs) have emerged as significant players in lipid metabolism that affects the risk of chronic disease. However, the impact of circulating APO concentrations on premature death remains undetermined. OBJECTIVES This study aimed to investigate the associations of serum APOs with all-cause, cardiovascular disease (CVD)-related, and cancer-related mortality. METHODS We included 340,737 participants who had serum APO measurements from the UK Biobank. Restricted cubic splines and multivariable Cox regression models were used to assess the associations between APOs and all-cause and cause-specific mortality by computing hazard ratios (HRs) and 95% confidence intervals (CIs). Based on 1-sample Mendelian randomization (MR) design, including 398,457 participants of White ancestry who had genotyping data from the UK Biobank, we performed instrumental variable analysis with 2-stage least squares regression to assess the association between genetically predicted APOs and mortality. RESULTS After adjusting for potential confounders including high-density and low-density lipoprotein particles, we observed nonlinear inverse relationships of APOA1 with all-cause, CVD-related, and cancer-related mortality (P-nonlinear < 0.001). By contrast, positive relationships were observed for APOB and all-cause (P-nonlinear < 0.001), CVD-related (P-linear < 0.001), and cancer-related (P-linear = 0.03) mortality. MR analysis showed consistent results, except that the association between APOB and cancer mortality was null. Furthermore, both observational and MR analyses found an inverse association between APOA1 and lung cancer-related mortality (HR comparing extreme deciles: 0.46; 95% CI: 0.26, 0.80; and HR: 0.78; 95% CI: 0.63, 0.97, respectively). CONCLUSIONS Our findings indicate that circulating APOA1 has potential beneficial effects on all-cause, CVD-related, and lung cancer-related death risk, whereas APOB may confer detrimental effects on all-cause and CVD-related death risk.
Collapse
Affiliation(s)
- Jiacong Li
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xianxiu Ge
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xinyi Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chengqu Fu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junyan Miao
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wei Zhao
- Center of Clinical Laboratory Science, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Lin Miao
- Medical Center for Digestive Diseases, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Dong Hang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine and China International Cooperation Center for Environment and Human Health, Gusu School, Nanjing Medical University, Nanjing, China
| |
Collapse
|
7
|
Lu J, Wang Z, Zhang J, Jiao F, Zou C, Han L, Jiang G. Causal association of blood lipids with all-cause and cause-specific mortality risk: a Mendelian randomization study. J Lipid Res 2024; 65:100528. [PMID: 38458338 PMCID: PMC10993189 DOI: 10.1016/j.jlr.2024.100528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 02/03/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
Dyslipidemia has long been implicated in elevating mortality risk; yet, the precise associations between lipid traits and mortality remained undisclosed. Our study aimed to explore the causal effects of lipid traits on both all-cause and cause-specific mortality. One-sample Mendelian randomization (MR) with linear and nonlinear assumptions was conducted in a cohort of 407,951 European participants from the UK Biobank. Six lipid traits, consisting of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides, apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), and lipoprotein(a), were included to investigate the causal associations with mortality. Two-sample MR was performed to replicate the association between each lipid trait and all-cause mortality. Univariable MR results showed that genetically predicted higher ApoA1 was significantly associated with a decreased all-cause mortality risk (HR[95% CI]:0.93 [0.89-0.97], P value = 0.001), which was validated by the two-sample MR analysis. Higher lipoprotein(a) was associated with an increased risk of all-cause mortality (1.03 [1.01-1.04], P value = 0.002). Multivariable MR confirmed the direct causal effects of ApoA1 and lipoprotein(a) on all-cause mortality. Meanwhile, nonlinear MR found no evidence for nonlinearity between lipids and all-cause mortality. Our examination into cause-specific mortality revealed a suggestive inverse association between ApoA1 and cancer mortality, a significant positive association between lipoprotein(a) and cardiovascular disease mortality, and a suggestive positive association between lipoprotein(a) and digestive disease mortality. High LDL-C was associated with an increased risk of cardiovascular disease mortality but a decreased risk of neurodegenerative disease mortality. The findings suggest that implementing interventions to raise ApoA1 and decrease lipoprotein(a) levels may improve overall health outcomes and mitigate cancer and digestive disease mortality.
Collapse
Affiliation(s)
- Jiawen Lu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenqian Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiaying Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng Jiao
- Guangzhou Centre for Applied Mathematics, Guangzhou University, Guangzhou, Guangdong, China
| | - Chenfeng Zou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liyuan Han
- Department of Global Health, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Guozhi Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
8
|
Sharma P, Roy A, Dhamija RK, Bhushan S, Baswal K, Kulandaisamy R, Yadav S, Kumar S, Inampudi KK. A comprehensive proteomic profiling of urinary exosomes and the identification of early non-invasive biomarker in patients with coronary artery disease. J Proteomics 2024; 293:105059. [PMID: 38151158 DOI: 10.1016/j.jprot.2023.105059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/29/2023]
Abstract
Urinary small extracellular vesicles or exosomes (uEVs) source could be an emerging trove of biomarkers in coronary artery disease (CAD). It is a chronic inflammatory disease having a long asymptomatic phase of fatty-fibrous development in arteries leading to angina, myocardial infarction, and death. Our study was aimed at identifying differential protein expression profiling of uEVs in CAD. We collected urine samples of CAD patients (n = 41) age 18-65 years and gender matched healthy controls (n = 41). We isolated uEVs using differential ultracentrifugation. Further, uEV samples were characterized by western blotting exosome markers (Flotillin, TSG, CD63, and CD9), nano tracking analysis, and transmission and scanning electron microscopy. A total of 508 proteins were identified by iTRAQ-based mass spectrometry. We observed protein expression levels of AZGP1, SEMG1/2, ORM1, IGL, SERPINA5, HSPG2, prosaposin, gelsolin, and CD59 were upregulated, and UMOD, KNG1, AMBP, prothrombin, and TF were downregulated. Protein-protein interactions, gene ontology and pathway analysis were performed to functionally annotate identified uEVs proteins. A novel uEVs differential protein signature is shown. On validating UMOD protein by ELISA in two clinically different CAD, stable-CAD patients had lower levels than healthy controls whereas recent myocardial infarction patients had lowest. Our findings suggest UMOD importance as early diagnostic biomarker. SIGNIFICANCE: Coronary artery disease is a chronic inflammatory disease caused by gradual deposition of cholesterol and fat along with other proteins to develop plaque inside arteries. This further leads to blockage of artery, heart attack and death. There are no identifiable early biomarkers to diagnose this. For the first time, we have identified the differentially expressed proteins isolated from non-invasive uEV of CAD patients compared to healthy controls by using MS Orbitrap and iTRAQ labelling of peptides. We have identified decreased levels of UMOD protein in CAD. These findings have been confirmed by ELISA. Furthermore, the levels of UMOD were observed as more highly decreased in recent myocardial infarction CAD patients, indicating the importance of this protein as an early diagnostic biomarker. Conclusively, our study represents a non-invasive urinary EVs trove of differentially expressed proteins in CAD. This will form a groundwork for understanding the pathophysiology of CAD and will help in future translational research utilizing uEVs.
Collapse
Affiliation(s)
- Pratibha Sharma
- Human Behaviour Department, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, New Delhi, India; Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| | - Ambuj Roy
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Rajinder K Dhamija
- Human Behaviour Department, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, New Delhi, India
| | - Sudha Bhushan
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Kamal Baswal
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | | | - Satyavir Yadav
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India; Department of Health Education and Technology, Luleå University of Technology, Sweden
| | | |
Collapse
|
9
|
Li S, Xie X, Zeng X, Wang S, Lan J. Serum apolipoprotein B to apolipoprotein A-I ratio predicts mortality in patients with heart failure. ESC Heart Fail 2024; 11:99-111. [PMID: 37822135 PMCID: PMC10804159 DOI: 10.1002/ehf2.14547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
AIMS Apolipoproteins have been reported to be involved in many cardiovascular diseases. The aim of our study was to investigate the prognostic value of apolipoprotein B (ApoB) to apolipoprotein A-I (ApoA-I) ratio (ApoB/ApoA-I) in patients with heart failure (HF). METHODS AND RESULTS We randomly assigned 2400 HF patients into the training cohort (n = 1400) and the validation cohort (n = 1000). Using a receiver operating characteristic curve, we identified the optimal cut-off value of the ApoB/ApoA-I in the training cohort as 0.69, which was further validated in the validation cohort. A propensity score matching (PSM) analysis was conducted to eliminate the imbalance in the baseline characteristics of the high and low ApoB/ApoA-I group. A total of 2242 HF patients were generated in the PSM cohort. We also validated our results with an independent cohort (n = 838). Univariate and multivariate analyses were conducted to explore the independent prognostic value of ApoB/ApoA-I in the training cohort (n = 1400), the validation cohort (n = 1000), the PSM cohort (n = 2242), and the independent cohort (n = 838). Patients with high ApoB/ApoA-I ratio had significantly poorer prognosis compared with those with low ApoB/ApoA-I ratio in the training cohort, the validation cohort, the PSM cohort, and the independent cohort (P < 0.05). Multivariate analysis indicated that the ApoB/ApoA-I was an independent prognostic factor for HF in the training cohort [hazard ratio (HR) = 1.637, 95% confidence interval (CI) = 1.201-2.231, P = 0.002], the validation cohort (HR = 1.54, 95% CI = 1.051-2.257, P = 0.027), the PSM cohort (HR = 1.645, 95% CI = 1.273-2.125, P < 0.001), and the independent cohort (HR = 1.987, 95% CI = 1.251-3.155, P = 0.004). CONCLUSIONS Serum ApoB/ApoA-I ratio is an independent predictor for the prognosis of HF patients.
Collapse
Affiliation(s)
- Shiyang Li
- Division of CardiologyPanzhihua Central HospitalPanzhihuaChina
- Panzhihua Central Hospital affiliated to Dali UniversityYunnanChina
| | - Xiaoshuang Xie
- Division of CardiologyPanzhihua Central HospitalPanzhihuaChina
| | - Xiaobin Zeng
- Division of CardiologyPanzhihua Central HospitalPanzhihuaChina
| | - Shihai Wang
- Division of CardiologyPanzhihua Central HospitalPanzhihuaChina
| | - Jianjun Lan
- Division of CardiologyPanzhihua Central HospitalPanzhihuaChina
| |
Collapse
|
10
|
Kazenwadel J, Berezhnoy G, Cannet C, Schäfer H, Geisler T, Rohlfing AK, Gawaz M, Merle U, Trautwein C. Stratification of hypertension and SARS-CoV-2 infection by quantitative NMR spectroscopy of human blood serum. COMMUNICATIONS MEDICINE 2023; 3:145. [PMID: 37845506 PMCID: PMC11081957 DOI: 10.1038/s43856-023-00365-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Diagnostic approaches like the nuclear magnetic resonance spectroscopy (NMR) based quantification of metabolites, lipoproteins, and inflammation markers has helped to identify typical alterations in the blood serum of COVID-19 patients. However, confounders such as sex, and comorbidities, which strongly influence the metabolome, were often not considered. Therefore, the aim of this NMR study was to consider sex, as well as arterial hypertension (AHT), when investigating COVID-19-positive serum samples in a large age-and sex matched cohort. METHODS NMR serum data from 329 COVID-19 patients were compared with 305 healthy controls. 134 COVID-19 patients were affected by AHT. These were analyzed together with NMR data from 58 hypertensives without COVID-19. In addition to metabolite, lipoprotein, and glycoprotein data from NMR, common laboratory parameters were considered. Sex was considered in detail for all comparisons. RESULTS Here, we show that several differences emerge from previous NMR COVID-19 studies when AHT is considered. Especially, the previously described triglyceride-rich lipoprotein profile is no longer observed in COVID-19 patients, nor an increase in ketone bodies. Further alterations are a decrease in glutamine, leucine, isoleucine, and lysine, citric acid, HDL-4 particles, and total cholesterol. Additionally, hypertensive COVID-19 patients show higher inflammatory NMR parameters than normotensive patients. CONCLUSIONS We present a more precise picture of COVID-19 blood serum parameters. Accordingly, considering sex and comorbidities should be included in future metabolomics studies for improved and refined patient stratification. Due to metabolic similarities with other viral infections, these results can be applied to other respiratory diseases in the future.
Collapse
Affiliation(s)
- Jasmin Kazenwadel
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Georgy Berezhnoy
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, Ettlingen, Germany
| | - Hartmut Schäfer
- Bruker BioSpin GmbH, Applied Industrial and Clinical Division, Ettlingen, Germany
| | - Tobias Geisler
- Department of Internal Medicine III, Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Internal Medicine III, Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Internal Medicine III, Cardiology and Angiology, University Hospital Tübingen, Tübingen, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department for Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
11
|
Sharma P, Dhamija RK, Nag TC, Roy A, Inampudi KK. Different Biofluids, Small Extracellular Vesicles or Exosomes: Structural Analysis in Atherosclerotic Cardiovascular Disease Using Electron Microscopy Techniques. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:1168-1177. [PMID: 37749667 DOI: 10.1093/micmic/ozad025] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/20/2023] [Indexed: 09/27/2023]
Abstract
Small extracellular vesicles (sEVs) or exosomes are secretory vesicles largely involved in cell-cell communications and found to play a role in development as well as diseases including atherosclerosis. They hold a huge potential for translational research by devising better clinical diagnostics, biomarker discovery, drug delivery, and therapeutic strategies. Variations terms of morphology and distribution are crucial to biological function integrity. Moreover, it is dependent on susceptibility to influential factors of the environment like cell stress, inflammation, and secretion by different cells in subsequent biofluids. We have observed the morphological variations in sEVs or exosomes freshly isolated from patients with atherosclerotic cardiovascular disease (AsCVD), in blood plasma, saliva, and urine biofluids compared to healthy controls. High-resolution images were obtained by transmission electron microscopy (TEM) and scanning electron microscopy (SEM) for the characterization of sEVs morphology. Western blotting and immuno-TEM gold labeling confirmed the presence of exosome markers. For the first time, we report size and shape variations, which suggest the existence of different functions of sEVs in the disease state. Morphological variations in sEVs were observed significantly in noninvasive AsCVD saliva and urine samples, important to understand the cell behavior and physiological state. These variations will be useful to investigate their possible role in the disease process.
Collapse
Affiliation(s)
- Pratibha Sharma
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
- Human Behaviour Department, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, Delhi 110095, India
| | - Rajinder K Dhamija
- Human Behaviour Department, Institute of Human Behaviour and Allied Sciences, Dilshad Garden, Delhi 110095, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Ambuj Roy
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Krishna Kishore Inampudi
- Department of Biophysics, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
12
|
Pillai JA, Bena J, Bekris L, Kodur N, Kasumov T, Leverenz JB, Kashyap SR. Metabolic syndrome biomarkers relate to rate of cognitive decline in MCI and dementia stages of Alzheimer's disease. Alzheimers Res Ther 2023; 15:54. [PMID: 36927447 PMCID: PMC10018847 DOI: 10.1186/s13195-023-01203-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The relationship between biomarkers of metabolic syndrome and insulin resistance, plasma triglyceride/HDL cholesterol (TG/HDL-C) ratio, on the rate of cognitive decline in mild cognitive impairment (MCI) and dementia stages of Alzheimer's disease (AD) is unknown. The role of peripheral and cerebrospinal fluid (CSF) levels of Apolipoprotein A1 (ApoA1), a key functional component of HDL, on cognitive decline also remains unclear among them. Here we evaluate baseline plasma TG/HDL-C ratio and CSF and plasma ApoA1 levels and their relation with cognitive decline in the MCI and Dementia stages of AD. PATIENTS AND METHODS A retrospective longitudinal study (156 participants; 106 MCI, 50 AD dementia) from the Alzheimer's Disease Neuroimaging Initiative, with an average of 4.0 (SD 2.8) years follow-up. Baseline plasma TG/HDL-C, plasma, and CSF ApoA1 and their relationship to inflammation and blood-brain barrier (BBB) biomarkers and longitudinal cognitive outcomes were evaluated. Multivariable linear mixed effect models were used to assess the effect of baseline analytes with longitudinal changes in Mini-Mental State Exam (MMSE), Clinical Dementia Rating-Sum of Boxes (CDR-SB), and Logical Memory delayed recall (LM) score after controlling for well-known covariates. RESULTS A total of 156 participants included 98 women, 63%; mean age was 74.9 (SD 7.3) years. At baseline, MCI and dementia groups did not differ significantly in TG/HDL-C (Wilcoxon W statistic = 0.39, p = 0.39) and CSF ApoA1 levels (W = 3642, p = 0.29), but the dementia group had higher plasma ApoA1 than the MCI group (W = 4615, p = 0.01). Higher TG/HDL-C ratio was associated with faster decline in CDR-SB among MCI and dementia groups. Higher plasma ApoA1 was associated with faster decline in MMSE and LM among MCI, while in contrast higher CSF ApoA1 levels related to slower cognitive decline in MMSE among MCI. CSF and plasma ApoA1 also show opposite directional correlations with biomarkers of BBB integrity. CSF but not plasma levels of ApoA1 positively correlated to inflammation analytes in the AGE-RAGE signaling pathway in diabetic complications (KEGG ID:KO04933). CONCLUSIONS Biomarkers of metabolic syndrome relate to rate of cognitive decline among MCI and dementia individuals. Elevated plasma TG/HDL-C ratio and plasma ApoA1 are associated with worse cognitive outcomes in MCI and dementia participants. CSF ApoA1 and plasma ApoA1 likely have different roles in AD progression in MCI stage.
Collapse
Affiliation(s)
- Jagan A Pillai
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave/U10, Cleveland, OH, 44195, USA. .,Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA. .,Department of Neurology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA. .,Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Lynn Bekris
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Nandan Kodur
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave/U10, Cleveland, OH, 44195, USA.,Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Department of Neurology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Sangeeta R Kashyap
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine New York Presbyterian, New York, NY, 10021, USA
| | | |
Collapse
|
13
|
Faaborg-Andersen CC, Liu C, Subramaniyam V, Desai SR, Sun YV, Wilson PWF, Sperling LS, Quyyumi AA. U-shaped relationship between apolipoprotein A1 levels and mortality risk in men and women. Eur J Prev Cardiol 2023; 30:293-304. [PMID: 36351048 PMCID: PMC10024919 DOI: 10.1093/eurjpc/zwac263] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/25/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Apolipoprotein A1 (ApoA1) is the principal protein component of high-density lipoprotein (HDL). Although low HDL cholesterol (HDL-C) levels are known to be associated with greater cardiovascular risk, recent studies have also shown heightened mortality risk at very high HDL-C levels. AIMS To investigate the sex-specific association between elevated ApoA1 levels and adverse outcomes, and their genetic basis. METHODS A prospective cohort study of United Kingdom Biobank participants without coronary artery disease at enrollment was performed. The primary exposure was serum ApoA1 levels. The primary and secondary outcome measures were cardiovascular and all-cause death, respectively. RESULTS In 402 783 participants followed for a median of 12.1 years, there was a U-shaped relationship between ApoA1 levels and both cardiovascular as well as all-cause mortality, after adjustment for traditional cardiovascular risk factors. Individuals in the highest decile of ApoA1 levels (1.91-2.50 g/L) demonstrated higher cardiovascular (HR 1.21, 95% CI 1.07-1.37, P < 0.0022) and all-cause mortality (HR 1.14, 95% CI 1.07-1.21, P < 0.0001) compared with those within the lowest risk eighth decile (1.67-1.75 g/L). The U-shaped relationship was present in both sexes, though more pronounced in men. Sensitivity analyses showed that cardiovascular mortality rates were higher in those with greater alcohol intake (P < 0.004). Adjustment for polygenic variation associated with higher ApoA1 levels did not attenuate the effect of very high ApoA1 levels on mortality. In the sub-group with very elevated HDL-C levels (> 80 mg/dL in men, > 100 mg/dL in women), there was no association between ApoA1 levels and mortality. CONCLUSION Both very low and very elevated ApoA1 levels are associated with higher cardiovascular and all-cause mortality.
Collapse
Affiliation(s)
- Christian C. Faaborg-Andersen
- Department of Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA 30322, USA
| | - Chang Liu
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | | | - Shivang R. Desai
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA 30322, USA
- Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Peter W. F. Wilson
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA 30322, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Laurence S. Sperling
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA 30322, USA
- Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Arshed A. Quyyumi
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, 1462 Clifton Road N.E. Suite 507, Atlanta, GA 30322, USA
- Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| |
Collapse
|
14
|
Kunutsor SK, Laukkanen JA. Further proof of a paradoxical relationship between high-density lipoprotein levels and adverse cardiovascular outcomes: are there implications for cardiovascular disease prevention? Eur J Prev Cardiol 2023; 30:290-292. [PMID: 36384026 DOI: 10.1093/eurjpc/zwac272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Setor K Kunutsor
- Diabetes Research Centre, Leicester General Hospital, University of Leicester, Gwendolen Road, Leicester LE5 4WP, UK
| | - Jari A Laukkanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1 C, Canthia building, B wing, FI-70211 Kuopio, Finland
- Department of Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio University Hospital, FI-70029 Kuopio, Finland
- Department of Medicine, Central Finland Health Care District, 40620 Jyväskylä, Finland
| |
Collapse
|
15
|
najafi S, Moshtaghie AA, Hassanzadeh F, Nayeri H, Jafari E. Design, synthesis, and biological evaluation of novel atorvastatin derivatives. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
|
16
|
Chalise U, Becirovic-Agic M, Rodriguez-Paar JR, Konfrst SR, de Morais SDB, Johnson CS, Flynn ER, Hall ME, Anderson DR, Cook LM, DeLeon-Pennell KY, Lindsey ML. Harnessing the Plasma Proteome to Mirror Current and Predict Future Cardiac Remodeling After Myocardial Infarction. J Cardiovasc Transl Res 2023; 16:3-16. [PMID: 36197585 PMCID: PMC9944212 DOI: 10.1007/s12265-022-10326-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 09/15/2022] [Indexed: 12/01/2022]
Abstract
To identify plasma proteins that mirror current and predict future remodeling after myocardial infarction (MI), we retrospectively interrogated plasma proteomes of day (D)0 control (n = 16) and D3 MI (n = 15) from C57BL/6 J mice (20 ± 1 months). A total of 165 unique proteins were correlated with cardiac physiology variables. We prospectively tested the hypothesis that candidates identified retrospectively would predict cardiac physiology at an extended timepoint (D7 MI) in a second cohort of mice (n = 4 ± 1 months). We also examined human plasma from healthy controls (n = 18) and patients 48 h after presentation for MI (n = 41). Retrospectively, we identified 5 strong reflectors of remodeling (all r ≥ 0.60 and p < 0.05). Prospectively, ApoA1, IgA, IL-17E, and TIMP-1 mirrored current and predicted future remodeling. In humans, cytokine-cytokine receptor signaling was the top enriched KEGG pathway for all candidates. In summary, we identified plasma proteins that serve as useful prognostic indicators of adverse remodeling and progression to heart failure.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Jocelyn R Rodriguez-Paar
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Sharon D B de Morais
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68198, USA
| | - Catherine S Johnson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, 68198, USA
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Daniel R Anderson
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kristine Y DeLeon-Pennell
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, 29425, USA
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, 29401, USA
| | - Merry L Lindsey
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr DB Todd Jr Blvd, Nashville, TN, 37208, USA.
- Nashville VA Medical Center, Nashville, TN, 37212, USA.
| |
Collapse
|
17
|
Bordeianu G, Mitu I, Stanescu RS, Ciobanu CP, Petrescu-Danila E, Marculescu AD, Dimitriu DC. Circulating Biomarkers for Laboratory Diagnostics of Atherosclerosis-Literature Review. Diagnostics (Basel) 2022; 12:diagnostics12123141. [PMID: 36553147 PMCID: PMC9777004 DOI: 10.3390/diagnostics12123141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is still considered a disease burden with long-term damaging processes towards the cardiovascular system. Evaluation of atherosclerotic stages requires the use of independent markers such as those already considered traditional, that remain the main therapeutic target for patients with atherosclerosis, together with emerging biomarkers. The challenge is finding models of predictive markers that are particularly tailored to detect and evaluate the evolution of incipient vascular lesions. Important advances have been made in this field, resulting in a more comprehensible and stronger linkage between the lipidic profile and the continuous inflammatory process. In this paper, we analysed the most recent data from the literature studying the molecular mechanisms of biomarkers and their involvement in the cascade of events that occur in the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
| | - Ivona Mitu
- Correspondence: (I.M.); (R.S.S.); Tel.: +40-75206-1747 (I.M.)
| | | | | | | | | | | |
Collapse
|
18
|
Weber P, Thomsen MN, Skytte MJ, Samkani A, Carl MH, Astrup A, Frystyk J, Holst JJ, Hartmann B, Madsbad S, Magkos F, Krarup T, Haugaard SB. Effects of Carbohydrate Restriction on Body Weight and Glycemic Control in Individuals with Type 2 Diabetes: A Randomized Controlled Trial of Efficacy in Real-Life Settings. Nutrients 2022; 14:5244. [PMID: 36558403 PMCID: PMC9788105 DOI: 10.3390/nu14245244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
A fully provided, hypocaloric, carbohydrate-reduced high-protein (CRHP) diet compared to a hypocaloric conventional diabetes (CD) diet for 6 weeks improved glycemic control to a greater extent in face of an intended 6% weight loss in individuals with type 2 diabetes mellitus (T2DM). The present 24-week extension of that study reports on the efficacy of CRHP and CD diets in a real-life setting. Sixty-five individuals with T2DM who completed the initial 6-week fully provided diet period (% energy from carbohydrate, protein, and fat was 30/30/40 in CRHP, and 50/17/33 in CD) continued a free-living, dietician guided 24-week period of which 59 individuals completed. The CRHP compared to CD group reported a 4% lower carbohydrate intake and had higher urea excretion by 22% (both p ≤ 0.05) at week 30, suggesting less difference in carbohydrate and protein intake between groups during the 24-week extension compared to week 6. The loss of body weight during the initial 6 weeks was maintained in both groups during the 24-week extension (-5.5 ± 4.5 and -4.6 ± 4.8 kg) as well as HbA1c (-8.4 ± 6.2 and -8.4 ± 6.9 mmol/mol) with no significant differences between groups. The additional benefits on glucoregulation harnessed by carbohydrate restriction under full diet provision for 6 weeks combined with titrated weight loss could not be maintained in a real-life setting of self-prepared diet aiming on similar diets for 6 months.
Collapse
Affiliation(s)
- Philip Weber
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
| | - Mads N. Thomsen
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
| | - Mads Juul Skytte
- Department of Forensic Medicine, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Amirsalar Samkani
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
| | - Martin Hansen Carl
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
| | - Arne Astrup
- Department of Obesity and Nutritional Sciences, Novo Nordisk Foundation, 2900 Hellerup, Denmark
| | - Jan Frystyk
- Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
- Department of Endocrinology, Odense University Hospital, 5000 Odense, Denmark
| | - Jens J. Holst
- NNF Centre for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bolette Hartmann
- NNF Centre for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Amager Hvidovre, 2650 Hvidovre, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise and Sports, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thure Krarup
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
- Department of Nutrition, Exercise and Sports, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Steen B. Haugaard
- Department of Endocrinology, Copenhagen University Hospital Bispebjerg, 2400 Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Zhang Q, Zhou X, Zhang J, Li Q, Qian Z. Selenium and vitamin B6 co-supplementation improve dyslipidemia and fatty liver syndrome by SIRT1/SREBP-1c pathway in hyperlipidemic Sprague-Dawley rats induced by high-fat diet. Nutr Res 2022; 106:101-118. [DOI: 10.1016/j.nutres.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
|
20
|
Karagiannidis E, Moysidis DV, Papazoglou AS, Panteris E, Deda O, Stalikas N, Sofidis G, Kartas A, Bekiaridou A, Giannakoulas G, Gika H, Theodoridis G, Sianos G. Prognostic significance of metabolomic biomarkers in patients with diabetes mellitus and coronary artery disease. Cardiovasc Diabetol 2022; 21:70. [PMID: 35525960 PMCID: PMC9077877 DOI: 10.1186/s12933-022-01494-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/06/2022] [Indexed: 12/18/2022] Open
Abstract
Background Diabetes mellitus (DM) and coronary artery disease (CAD) constitute inter-related clinical entities. Biomarker profiling emerges as a promising tool for the early diagnosis and risk stratification of either DM or CAD. However, studies assessing the predictive capacity of novel metabolomics biomarkers in coexistent CAD and DM are scarce. Methods This post-hoc analysis of the CorLipid trial (NCT04580173) included 316 patients with CAD and comorbid DM who underwent emergency or elective coronary angiography due to acute or chronic coronary syndrome. Cox regression analyses were performed to identify metabolomic predictors of the primary outcome, which was defined as the composite of major adverse cardiovascular or cerebrovascular events (MACCE: cardiovascular death, myocardial infarction, stroke, major bleeding), repeat unplanned revascularizations and cardiovascular hospitalizations. Linear regression analyses were also performed to detect significant predictors of CAD complexity, as assessed by the SYNTAX score. Results After a median 2-year follow up period (IQR = 0.7 years), the primary outcome occurred in 69 (21.8%) of patients. Acylcarnitine ratio C4/C18:2, apolipoprotein (apo) B, history of heart failure (HF), age > 65 years and presence of acute coronary syndrome were independent predictors of the primary outcome in diabetic patients with CAD (aHR = 1.89 [1.09, 3.29]; 1.02 [1.01, 1.04]; 1.28 [1.01, 1.41]; 1.04 [1.01, 1.05]; and 1.12 [1.05–1.21], respectively). Higher levels of ceramide ratio C24:1/C24:0, acylcarnitine ratio C4/C18:2, age > 65 and peripheral artery disease were independent predictors of higher CAD complexity (adjusted β = 7.36 [5.74, 20.47]; 3.02 [0.09 to 6.06]; 3.02 [0.09, 6.06], respectively), while higher levels of apoA1 were independent predictors of lower complexity (adjusted β= − 0.65 [− 1.31, − 0.02]). Conclusions In patients with comorbid DM and CAD, novel metabolomic biomarkers and metabolomics-based prediction models could be recruited to predict clinical outcomes and assess the complexity of CAD, thereby enabling the integration of personalized medicine into routine clinical practice. These associations should be interpreted taking into account the observational nature of this study, and thus, larger trials are needed to confirm its results and validate them in different and larger diabetic populations.
Collapse
Affiliation(s)
- Efstratios Karagiannidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece.
| | - Dimitrios V Moysidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - Andreas S Papazoglou
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - Eleftherios Panteris
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.,Biomic_Auth, Bioanalysis and Omics Lab, Centre for Interdisciplinary Research of Aristotle, University of Thessaloniki, Innovation Area of Thessaloniki, 57001, Thermi, Greece
| | - Olga Deda
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.,Biomic_Auth, Bioanalysis and Omics Lab, Centre for Interdisciplinary Research of Aristotle, University of Thessaloniki, Innovation Area of Thessaloniki, 57001, Thermi, Greece
| | - Nikolaos Stalikas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - Georgios Sofidis
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - Anastasios Kartas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - Alexandra Bekiaridou
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - George Giannakoulas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece
| | - Helen Gika
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.,Biomic_Auth, Bioanalysis and Omics Lab, Centre for Interdisciplinary Research of Aristotle, University of Thessaloniki, Innovation Area of Thessaloniki, 57001, Thermi, Greece
| | - George Theodoridis
- Biomic_Auth, Bioanalysis and Omics Lab, Centre for Interdisciplinary Research of Aristotle, University of Thessaloniki, Innovation Area of Thessaloniki, 57001, Thermi, Greece.,Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Sianos
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, St. Kiriakidi 1, 54636, Thessaloniki, Greece.
| |
Collapse
|
21
|
Diab A, Valenzuela Ripoll C, Guo Z, Javaheri A. HDL Composition, Heart Failure, and Its Comorbidities. Front Cardiovasc Med 2022; 9:846990. [PMID: 35350538 PMCID: PMC8958020 DOI: 10.3389/fcvm.2022.846990] [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: 12/31/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Although research on high-density lipoprotein (HDL) has historically focused on atherosclerotic coronary disease, there exists untapped potential of HDL biology for the treatment of heart failure. Anti-oxidant, anti-inflammatory, and endothelial protective properties of HDL could impact heart failure pathogenesis. HDL-associated proteins such as apolipoprotein A-I and M may have significant therapeutic effects on the myocardium, in part by modulating signal transduction pathways and sphingosine-1-phosphate biology. Furthermore, because heart failure is a complex syndrome characterized by multiple comorbidities, there are complex interactions between heart failure, its comorbidities, and lipoprotein homeostatic mechanisms. In this review, we will discuss the effects of heart failure and associated comorbidities on HDL, explore potential cardioprotective properties of HDL, and review novel HDL therapeutic targets in heart failure.
Collapse
|
22
|
Haji Aghajani M, Madani Neishaboori A, Ahmadzadeh K, Toloui A, Yousefifard M. The association between apolipoprotein A-1 plasma level and premature coronary artery disease: A systematic review and meta-analysis. Int J Clin Pract 2021; 75:e14578. [PMID: 34181800 DOI: 10.1111/ijcp.14578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/25/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Apolipoprotein A-1 (Apo A-1) is a constituent of high-density lipoprotein (HDL) and emerging evidences put forward a potential association between Apo A-1 plasma levels and premature coronary artery disease (pCAD). The aim of the present study is to gather relative literature and perform a systematic review and meta-analysis regarding the association between serum ApoA-1 levels and pCAD. METHODS Medline (via PubMed), Scopus, Embase and Web of Science databases were searched from the inception of databases until December 7, 2020. All articles reporting the plasma levels of ApoA-1 in patients with pCAD and the control group were included. A meta-analysis with pooled standardised mean difference (SMD) and 95% confidence interval (95% CI) was reported. Subgroup analyses were done based on the observed heterogeneity in results. RESULTS Seventeen case-control studies were included. ApoA-1 plasma level was calculated to be lower in pCAD patients compared with the control group (SMD: -0.67; 95% CI: -0.48 to -0.86; P < .001). The subgroup analysis and meta-regression showed that the variation in gender distribution, the development level of the target population's country and quality score of included studies were the main sources of heterogeneity. It was observed that the relationship was only significant in the developed countries (P < .001). Also, the heterogeneity was reduced when the analysis was limited to males (I2 = 57.2%) and females only (I2 = 26.0%). CONCLUSION In conclusion, there seems to be a significant association between the serum levels of ApoA-1 and pCAD. However, all of the included studies had a case-control design and since there is no good quality and prospective cohort studies included, reliability of the current evidence is debatable. Therefore, further well-designed cohort studies are required to assess the impact of serum ApoA-1 reduction on pCAD onset.
Collapse
Affiliation(s)
- Mohammad Haji Aghajani
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Cardiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Koohyar Ahmadzadeh
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amirmohammad Toloui
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Kumar V, Rai C, Kumari B, Srivastava S, Yanamandra U, Singh J, Ganju L, Varshney R, Garg I. Association of APOA1 gene polymorphisms (G-75A and C+83T) with deep vein -thrombosis: An Indian study. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
24
|
The effect of cranberry consumption on lipid metabolism and inflammation in human apo A-I transgenic mice fed a high-fat and high-cholesterol diet. Br J Nutr 2021; 126:183-190. [PMID: 33059793 DOI: 10.1017/s0007114520004080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lipid metabolism and inflammation contribute to CVD development. This study investigated whether the consumption of cranberries (CR; Vaccinium macrocarpon) can alter HDL metabolism and prevent inflammation in mice expressing human apo A-I transgene (hApoAITg), which have similar HDL profiles to those of humans. Male hApoAITg mice were fed a modified American Institute of Nutrition-93M high-fat/high-cholesterol diet (16 % fat, 0·25 % cholesterol, w/w; n 15) or the high-fat/high-cholesterol diet containing CR (5 % dried CR powder, w/w, n 16) for 8 weeks. There were no significant differences in body weight between the groups. Serum total cholesterol, non-HDL-cholesterol and TAG concentrations were significantly lower in the control than CR group with no significant differences in serum HDL-cholesterol and apoA-I. Mice fed CR showed significantly lower serum lecithin-cholesterol acyltransferase activity than the control. Liver weight and steatosis were not significantly different between the groups, but hepatic expression of genes involved in cholesterol metabolism was significantly lower in the CR group. In the epididymal white adipose tissue (eWAT), the CR group showed higher weights with decreased expression of genes for lipogenesis and fatty acid oxidation. The mRNA abundance of F4/80, a macrophage marker and the numbers of crown-like structures were less in the CR group. In the soleus muscle, the CR group also demonstrated higher expression of genes for fatty acid β-oxidation and mitochondrial biogenesis than those of the control. In conclusion, although CR consumption elicited minor effects on HDL metabolism, it prevented obesity-induced inflammation in eWAT with concomitant alterations in soleus muscle energy metabolism.
Collapse
|
25
|
Read RW, Schlauch KA, Lombardi VC, Cirulli ET, Washington NL, Lu JT, Grzymski JJ. Genome-Wide Identification of Rare and Common Variants Driving Triglyceride Levels in a Nevada Population. Front Genet 2021; 12:639418. [PMID: 33763119 PMCID: PMC7982958 DOI: 10.3389/fgene.2021.639418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/12/2021] [Indexed: 01/08/2023] Open
Abstract
Clinical conditions correlated with elevated triglyceride levels are well-known: coronary heart disease, hypertension, and diabetes. Underlying genetic and phenotypic mechanisms are not fully understood, partially due to lack of coordinated genotypic-phenotypic data. Here we use a subset of the Healthy Nevada Project, a population of 9,183 sequenced participants with longitudinal electronic health records to examine consequences of altered triglyceride levels. Specifically, Healthy Nevada Project participants sequenced by the Helix Exome+ platform were cross-referenced to their electronic medical records to identify: (1) rare and common single-variant genome-wide associations; (2) gene-based associations using a Sequence Kernel Association Test; (3) phenome-wide associations with triglyceride levels; and (4) pleiotropic variants linked to triglyceride levels. The study identified 549 significant single-variant associations (p < 8.75 × 10-9), many in chromosome 11's triglyceride hotspot: ZPR1, BUD13, APOC3, APOA5. A well-known protective loss-of-function variant in APOC3 (R19X) was associated with a 51% decrease in triglyceride levels in the cohort. Sixteen gene-based triglyceride associations were identified; six of these genes surprisingly did not include a single variant with significant associations. Results at the variant and gene level were validated with the UK Biobank. The combination of a single-variant genome-wide association, a gene-based association method, and phenome wide-association studies identified rare and common variants, genes, and phenotypes associated with elevated triglyceride levels, some of which may have been overlooked with standard approaches.
Collapse
Affiliation(s)
- Robert W. Read
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Karen A. Schlauch
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
| | - Vincent C. Lombardi
- Department of Microbiology and Immunology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | | | | | - James T. Lu
- Helix Opco, LLC., San Mateo, CA, United States
| | - Joseph J. Grzymski
- Center for Genomic Medicine, Desert Research Institute, Reno, NV, United States
- Renown Health, Reno, NV, United States
| |
Collapse
|
26
|
Patel VK, Williams H, Li SCH, Fletcher JP, Medbury HJ. Monocyte Subset Recruitment Marker Profile Is Inversely Associated With Blood ApoA1 Levels. Front Immunol 2021; 12:616305. [PMID: 33717107 PMCID: PMC7952433 DOI: 10.3389/fimmu.2021.616305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/12/2021] [Indexed: 12/30/2022] Open
Abstract
Dyslipidemia promotes development of the atherosclerotic plaques that characterise cardiovascular disease. Plaque progression requires the influx of monocytes into the vessel wall, but whether dyslipidemia is associated with an increased potential of monocytes to extravasate is largely unknown. Here (using flow cytometry) we examined recruitment marker expression on monocytes from generally healthy individuals who differed in lipid profile. Comparisons were made between monocyte subsets, participants and relative to participants’ lipid levels. Monocyte subsets differed significantly in their expression of recruitment markers, with highest expression being on either the classical or non-classical subsets. However, these inter-subset differences were largely overshadowed by considerable inter-participant differences with some participants having higher levels of recruitment markers on all three monocyte subsets. Furthermore, when the expression of one recruitment marker was high, so too was that of most of the other markers, with substantial correlations evident between the markers. The inter-participant differences were explained by lipid levels. Most notably, there was a significant inverse correlation for most markers with ApoA1 levels. Our results indicate that dyslipidemia, in particular low levels of ApoA1, is associated with an increased potential of all monocyte subsets to extravasate, and to do so using a wider repertoire of recruitment markers than currently appreciated.
Collapse
Affiliation(s)
- Vyoma K Patel
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| | - Helen Williams
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| | - Stephen C H Li
- Western Sydney University, Blacktown/Mt Druitt Clinical School, Blacktown Hospital, Blacktown, NSW, Australia.,Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - John P Fletcher
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| | - Heather J Medbury
- Vascular Biology Research Centre, Department of Surgery, Westmead Hospital, Westmead, NSW, Australia.,The University of Sydney, Westmead Clinical School, Westmead, NSW, Australia
| |
Collapse
|
27
|
Mesgari-Abbasi M, Mahmoudinezhad M, Farhangi MA. Soluble P-selectin, procalcitonin, transforming growth factor (TGF)-β and apo-proteins in association with the components of metabolic syndrome in obese individuals. Clin Nutr ESPEN 2021; 41:386-390. [PMID: 33487294 DOI: 10.1016/j.clnesp.2020.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/07/2020] [Accepted: 10/25/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND & AIMS Metabolic syndrome (MetS) is a clinical disorder with widespread prevalence. In the current study, we aimed to investigate the association between serum soluble P-selectin, procalcitonin, transforming growth factor (TGF)-β and apo-proteins with the components of metabolic syndrome in obese individuals. MATERIAL AND METHODS Sixty two obese patients with MetS and sixty five obese apparently healthy controls were participated in the current case-control study. The participants' anthropometric assessments and systolic and diastolic blood pressure (SBP and DBP) were measured. Serum lipids and the concentrations of ox-LDL, P-selectin, procalcitonin, TGF-β and apo-proteins were measured with commercial ELIZA kits. RESULTS Serum TG and TC were significantly higher in obese subjects with MetS; while TGF-β, procalcitonin, apoprotein B and insulin concentrations were higher in obese non- MetS group. In obese subjects with MetS, procalcitonin was in positive association with ox-LDL and apoprotein-B was in positive association with HDL. In obese subjects without MetS, apoprotein -B was in positive association with WC and HDL and WC. CONCLUSION The current study found several associations between serum lipids and PCT and serum apo-proteins in obese individuals either with or without MetS. Further studies with large sample size are warranted to better elucidate the observed relationships and underlying mechanism. TRIAL REGISTRATION Not Applicable.
Collapse
|
28
|
Edes A, Brown J, Edwards K. Testing lipid markers as predictors of all-cause morbidity, cardiac disease, and mortality risk in captive western lowland gorillas ( Gorilla gorilla gorilla). Primate Biol 2020; 7:41-59. [PMID: 33553576 PMCID: PMC7852406 DOI: 10.5194/pb-7-41-2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 10/19/2020] [Indexed: 12/30/2022] Open
Abstract
Great apes and humans develop many of the same health conditions, including cardiac disease as a leading cause of death. In humans, lipid markers are strong predictors of morbidity and mortality risk. To determine if they similarly predict risk in gorillas, we measured five serum lipid markers and calculated three lipoprotein ratios from zoo-housed western lowland gorillas (aged 6-52 years, n = 61 , subset with routine immobilizations only: n = 47 ): total cholesterol (TC), triglycerides (TGs), high-density lipoprotein (HDL), low-density lipoprotein (LDL), apolipoprotein A1 (apoA1), TC / HDL , LDL / HDL , and TG / HDL . We examined each in relation to age and sex, then analyzed whether they predicted all-cause morbidity, cardiac disease, and mortality using generalized linear models (GLMs). Older age was significantly associated with higher TG, TC / HDL , LDL / HDL , and TG / HDL , and lower HDL and apoA1. With all ages combined, compared to females, males had significantly lower TG, TC / HDL , LDL / HDL , and TG / HDL , and higher HDL. Using GLMs, age, sex, and lower LDL / HDL were significant predictors of all-cause morbidity; this is consistent with research demonstrating lower LDL in humans with arthritis, which was the second most prevalent condition in this sample. In contrast to humans, lipid markers were not better predictors of cardiac disease and mortality risk in gorillas, with cardiac disease best predicted by age and sex alone, and mortality risk only by age. Similar results were observed when multimodel inference was used as an alternative analysis strategy, suggesting it can be used in place of or in addition to traditional methods for predicting risk.
Collapse
Affiliation(s)
- Ashley N. Edes
- Center for Species Survival, Smithsonian Conservation Biology
Institute, 1500 Remount Rd., Front Royal, VA 22630, USA
- currently at: Department of Reproductive and
Behavioral Sciences, Saint Louis Zoo, St. Louis, MO 63110, USA
| | - Janine L. Brown
- Center for Species Survival, Smithsonian Conservation Biology
Institute, 1500 Remount Rd., Front Royal, VA 22630, USA
| | - Katie L. Edwards
- Center for Species Survival, Smithsonian Conservation Biology
Institute, 1500 Remount Rd., Front Royal, VA 22630, USA
- North of England Zoological Society, Chester Zoo, Upton by Chester,
CH2 1LH, UK
| |
Collapse
|
29
|
Negi P, Heikkilä T, Tallgren T, Malmi P, Lund J, Sinisalo J, Metso J, Jauhiainen M, Pettersson K, Lamminmäki U, Lövgren J. Three two-site apoA-I immunoassays using phage expressed detector antibodies - Preliminary clinical evaluation with cardiac patients. J Pharm Biomed Anal 2020; 194:113772. [PMID: 33309125 DOI: 10.1016/j.jpba.2020.113772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 11/25/2022]
Abstract
High density lipoproteins (HDL) are a heterogenous group of subpopulations differing in protein/lipid composition and in their anti-atherogenic function. There is a lack of specific and robust assays which can target the functionality of HDL with respect to atherosclerosis. With recently generated CAD HDL targeted, single chain recombinant antibodies (scFvs) we set out to design and optimize apo A-I tests to compare it with conventional HDL-C and apo A-I analyses for diagnosis and risk assessment of coronary artery disease (CAD) and its outcome. Three highly sensitive two-site apo A-I assays: 022-454, 109-121 and 110-525 were optimized. A preliminary clinical evaluation of these assays, after proper sample dilution procedure, was performed using samples derived from 195 chest pain patients (myocardial infarction (MI), n = 86 and non-MI, n = 109), collected at the time of admission and at discharge from hospital (hospital stay ≤ 24 h). The clinical performance of the assays was compared with apo A-I measured with polyclonal anti-apo A-I antibody using conventional ELISA. Apo A-I data was in addition compared with HDL-C concentration of the samples. The concentration of apo A-I was significantly lower in MI patients than in non-MI individuals with assay 022-454 (admission and discharge samples, P < 0.0001 and = 0.004); assay 109-121 (admission and discharge samples, P = 0.04 and 0.0009), and, ELISA based apo A-I test (admission and discharge samples, P = 0.008 and < 0.0001). HDL-C (admission and discharge samples, P = 0.002 and P = 0.01) was also significantly lower in MI patients. In Kaplan- Meier analysis, two-site assay 109-121 assay predicted mortality from admission samples at 1.5 yrs (whole cohort, P = 0.01 and in MI patients, P = 0.05) and at 6 months (whole cohort, P = 0.04). Assay 110-525 predicted mortality at 1.5 yrs from admission samples of non-MI patients (P = 0.01) and at 6 months from whole discharge sample cohort (P = 0.04). Polyclonal anti-apo A-I based conventional assay predicted mortality at 1.5 yrs from admission samples of whole cohort (P = 0.03). Two-site apo A-I assay 022-454 and HDL-C provided no capability of predicting mortality in the whole cohort or any sub-group. In conclusion, two of the tested recombinant apo A-I antibody combinations (sc 109-121 and sc 110-525) display promising outcome to improve diagnosis and prediction of future cardiac events in cardiac patients over polyclonal apo A-I ELISA and HDL-C assays. The noted differences, while interesting, are preliminary and need however to be verified in extensive cohorts of pathological cardiac conditions and healthy controls.
Collapse
Affiliation(s)
- Priyanka Negi
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland.
| | - Taina Heikkilä
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Terhi Tallgren
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Päivi Malmi
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Juha Lund
- Heart Center, Turku University Hospital, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Jari Metso
- Minerva Foundation Institute for Medical Research, Biomedicum, Helsinki, Finland; National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum, Helsinki, Finland
| | - Matti Jauhiainen
- Minerva Foundation Institute for Medical Research, Biomedicum, Helsinki, Finland; National Institute for Health and Welfare, Genomics and Biomarkers Unit, Biomedicum, Helsinki, Finland
| | - Kim Pettersson
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Urpo Lamminmäki
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| | - Janita Lövgren
- Department of Biochemistry, Division of Biotechnology, University of Turku, Turku, Finland
| |
Collapse
|
30
|
Dziedzic A, Miller E, Bijak M, Przyslo L, Saluk-Bijak J. Increased Pro-Thrombotic Platelet Activity Associated with Thrombin/PAR1-Dependent Pathway Disorder in Patients with Secondary Progressive Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21207722. [PMID: 33086557 PMCID: PMC7589910 DOI: 10.3390/ijms21207722] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022] Open
Abstract
Epidemiological studies confirm the high risk of ischemic events in multiple sclerosis (MS) that are associated with increased pro-thrombotic activity of blood platelets. The most potent physiological platelet agonist is thrombin, which activates platelets via cleavage of specific protease-activated receptors (PARs). Our current study is aimed to determine the potential genetics and proteomic abnormalities of PAR1 in both platelets and megakaryocytes, which may have thromboembolic consequences in the course of MS. The obtained results were correlated with the expression level of platelet and megakaryocyte transcripts for APOA1 and A2M genes encoding atherosclerosis biomarkers: apolipoprotein A1 (ApoA1) and α-2-macroglobulin (α2M), respectively. Moreover, PAR1 functionality in MS platelets was assessed by flow cytometry, determining the level of platelet–platelet and platelet–leukocyte aggregates, platelet microparticles and surface expression of P-selectin. As a PAR1 agonist, the synthetic TRAP-6 peptide was used, which made it possible to achieve platelet activation in whole blood without triggering clotting. Comparative analyses showed an elevated level of platelet activation markers in the blood of MS patients compared to controls. The mRNA expression of gene coding α2M was upregulated, whilst ApoA1 was down-regulated, both in platelets and megakaryocytes from MS patients. Furthermore, we observed an increase in both mRNA expression and surface density of PAR1 in platelets and megakaryocytes in MS compared to controls. Both the level of platelet activation markers and PAR1 expression showed a high correlation with the expression of transcripts for APOA1 and A2M genes.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Lukasz Przyslo
- Department of Developmental Neurology and Epileptology, Research Institute of Polish Mother’s Memorial Hospital, Rzgowska 281/289, 93-338 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
- Correspondence: ; Tel./Fax: +48-42-635-4336
| |
Collapse
|
31
|
Froyen E, Burns-Whitmore B. The Effects of Linoleic Acid Consumption on Lipid Risk Markers for Cardiovascular Disease in Healthy Individuals: A Review of Human Intervention Trials. Nutrients 2020; 12:E2329. [PMID: 32759714 PMCID: PMC7469037 DOI: 10.3390/nu12082329] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. Risk factors for developing this disease include high serum concentrations of total cholesterol, triglycerides, low-density lipoproteins, very-low density lipoproteins, and low concentrations of high-density lipoproteins. One proposed dietary strategy for decreasing risk factors involves replacing a portion of dietary saturated fatty acids with mono- and polyunsaturated fatty acids (PUFAs). The essential omega-6 PUFA, linoleic acid (LA), is suggested to decrease the risk for CVD by affecting these lipid risk markers. Reviewing human intervention trials will provide further evidence of the effects of LA consumption on risk factors for CVD. PubMed was used to search for peer-reviewed articles. The purpose of this review was: (1) To summarize human intervention trials that studied the effects of LA consumption on lipid risk markers for CVD in healthy individuals, (2) to provide mechanistic details, and (3) to provide recommendations regarding the consumption of LA to decrease the lipid risk markers for CVD. The results from this review provided evidence that LA consumption decreases CVD lipid risk markers in healthy individuals.
Collapse
Affiliation(s)
- Erik Froyen
- Department of Nutrition and Food Science, Huntley College of Agriculture, California State Polytechnic University, Pomona, CA 91768, USA;
| | | |
Collapse
|
32
|
Berkley A, Ferro A. Changes in C-reactive protein in response to anti-inflammatory therapy as a predictor of cardiovascular outcomes: A systematic review and meta-analysis. JRSM Cardiovasc Dis 2020; 9:2048004020929235. [PMID: 32547736 PMCID: PMC7273624 DOI: 10.1177/2048004020929235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/20/2020] [Accepted: 05/04/2020] [Indexed: 11/17/2022] Open
Abstract
Background Despite the availability of aggressive lipid-lowering strategies, many patients remain at risk of cardiovascular events. C-reactive protein is a marker of inflammation elevated in patients at high risk of cardiovascular events. C-reactive protein has demonstrated value as a predictor of cardiovascular risk; however, it is unclear whether targeting C-reactive protein levels improves outcomes. This systematic review aimed to characterise the relationship between C-reactive protein and cardiovascular outcomes and to assess whether the magnitude of C-reactive protein reduction correlates to the extent of cardiovascular risk reduction. Methods A systematic review was conducted to identify randomised controlled trials that measured C-reactive protein before and after administration of therapies for cardiovascular disease and measured incidence of cardiovascular events. A meta-analysis of placebo-controlled studies assessed the relationship between extent of C-reactive protein reduction and cardiovascular risk reduction. Placebo-controlled studies where low-density lipoprotein and triglyceride data were available were also included in a meta-regression to assess the influence of these established risk factors on the efficacy of treatment when compared to C-reactive protein. Results Fifteen studies met the criteria for inclusion in this review, of which six were active comparator studies and nine were placebo controlled. Six placebo-controlled studies had data available for meta-regression. Eight studies demonstrated a reduction in events that could be explained by changes in lipid levels, whereas the results of five studies suggested that the association between C-reactive protein reduction and event rates cannot be explained by changes in lipid levels alone. No correlation was found between magnitude of C-reactive protein reduction and cardiovascular risk reduction. A strong correlation was found between C-reactive protein and low-density lipoprotein reduction (adjusted r 2 = 0.8). Conclusions Targeting C-reactive protein does not offer additional benefit over targeting low-density lipoprotein across the general population in terms of cardiovascular risk reduction. However, there is value in targeting C-reactive protein in patients at high residual inflammatory risk despite non-elevated lipid levels or use of lipid-lowering therapy.
Collapse
Affiliation(s)
- Annie Berkley
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, UK
| | - Albert Ferro
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, UK
| |
Collapse
|
33
|
Garrett-Bakelman FE, Darshi M, Green SJ, Gur RC, Lin L, Macias BR, McKenna MJ, Meydan C, Mishra T, Nasrini J, Piening BD, Rizzardi LF, Sharma K, Siamwala JH, Taylor L, Vitaterna MH, Afkarian M, Afshinnekoo E, Ahadi S, Ambati A, Arya M, Bezdan D, Callahan CM, Chen S, Choi AMK, Chlipala GE, Contrepois K, Covington M, Crucian BE, De Vivo I, Dinges DF, Ebert DJ, Feinberg JI, Gandara JA, George KA, Goutsias J, Grills GS, Hargens AR, Heer M, Hillary RP, Hoofnagle AN, Hook VYH, Jenkinson G, Jiang P, Keshavarzian A, Laurie SS, Lee-McMullen B, Lumpkins SB, MacKay M, Maienschein-Cline MG, Melnick AM, Moore TM, Nakahira K, Patel HH, Pietrzyk R, Rao V, Saito R, Salins DN, Schilling JM, Sears DD, Sheridan CK, Stenger MB, Tryggvadottir R, Urban AE, Vaisar T, Van Espen B, Zhang J, Ziegler MG, Zwart SR, Charles JB, Kundrot CE, Scott GBI, Bailey SM, Basner M, Feinberg AP, Lee SMC, Mason CE, Mignot E, Rana BK, Smith SM, Snyder MP, Turek FW. The NASA Twins Study: A multidimensional analysis of a year-long human spaceflight. Science 2019; 364:364/6436/eaau8650. [PMID: 30975860 DOI: 10.1126/science.aau8650] [Citation(s) in RCA: 465] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022]
Abstract
To understand the health impact of long-duration spaceflight, one identical twin astronaut was monitored before, during, and after a 1-year mission onboard the International Space Station; his twin served as a genetically matched ground control. Longitudinal assessments identified spaceflight-specific changes, including decreased body mass, telomere elongation, genome instability, carotid artery distension and increased intima-media thickness, altered ocular structure, transcriptional and metabolic changes, DNA methylation changes in immune and oxidative stress-related pathways, gastrointestinal microbiota alterations, and some cognitive decline postflight. Although average telomere length, global gene expression, and microbiome changes returned to near preflight levels within 6 months after return to Earth, increased numbers of short telomeres were observed and expression of some genes was still disrupted. These multiomic, molecular, physiological, and behavioral datasets provide a valuable roadmap of the putative health risks for future human spaceflight.
Collapse
Affiliation(s)
- Francine E Garrett-Bakelman
- Weill Cornell Medicine, New York, NY, USA.,University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Manjula Darshi
- Center for Renal Precision Medicine, University of Texas Health, San Antonio, TX, USA
| | | | - Ruben C Gur
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ling Lin
- Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Cem Meydan
- Weill Cornell Medicine, New York, NY, USA.,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, USA
| | | | - Jad Nasrini
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | - Kumar Sharma
- Center for Renal Precision Medicine, University of Texas Health, San Antonio, TX, USA
| | | | - Lynn Taylor
- Colorado State University, Fort Collins, CO, USA
| | | | | | - Ebrahim Afshinnekoo
- Weill Cornell Medicine, New York, NY, USA.,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, USA
| | - Sara Ahadi
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aditya Ambati
- Stanford University School of Medicine, Palo Alto, CA, USA
| | | | - Daniela Bezdan
- Weill Cornell Medicine, New York, NY, USA.,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, USA
| | | | - Songjie Chen
- Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | - Marisa Covington
- National Aeronautics and Space Administration (NASA), Houston, TX, USA
| | - Brian E Crucian
- National Aeronautics and Space Administration (NASA), Houston, TX, USA
| | | | - David F Dinges
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | - Ryan P Hillary
- Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | - Peng Jiang
- Northwestern University, Evanston, IL, USA
| | | | | | | | | | | | | | | | - Tyler M Moore
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Hemal H Patel
- University of California, San Diego, La Jolla, CA, USA
| | | | - Varsha Rao
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - Rintaro Saito
- University of California, San Diego, La Jolla, CA, USA
| | - Denis N Salins
- Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | | | - Michael B Stenger
- National Aeronautics and Space Administration (NASA), Houston, TX, USA
| | | | | | | | | | - Jing Zhang
- Stanford University School of Medicine, Palo Alto, CA, USA
| | | | - Sara R Zwart
- University of Texas Medical Branch, Galveston, TX, USA
| | - John B Charles
- National Aeronautics and Space Administration (NASA), Houston, TX, USA.
| | - Craig E Kundrot
- Space Life and Physical Sciences Division, NASA Headquarters, Washington, DC, USA.
| | - Graham B I Scott
- National Space Biomedical Research Institute, Baylor College of Medicine, Houston, TX, USA.
| | | | - Mathias Basner
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | | | | | - Christopher E Mason
- Weill Cornell Medicine, New York, NY, USA. .,The Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, New York, NY, USA.,The Feil Family Brain and Mind Research Institute, New York, NY, USA.,The WorldQuant Initiative for Quantitative Prediction, New York, NY, USA
| | | | - Brinda K Rana
- University of California, San Diego, La Jolla, CA, USA.
| | - Scott M Smith
- National Aeronautics and Space Administration (NASA), Houston, TX, USA.
| | | | | |
Collapse
|
34
|
Kurgan N, Noaman N, Pergande MR, Cologna SM, Coorssen JR, Klentrou P. Changes to the Human Serum Proteome in Response to High Intensity Interval Exercise: A Sequential Top-Down Proteomic Analysis. Front Physiol 2019; 10:362. [PMID: 31001142 PMCID: PMC6454028 DOI: 10.3389/fphys.2019.00362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/14/2019] [Indexed: 12/19/2022] Open
Abstract
Exercise has been shown to improve health status and prevent chronic diseases. In contrast, overtraining can lead to maladaptation and detrimental health outcomes. These outcomes appear to be mediated in part by released peptides and, potentially, alterations in protein abundances and their modified forms, termed proteoforms. Proteoform biomarkers that either predict the beneficial effects of exercise or indicate (mal)adaptation are yet to be elucidated. Thus, we assessed the influence of high-intensity interval exercise (HIIE) on the human serum proteome to identify novel exercise-regulated proteoforms. To this end, a top-down proteomics approach was used, whereby two-dimensional gel electrophoresis was used to resolve and differentially profile intact proteoforms, followed by protein identification via liquid chromatography-tandem mass spectrometry. Blood was collected from six young-adult healthy males, pre-exercise and 5 min and 1 h post-exercise. Exercise consisted of a maximal cycle ergometer test followed by 8 min × 1 min high-intensity intervals at 90% W max, with 1 min non-active recovery between intervals. Twenty resolved serum proteoforms changed significantly in abundance at 5 min and/or 1 h post-HIIE, including apolipoproteins, serpins (protease inhibitors), and immune system proteins, known to have broad anti-inflammatory and antioxidant effects, involvement in lipid clearance, and cardio-/neuro-protective effects. This initial screening for potential biomarkers indicates that a top-down analytical proteomic approach may prove useful in further characterizing the response to exercise and in understanding the molecular mechanisms that lead to health benefits, as well as identifying novel biomarkers for exercise (mal)adaptation.
Collapse
Affiliation(s)
- Nigel Kurgan
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Nour Noaman
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
- Molecular Medicine Research Group, Department of Molecular Physiology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Melissa R. Pergande
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Stephanie M. Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, United States
| | - Jens R. Coorssen
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| | - Panagiota Klentrou
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
35
|
The effect of chronic kidney disease on lipid metabolism. Int Urol Nephrol 2018; 51:265-277. [DOI: 10.1007/s11255-018-2047-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/27/2018] [Indexed: 12/26/2022]
|
36
|
Panth N, Abbott KA, Dias CB, Wynne K, Garg ML. Differential effects of medium- and long-chain saturated fatty acids on blood lipid profile: a systematic review and meta-analysis. Am J Clin Nutr 2018; 108:675-687. [PMID: 30239550 DOI: 10.1093/ajcn/nqy167] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/29/2018] [Indexed: 12/23/2022] Open
Abstract
Background Medium-chain saturated fatty acids (MCFAs) may affect circulating lipids and lipoproteins differently than long-chain saturated fatty acids (LCSFAs), but the results from human intervention trials have been equivocal. Objective The aim of this study was to determine whether MCFAs and LCSFAs have differential impacts on blood lipids and lipoproteins. Design Five databases were searched (EMBASE, MEDLINE, CINAHL, Cochrane, and Scopus) until April 2018, and published clinical trials investigating the differential effects of dietary MCFAs and LCSFAs on blood lipids were included. Searches were limited to the English language and to studies with adults aged >18 y. Where possible, studies were pooled for meta-analysis using RevMan 5.2. The principle summary measure was the mean difference between groups calculated using the random-effects model. Results Eleven eligible crossover and 1 parallel trial were identified with a total of 299 participants [weighted mean ± SD age: 38 ± 3 y; weighted mean ± SD body mass index (kg/m2): 24 ± 2]. All studies were pooled for the meta-analysis. Diets enriched with MCFAs led to significantly higher high-density lipoprotein (HDL) cholesterol concentrations than diets enriched with LCSFAs (0.11 mmol/L; 95% CI: 0.07, 0.15 mmol/L) with no effect on triglyceride, low-density lipoprotein (LDL) cholesterol, and total cholesterol concentrations. Consumption of diets rich in MCFAs significantly increased apolipoprotein A-I (apoA-I) concentrations compared with diets rich in LCSFAs (0.08 g/L; 95% CI: 0.02, 0.14 g/L). There was no evidence of statistical heterogeneity for HDL cholesterol, apoA-I, and triglyceride concentrations; however, significant heterogeneity was observed for the total cholesterol (I2 = 49%) and LDL cholesterol analysis (I2 = 58%). Conclusion The findings of this research demonstrate a differential effect of MCFAs and LCSFAs on HDL cholesterol concentrations. Further investigations are warranted to elucidate the mechanism by which the lipid profile is altered. This trial was registered at www.crd.york.ac.uk/PROSPERO as CRD42017078277.
Collapse
Affiliation(s)
- Nisha Panth
- Nutraceuticals Research Program, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Kylie A Abbott
- Nutraceuticals Research Program, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Cintia B Dias
- Nutraceuticals Research Program, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Katie Wynne
- Department of Diabetes and Endocrinology, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, New Lambton, New South Wales, Australia
| | - Manohar L Garg
- Nutraceuticals Research Program, School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
37
|
Yu LL, Yu HH, Liang XF, Li N, Wang X, Li FH, Wu XF, Zheng YH, Xue M, Liang XF. Dietary butylated hydroxytoluene improves lipid metabolism, antioxidant and anti-apoptotic response of largemouth bass (Micropterus salmoides). FISH & SHELLFISH IMMUNOLOGY 2018; 72:220-229. [PMID: 29108969 DOI: 10.1016/j.fsi.2017.10.054] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/18/2017] [Accepted: 10/28/2017] [Indexed: 06/07/2023]
Abstract
A 10-week growth trail was conducted to investigate the efficacy and tolerance of dietary butylated hydroxytoluene (BHT) by evaluating inflammation, apoptosis and hepatic disease related to oxidative stress in largemouth bass (Micropterus salmoides). Four experimental diets were prepared with BHT supplement levels of 0 (B0), 150 (B150), 300 (B300) and 1500 (B1500) mg/kg, in which B150 was at the maximum recommended level established by European Union Regulation, and the B300 and B1500 levels were 2 and 10-fold of B150, respectively. Each diet was fed to 6 replicates with 30 largemouth bass (initial body weight, IBW = 6.20 ± 0.01 g) in each tank. The BHT inclusion level did not affect the specific growth rate, but fish in the B150 group showed the lowest feed conversion rate (P < 0.05). BHT inclusion significantly decreased the levels of plasma TC, TG, LDL, ALT and AKP, and increased the (HDL-C)/TC ratio (P < 0.05). Plasma MDA was significantly decreased in the B150 group and GSH-Px was extremely enhanced in each BHT inclusion group (P < 0.05). Hepatic T-AOC was significantly enhanced and O2- was significantly decreased in each BHT inclusion group compared to the B0 group (P < 0.05), as well as hepatic MDA was significantly decreased in B1500 group (P < 0.05). Dietary BHT inclusion down-regulated the hepatic mRNA levels of inflammation, apoptosis and fibrosis related genes, including TNFα, TGF-β1, α-SMA, IL8, IL11β and caspase-9. Moreover, BHT could improve hepatic lipid metabolism via up-regulating the mRNA levels of APOA1, CYP7A1, CYP8B1, and down-regulating the mRNA levels of PPAR-γ and APOB. Histological examination of the liver morphology with H&E and Sirius Red staining showed that BHT inclusion decreased necrotic degenerative changes and collagen deposition in largemouth bass. An immunofluorescence examination revealed significantly decreased cleaved caspase-3 signals in the BHT groups. In conclusion, the results demonstrated that ROS induces hepatic cell apoptosis and fibrosis via the intrinsic pathway of apoptosis by activating caspase-9 in the mitochondria and then initiates apoptosis by activating caspase-3. Consuming 2.32-23.80 mg/kg·bw/d (150-1500 mg/kg in diet) of BHT effectively improved the plasma and hepatic lipid metabolism, antioxidant response as well as reduced ROS production, protecting hepatic cells from injury. It is implied that even a 10-fold increase of the maximum level of BHT (150 mg/kg) is safe for the largemouth bass.
Collapse
Affiliation(s)
- L L Yu
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - H H Yu
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - X F Liang
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - N Li
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - X Wang
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - F H Li
- Beijing General Station of Animal Husbandry Senior Veterinary, 100107, China
| | - X F Wu
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Y H Zheng
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - M Xue
- National Aquafeed Safety Assessment Center, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Key Laboratory of Feed Biotechnology of Ministry of Agriculture, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China; Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, 430070, China.
| | - X F Liang
- Freshwater Aquaculture Collaborative Innovation Center of Hubei Province, Wuhan, 430070, China; College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
38
|
Lee CL, Wen JY, Hsu YW, Pan TM. The blood lipid regulation of Monascus-produced monascin and ankaflavin via the suppression of low-density lipoprotein cholesterol assembly and stimulation of apolipoprotein A1 expression in the liver. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2016; 51:27-37. [PMID: 27422746 DOI: 10.1016/j.jmii.2016.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 10/26/2015] [Accepted: 06/13/2016] [Indexed: 10/21/2022]
Abstract
BACKGROUND/PURPOSES Monascin (MS) and ankaflavin (AK) produced by Monascus purpureus NTU 568 were proven to show excellent hypolipidemic effects in our previous studies; however, the mechanism is still unclear. METHODS This study used MS, AK, and monacolin K as test substances and performed tests on rats fed high-fat and high-cholesterol diet for 8 weeks. The lipid levels and the related protein levels of the rats were assessed to understand the effects of MS, AK, and monacolin K on lipid metabolism. RESULTS MS and AK lowered low-density lipoprotein cholesterol (LDL-C) and preserved high-density lipoprotein cholesterol contents. MS and AK inhibited acetyl-coenzyme A acetyltransferase, microsomal triglyceride transfer protein, and apolipoprotein (apo) B-100 expression, thereby preventing LDL assembly. In addition, enhanced LDL-receptor expression increased the transport of LDL-C to the liver for metabolism. MS and AK also significantly increase apo A1 expression, which facilitates high-density lipoprotein cholesterol formation. CONCLUSION Monascus-fermented MS and AK can perform blood lipid regulation via the suppression of LDL-C assembly and stimulation of apo A1 expression in liver.
Collapse
Affiliation(s)
- Chun-Lin Lee
- Department of Life Science, National Taitung University, Taitung, Taiwan, ROC
| | - Ja-Yan Wen
- Department of Life Science, National Taitung University, Taitung, Taiwan, ROC
| | - Ya-Wen Hsu
- R&D Division, SunWay Biotechnology Company Limited, Taipei, Taiwan, ROC
| | - Tzu-Ming Pan
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
39
|
Ford J, Lawson M, Fowler D, Maruyama N, Mito S, Tomiyasu K, Kinoshita S, Suzuki C, Kawaguchi A, Round P, Boyce M, Warrington S, Weber W, van Deventer S, Kastelein JJP. Tolerability, pharmacokinetics and pharmacodynamics of TA-8995, a selective cholesteryl ester transfer protein (CETP) inhibitor, in healthy subjects. Br J Clin Pharmacol 2015; 78:498-508. [PMID: 24628035 DOI: 10.1111/bcp.12380] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 03/11/2014] [Indexed: 12/20/2022] Open
Abstract
AIMS Two double-blind, randomized studies were conducted to assess the tolerability, pharmacokinetics and pharmacodynamics of oral TA-8995, a new cholesteryl ester transfer protein (CETP) inhibitor, in healthy subjects. METHODS Study 1: Subjects received single doses of TA-8995 or placebo (fasted). Doses were 5, 10, 25, 50 (fed/fasted), 100 and 150 mg (Caucasian males, 18-55 years), 25 mg (Caucasian males, > 65 years and Caucasian females, 18-55 years), 25, 50, 100 and 150 mg (Japanese males, 18-55 years). Study 2: Caucasian males (18-55 years) received 1, 2.5, 10 or 25 mg once daily TA-8995 or placebo for 21-28 days. Blood and urine for pharmacokinetics and/or pharmacodynamics were collected. Tolerability was assessed by adverse events, vital signs, electrocardiograms and laboratory safety tests. RESULTS Peak TA-8995 concentrations occurred approximately 4 h post-dose. Mean half-lives ranged from 81 to 166 h, without an obvious dose relationship. Exposure increased less than proportionally to dose. TA-8995 was not excreted in urine. Following 2.5 to 25 mg once daily dosing, TA-8995 demonstrated nearly complete inhibition of CETP activity (92-99%), increased high density lipoprotein-cholesterol (HDL-C) by 96 to 140% and decreased low density liporotein-cholesterol (LDL-C) by 40% to 53%. There were dose-related increases in apolipoproteins A-1 and E, HDL2-C and HDL3-C, and decreases in apolipoprotein B and lipoprotein A. There was no evidence of significant effects of age, gender, ethnicity or food on pharmacokinetics or pharmacodynamics. All doses were well tolerated. CONCLUSIONS TA-8995 is a potent CETP inhibitor and warrants further investigation.
Collapse
Affiliation(s)
- John Ford
- Dezima Pharma BV, Naarden, The Netherlands; Xention Ltd, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kaldmäe M, Viigimaa M, Zemtsovskaja G, Kaart T, Abina J, Annuk M. Prevalence and determinants of hypertension in Estonian adults. Scand J Public Health 2014; 42:504-10. [DOI: 10.1177/1403494814532565] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Although Eastern Europe, including Estonia, has one of the highest morbidity and mortality rates associated with hypertension, there is little information in the literature concerning the biochemical risk factor profile or its association with hypertension in Estonia. This study examined the cross-sectional gender-stratified association between biochemical risk markers and hypertension in a population-based sample of adults in Estonia. Methods: The study was carried out in Tallinn, Estonia and consisted of 511 men and 600 women with a mean age of 46 years. Physiological measurements were taken and blood samples drawn to measure the following markers: cholesterol, high- and low-density lipoprotein cholesterol, apolipoproteins A-1 and B, lipoprotein(a), triglycerides, glucose, fibrinogen, high-sensitivity C-reactive protein and homocysteine. Results: Overall, 36% of participants had hypertension, with approximately 80% being aware of their condition. A total of 40% of participants reported taking antihypertensive medication. Multivariate binary logistic regression analysis showed that a decrease in high-density lipoprotein cholesterol and increases in age, body mass index, apolipoprotein B, triglyceride and homocysteine levels were associated with an increased probability of hypertension. Conclusions: Elevations in biochemical markers and cardiovascular risk factors are associated with hypertension. Increasing body mass index, triglyceride, apolipoprotein B and homocysteine levels with decreasing high-density lipoprotein cholesterol level should be investigated and monitored in Estonian adults.
Collapse
Affiliation(s)
- Margit Kaldmäe
- Tallinn University, Institute of Mathematics and Natural Sciences, Tallinn, Estonia
| | - Margus Viigimaa
- North Estonia Medical Centre, Centre of Cardiology, Tallinn, Estonia
- Tallinn University of Technology, Institute of Biomedical Engineering, Tallinn, Estonia
| | | | - Tanel Kaart
- Estonian University of Life Sciences, Institute of Veterinary Medicine and Animal Sciences, Tartu, Estonia
| | - Jelena Abina
- Tallinn University of Technology, Institute of Biomedical Engineering, Tallinn, Estonia
| | - Margus Annuk
- Tallinn University, Institute of Mathematics and Natural Sciences, Tallinn, Estonia
| |
Collapse
|
41
|
Wang SY, Hsieh MC, Tu ST, Chuang CS. New frontiers in the treatment of diabetic dyslipidemia. Rev Diabet Stud 2013; 10:204-12. [PMID: 24380093 DOI: 10.1900/rds.2013.10.204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Dyslipidemia is a major risk factor for cardiovascular complications in people with diabetes. Lowering low-density lipoprotein cholesterol (LDL-C) levels is effective in the primary and secondary prevention of diabetic vascular complications. However, LDL-C levels do not reflect all aspects of diabetic dyslipidemia, which is characterized by hypertriglyceridemia and low high-density lipoprotein cholesterol (HDL-C). Statins, nicotinic acid, and fibrates play a role in treating diabetic dyslipidemia. Atherosclerosis is a major disorder of the blood vessel wall in patients with diabetes. A number of antihyperlipidemic agents may be beneficial and exhibit effects at the actual site of vascular disease and not only on plasma lipoprotein concentrations. Several novel therapeutic compounds are currently being developed. These include additional therapeutics for LDL-C, triglycerides, HDL-C, and modulators of inflammation that can be used as possible synergic agents for the treatment of atherosclerosis and irregularities in plasma lipoprotein concentrations.
Collapse
Affiliation(s)
- Shu-Yi Wang
- Departments of Endocrinology and Metabolism, Changhua Christian Hospital, Changhua, Taiwan
| | - Ming-Chia Hsieh
- Departments of Endocrinology and Metabolism, Changhua Christian Hospital, Changhua, Taiwan
| | - Shih-Te Tu
- Departments of Endocrinology and Metabolism, Changhua Christian Hospital, Changhua, Taiwan
| | - Chieh-Sen Chuang
- Departments of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| |
Collapse
|
42
|
ApoB/apoA1 is an effective predictor of coronary heart disease risk in overweight and obesity. J Biomed Res 2013; 25:266-73. [PMID: 23554700 PMCID: PMC3597070 DOI: 10.1016/s1674-8301(11)60036-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 02/18/2011] [Accepted: 05/18/2011] [Indexed: 01/09/2023] Open
Abstract
We investigated the relationship of apoB/apoA1 ratio and coronary heart disease (CHD) in persons who were overweight or obese. The subjects were divided by the body mass indexes (BMI) into the normal weight group (n=397, BMI<24 kg/m(2)) and the overweight group (n=400, BMI>24 kg/m(2)). Our results showed that the over-weight group had higher blood pressure [(130.15±19.01) mmHg vs (123.66±18.70) mmHg] and higher levels of blood sugar [(7.09±2.89) mmol/L vs (6.21±2.59) mmol/L], triglyceride [(1.93±1.19) mmol/L vs (1.44±0.85) mmol/L], total cholesterol [(4.26±1.06) mmol/L vs (4.09±0.99) mmol/L], low-density lipoprotein cholesterol (LDL-C) [(2.56±0.75) mmol/L vs (2.39±0.72) mmol/L], and apoB [(0.83±0.27) mg/L vs (0.78±0.23) mg/L], and a higher apoB/apoA1 ratio (0.83±0.27 vs 0.75±0.25) and lower levels high-density lipoprotein cholesterol [(1.10±0.26) mmol/L vs (1.21±0.31) mmol/L] and apoA1 [(1.04±0.20) mg/L vs (1.08±0.22) mg/L] than those of the normal weight group (all P < 0.05). The prevalence of CHD in the over-weight group in the lowest LDL quartile was almost twice greater than that of the highest apoB/apoA1 quartile, compared with the subjects in the lowest apoB/apoA1 quartile. The higher apoB/apoA1 quartile was in agreement with the higher prevalence of CHD. In the overweight and obesity group, the area under ROC curve (AUC) was the highest for apoB/apoA1 (0.655). The cut-off point of apoB/apoA1 for optimal sensitivity and specificity was at 0.80, with a sensitivity of 57.19% and a specificity of 71.72%. In conclusion, apoB and apoA1 were simple clinical indicators, and the apoB/apoA1 ratio was closely related with CHD in overweight and obese patients. The apoB/apoA1 ratio may provide some useful information in the differential diagnosis.
Collapse
|
43
|
Lee CM, Mudaliar MAV, Haggart DR, Wolf CR, Miele G, Vass JK, Higham DJ, Crowther D. Simultaneous non-negative matrix factorization for multiple large scale gene expression datasets in toxicology. PLoS One 2012; 7:e48238. [PMID: 23272042 PMCID: PMC3522745 DOI: 10.1371/journal.pone.0048238] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 09/25/2012] [Indexed: 01/02/2023] Open
Abstract
Non-negative matrix factorization is a useful tool for reducing the dimension of large datasets. This work considers simultaneous non-negative matrix factorization of multiple sources of data. In particular, we perform the first study that involves more than two datasets. We discuss the algorithmic issues required to convert the approach into a practical computational tool and apply the technique to new gene expression data quantifying the molecular changes in four tissue types due to different dosages of an experimental panPPAR agonist in mouse. This study is of interest in toxicology because, whilst PPARs form potential therapeutic targets for diabetes, it is known that they can induce serious side-effects. Our results show that the practical simultaneous non-negative matrix factorization developed here can add value to the data analysis. In particular, we find that factorizing the data as a single object allows us to distinguish between the four tissue types, but does not correctly reproduce the known dosage level groups. Applying our new approach, which treats the four tissue types as providing distinct, but related, datasets, we find that the dosage level groups are respected. The new algorithm then provides separate gene list orderings that can be studied for each tissue type, and compared with the ordering arising from the single factorization. We find that many of our conclusions can be corroborated with known biological behaviour, and others offer new insights into the toxicological effects. Overall, the algorithm shows promise for early detection of toxicity in the drug discovery process.
Collapse
Affiliation(s)
- Clare M. Lee
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, United Kingdom
| | | | | | - C. Roland Wolf
- University of Dundee Medical Research Institute, Ninewells Hospital & Medical School, Dundee, United Kingdom
- CXR Biosciences Ltd, Dundee, United Kingdom
| | - Gino Miele
- Epsistem Ltd., Manchester, United Kingdom
| | - J. Keith Vass
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, United Kingdom
| | - Desmond J. Higham
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, United Kingdom
- * E-mail:
| | | |
Collapse
|
44
|
Abstract
3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) are established first line treatments for hypercholesterolaemia. In addition to the direct effects of statins in reducing concentrations of atherogenic low density lipoprotein cholesterol (LDL-C), several studies have indicated that the beneficial effects of statins may be due to some of their cholesterol-independent, multiple (pleiotropic) effects which may differ between different members of the class. Pitavastatin is a novel synthetic lipophilic statin that has a number of pharmacodynamic and pharmacokinetic properties distinct from those of other statins, which may underlie its potential pleiotropic benefits in reducing cardiovascular risk factors. This review examines the principal pleiotropic effects of pitavastatin on endothelial function, vascular inflammation, oxidative stress and thrombosis. The article is based on a systematic literature search carried out in December 2010, together with more recent relevant publications where appropriate. The available data from clinical trials and in vitro and animal studies suggest that pitavastatin is not only effective in reducing LDL-C and triglycerides, but also has a range of other effects. These include increasing high density lipoprotein cholesterol, decreasing markers of platelet activation, improving cardiac, renal and endothelial function, and reducing endothelial stress, lipoprotein oxidation and, ultimately, improving the signs and symptoms of atherosclerosis. It is concluded that the diverse pleiotropic actions of pitavastatin may contribute to reducing cardiovascular morbidity and mortality beyond that achieved through LDL-C reduction.
Collapse
Affiliation(s)
- Jean Davignon
- Hyperlipidemia and Atherosclerosis Research Group, Clinical Research Institute of Montréal (IRCM) and University of Montréal, QC, Canada.
| |
Collapse
|
45
|
Da Costa LA, Arora P, García-Bailo B, Karmali M, El-Sohemy A, Badawi A. The association between obesity, cardiometabolic disease biomarkers, and innate immunity-related inflammation in Canadian adults. Diabetes Metab Syndr Obes 2012; 5:347-55. [PMID: 23055759 PMCID: PMC3468056 DOI: 10.2147/dmso.s35115] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Obesity is associated with a state of chronic inflammation, and increased cardiometabolic disease risk. The present study examined the relationship between body mass index (BMI) and cardiometabolic and inflammatory biomarkers among normal weight, overweight, and obese Canadian adults. METHODS Subjects (n = 1805, aged 18 to 79 years) from the Canadian Health Measures Survey (CHMS) were examined for associations between BMI, cardiometabolic markers (apolipoprotein [Apo] A1, ApoB, low-density lipoprotein cholesterol [LDL-C], high-density lipoprotein cholesterol [HDL-C], total cholesterol, total cholesterol/HDL ratio [total:HDL-C ratio], triglycerides, and glycosylated hemoglobin [HbA(1c)]), inflammatory factors (C-reactive protein [CRP], fibrinogen, and homocysteine), and 25-hydroxyvitamin D [25(OH)D]. Bootstrap weights for variance and sampling weights for point estimates were applied to account for the complex survey design. Linear regression models adjusted for age, sex, physical activity, smoking status, and ethnicity (in addition to season of clinic visit, for vitamin D analyses only) were used to examine the association between cardiometabolic markers, inflammatory factors, and BMI in Canadian adults. RESULTS All biomarkers were significantly associated with BMI (P ≤ 0.001). ApoA1 (β = -0.31, P < 0.0001), HDL-C (β = -0.61, P < 0.0001), and 25(OH)D (β = -0.25, P < 0.0001) were inversely associated with BMI, while all other biomarkers showed positive linear associations. Distinct patterns of association were noted among normal weight, overweight, and obese groups, excluding CRP which showed a significant positive association with BMI in the overall population (β = 2.80, P < 0.0001) and in the normal weight (β = 3.20, P = 0.02), overweight (β = 3.53, P = 0.002), and obese (β = 2.22, P = 0.0002) groups. CONCLUSIONS There is an apparent profile of cardiometabolic and inflammatory biomarkers that emerges as BMI increases from normal weight to obesity. Understanding these profiles may permit developing an effective approach for early risk prediction for cardiometabolic disease.
Collapse
Affiliation(s)
- Laura A Da Costa
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Office of Biotechnology and Population Health, Public Health Agency of Canada, Toronto, Ontario, Canada
| | - Paul Arora
- Office of Biotechnology and Population Health, Public Health Agency of Canada, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Bibiana García-Bailo
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Office of Biotechnology and Population Health, Public Health Agency of Canada, Toronto, Ontario, Canada
| | - Mohamed Karmali
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
- Office of Biotechnology and Population Health, Public Health Agency of Canada, Toronto, Ontario, Canada
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Alaa Badawi
- Office of Biotechnology and Population Health, Public Health Agency of Canada, Toronto, Ontario, Canada
- Correspondence: Alaa Badawi, Office for Biotechnology, Genomics and Population Health, Public Health Agency of Canada, 180 Queen Street West, 11th floor, Toronto, Ontario, M5V 3L7, Canada, Email
| |
Collapse
|
46
|
Park KH, Cho KH. High-density lipoprotein (HDL) from elderly and reconstituted HDL containing glycated apolipoproteins A-I share proatherosclerotic and prosenescent properties with increased cholesterol influx. J Gerontol A Biol Sci Med Sci 2011; 66:511-20. [PMID: 21415260 DOI: 10.1093/gerona/glr016] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
High-density lipoprotein (HDL) is a strong antioxidant, anti-inflammatory, and antisenescence molecule. However, in the current study, HDL from the elderly group (E-HDL) exhibited increased glycation with apolipoprotein (apo) A-I multimerization and decreased phospholipid content. Similarly, glycated apoA-I (gA-I) by fructosylation has a covalently multimerized band without a crosslinker and impaired phospholipid-binding ability. Treatment of human dermal fibroblasts and macrophages with E-HDL and gA-I caused more severe cellular senescence and foam cell formation, respectively; however, treatment with HDL from a young group (Y-HDL) and native apoA-I (nA-I) suppressed senescence and atherosclerosis. E-HDL(3) and reconstituted HDL (rHDL) containing gA-I showed enhanced cholesterol influx into macrophages compared with Y-HDL(3) and nA-I-rHDL. In conclusion, E-HDL and gA-I-rHDL share similar physiologic properties in macrophages and human dermal fibroblasts. E-HDL and gA-I-rHDL exacerbated cellular senescence and atherosclerosis with increased cellular cholesterol influx.
Collapse
Affiliation(s)
- Ki-Hoon Park
- School of Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | | |
Collapse
|
47
|
Ahmed K, Rask P, Hurtig-Wennlof A. Serum apolipoproteins, apoB/apoA-I ratio and objectively measured physical activity in elderly. SCAND CARDIOVASC J 2011; 45:105-11. [DOI: 10.3109/14017431.2011.553966] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Urbonavicius S, Lindholt JS, Delbosc S, Urbonaviciene G, Henneberg EW, Vorum H, Meilhac O, Honoré B. Proteins associated with the size and expansion rate of the abdominal aortic aneurysm wall as identified by proteomic analysis. Interact Cardiovasc Thorac Surg 2010; 11:433-41. [PMID: 20675398 DOI: 10.1510/icvts.2010.238139] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVES Identification of biomarkers for the natural history of abdominal aortic aneurysms (AAA) holds the key to non-surgical intervention and improved selection for AAA repair. We aimed to associate the basic proteomic composition of AAA wall tissue with the expansion rate and size in patients with AAA. METHODS A proteomic approach was used, consisting of two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and mass spectrometry (MS) to identify differentially expressed proteins in AAA tissue. Relevant protein spots were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS Spearman's correlation analysis revealed 16 protein spots were strongly correlated with AAA expansion rate (ρ>±0.75). Nine protein spots were identified. Six protein spots showed correlation with AAA size (ρ>±0.5). Three protein spots were identified: vitronectin with traces of calreticulin, albumin and a spot containing two proteins: collagen α-3(VI) chain and vitamin D binding protein. Interestingly, in our previous study vitronectin was shown to be down-regulated in a ruptured AAA group compared with non-ruptured AAA. Western blot analysis in the present study confirmed a correlation of vitronectin bands with AAA size in aortic aneurysm tissue. CONCLUSION A proteomic approach seems valuable, and identified several candidates not previously associated with AAA. Larger studies are required to confirm the potential and clinical role of the identified proteins.
Collapse
Affiliation(s)
- Sigitas Urbonavicius
- Department of Vascular Surgery, Vascular Research Unit, Viborg Hospital, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lavie CJ, Milani RV, O'Keefe JH. To B or not to B: is non-high-density lipoprotein cholesterol an adequate surrogate for apolipoprotein B? Mayo Clin Proc 2010; 85:446-50. [PMID: 20435838 PMCID: PMC2861974 DOI: 10.4065/mcp.2010.0058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Carl J Lavie
- Cardiac Rehabilitation and Exercise Laboratories, John Ochsner Heart and Vascular Institute, Ochsner Medical Center, 1514 Jefferson Hwy, New Orleans, LA 70121-2483, USA.
| | | | | |
Collapse
|
50
|
Samouilidou EC, Grapsa E. Relationship of Serum Cystatin C with C-Reactive Protein and Apolipoprotein A1 in Patients on Hemodialysis. Ren Fail 2009; 30:711-5. [DOI: 10.1080/08860220802212973] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|