1
|
Shi Y, Bao L, Li Y, Ou D, Li J, Liu X, Deng N, Deng C, Huang X, Zhang W, Ding H. Multi-omics combined to investigate potential druggable therapeutic targets for stroke: A systematic Mendelian randomization study and transcriptome verification. J Affect Disord 2024; 366:196-209. [PMID: 39214372 DOI: 10.1016/j.jad.2024.08.182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/15/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Stroke is a highly prevalent and disabling disease whose disease mechanisms are not fully understood. The discovery of disease-associated proteins with genetic evidence of pathogenicity provides an opportunity to identify new therapeutic targets. METHOD We examined the observed and causal associations of thousands of plasma and inflammatory proteins that were measured using affinity-based proteomic assays. First, we pooled >3000 relevant proteins using a fixed-effects meta-analysis of 2 population-based studies involving 48,383 participants, then investigated the causal effects of stroke and its subtype-associated proteins by forward Mendelian randomization using cis-protein quantitative locus genetic tools identified from genome-wide association studies of these >48,000 individuals. To improve the accuracy of causal estimation, we implemented a systematic Mendelian randomization model that accounts for cascading imbalances between instruments and tested the robustness of causal estimation through multi-method analyses. To further validate the hypothesis that ginsenoside Rg1 monomer acts on the five protein targets screened for drug-targeted regulation, we conducted a comparative analysis of the mRNA (gene) expression levels of a limited number of genes in the brain tissues of different groups of SD rats. The druggability of the candidate proteins was investigated and the mechanism of action and potential targeting side effects were explored by Phenome-wide MR. RESULTS Six circulating proteins were identified to have a significant genetic association with stroke (PFDR < 0.05). For example, in patients with cardioembolic stroke, higher genetically predicted APRT was associated with a lower risk of cardioembolic stroke (ORivw [95 % CI] = 0.641 [0.517, 0.795]; P = 5.25 × 10-5, ORSMR [95 % CI] = 0.572, [0.397, 0.825], PSMR = 0.003). Mediation analyses suggested that atrial fibrillation, angina pectoris, and heart failure may mediate the association of CD40L, LIFR, and UPA with stroke. Molecular docking revealed promising interactions between the identified proteins and glycosides. Transcriptomic sequencing in animal models indicated that ginsenoside Rg1 may act through APRT, IL15RA, and VSIR pathways, with APRT showing significant variability in mRNA sequencing expression. Phenome-wide MR of the six target proteins showed an overwhelming predominance of PFDR > 0.05, indicating less toxicity. CONCLUSIONS The present study provides genetic evidence to support the potential efficacy of targeting the three druggable protein targets for the treatment of stroke. This is achieved by triangulating population genomic and proteomic data. Furthermore, the study validates the pathway mechanisms by which APRT, IL15RA, and VSIR dock ginsenoside Rg1 in animal models. This will help to prioritize stroke drug development.
Collapse
Affiliation(s)
- Yiming Shi
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Le Bao
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Yanling Li
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Dian Ou
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Jiating Li
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Xiaodan Liu
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Nujiao Deng
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Changqing Deng
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Xiaoping Huang
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China.
| | - Wei Zhang
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China.
| | - Huang Ding
- Hunan University of Chinese Medicine, Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China.
| |
Collapse
|
2
|
Bellos I, Marinaki S, Lagiou P, Benetou V. Association of soluble suppression of tumorigenicity 2 with mortality and adverse outcomes in chronic kidney disease: a systematic review and meta-analysis. Clin Exp Nephrol 2024; 28:988-1003. [PMID: 38678167 PMCID: PMC11493800 DOI: 10.1007/s10157-024-02506-6] [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: 02/12/2024] [Accepted: 04/11/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Early risk stratification is necessary to prevent chronic kidney disease progression and complications. This systematic review aims to evaluate the association of soluble suppression of tumorigenicity 2 (sST2), a member of the interleukin-1 receptor family, with all-cause mortality, cardiovascular disease and renal function deterioration among chronic kidney disease patients. METHODS PubMed, Scopus, Web of Science, CENTRAL and Google Scholar were systematically searched from inception to December 20, 2023. Cohort studies examining the prognostic role of sST2 levels in pre-dialysis and dialysis patients were included. In case of 3 or more studies per outcome, conventional and dose-response meta-analyses were conducted. RESULTS Overall, 21 studies were included comprising 15,100 patients. In pre-dialysis patients, the qualitative synthesis of studies suggested that high sST2 is associated with significantly increased all-cause mortality, while evidence regarding cardiovascular events or kidney disease progression was conflicting. In the dialysis population, high sST2 was linked to an elevated risk of all-cause (Hazard ratio-HR: 3.00, 95% confidence intervals-CI: 1.95-4.61) and cardiovascular (HR: 2.38, 95% CI: 1.69-3.34) mortality. Dose-response meta-analysis suggested a log-linear association of sST2 with both all-cause (χ2: 34.65, p value < 0.001) and cardiovascular (χ2: 29.14, p value < 0.001) mortality, whereas findings regarding cardiovascular events were limited with mixed results. CONCLUSIONS High sST2 values are associated with an increased risk of all-cause mortality in pre-dialysis and dialysis patients, as well as with an elevated risk of cardiovascular mortality in the dialysis population. Further studies are needed to elucidate its potential association with cardiovascular events and kidney disease progression.
Collapse
Affiliation(s)
- Ioannis Bellos
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece.
- Department of Nephrology and Renal Transplantation, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Smaragdi Marinaki
- Department of Nephrology and Renal Transplantation, Medical School, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Pagona Lagiou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece
| | - Vassiliki Benetou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str, 115 27, Athens, Greece
| |
Collapse
|
3
|
Han S, Wu X, Peng X, Zhang C. Association of asthma with the risk of cardiovascular disease: A Mendelian randomization study. Exp Gerontol 2024; 195:112549. [PMID: 39159834 DOI: 10.1016/j.exger.2024.112549] [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: 06/27/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Association of asthma with the risk of cardiovascular disease has not been fully elucidated. So, this study tried to explore the genetic effect of asthma on five cardiovascular diseases and 90 peripheral cardiovascular proteins to answer the above topic. METHODS Instrumental variables predicting asthma was extracted from its genome-wide association study data. Two-sample and multivariate MR approaches were used to assess the genetic association of exposure factor (i.e., asthma) with outcome factors (i.e., hypertension, atrial fibrillation, angina pectoris, myocardial infarction, heart failure, and 90 peripheral cardiovascular proteins). RESULTS First, asthma nominally increased the risk of hypertension and atrial fibrillation (OR = 1.009, 95%CI = 1.003-1.016, P = 0.004; OR = 1.074, 95%CI = 1.024-1.127, P = 0.003). Second, of the 90 cardiovascular proteins, asthma was associated with the increased levels of tumor necrosis factor ligand superfamily member 14 and CC motif chemokine 4 (β = 0.145, 95%CI = 0.077-0.212, P = 2.936e-05; β = 0.128, 95%CI = 0.063-0.193, P = 1.036e-04). Third, CC motif chemokine 4 increased the risk of hypertension (P = 0.043); and after adjusting for this protein, asthma still increased the risk of hypertension, but the strength of its P-value changed from 0.004 to 0.011. CONCLUSION Asthma was a risk factor for hypertension and atrial fibrillation at the genetic level, and CC motif chemokine 4 might play a mediating role in the mechanism by which asthma promoted hypertension. Thus, effective control of asthma may help reduce the risk of some cardiovascular diseases in older adults.
Collapse
Affiliation(s)
- Shuang Han
- Department of Respiratory and Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, Shandong Province, China
| | - Xiao Wu
- Department of Respiratory and Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, Shandong Province, China.
| | - Xiufa Peng
- Medical Record Management Center, the Affiliated Hospital of Qingdao University, Qingdao 266042, Shandong Province, China
| | - Chunling Zhang
- Department of Respiratory and Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao 266042, Shandong Province, China.
| |
Collapse
|
4
|
Luo H, Petrera A, Hauck SM, Rathmann W, Herder C, Gieger C, Hoyer A, Peters A, Thorand B. Association of plasma proteomics with mortality in individuals with and without type 2 diabetes: Results from two population-based KORA cohort studies. BMC Med 2024; 22:420. [PMID: 39334377 PMCID: PMC11438072 DOI: 10.1186/s12916-024-03636-0] [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: 04/03/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Protein biomarkers may contribute to the identification of vulnerable subgroups for premature mortality. This study aimed to investigate the association of plasma proteins with all-cause and cause-specific mortality among individuals with and without baseline type 2 diabetes (T2D) and evaluate their impact on the prediction of all-cause mortality in two prospective Cooperative Health Research in the Region of Augsburg (KORA) studies. METHODS The discovery cohort comprised 1545 participants (median follow-up 15.6 years; 244 with T2D: 116 total, 62 cardiovascular, 31 cancer-related and 23 other-cause deaths; 1301 without T2D: 321 total, 114 cardiovascular, 120 cancer-related and 87 other-cause deaths). The validation cohort comprised 1031 participants (median follow-up 6.9 years; 203 with T2D: 76 total, 45 cardiovascular, 19 cancer-related and 12 other-cause deaths; 828 without T2D: 169 total, 74 cardiovascular, 39 cancer-related and 56 other-cause deaths). We used Cox regression to examine associations of 233 plasma proteins with all-cause and cause-specific mortality and Lasso regression to construct prediction models for all-cause mortality stratifying by baseline T2D. C-index, category-free net reclassification index (cfNRI), and integrated discrimination improvement (IDI) were conducted to evaluate the predictive performance of built prediction models. RESULTS Thirty-five and 62 proteins, with 29 overlapping, were positively associated with all-cause mortality in the group with and without T2D, respectively. Out of these, in the group with T2D, 35, eight, and 26 were positively associated with cardiovascular, cancer-related, and other-cause mortality, while in the group without T2D, 55, 41, and 47 were positively associated with respective cause-specific outcomes in the pooled analysis of both cohorts. Regulation of insulin-like growth factor (IGF) transport and uptake by IGF-binding proteins emerged as a unique pathway enriched for all-cause and cardiovascular mortality in individuals with T2D. The combined model containing the selected proteins (five and 12 proteins, with four overlapping, in the group with and without T2D, respectively) and clinical risk factors improved the prediction of all-cause mortality by C-index, cfNRI, and IDI. CONCLUSIONS This study uncovered shared and unique mortality-related proteins in persons with and without T2D and emphasized the role of proteins in improving the prediction of mortality in different T2D subgroups.
Collapse
Affiliation(s)
- Hong Luo
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Partner Düsseldorf, Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Annika Hoyer
- Biostatistics and Medical Biometry, Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, Pettenkofer School of Public Health, LMU Munich, Munich, Germany.
- German Center for Diabetes Research (DZD), Partner München-Neuherberg, Neuherberg, Germany.
| |
Collapse
|
5
|
Schmidt IM, Surapaneni AL, Zhao R, Upadhyay D, Yeo WJ, Schlosser P, Huynh C, Srivastava A, Palsson R, Kim T, Stillman IE, Barwinska D, Barasch J, Eadon MT, El-Achkar TM, Henderson J, Moledina DG, Rosas SE, Claudel SE, Verma A, Wen Y, Lindenmayer M, Huber TB, Parikh SV, Shapiro JP, Rovin BH, Stanaway IB, Sathe NA, Bhatraju PK, Coresh J, Rhee EP, Grams ME, Waikar SS. Plasma proteomics of acute tubular injury. Nat Commun 2024; 15:7368. [PMID: 39191768 PMCID: PMC11349760 DOI: 10.1038/s41467-024-51304-x] [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: 02/13/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
The kidney tubules constitute two-thirds of the cells of the kidney and account for the majority of the organ's metabolic energy expenditure. Acute tubular injury (ATI) is observed across various types of kidney diseases and may significantly contribute to progression to kidney failure. Non-invasive biomarkers of ATI may allow for early detection and drug development. Using the SomaScan proteomics platform on 434 patients with biopsy-confirmed kidney disease, we here identify plasma biomarkers associated with ATI severity. We employ regional transcriptomics and proteomics, single-cell RNA sequencing, and pathway analysis to explore biomarker protein and gene expression and enriched biological pathways. Additionally, we examine ATI biomarker associations with acute kidney injury (AKI) in the Kidney Precision Medicine Project (KPMP) (n = 44), the Atherosclerosis Risk in Communities (ARIC) study (n = 4610), and the COVID-19 Host Response and Clinical Outcomes (CHROME) study (n = 268). Our findings indicate 156 plasma proteins significantly linked to ATI with osteopontin, macrophage mannose receptor 1, and tenascin C showing the strongest associations. Pathway analysis highlight immune regulation and organelle stress responses in ATI pathogenesis.
Collapse
Affiliation(s)
- Insa M Schmidt
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Section of Nephrology, Boston Medical Center, Boston, MA, USA.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Aditya L Surapaneni
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Runqi Zhao
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Boston Medical Center, Boston, MA, USA
| | - Dhairya Upadhyay
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Wan-Jin Yeo
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Pascal Schlosser
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center - University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Courtney Huynh
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Boston Medical Center, Boston, MA, USA
| | - Anand Srivastava
- Division of Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Ragnar Palsson
- Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Taesoo Kim
- Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Isaac E Stillman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daria Barwinska
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonathan Barasch
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael T Eadon
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tarek M El-Achkar
- Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel Henderson
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Dennis G Moledina
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Sylvia E Rosas
- Kidney and Hypertension Unit, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Sophie E Claudel
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Boston Medical Center, Boston, MA, USA
| | - Ashish Verma
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Boston Medical Center, Boston, MA, USA
| | - Yumeng Wen
- Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maja Lindenmayer
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samir V Parikh
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - John P Shapiro
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brad H Rovin
- Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ian B Stanaway
- Kidney Research Institute, Division of Nephrology, University of Washington School of Medicine, Seattle, WA, USA
| | - Neha A Sathe
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Pavan K Bhatraju
- Kidney Research Institute, Division of Nephrology, University of Washington School of Medicine, Seattle, WA, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Josef Coresh
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Eugene P Rhee
- Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
| | - Morgan E Grams
- Department of Medicine, New York University Langone School of Medicine, New York, NY, USA
| | - Sushrut S Waikar
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Section of Nephrology, Boston Medical Center, Boston, MA, USA
| |
Collapse
|
6
|
Sircuța AF, Grosu ID, Schiller A, Petrica L, Ivan V, Schiller O, Bodea M, Mircea MN, Goleț I, Bob F. The Relationship between Circulating Kidney Injury Molecule-1 and Cardiovascular Morbidity and Mortality in Hemodialysis Patients. Biomedicines 2024; 12:1903. [PMID: 39200365 PMCID: PMC11352197 DOI: 10.3390/biomedicines12081903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND The importance of identifying mortality biomarkers in chronic kidney disease (CKD), and especially in patients treated with hemodialysis (HD), has become evident. In addition to being a marker of tubulointerstitial injury, plasma kidney injury molecule-1 (KIM-1) has been mentioned in regard to HD patients as a risk marker for cardiovascular (CV) mortality and coronary artery calcification. The aim of this study was to assess the level of plasma KIM-1 as a marker of cardiovascular disease (CVD) and mortality in CKD5-HD patients (patients with CKD stage G5D treated with hemodialysis). METHODS We conducted a prospective case-control study that included 63 CKD5-HD patients (HD for 1-5 years) followed up for 48 months and a control group consisting of 52 non-dialysis patients diagnosed with CKD stages G1-G5 (ND-CKD). All patients had a CVD baseline assessment including medical history, echocardiography, and electrocardiography (ECG). Circulating plasma KIM-1 levels were determined with single-molecule counting immunoassay technology using an enzyme-linked immunosorbent assay. We obtained the following parameters: serum creatinine and urea; the inflammation markers CRP (C-reactive protein) and IL-6 (interleukin-6); and the anemia markers complete blood count, serum ferritin, and transferrin saturation (TSAT). RESULTS The mean plasma KIM-1 level was 403.8 ± 546.8 pg/mL, showing a statistically significant correlation with inflammation (CRP, R = 0.28, p = 0.02; IL-6, R = 0.36, p = 0.005) and with anemia (hematocrit, R = -0.5, p = -0.0316; hemoglobin (Hb), R = -0.5, p = 0.02). We found that patients with left ventricular hypertrophy (LVH) on echocardiography (59.7%) had significantly lower mean levels of plasma KIM-1 than patients from the control group (155.51 vs. 432.12 pg/mL; p = 0.026). Regarding the patients' follow-up, we assessed all-cause mortality as an endpoint. After 24 months of follow-up, we found a mortality rate of 22.23%, while after 48 months, the mortality rate was 50.73%. A plasma KIM-1 level < 82.98 pg/mL was significantly associated with decreased survival in hemodialysis patients (p < 0.001). CONCLUSIONS In patients treated with hemodialysis, low levels of plasma KIM-1 were associated with cardiovascular changes and an increased risk of mortality. Plasma KIM-1 levels were significantly higher in HD patients compared to ND-CKD patients.
Collapse
Affiliation(s)
- Alexandru Florin Sircuța
- Department of Internal Medicine II—Nephrology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.F.S.); (A.S.); (L.P.); (M.B.); (F.B.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- County Emergency Hospital, L. Rebreanu Street, Nr. 156, 300723 Timișoara, Romania;
| | - Iulia Dana Grosu
- Department of Internal Medicine II—Nephrology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.F.S.); (A.S.); (L.P.); (M.B.); (F.B.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- County Emergency Hospital, L. Rebreanu Street, Nr. 156, 300723 Timișoara, Romania;
| | - Adalbert Schiller
- Department of Internal Medicine II—Nephrology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.F.S.); (A.S.); (L.P.); (M.B.); (F.B.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Ligia Petrica
- Department of Internal Medicine II—Nephrology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.F.S.); (A.S.); (L.P.); (M.B.); (F.B.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- County Emergency Hospital, L. Rebreanu Street, Nr. 156, 300723 Timișoara, Romania;
| | - Viviana Ivan
- County Emergency Hospital, L. Rebreanu Street, Nr. 156, 300723 Timișoara, Romania;
- Department of Internal Medicine II—Cardiology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Oana Schiller
- B Braun Avitum Dialysis Centre, 300417 Timișoara, Romania;
| | - Madalina Bodea
- Department of Internal Medicine II—Nephrology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.F.S.); (A.S.); (L.P.); (M.B.); (F.B.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- County Emergency Hospital, L. Rebreanu Street, Nr. 156, 300723 Timișoara, Romania;
| | | | - Ionuţ Goleț
- Department of Management, Faculty of Economics and Business Administration, University of the West, 300115 Timișoara, Romania;
| | - Flaviu Bob
- Department of Internal Medicine II—Nephrology University Clinic, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania; (A.F.S.); (A.S.); (L.P.); (M.B.); (F.B.)
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- County Emergency Hospital, L. Rebreanu Street, Nr. 156, 300723 Timișoara, Romania;
| |
Collapse
|
7
|
Figuer A, Santos FM, Ciordia S, Valera G, Martín-Jouve B, Hernández-Fonseca JP, Bodega G, Ceprián N, Ramírez R, Carracedo J, Alique M. Proteomic analysis of endothelial cells and extracellular vesicles in response to indoxyl sulfate: Mechanisms of endothelial dysfunction in chronic kidney disease. Life Sci 2024; 351:122810. [PMID: 38871114 DOI: 10.1016/j.lfs.2024.122810] [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: 04/03/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
AIMS Cardiovascular pathology is the main cause of death in chronic kidney disease (CKD) patients. CKD is associated with the accumulation of uremic toxins in the bloodstream, and indoxyl sulfate (IS) is one of the most abundant uremic toxins found in the blood of CKD patients. We conducted an in vitro study to assess the mechanisms underlying the IS-induced endothelial dysfunction that could lead to cardiovascular diseases. We also studied their extracellular vesicles (EVs) owing to their capacity to act as messengers that transmit signals through their cargo. MAIN METHODS EVs were characterized by nanoparticle tracking analysis, transmission electron microscopy, flow cytometry, and tetraspanin expression. Cell lysates and isolated EVs were analyzed using liquid chromatography coupled with mass spectrometry, followed by Gene Set Enrichment Analysis to identify the altered pathways. KEY FINDINGS Proteomic analysis of endothelial cells revealed that IS causes an increase in proteins related to adipogenesis, inflammation, and xenobiotic metabolism and a decrease in proliferation. Extracellular matrix elements, as well as proteins associated with myogenesis, response to UV irradiation, and inflammation, were found to be downregulated in IS-treated EVs. Fatty acid metabolism was also found to be increased along with adipogenesis and inflammation observed in cells. SIGNIFICANCE The treatment of endothelial cells with IS increased the expression of proteins related to adipogenesis, inflammation, and xenobiotic metabolism and was less associated with proliferation. Furthermore, EVs from cells treated with IS may mediate endothelial dysfunction, since they present fewer extracellular matrix elements, myogenesis, inflammatory factors, and proteins downregulated in response to UV radiation.
Collapse
Affiliation(s)
- Andrea Figuer
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcala de Henares, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Fátima M Santos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain; Functional Proteomics Laboratory, Centro Nacional de Biotecnología, CSIC, Calle Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Sergio Ciordia
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología, CSIC, Calle Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Gemma Valera
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
| | - Beatriz Martín-Jouve
- Electron Microscopy Unit, Centro Nacional de Biotecnología, CSIC, Calle Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Juan Pablo Hernández-Fonseca
- Electron Microscopy Unit, Centro Nacional de Biotecnología, CSIC, Calle Darwin 3, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Universidad de Alcalá, 28871 Alcala de Henares, Madrid, Spain
| | - Noemí Ceprián
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense, 28040 Madrid, Spain
| | - Rafael Ramírez
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcala de Henares, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense, 28040 Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain.
| | - Matilde Alique
- Departamento de Biología de Sistemas, Universidad de Alcalá, 28871 Alcala de Henares, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain.
| |
Collapse
|
8
|
Li L, Liu H, Zhang Q, Jin H, Tao H, Chen H, Zhou Z. Pentraxin-3 and Outcomes in CKD: A Systematic Review and Meta-analysis. Kidney Med 2024; 6:100800. [PMID: 38645733 PMCID: PMC11026967 DOI: 10.1016/j.xkme.2024.100800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2024] Open
Abstract
Rationale & Objective Long pentraxin-3 (PTX-3) serves as a biomarker for prognosticating adverse clinical outcomes in individuals with chronic kidney disease (CKD). The objective of the current meta-analysis was to evaluate the prognostic efficacy of PTX-3 in patients with CKD. In addition, we compared the prognostic effectiveness of PTX-3 and the short pentraxin C-reactive protein (CRP) in the identical cohort of patients with CKD. Study Design A systematic review and meta-analysis. Setting & Participants Patients with CKD treated with or without dialysis. Selection Criteria for Studies A cohort study with a minimum 1-year follow-up. Data Extraction Risk measurements, adjusted hazard risk with 95% CI, and modified variables. Analytical Approach To aggregate the adjusted effect estimates, a fixed-effects or random-effects model was employed. Results Nine studies covering 1,825 patients with CKD were selected in the present review. Six of the 9 studies exclusively included patients receiving hemodialysis. The collected findings indicated that patients with CKD in the highest tertile of PTX-3 demonstrated significantly higher risks of all-cause mortality (HR, 1.92; 95% CI, 1.44-2.56), cardiovascular death (HR, 1.98; 95% CI, 1.28-3.05), infectious death (HR, 5.26; 95% CI, 1.60-17.31), and fatal and nonfatal cardiovascular events (HR, 1.81; 95% CI, 1.35-2.42), as compared with those in the lowest tertile. These significant associations with risk were also observed when effect estimates were presented as per unit change in the PTX-3. Moreover, when comparing the prognostic value of PTX-3 and CRP in the same individuals (5 studies covering 904 patients), PTX-3 proved to be a satisfactory predictor of adverse events in these patients, whereas CRP failed to exhibit such predictive capability, regardless of the type of effect estimate used. Limitations A relatively small sample size and some heterogeneity. Conclusions Pentraxin 3 is associated with adverse events in individuals with CKD and may be a more reliable predictor of adverse clinical events than CRP in this population.
Collapse
Affiliation(s)
- Li Li
- Department of Clinical Laboratory, Binhai County People’s Hospital, Binhai, Jiangsu, China
| | - Hongli Liu
- Department of Clinical Laboratory, Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Jiangsu, China
| | - Qinglin Zhang
- Department of Blood Transfusion, Affiliated Hospital 6 of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, Jiangsu, China
| | - Hao Jin
- Department of Blood Transfusion, Affiliated Hospital 6 of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, Jiangsu, China
| | - Hui Tao
- Department of Blood Transfusion, Affiliated Hospital 6 of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, Jiangsu, China
| | - Hongmei Chen
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, Jiangsu, China
| | - Zhongwei Zhou
- Department of Clinical Laboratory, Affiliated Hospital 6 of Nantong University, The Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng Third People's Hospital, Jiangsu, China
| |
Collapse
|
9
|
Hayward SJ, Chesnaye NC, Hole B, Aylward R, Meuleman Y, Torino C, Porto G, Szymczak M, Drechsler C, Dekker FW, Evans M, Jager KJ, Wanner C, Caskey FJ. Protein Biomarkers and Major Cardiovascular Events in Older People With Advanced CKD: The European Quality (EQUAL) Study. Kidney Med 2024; 6:100745. [PMID: 38162538 PMCID: PMC10757029 DOI: 10.1016/j.xkme.2023.100745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Rationale & Objective Cardiovascular disease is the leading cause of morbidity and mortality in chronic kidney disease (CKD). We investigated 184 inflammatory and cardiovascular proteins to determine their potential as biomarkers for major cardiovascular events (MACEs). Study Design The European Quality (EQUAL) is an observational cohort study that enrolled people aged ≥65 years with an estimated glomerular filtration rate ≤20 mL/min/1.73 m2. Setting & Participants Recruited participants were split into the discovery (n = 611) and replication cohorts (n = 292). Exposure Levels of 184 blood proteins were measured at the baseline visit, and each protein was analyzed individually. Outcome MACE. Analytical Approach Cox proportional hazard models adjusted for age, sex, estimated glomerular filtration rate, previous MACE, and country were used to determine the risk of MACE. Proteins with false discovery rate adjusted P values of <0.05 in the discovery cohort were tested in the replication cohort. Sensitivity analyses were performed by adjusting for traditional risk factors, CKD-specific risk factors, and level of proteinuria and segregating atherosclerotic and nonatherosclerotic MACE. Results During a median follow-up of 2.9 years, 349 people (39%) experienced a MACE. Forty-eight proteins were associated with MACE in the discovery cohort; 9 of these were reproduced in the replication cohort. Three of these proteins maintained a strong association with MACE after adjustment for traditional and CKD-specific risk factors and proteinuria. Tenascin (TNC), fibroblast growth factor-23 (FGF-23), and V-set and immunoglobulin domain-containing protein 2 (VSIG2) were associated with both atherosclerotic and nonatherosclerotic MACE. All replicated proteins except carbonic anhydrase 1 and carbonic anhydrase 3 were associated with nonatherosclerotic MACE. Limitations Single protein concentration measurements and limited follow-up time. Conclusions Our findings corroborate previously reported relationships between FGF-23, vascular cell adhesion protein-1, TNC, and placental growth factor with cardiovascular outcomes in CKD. We identify 5 proteins not previously linked with MACE in CKD that may be targets for future therapies. Plain-Language Summary Kidney disease increases the risk of heart disease, stroke, and other vascular conditions. Blood tests that predict the likelihood of these problems may help to guide treatment, but studies are needed in people with kidney disease. We analyzed blood tests from older people with kidney disease, looking for proteins associated with higher risk of these conditions. Nine proteins were identified, of which 3 showed a strong effect after all other information was considered. This work supports previous research regarding 4 of these proteins and identifies 5 additional proteins that may be associated with higher risk. Further work is needed to confirm our findings and to determine whether these proteins can be used to guide treatment.
Collapse
Affiliation(s)
- Samantha J.L. Hayward
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
| | - Nicholas C. Chesnaye
- Amsterdam UMC, University of Amsterdam, ERA Registry, Medical Informatics, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
| | - Barnaby Hole
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ryan Aylward
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Yvette Meuleman
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia Torino
- Institute of Clinical Physiology, National Research Council, Reggio Calabria, Italy
| | - Gaetana Porto
- GOM Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Maciej Szymczak
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
| | | | - Friedo W. Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Evans
- Department of Clinical Sciences Intervention and Technology, Karolinska Institutet and Karolinska, Stockholm, Sweden
| | - Kitty J. Jager
- Amsterdam UMC, University of Amsterdam, ERA Registry, Medical Informatics, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
| | - Christoph Wanner
- Division of Nephrology, University Hospital of Wurzburg, Wurzburg, Germany
| | - Fergus J. Caskey
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - EQUAL investigators
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
- Amsterdam UMC, University of Amsterdam, ERA Registry, Medical Informatics, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Clinical Physiology, National Research Council, Reggio Calabria, Italy
- GOM Bianchi Melacrino Morelli, Reggio Calabria, Italy
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
- Division of Nephrology, University Hospital of Wurzburg, Wurzburg, Germany
- Department of Clinical Sciences Intervention and Technology, Karolinska Institutet and Karolinska, Stockholm, Sweden
| |
Collapse
|
10
|
Lanktree MB, Collister D, Pigyere M, Mazzetti A, Paré G, Walsh M. Proteome-Wide Changes in Blood Biomarkers During Hemodialysis. Kidney Int Rep 2024; 9:177-181. [PMID: 38312796 PMCID: PMC10831367 DOI: 10.1016/j.ekir.2023.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 02/06/2024] Open
Affiliation(s)
- Matthew B. Lanktree
- Population Health Research Institute, Hamilton, Ontario, Canada
- Division of Nephrology, Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - David Collister
- Division of Nephrology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Marie Pigyere
- Population Health Research Institute, Hamilton, Ontario, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Andrea Mazzetti
- Population Health Research Institute, Hamilton, Ontario, Canada
- Division of Nephrology, Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Population Health Research Institute, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michael Walsh
- Population Health Research Institute, Hamilton, Ontario, Canada
- Division of Nephrology, Department of Medicine, McMaster University and St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Ren Y, Ruan P, Segal M, Dobre M, Schelling JR, Banerjee U, Shafi T, Ganz P, Dubin RF. Evaluation of a large-scale aptamer proteomics platform among patients with kidney failure on dialysis. PLoS One 2023; 18:e0293945. [PMID: 38079395 PMCID: PMC10712847 DOI: 10.1371/journal.pone.0293945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Patients with kidney failure suffer high mortality, and we currently lack markers for risk stratification for these patients. We carried out a quality control study of a modified aptamer assay (SomaScan v.4.0) that measures ~ 5000 proteins, in preparation for a larger study using this platform in cohorts with kidney failure. METHODS Forty participants from the Cardiac, Endothelial Function and Arterial Stiffness in End-Stage Renal Disease (CERES study) were selected to analyze technical and short-term biological variability, orthogonal correlations and differential protein expression in plasma from patients who died during 2.5 year follow-up. Long-term (one year) variability was studied in 421 participants in the Chronic Renal Insufficiency Cohort. We evaluated 4849 aptamers (4607 unique proteins) using data formats including raw data and data formatted using Adaptive Normalization by Maximum Likelihood (ANML), an algorithm developed for SomaScan data in individuals with normal kidney function. RESULTS In ANML format, median[IQR] intra-assay coefficient of variation (CV) was 2.38%[1.76, 3.40] and inter-assay CV was 7.38%[4.61, 13.12]. Short-term within-subject CV was 5.76% [3.35, 9.72]; long-term CV was 8.71%[5.91, 13.37]. Spearman correlations between aptamer and traditional assays for PTH, NT-proBNP, FGF-23 and CRP were all > 0.7. Fold-change (FC) in protein levels among non-survivors, significant after Bonferroni correction, included SVEP1 (FC[95% CI] 2.14 [1.62, 2.82]), keratocan (1.74 [1.40, 2.15]) and LanC-like protein 1 (0.56 [0.45, 0.70]). Compared to raw aptamer data, technical and short-term biological variability in paired samples was lower in ANML-formatted data. ANML formatting had minimal impact on orthogonal correlations with traditional assays or the associations of proteins with the phenotype of mortality. CONCLUSIONS SomaScan had excellent technical variability and low within-subject short-term variability. ANML formatting could facilitate comparison of biomarker results with other studies that utilize this format. We expect SomaScan to provide novel and reproducible information in patients with kidney failure on dialysis.
Collapse
Affiliation(s)
- Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Peifeng Ruan
- Peter O’Donnell Jr School of Public Health, UT Southwestern, Dallas, Texas, United States of America
| | - Mark Segal
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, United States of America
| | - Mirela Dobre
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Jeffrey R. Schelling
- Department of Physiology & Biophysics, Case Western Reserve University of School of Medicine, Cleveland, Ohio, United States of America
| | - Upasana Banerjee
- Department of Internal Medicine, Hurley Medical Center/Michigan State University, Flint, Michigan, United States of America
| | - Tariq Shafi
- Division of Kidney Diseases, Hypertension and Transplantation, Houston Methodist Hospital, Houston, Texas, United States of America
| | - Peter Ganz
- Division of Cardiology, University of California, San Francisco, San Francisco, California, United States of America
| | - Ruth F. Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | | |
Collapse
|
12
|
Wändell P, Carlsson AC, Larsson A, Ärnlöv J, Feldreich T, Ruge T. The C-reactive protein Albumin ratio was not consistently associated with cardiovascular and all-cause mortality in two community-based cohorts of 70-year-olds. Scand J Clin Lab Invest 2023; 83:439-443. [PMID: 37702518 DOI: 10.1080/00365513.2023.2255971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/03/2023] [Indexed: 09/14/2023]
Abstract
C-reactive protein (CRP)/Albumin ratio (CAR) seems to mirror disease severity and prognosis in several acute disorders particularly in elderly patients, which we aimed to study. As method we use a prospective study design; the Prospective Investigation of the Vasculature in Uppsala Seniors (PIVUS; n = 912, women 50%; mean age 70 years, baseline 2001 and 2004, median follow-up 15.0 years, end of follow-up 2019) and the Uppsala Longitudinal Study of Adult Men (ULSAM, n = 924 mean age 71 years, baseline 1991-1995, median follow-up 15.6 years, end of follow-up 2016). Serum samples were used for analyses of CRP and Albumin. Cox regression analyses were performed for cardiovascular and all-cause mortality in models adjusting for several factors (age; physical activity; Interleukin-6; cardiovascular (CVD) risk factors: smoking, BMI level, systolic blood pressure, LDL-cholesterol, and diabetes), with 95% confidence interval (CI). When adjusting for age and CVD risk factors, CAR was significantly associated with cardiovascular mortality for meta-analyzed results from PIVUS and ULSAM, HR 1.09 (95% 1.01-1.18), but neither in PIVUS (HR 1.14, 95% CI 0.99-1.31) nor in ULSAM (1.07, 95% CI 0.98-1.17). Additionally, CAR was significantly associated with all-cause mortality in ULSAM 1.31 (95% CI 1.12-1.54) but not in PIVUS HRs 1.01 (95% 0.089-1.15). The predictive value of CAR was similar to CRP alone in PIVUS and ULSAM and slightly better than albumin for the prediction of CVD-mortality in ULSAM. In conclusion, CAR was not consistently associated with cardiovascular and all-cause mortality in the two cohorts. The prognostic value of CAR for long-term CVD-mortality was similar to CRP.
Collapse
Affiliation(s)
- Per Wändell
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Axel Carl Carlsson
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Academic Primary Health Care Centre, Stockholm, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- School of Health and Welfare, Dalarna University, Falun, Sweden
| | | | - Toralph Ruge
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
- Department of Emergency and Internal Medicine, Skånes University Hospital, Malmö, Sweden
- Department of Clinical Sciences Malmö, Lund University & Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
13
|
Srialluri N, Surapaneni A, Schlosser P, Chen TK, Schmidt IM, Rhee EP, Coresh J, Grams ME. Circulating Proteins and Mortality in CKD: A Proteomics Study of the AASK and ARIC Cohorts. Kidney Med 2023; 5:100714. [PMID: 37711886 PMCID: PMC10498294 DOI: 10.1016/j.xkme.2023.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023] Open
Abstract
Rationale & Objective Proteomics could provide pathophysiologic insight into the increased risk of mortality in patients with chronic kidney disease (CKD). This study aimed to investigate associations between the circulating proteome and all-cause mortality among patients with CKD. Study Design Observational cohort study. Setting & Participants Primary analysis in 703 participants in the African American Study of Kidney Disease and Hypertension (AASK) and validation in 1,628 participants with CKD in the Atherosclerosis Risk in Communities (ARIC) study who attended visit 5. Exposure Circulating proteins. Outcome All-cause mortality. Analytical Approach Among AASK participants, we evaluated the associations of 6,790 circulating proteins with all-cause mortality using multivariable Cox proportional hazards models. Proteins with significant associations were further studied in ARIC Visit 5 participants with CKD. Results In the AASK cohort, the mean age was 54.5 years, 271 (38.5%) were women, and the mean measured glomerular filtration rate (GFR) was 46 mL/min/1.73 m2. The median follow-up was 9.6 years, and 7 distinct proteins were associated with all-cause mortality at the Bonferroni-level threshold (P < 0.05 of the 6,790) after adjustment for demographics and clinical factors, including baseline measured estimated GFR and proteinuria. In the ARIC visit 5 cohort, the mean age was 77.2 years, 903 (55.5%) were women, the mean estimated GFR was 54 mL/min/1.73 m2 and median follow-up was 6.9 years. Of the 7 proteins found in AASK, 3 (β2-microglobulin, spondin-1, and N-terminal pro-brain natriuretic peptide) were available in the ARIC data, with all 3 significantly associated with death in ARIC. Limitations Possibility of unmeasured confounding. Cause of death was not known. Conclusions Using large-scale proteomic analysis, proteins were reproducibly associated with mortality in 2 cohorts of participants with CKD. Plain-Language Summary Patients with chronic kidney disease (CKD) have a high risk of premature death, with various pathophysiological processes contributing to this increased risk of mortality. This observational cohort study aimed to investigate the associations between circulating proteins and all-cause mortality in patients with CKD using large-scale proteomic analysis. The study analyzed data from the African American Study of Kidney Disease and Hypertension (AASK) study and validated the findings in the Atherosclerosis Risk in Communities (ARIC) Study. A total of 6,790 circulating proteins were evaluated in AASK, and 7 proteins were significantly associated with all-cause mortality. Three of these proteins (β2-microglobulin, spondin-1, and N-terminal pro-brain natriuretic peptide (BNP)) were also measured in ARIC and were significantly associated with death. Additional studies assessing biomarkers associated with mortality among patients with CKD are needed to evaluate their use in clinical practice.
Collapse
Affiliation(s)
- Nityasree Srialluri
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland
| | - Aditya Surapaneni
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York
| | - Pascal Schlosser
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Teresa K. Chen
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Kidney Health Research Collaborative; Division of Nephrology, Department of Medicine, University of California San Francisco and San Francisco VA Health Care System, San Francisco, California
| | - Insa M. Schmidt
- Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Eugene P. Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts
| | - Josef Coresh
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Morgan E. Grams
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Division of Precision Medicine, Department of Medicine, New York University, New York, New York
| |
Collapse
|
14
|
Kamyshnikova LA, Gorbachevskaya KS, Efremova OA, Obolonkova NI, Bolkhovitina OA. Biomarkers of Adverse Cardiovascular Events in Kidney Disease. THE RUSSIAN ARCHIVES OF INTERNAL MEDICINE 2023; 13:253-262. [DOI: 10.20514/2226-6704-2023-13-4-253-262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Based on domestic and international literature the review refers to the analysis of the research data on risk factors and biomarkers for the development of adverse cardiovascular events in patients with chronic kidney disease and acute kidney injury. Biomarker studies are important, especially in the early stages of chronic kidney disease, that is, in patients with creatinine clearance above 60 ml/min/1.73 m2, when preventive and therapeutic measures work more effectively. Among the potential predictors of adverse cardiovascular events, the biomarkers related to the following pathological processes (conditions) should be noted: oxidative stress (malondialdehyde, ischemic-modified albumin; superoxide dismutase), inflammation (interleukin-6, interleukin-18), acute kidney injury (kidney injury molecule 1; neutrophil gelatinase-associated lipocalin), cardiospecific biomarkers (highly sensitive troponin) and circulating microribonucleic acids (specific miRNA-133a, miRNA-21), as well as the prospects for further study of some biomarkers in cardionephrology are discussed. A separate emphasis is placed on the need to establish threshold values for various molecules in chronic kidney disease, depending on the degree of decline in kidney function, which will allow these indicators to be effectively used in clinical practice as diagnostic and prognostic biomarkers for cardiovascular diseases, since their usual reference values are used in the general population, will be higher in kidney disease. Currently, only for troponin and natriuretic peptides, certain reference values are established, which are less clear-cut in the population with chronic kidney disease than in the general population, and for all other biomarkers, cut-off values are not yet known.
Collapse
|
15
|
Kuang N, Shu B, Yang F, Li S, Zhang M. TRAIL or TRAIL-R2 as a Predictive Biomarker for Mortality or Cardiovascular Events: A Systematic Review and Meta-analysis. J Cardiovasc Pharmacol 2023; 81:348-354. [PMID: 36888983 DOI: 10.1097/fjc.0000000000001415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/10/2023] [Indexed: 03/10/2023]
Abstract
ABSTRACT Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and TRAIL-receptor-2 (TRAIL-R2) are associated with atherosclerosis. This meta-analysis aimed to investigate the potential association between TRAIL/TRAIL-R2 with mortality or cardiovascular (CV) events. PubMed, Embase, and Cochrane Library were searched for reports published up to May 2021. Reports were included when the association between TRAIL or TRAIL-R2 and mortality or CV events was reported. Considering the heterogeneity between studies, we used the random-effects model for all analyses. Ultimately, the meta-analysis included 18 studies (16,295 patients). The average follow-up ranged from 0.25 to 10 years. Decreased TRAIL levels were negatively associated with all-cause mortality [rank variable, hazard ratio (HR), 95% CI, 2.93, 1.94-4.42; I2 = 0.0%, Pheterogeneity = 0.835]. Increased TRAIL-R2 levels were positively associated with all-cause mortality (continuous variable, HR, 95% CI, 1.43, 1.23-1.65; I2 = 0.0%, Pheterogeneity = 0.548; rank variable, HR, 95% CI, 7.08, 2.70-18.56; I2 = 46.5%, Pheterogeneity = 0.154), CV mortality (continuous variable, HR, 95% CI, 1.33, 1.14-1.57; I2 = 0.0%, Pheterogeneity = 0.435), myocardial infarction (continuous variable, HR, 95% CI, 1.23, 1.02-1.49; rank variable, HR, 95% CI, 1.49, 1.26-1.76; I2 = 0.7%, Pheterogeneity = 0.402), and new-onset heart failure (rank variable, HR, 95% CI, 3.23, 1.32-7.87; I2 = 83.0%, Pheterogeneity = 0.003). In conclusion, decreased TRAIL was negatively associated with all-cause mortality, and increased TRAIL-R2 was positively associated with all-cause mortality, CV mortality, myocardial infarction, and heart failure.
Collapse
Affiliation(s)
- Na Kuang
- Department of Cardiology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | | | | | | | | |
Collapse
|
16
|
Kelland E, Patil MS, Patel S, Cartland SP, Kavurma MM. The Prognostic, Diagnostic, and Therapeutic Potential of TRAIL Signalling in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24076725. [PMID: 37047698 PMCID: PMC10095395 DOI: 10.3390/ijms24076725] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL) was originally discovered, almost 20 years ago, for its ability to kill cancer cells. More recent evidence has described pleiotropic functions, particularly in the cardiovascular system. There is potential for TRAIL concentrations in the circulation to act as prognostic and/or diagnostic factors for cardiovascular diseases (CVD). Pre-clinical studies also describe the therapeutic capacity for TRAIL signals, particularly in the context of atherosclerotic disease and diseases of the myocardium. Because diabetes mellitus significantly contributes to the progression and pathogenesis of CVDs, in this review we highlight recent evidence for the prognostic, diagnostic, and therapeutic potential of TRAIL signals in CVDs, and where relevant, the impact of diabetes mellitus. A greater understanding of how TRAIL signals regulate cardiovascular protection and pathology may offer new diagnostic and therapeutic avenues for patients suffering from CVDs.
Collapse
Affiliation(s)
- Elaina Kelland
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| | - Manisha S. Patil
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
- Royal Prince Alfred Hospital, Sydney 2006, Australia
| | - Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| | - Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney 2042, Australia
| |
Collapse
|
17
|
Chen Z, Wang Y. Interleukin-6 levels can be used to estimate cardiovascular and all-cause mortality risk in dialysis patients: A meta-analysis and a systematic review. Immun Inflamm Dis 2023; 11:e818. [PMID: 37102647 PMCID: PMC10132186 DOI: 10.1002/iid3.818] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Although previous studies have explored the correlation of interleukin (IL)-6 with mortality risk in dialysis patients, the findings have been conflicting. Hence, this meta-analysis aimed to comprehensively assess the use of IL-6 measurement for estimating cardiovascular mortality and all-cause mortality in dialysis patients. METHODS The Embase, PubMed, Web of Science, and MEDLINE databases were searched to identify relevant studies. After screening out the eligible studies, the data were extracted. RESULTS Twenty-eight eligible studies with 8370 dialysis patients were included. Pooled analyses revealed that higher IL-6 levels were related to increased cardiovascular mortality risk (hazard ratio [HR] = 1.55, 95% confidence interval [CI]: 1.20-1.90) and all-cause mortality risk (HR = 1.11, 95% CI: 1.05-1.17) in dialysis patients. Further subgroup analyses suggested that higher IL-6 levels were associated with elevated cardiovascular mortality in hemodialysis patients (HR = 1.59, 95% CI: 1.36-1.81) but not in peritoneal dialysis patients (HR = 1.56, 95% CI: 0.46-2.67). Moreover, sensitivity analyses indicated that the results were robust. Egger's test revealed potential publication bias among studies exploring the correlation of IL-6 levels with cardiovascular mortality (p = .004) and all-cause mortality (p < .001); however, publication bias was not observed when using Begg's test (both p > .05). CONCLUSIONS This meta-analysis reveals that higher IL-6 levels could indicate higher risks of cardiovascular mortality and all-cause mortality in dialysis patients. These findings suggest that monitoring IL-6 cytokine may help to enhance dialysis management and improve the general prognosis of patients.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of CardiologyThe First People's Hospital of ZiyangZiyangChina
| | - Yan Wang
- Department of NephrologyThe First People's Hospital of ZiyangZiyangChina
| |
Collapse
|
18
|
Huang J, Caliskan Guzelce E, Gholami SK, Gawelek KL, Mitchell RN, Pojoga LH, Romero JR, Williams GH, Adler GK. Effects of Mineralocorticoid Receptor Blockade and Statins on Kidney Injury Marker 1 (KIM-1) in Female Rats Receiving L-NAME and Angiotensin II. Int J Mol Sci 2023; 24:6500. [PMID: 37047470 PMCID: PMC10095483 DOI: 10.3390/ijms24076500] [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: 02/08/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Kidney injury molecule-1 (KIM-1) is a biomarker of renal injury and a predictor of cardiovascular disease. Aldosterone, via activation of the mineralocorticoid receptor, is linked to cardiac and renal injury. However, the impact of mineralocorticoid receptor activation and blockade on KIM-1 is uncertain. We investigated whether renal KIM-1 is increased in a cardiorenal injury model induced by L-NAME/ANG II, and whether mineralocorticoid receptor blockade prevents the increase in KIM-1. Since statin use is associated with lower aldosterone, we also investigated whether administering eiSther a lipophilic statin (simvastatin) or a hydrophilic statin (pravastatin) prevents the increase in renal KIM-1. Female Wistar rats (8-10 week old), consuming a high salt diet (1.6% Na+), were randomized to the following conditions for 14 days: control; L-NAME (0.2 mg/mL in drinking water)/ANG II (225 ug/kg/day on days 12-14); L-NAME/ANG II + eplerenone (100 mg/kg/day p.o.); L-NAME/ANG II + pravastatin (20 mg/kg/day p.o.); L-NAME/ANG II + simvastatin (20 mg/kg/day p.o.). Groups treated with L-NAME/ANG II had significantly higher blood pressure, plasma and urine aldosterone, cardiac injury/stroke composite score, and renal KIM-1 than the control group. Both eplerenone and simvastatin reduced 24-h urinary KIM-1 (p = 0.0046, p = 0.031, respectively) and renal KIM-1 immunostaining (p = 0.004, p = 0.037, respectively). Eplerenone also reduced renal KIM-1 mRNA expression (p = 0.012) and cardiac injury/stroke composite score (p = 0.04). Pravastatin did not affect these damage markers. The 24-h urinary KIM-1, renal KIM-1 immunostaining, and renal KIM-1 mRNA expression correlated with cardiac injury/stroke composite score (p < 0.0001, Spearman ranked correlation = 0.69, 0.66, 0.59, respectively). In conclusion, L-NAME/ANG II increases renal KIM-1 and both eplerenone and simvastatin blunt this increase in renal KIM-1.
Collapse
Affiliation(s)
- Jiayan Huang
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ezgi Caliskan Guzelce
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shadi K. Gholami
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kara L. Gawelek
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Richard N. Mitchell
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Luminita H. Pojoga
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jose R. Romero
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon H. Williams
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gail K. Adler
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Sun Z, Gao Z, Li X, Zheng X, Wang W, Qiao P. Perfusate Neutrophil Gelatinase-Associated Lipocalin, Kidney Injury Molecular-1, Liver-Type Fatty Acid Binding Protein, and Interleukin-18 as Potential Biomarkers to Predict Delayed Graft Function and Long-Term Prognosis in Kidney Transplant Recipients: A Single-Center Retrospective Study. Med Sci Monit 2023; 29:e938758. [PMID: 36869580 PMCID: PMC9993774 DOI: 10.12659/msm.938758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Delayed graft function (DGF) caused by ischemia-reperfusion injury is a common pathophysiological process that should be monitored by specific biomarkers in addition to serum creatinine. Thus, this single-center retrospective study aimed to investigate the association between levels of neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecular-1 (KIM-1), liver-type fatty acid binding protein (L-FABP), and interleukin-18 (IL-18) in DGF associated with acute kidney injury in kidney transplant recipients (KTRs) and estimated glomerular filtration rate (eGFR) at 3 years post-transplant. MATERIAL AND METHODS A total of 102 KTRs [14(13.7%) of DGF and 88(86.3%) of NON-DGF] were enrolled. DGF was defined as "dialysis is needed within 1 week after kidney transplantation". NGAL, KIM-1, L-FABP, and IL-18 were obtained from perfusate samples of donation-after-cardiac-death (DCD) kidneys, and measured by ELISA. RESULTS Compared to the NON-DGF group, KTRs in the DGF group had a statistically significant increase in levels of NGAL (P<0.001) and KIM-1 (P<0.001). Multiple logistic regression analyses showed that NGAL (OR=1.204, 95% CI 1.057-1.372, P=0.005) and KIM-1 (OR=1.248, CI=1.065-1.463, P=0.006) could be regarded as independent risk factors. The accuracy of NGAL and KIM-1 was 83.3% and 82.1%, respectively, calculated using the area under the receiver operating characteristic curve. Furthermore, the eGFR at 3 years post-transplant had a moderate negative correlation with NGAL (r=-0.208, P=0.036) and KIM-1 (r=-0.260, P=0.008). CONCLUSIONS Our results support those from previous studies showing that perfusate levels of NGAL and KIM-1 are associated with DGF in KTRs and also with reduced eGFR at 3 years post-transplant.
Collapse
Affiliation(s)
- Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Zihao Gao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Xin Li
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Xiang Zheng
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| | - Peng Qiao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China (mainland)
| |
Collapse
|
20
|
Png G, Gerlini R, Hatzikotoulas K, Barysenka A, Rayner NW, Klarić L, Rathkolb B, Aguilar-Pimentel JA, Rozman J, Fuchs H, Gailus-Durner V, Tsafantakis E, Karaleftheri M, Dedoussis G, Pietrzik C, Wilson JF, Angelis MH, Becker-Pauly C, Gilly A, Zeggini E. Identifying causal serum protein-cardiometabolic trait relationships using whole genome sequencing. Hum Mol Genet 2022; 32:1266-1275. [PMID: 36349687 PMCID: PMC10077504 DOI: 10.1093/hmg/ddac275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2022] Open
Abstract
Cardiometabolic diseases, such as type 2 diabetes and cardiovascular disease, have a high public health burden. Understanding the genetically-determined regulation of proteins that are dysregulated in disease can help to dissect the complex biology underpinning them. Here, we perform a protein quantitative trait locus (pQTL) analysis of 255 serum proteins relevant to cardiometabolic processes in 2893 individuals. Meta-analysing whole-genome sequencing (WGS) data from two Greek cohorts, MANOLIS (n = 1356; 22.5x WGS) and Pomak (n = 1537; 18.4x WGS), we detect 302 independently-associated pQTL variants for 171 proteins, including 12 rare variants (minor allele frequency [MAF] < 1%). We additionally find 15 pQTL variants that are rare in non-Finnish European populations, but have drifted up in frequency in the discovery cohorts here. We identify proteins causally associated with cardiometabolic traits, including MEP1B for high-density lipoprotein levels; and describe a knock-out Mep1b mouse model. Our findings furnish insights into the genetic architecture of the serum proteome, identify new protein-disease relationships, and demonstrate the importance of isolated populations in pQTL analysis.
Collapse
Affiliation(s)
- Grace Png
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Technical University of Munich (TUM), School of Medicine, Munich, Germany
| | - Raffaele Gerlini
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Andrei Barysenka
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - N William Rayner
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Lucija Klarić
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Juan A Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Jan Rozman
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Institute of Molecular Genetics of the Czech Academy of Sciences, Czech Centre for Phenogenomics, Vestec, Czech Republic
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | | | | | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Claus Pietrzik
- Institute for Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - James F Wilson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin Hrabe Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany.,Chair of Experimental Genetics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Christoph Becker-Pauly
- Institute of Biochemistry, Unit for Degradomics of the Protease Web, University of Kiel, Kiel, Germany
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany
| |
Collapse
|
21
|
Chen L, Peters JE, Prins B, Persyn E, Traylor M, Surendran P, Karthikeyan S, Yonova-Doing E, Di Angelantonio E, Roberts DJ, Watkins NA, Ouwehand WH, Danesh J, Lewis CM, Bronson PG, Markus HS, Burgess S, Butterworth AS, Howson JMM. Systematic Mendelian randomization using the human plasma proteome to discover potential therapeutic targets for stroke. Nat Commun 2022; 13:6143. [PMID: 36253349 PMCID: PMC9576777 DOI: 10.1038/s41467-022-33675-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 09/28/2022] [Indexed: 02/02/2023] Open
Abstract
Stroke is the second leading cause of death with substantial unmet therapeutic needs. To identify potential stroke therapeutic targets, we estimate the causal effects of 308 plasma proteins on stroke outcomes in a two-sample Mendelian randomization framework and assess mediation effects by stroke risk factors. We find associations between genetically predicted plasma levels of six proteins and stroke (P ≤ 1.62 × 10-4). The genetic associations with stroke colocalize (Posterior Probability >0.7) with the genetic associations of four proteins (TFPI, TMPRSS5, CD6, CD40). Mendelian randomization supports atrial fibrillation, body mass index, smoking, blood pressure, white matter hyperintensities and type 2 diabetes as stroke risk factors (P ≤ 0.0071). Body mass index, white matter hyperintensity and atrial fibrillation appear to mediate the TFPI, IL6RA, TMPRSS5 associations with stroke. Furthermore, thirty-six proteins are associated with one or more of these risk factors using Mendelian randomization. Our results highlight causal pathways and potential therapeutic targets for stroke.
Collapse
Affiliation(s)
- Lingyan Chen
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - James E Peters
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| | - Bram Prins
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Elodie Persyn
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Matthew Traylor
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN, Cambridge, UK
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ekaterina Yonova-Doing
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Human Technopole, Milan, Italy
| | - David J Roberts
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant-Oxford Centre, Level 2, John Radcliffe Hospital, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Nicholas A Watkins
- NHS Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge, UK
| | - Willem H Ouwehand
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Wellcome Sanger Institute, Hinxton, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Cathryn M Lewis
- Department of Medical and Molecular Genetics, King's College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK
| | | | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK.
| |
Collapse
|
22
|
Stopic B, Medic-Brkic B, Savic-Vujovic K, Davidovic Z, Todorovic J, Dimkovic N. Biomarkers and Predictors of Adverse Cardiovascular Events in Different Stages of Chronic Kidney Disease. Dose Response 2022; 20:15593258221127568. [PMID: 36118679 PMCID: PMC9478703 DOI: 10.1177/15593258221127568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Chronic kidney disease (CKD) is an important factor that contributes to the
increase of all-cause morbidity and mortality in the group of non-communicable
diseases, and it is also recognized as a strong and independent risk factor that
contributes to cardiovascular disease (CVD). CVDs are a consequence of the
action of a large number of risk factors among which are traditional and
non-traditional. These risk factors have been the subject of a large number of
studies which partially explained the unfavorable cardiovascular (CV) outcome of
CKD patients. Therefore, valid studies about clinical and biohumoral predictors
are of particular importance, especially in the early stages of renal disease,
that is, in patients with creatinine clearance below
60 ml/min/1.73 m2 when preventive measures are most effective.
Among potential predictors of adverse CV outcome are biomarkers of inflammation
(Interleukin-18—IL-18), oxidative stress (ischemia-modified albumin—IMA;
superoxide dismutase—SOD), acute kidney injury (kidney injury
molecule-1—KIM-1; neutrophil gelatinase–associated
lipocalin—NGAL), and microribonucleic acids (specific microRNA-133a). In this
review, we tried to confirm the relationship between risk factors of CKD and CVD
and newer, less frequently examined biomarkers with the occurrence of incidental
CV events in renal patients.
Collapse
Affiliation(s)
- Bojan Stopic
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Branislava Medic-Brkic
- Faculty of Medicine, Department of Pharmacology, University of Belgrade, Belgrade, Serbia
| | - Katarina Savic-Vujovic
- Faculty of Medicine, Department of Pharmacology, University of Belgrade, Belgrade, Serbia
| | - Zeljko Davidovic
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia
| | - Jovana Todorovic
- Faculty of Medicine, Institute of Social Medicine, University of Belgrade, Belgrade, Serbia
| | - Nada Dimkovic
- Clinical Department for Nephrology, Zvezdara University Medical Center, Belgrade, Serbia
| |
Collapse
|
23
|
Wu PH, Glerup RI, Svensson MHS, Eriksson N, Christensen JH, de Laval P, Soveri I, Westerlund M, Linde T, Ljunggren Ö, Fellström B. Novel Biomarkers Detected by Proteomics Predict Death and Cardiovascular Events in Hemodialysis Patients. Biomedicines 2022; 10:biomedicines10040740. [PMID: 35453489 PMCID: PMC9026983 DOI: 10.3390/biomedicines10040740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/06/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
End-stage kidney disease increases mortality and the risk of cardiovascular (CV) disease. It is crucial to explore novel biomarkers to predict CV disease in the complex setting of patients receiving hemodialysis (HD). This study investigated the association between 92 targeted proteins with all-cause death, CV death, and composite vascular events (CVEs) in HD patients. From December 2010 to March 2011, 331 HD patients were included and followed prospectively for 5 years. Serum was analyzed for 92 CV-related proteins using Proseek Multiplex Cardiovascular I panel, a high-sensitivity assay based on proximity extension assay (PEA) technology. The association between biomarkers and all-cause death, CV death, and CVEs was evaluated using Cox-regression analyses. Of the PEA-based proteins, we identified 20 proteins associated with risk of all-cause death, 7 proteins associated with risk of CV death, and 17 proteins associated with risk of CVEs, independent of established risk factors. Interleukin-8 (IL-8), T-cell immunoglobulin and mucin domain 1 (TIM-1), and C-C motif chemokine 20 (CCL20) were associated with increased risk of all-cause death, CV death, and CVE in multivariable-adjusted models. Stem cell factor (SCF) and Galanin peptides (GAL) were associated with both decreased risk of all-cause death and CV death. In conclusion, IL-8, TIM-1, and CCL20 predicted death and CV outcomes in HD patients. Novel findings were that SCF and GAL were associated with a lower risk of all-cause death and CV death. The SCF warrants further study with regard to its possible biological effect in HD patients.
Collapse
Affiliation(s)
- Ping-Hsun Wu
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Rie Io Glerup
- Department of Nephrology, Aalborg University Hospital, 9000 Aalborg, Denmark; (R.I.G.); (J.H.C.)
| | - My Hanna Sofia Svensson
- Division of Medicine, Department of Nephrology, Akershus University Hospital, 1478 Oslo, Norway;
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, 75185 Uppsala, Sweden;
| | | | - Philip de Laval
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Inga Soveri
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Magnus Westerlund
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Torbjörn Linde
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Bengt Fellström
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Correspondence: ; Tel.: +46-18-6114348
| |
Collapse
|
24
|
Wessman T, Tofik R, Ruge T, Melander O. Associations between biomarkers of multimorbidity burden and mortality risk among patients with acute dyspnea. Intern Emerg Med 2022; 17:559-567. [PMID: 34417729 PMCID: PMC8964555 DOI: 10.1007/s11739-021-02825-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/29/2021] [Indexed: 11/10/2022]
Abstract
The patients' burden of comorbidities is a cornerstone in risk assessment, clinical management and follow-up. The aim of this study was to evaluate if biomarkers associated with comorbidity burden can predict outcome in acute dyspnea patients. We included 774 patients with dyspnea admitted to an emergency department and measured 80 cardiovascular protein biomarkers in serum collected at admission. The number of comorbidities for each patient were added, and a multimorbidity score was created. Eleven of the 80 biomarkers were independently associated with the multimorbidity score and their standardized and weighted values were summed into a biomarker score of multimorbidities. The biomarker score and the multimorbidity score, expressed per standard deviation increment, respectively, were related to all-cause mortality using Cox Proportional Hazards Model. During long-term follow-up (2.4 ± 1.5 years) 45% of the patients died and during short-term follow-up (90 days) 12% died. Through long-term follow-up, in fully adjusted models, the HR (95% CI) for mortality concerning the biomarker score was 1.59 (95% CI 1348-1871) and 1.18 (95% CI 1035-1346) for the multimorbidity score. For short-term follow-up, in the fully adjusted model, the biomarker score was strongly related to 90-day mortality (HR 1.98, 95% CI 1428-2743), whereas the multimorbidity score was not significant. Our main findings suggest that the biomarker score is superior to the multimorbidity score in predicting long and short-term mortality. Measurement of the biomarker score may serve as a biological fingerprint of the multimorbidity score at the emergency department and, therefore, be helpful for risk prediction, treatment decisions and need of follow-up both in hospital and after discharge from the emergency department.
Collapse
Affiliation(s)
- Torgny Wessman
- grid.411843.b0000 0004 0623 9987Department of Emergency Medicine, Skåne University Hospital, Ruth Lundskogs gata 3, 20502 Malmö, Sweden
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Rafid Tofik
- grid.411843.b0000 0004 0623 9987Department of Emergency Medicine, Skåne University Hospital, Ruth Lundskogs gata 3, 20502 Malmö, Sweden
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Thoralph Ruge
- grid.411843.b0000 0004 0623 9987Department of Emergency Medicine, Skåne University Hospital, Ruth Lundskogs gata 3, 20502 Malmö, Sweden
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Olle Melander
- grid.411843.b0000 0004 0623 9987Department of Internal Medicine, Skåne University Hospital, Malmö, Sweden
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
25
|
Blood DNA Methylation Predicts Diabetic Kidney Disease Progression in High Fat Diet-Fed Mice. Nutrients 2022; 14:nu14040785. [PMID: 35215435 PMCID: PMC8880442 DOI: 10.3390/nu14040785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/29/2022] Open
Abstract
Diabetic kidney disease (DKD) progresses at different rates among patients with type 2 diabetes mellitus (T2D). Early identification of patients with a higher risk of DKD progression is essential to improve prognosis. Epigenetic modifications, particularly DNA methylation, have been independently implicated in T2D and chronic kidney disease. The current study aimed to determine changes in blood DNA methylation that reflects and predicts DKD progression. C57BL/6 mice were fed a high-fat diet (HFD) from weaning and subclassified into two groups, HFD-1 and HFD-2, according to urinary kidney injury marker KIM-1/creatinine ratios (low vs. high) and histological abnormalities (mild–moderate vs. advanced). DNA methylation profiles were determined by reduced representative bisulfide sequencing (RRBS). Our results confirmed early and established DKD at week 9 and week 32, respectively. At week 32, advanced kidney injury was associated with dysregulation of methylation and demethylation enzymes in the kidney. Blood RRBS revealed 579 and 203 differentially methylated sites (DMS) between HFD-1 and HFD-2 animals at week 32 and week 9, respectively, among which 11 were common. The DMS in blood and kidney at week 32 were both related to organ development, neurogenesis, cell junction, and Wnt signalling, while the DMS in blood at week 9 suggested a specific enrichment of kidney development processes. In conclusion, our data strongly support the implication of early blood DNA methylation modifications and DKD progression in T2D that could be used to improve the disease’s prognostication.
Collapse
|
26
|
Schmidt IM, Sarvode Mothi S, Wilson PC, Palsson R, Srivastava A, Onul IF, Kibbelaar ZA, Zhuo M, Amodu A, Stillman IE, Rennke HG, Humphreys BD, Waikar SS. Circulating Plasma Biomarkers in Biopsy-Confirmed Kidney Disease. Clin J Am Soc Nephrol 2022; 17:27-37. [PMID: 34759008 PMCID: PMC8763150 DOI: 10.2215/cjn.09380721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/02/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Biomarkers for noninvasive assessment of histopathology and prognosis are needed in patients with kidney disease. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Using a proteomics assay, we measured a multimarker panel of 225 circulating plasma proteins in a prospective cohort study of 549 individuals with biopsy-confirmed kidney diseases and semiquantitative assessment of histopathology. We tested the associations of each biomarker with histopathologic lesions and the risks of kidney disease progression (defined as ≥40% decline in eGFR or initiation of KRT) and death. RESULTS After multivariable adjustment and correction for multiple testing, 46 different proteins were associated with histopathologic lesions. The top-performing markers positively associated with acute tubular injury and interstitial fibrosis/tubular atrophy were kidney injury molecule-1 (KIM-1) and V-set and Ig domain-containing protein 2 (VSIG2), respectively. Thirty proteins were significantly associated with kidney disease progression, and 35 were significantly associated with death. The top-performing markers for kidney disease progression were placental growth factor (hazard ratio per doubling, 5.4; 95% confidence interval, 3.4 to 8.7) and BMP and activin membrane-bound inhibitor (hazard ratio, 3.0; 95% confidence interval, 2.1 to 4.2); the top-performing markers for death were TNF-related apoptosis-inducing ligand receptor-2 (hazard ratio, 2.9; 95% confidence interval, 2.0 to 4.0) and CUB domain-containing protein-1 (hazard ratio, 2.4; 95% confidence interval, 1.8 to 3.3). CONCLUSION We identified several plasma protein biomarkers associated with kidney disease histopathology and adverse clinical outcomes in individuals with a diverse set of kidney diseases. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2021_12_28_CJN09380721.mp3.
Collapse
Affiliation(s)
- Insa M. Schmidt
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Suraj Sarvode Mothi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Parker C. Wilson
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Ragnar Palsson
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ingrid F. Onul
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Zoe A. Kibbelaar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Min Zhuo
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Afolarin Amodu
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Isaac E. Stillman
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Helmut G. Rennke
- Department of Pathology, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University, St. Louis, Missouri
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
27
|
Glerup R, Svensson M, Jakobsen LH, Fellstrøm B, Jensen JD, Christensen JH. Multiplex proteomics as risk predictor of infection in patients treated with hemodialysis-A prospective multicenter study. Hemodial Int 2021; 26:191-201. [PMID: 34964538 DOI: 10.1111/hdi.12987] [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: 04/21/2021] [Revised: 09/11/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Severe infection is a major problem in hemodialysis patients. Multiplex proteomics might reveal novel insights into disease mechanisms increasing the risk of infection and might also be used as a risk prediction tool. The aims of this study were (1) to evaluate associations between 92 proteins assessed by a proximity extension assay and the development of severe infection in patients on hemodialysis and (2) to develop a risk prediction model for severe infection using prespecified clinical variables and proteomics. METHODS Prospective, observational multicenter cohort study with 5-year follow-up. Patients receiving in-center hemodialysis in five facilities in Denmark were included. The primary composite endpoint was death caused by infection, bacteremia, and infections requiring hospitalization of at least 2 days or prolonging a hospital stay. FINDINGS Of 331 patients included 210 patients reached the primary endpoint during follow-up. In adjusted Cox regression analyses, 14 plasma proteins were associated with severe infection. Correcting for multiple testing revealed only cathepsin-L1 and interleukin-6 significantly associated with the primary outcome. Cathepsin-L1-hazard ratio: 1.64 (95% confidence interval [CI] 1.24-2.17) and interleukin-6-hazard ratio: 1.16 (95% CI 1.05-1.29). Apparent C-statistics of the risk prediction model using clinical variables was 0.605, addition of cathepsin-L1 and interleukin-6 to the model improved discrimination slightly: C = 0.625. DISCUSSION Proteomic profiling identified cathepsin-L1 and interleukin-6 as markers for infectious risk in hemodialysis patients. Further studies are needed to replicate the results and to examine possible causality. The developed risk prediction models need considerable improvement before implementation in clinical practice is meaningful.
Collapse
Affiliation(s)
- Rie Glerup
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - My Svensson
- Department of Renal Medicine, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lasse H Jakobsen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Bengt Fellstrøm
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jens D Jensen
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jeppe H Christensen
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark.,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
28
|
Tracz J, Luczak M. Applying Proteomics and Integrative "Omics" Strategies to Decipher the Chronic Kidney Disease-Related Atherosclerosis. Int J Mol Sci 2021; 22:7492. [PMID: 34299112 PMCID: PMC8305100 DOI: 10.3390/ijms22147492] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk of atherosclerosis and premature mortality, mainly due to cardiovascular events. However, well-known risk factors, which promote "classical" atherosclerosis are alone insufficient to explain the high prevalence of atherosclerosis-related to CKD (CKD-A). The complexity of the molecular mechanisms underlying the acceleration of CKD-A is still to be defied. To obtain a holistic picture of these changes, comprehensive proteomic approaches have been developed including global protein profiling followed by functional bioinformatics analyses of dysregulated pathways. Furthermore, proteomics surveys in combination with other "omics" techniques, i.e., transcriptomics and metabolomics as well as physiological assays provide a solid ground for interpretation of observed phenomena in the context of disease pathology. This review discusses the comprehensive application of various "omics" approaches, with emphasis on proteomics, to tackle the molecular mechanisms underlying CKD-A progression. We summarize here the recent findings derived from global proteomic approaches and underline the potential of utilizing integrative systems biology, to gain a deeper insight into the pathogenesis of CKD-A and other disorders.
Collapse
Affiliation(s)
| | - Magdalena Luczak
- European Centre for Bioinformatics and Genomics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland;
| |
Collapse
|
29
|
Néri AK, da S Junior GB, Meneses GC, Martins AM, F Daher ED, da C Lino DO, Silva RP, Psf Nunes MD, Alencar RL, Rodrigues MS, Saraiva IP. Cardiovascular risk assessment and association with novel biomarkers in patients with Type 2 diabetes mellitus. Biomark Med 2021; 15:561-576. [PMID: 33988460 DOI: 10.2217/bmm-2020-0611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aim: To investigate the association between cardiovascular risk and biomarkers in patients with Type 2 diabetes (T2DM). Methods: Cross-sectional study, with evaluation of traditional and new biomarkers (serum FGF-23, Syndecan-1 [Sdc-1] and vascular cell adhesion molecule-1 [VCAM-1] and urinary VEGF and kidney injury molecule-1 [KIM-1]) and risk scores (Framingham-FRS and UK Prospective Diabetes Study [UKPDS]). Results: 128 diabetics were included, with predominance of high risk by FRS and low risk by UKPDS. There was an independent association of VCAM-1 and VEGF with higher risk by FRS-lipids and UKPDS. Conclusion: There was an independent association of VCAM-1 and VEGF with higher cardiovascular risk, showing a subclinical endothelial dysfunction in T2DM. The inclusion of novel biomarkers to risk scores may increase accuracy when assessing cardiovascular risk of diabetic individuals.
Collapse
Affiliation(s)
- Ane Km Néri
- Postgraduate Program in Collective Health, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil.,Cardiology Service, Walter Cantídio Teaching Hospital, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.,School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Geraldo B da S Junior
- Postgraduate Program in Collective Health, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil.,Postgraduate Program in Medical Sciences, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil.,School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Gdayllon C Meneses
- Postgraduate Program in Medical Sciences, Department of Clinical Medicine, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Postgraduate Program in Pharmacology, Department of Medicine, Physiology & Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Alice Mc Martins
- Postgraduate Program in Pharmacology, Department of Medicine, Physiology & Pharmacology, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Postgraduate Program in Pharmaceutical Sciences, School of Pharmacy, Dentistry & Nursing, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Elizabeth De F Daher
- Postgraduate Program in Medical Sciences, Department of Clinical Medicine, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Danielli O da C Lino
- Postgraduate Program in Collective Health, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Ricardo P Silva
- Cardiology Service, Walter Cantídio Teaching Hospital, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil.,Postgraduate Program in Cardiovascular Sciences, Department of Clinical Medicine, School of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Marina de Psf Nunes
- School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Renan L Alencar
- School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Mariane S Rodrigues
- School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| | - Igor P Saraiva
- School of Medicine, Health Sciences Center, University of Fortaleza, Fortaleza, Ceará, Brazil
| |
Collapse
|
30
|
Letunica N, Cai T, Cameron F, Monagle P, Ignjatovic V, Attard C. Investigating potential protein markers of cardiovascular disease in children with type 1 diabetes mellitus. Proteomics Clin Appl 2021; 15:e2000060. [PMID: 33587825 DOI: 10.1002/prca.202000060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/01/2020] [Accepted: 12/17/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a metabolic disease characterized by dysglycaemia. Cardiovascular disease (CVD) is a major complication among T1DM patients and the leading cause of mortality later in life. METHODS The study subjects consisted of T1DM children with poor glycemic control (HbA1c > 7.5%) and healthy age and gender matched controls. Venous blood samples were collected and tested by utilizing a novel immunoassay panel with 96 protein biomarkers. Data were analyzed using non-linear regression analysis and the expression of biomarkers was compared between T1DM and healthy control groups using an unpaired student's t-test. Dynamic principal component analysis (PCA) was operated based on the differentially expressed proteins. RESULTS Ten T1DM children and 10 healthy controls were analyzed. Twelve CVD markers show significant differential expression between T1DM patients and healthy controls (p < 0.05). Dynamic PCA clustering based on differentially expressed proteins demonstrated an obvious clustering between the two populations. CONCLUSIONS This preliminary study reveals the feasibility of utilizing a novel immunoassay panel to investigate potential biomarkers for predicting incipient CVD in children with T1DM. In future, longitudinal studies are required to track the relationships between measurements of the selected protein markers and the development of CVD in T1DM patients.
Collapse
Affiliation(s)
- Natasha Letunica
- Haematology Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Tengyi Cai
- Haematology Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Fergus Cameron
- Haematology Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Australia
- Department of Endocrinology and Diabetes and Centre for Hormone Research, Royal Children's Hospital, Parkville, Australia
| | - Paul Monagle
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Australia
- Department of Clinical Haematology, Royal Children's Hospital, Parkville, Australia
| | - Vera Ignjatovic
- Haematology Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Australia
| | - Chantal Attard
- Haematology Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Australia
| |
Collapse
|
31
|
Kakareko K, Rydzewska-Rosołowska A, Zbroch E, Hryszko T. TRAIL and Cardiovascular Disease-A Risk Factor or Risk Marker: A Systematic Review. J Clin Med 2021; 10:jcm10061252. [PMID: 33803523 PMCID: PMC8002847 DOI: 10.3390/jcm10061252] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a pro-apoptotic protein showing broad biological functions. Data from animal studies indicate that TRAIL may possibly contribute to the pathophysiology of cardiomyopathy, atherosclerosis, ischemic stroke and abdominal aortic aneurysm. It has been also suggested that TRAIL might be useful in cardiovascular risk stratification. This systematic review aimed to evaluate whether TRAIL is a risk factor or risk marker in cardiovascular diseases (CVDs) focusing on major adverse cardiovascular events. Two databases (PubMed and Cochrane Library) were searched until December 2020 without a year limit in accordance to the PRISMA guidelines. A total of 63 eligible original studies were identified and included in our systematic review. Studies suggest an important role of TRAIL in disorders such as heart failure, myocardial infarction, atrial fibrillation, ischemic stroke, peripheral artery disease, and pulmonary and gestational hypertension. Most evidence associates reduced TRAIL levels and increased TRAIL-R2 concentration with all-cause mortality in patients with CVDs. It is, however, unclear whether low TRAIL levels should be considered as a risk factor rather than a risk marker of CVDs. Further studies are needed to better define the association of TRAIL with cardiovascular diseases.
Collapse
Affiliation(s)
- Katarzyna Kakareko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland; (A.R.-R.); (T.H.)
- Correspondence:
| | - Alicja Rydzewska-Rosołowska
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland; (A.R.-R.); (T.H.)
| | - Edyta Zbroch
- Department of Internal Medicine and Hypertension, Medical University of Białystok, 15-276 Białystok, Poland;
| | - Tomasz Hryszko
- 2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland; (A.R.-R.); (T.H.)
| |
Collapse
|
32
|
Proteomic profiling identifies key differences between inter-stage infants with single ventricle heart disease and healthy controls. Transl Res 2021; 229:24-37. [PMID: 33045409 PMCID: PMC8191179 DOI: 10.1016/j.trsl.2020.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
Despite significant morbidity among infants with single ventricle heart disease (SVHD), clinical monitoring is limited by poor understanding of the underlying pathobiology. Proteomics can identify novel biomarkers and important pathways in complex disease. No prior study has evaluated whether the proteome of SVHD infants differs from healthy controls, how it shifts after stage 2 palliation, or whether differences can predict post-operative outcomes. We present a prospective cohort study of cardiovascular proteomic phenotyping in infants with SVHD undergoing stage 2 palliation. Twenty-nine pre-stage-2 SVHD infants and 25 healthy controls were enrolled. Outcomes included postoperative hypoxemia and endotracheal intubation time. Serum samples were drawn pre-operatively (systemic and pulmonary vein) and at 24 hours postoperation. Targeted cardiovascular proteomic analysis included 184 proteins. Partial least squares discriminant analysis distinguished cases from controls (Accuracy = 0.98, R2 = 0.93, Q2 = 0.81) with decreased inflammatory mediators and increased modulators of vascular tone. Partial least squares discriminant analysis also distinguished cases pre-operation vs. post-operation (Accuracy=0.98, R2=0.99, Q2 = 0.92) with postoperative increase in both inflammatory and vascular tone mediators. Pre-operation pulmonary vein tissue inhibitor of metalloproteinase-1 (1.8x-fold, p=1.6 × 10-4) and nidogen-1 (1.5x-fold, p=1.7 × 10-4) were higher in subjects with longer endotracheal intubation time. Postoperation matrix metalloproteinase 7 levels were higher in subjects with greater postoperative hypoxemia (1.5x-fold, P= 1.97 × 10-5). Proteomic analysis identifies significant changes among SVHD infants pre- and post-stage 2, and healthy controls. Tissue inhibitor of metalloproteinase-1, nidogen-1, and matrix metalloproteinase 7 levels are higher in SVHD cases with greater morbidity suggesting an important role for regulation of extracellular matrix production. Proteomic profiling may identify high-risk SVHD infants.
Collapse
|
33
|
Prognostic Value of Soluble Suppression of Tumorigenicity 2 in Chronic Kidney Disease Patients: A Meta-Analysis. DISEASE MARKERS 2021; 2021:8881393. [PMID: 33574967 PMCID: PMC7857877 DOI: 10.1155/2021/8881393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/31/2020] [Accepted: 01/14/2021] [Indexed: 01/11/2023]
Abstract
Objective Previous studies have controversial results about the prognostic role of soluble suppression of tumorigenicity 2 (sST2) in chronic kidney disease (CKD). Therefore, we conduct this meta-analysis to access the association between sST2 and all-cause mortality, cardiovascular disease (CVD) mortality, and CVD events in patients with CKD. Methods The publication studies on the association of sST2 with all-cause mortality, CVD mortality, and CVD events from PubMed and Embase were searched through August 2020. We pooled the hazard ratio (HR) comparing high versus low levels of sST2 and subgroup analysis based on treatment, continent, and diabetes mellitus (DM) proportion, and sample size was also performed. Results There were 15 eligible studies with 11,063 CKD patients that were included in our meta-analysis. Elevated level of sST2 was associated with increased risk of all-cause mortality (HR 2.05; 95% confidence interval (CI), 1.51-2.78), CVD mortality (HR 1.68; 95% CI, 1.35-2.09), total CVD events (HR 1.88; 95% CI, 1.26-2.80), and HF (HR 1.35; 95% CI, 1.11-1.64). Subgroup analysis based on continent, DM percentage, and sample size showed that these factors did not influence the prognostic role of sST2 levels to all-cause mortality. Conclusions Our results show that high levels of sST2 could predict the all-cause mortality, CVD mortality, and CVD events in CKD patients.
Collapse
|
34
|
Qian Y, Li L, Sun Z, Liu J, Yuan W, Wang Z. A multi-omics view of the complex mechanism of vascular calcification. Biomed Pharmacother 2021; 135:111192. [PMID: 33401220 DOI: 10.1016/j.biopha.2020.111192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification is a high incidence and high risk disease with increasing morbidity and high mortality, which is considered the consequence of smooth muscle cell transdifferentiation initiating the mechanism of accumulation of hydroxyl calcium phosphate. Vascular calcification is also thought to be strongly associated with poor outcomes in diabetes and chronic kidney disease. Numerous studies have been accomplished; however, the specific mechanism of the disease remains unclear. Development of the genome project enhanced the understanding of life science and has entered the post-genomic era resulting in a variety of omics techniques used in studies and a large amount of available data; thus, a new perspective on data analysis has been revealed. Omics has a broader perspective and is thus advantageous over a single pathway analysis in the study of complex vascular calcification mechanisms. This paper reviews in detail various omics studies including genomics, proteomics, transcriptomics, metabolomics and multiple group studies on vascular calcification. Advances and deficiencies in the use of omics to study vascular calcification are presented in a comprehensive view. We also review the methodology of the omics studies and omics data analysis and processing. In addition, the methodology and data processing presented here can be applied to other areas. An omics landscape perspective across the boundaries between genomics, transcriptomics, proteomics and metabolomics is used to examine the mechanisms of vascular calcification. The perspective combined with various technologies also provides a direction for the subsequent exploration of clinical significance.
Collapse
Affiliation(s)
- Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Jia Liu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China.
| |
Collapse
|
35
|
Bellasi A, Raggi P, Bover J, Bushinsky DA, Chertow GM, Ketteler M, Rodriguez M, Sinha S, Salcedo C, Garg R, Gold A, Perelló J. Trial design and baseline characteristics of CaLIPSO: a randomized, double-blind placebo-controlled trial of SNF472 in patients receiving haemodialysis with cardiovascular calcification. Clin Kidney J 2021; 14:366-374. [PMID: 33564440 PMCID: PMC7857813 DOI: 10.1093/ckj/sfz144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/13/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The objective of CaLIPSO, a Phase 2b, randomized, double-blind, placebo-controlled clinical trial, is to test the hypothesis that myo-inositol hexaphosphate (SNF472) attenuates the progression of cardiovascular calcification in patients receiving maintenance haemodialysis. Here we report the trial design and baseline characteristics of trial participants. METHODS Adult patients on maintenance haemodialysis (≥6 months) with an Agatston coronary artery calcium score, as measured by a multidetector computed tomography scanner, of 100-3500 U were enrolled. Patients were stratified by Agatston score (100-<400, 400-1000 or >1000 U) and randomized in a 1:1:1 ratio to receive placebo, SNF472 300 mg or SNF472 600 mg administered intravenously three times weekly during each haemodialysis session. RESULTS Overall, 274 patients were randomized. The mean age of trial participants was 63.6 (standard deviation 8.9) years and 39% were women. The coronary artery, aorta and aortic valve median (25th-75th percentile) Agatston scores at baseline were 730 U (315-1435), 1728 U (625-4978) and 103 U (31-262), respectively, and the median (25th-75th percentile) calcium volume scores at baseline were 666 (310-1234), 1418 (536-4052) and 107 (38-278), respectively. Older age and diabetes mellitus were associated with higher calcium scores at baseline. CONCLUSIONS The CaLIPSO trial enrolled patients on haemodialysis with pre-existent cardiovascular calcification to test the hypothesis that SNF472 attenuates its progression in the coronary arteries, aorta and aortic valve.
Collapse
Affiliation(s)
- Antonio Bellasi
- Research, Innovation and Brand Reputation Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Raggi
- Department of Medicine, Mazankowski Alberta Heart Institute and University of Alberta, Edmonton, AB, Canada
| | - Jordi Bover
- Department of Nephrology, Fundació Puigvert and Universitat Autònoma, IIB Sant Pau, REDinREN, Barcelona, Spain
| | - David A Bushinsky
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Glenn M Chertow
- Department of Medicine, Stanford University, Palo Alto, CA, USA
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Mariano Rodriguez
- Nephrology Unit, Hospital Universitario Reina Sofia, IMIBIC, REDinREN, Córdoba, Spain
| | - Smeeta Sinha
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK
| | | | - Rekha Garg
- Research and Development, Sanifit Therapeutics, San Diego, CA, USA
| | - Alex Gold
- Research and Development, Sanifit Therapeutics, San Diego, CA, USA
| | - Joan Perelló
- Research and Development, Sanifit Therapeutics, Palma, Spain
| |
Collapse
|
36
|
Edfors R, Lindhagen L, Spaak J, Evans M, Andell P, Baron T, Mörtberg J, Rezeli M, Salzinger B, Lundman P, Szummer K, Tornvall P, Wallén HN, Jacobson SH, Kahan T, Marko-Varga G, Erlinge D, James S, Lindahl B, Jernberg T. Use of proteomics to identify biomarkers associated with chronic kidney disease and long-term outcomes in patients with myocardial infarction. J Intern Med 2020; 288:581-592. [PMID: 32638487 DOI: 10.1111/joim.13116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/30/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) have poor outcomes following myocardial infarction (MI). We performed an untargeted examination of 175 biomarkers to identify those with the strongest association with CKD and to examine the association of those biomarkers with long-term outcomes. METHODS A total of 175 different biomarkers from MI patients enrolled in the Swedish Web-System for Enhancement and Development of Evidence-Based Care in Heart Disease Evaluated According to Recommended Therapies (SWEDEHEART) registry were analysed either by a multiple reaction monitoring mass spectrometry assay or by a multiplex assay (proximity extension assay). Random forests statistical models were used to assess the predictor importance of biomarkers, CKD and outcomes. RESULTS A total of 1098 MI patients with a median estimated glomerular filtration rate of 85 mL min-1 /1.73 m2 were followed for a median of 3.2 years. The random forests analyses, without and with adjustment for differences in demography, comorbidities and severity of disease, identified six biomarkers (adrenomedullin, TNF receptor-1, adipocyte fatty acid-binding protein-4, TNF-related apoptosis-inducing ligand receptor 2, growth differentiation factor-15 and TNF receptor-2) to be strongly associated with CKD. All six biomarkers were also amongst the 15 strongest predictors for death, and four of them were amongst the strongest predictors of subsequent MI and heart failure hospitalization. CONCLUSION In patients with MI, a proteomic approach could identify six biomarkers that best predicted CKD. These biomarkers were also amongst the most important predictors of long-term outcomes. Thus, these biomarkers indicate underlying mechanisms that may contribute to the poor prognosis seen in patients with MI and CKD.
Collapse
Affiliation(s)
- R Edfors
- From the, Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden.,Bayer AB, Solna, Sweden
| | - L Lindhagen
- Uppsala Clinical Research Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - J Spaak
- From the, Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - M Evans
- Department of Clinical Science, Intervention and Technology (CLINTEC), Division of Renal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - P Andell
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - T Baron
- Uppsala Clinical Research Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - J Mörtberg
- Department of Clinical Sciences, Division of Renal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - M Rezeli
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - B Salzinger
- Department of Clinical Sciences, Division of Renal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - P Lundman
- From the, Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - K Szummer
- Department of Medicine, Unit of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - P Tornvall
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - H N Wallén
- From the, Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - S H Jacobson
- Department of Clinical Sciences, Division of Renal Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - T Kahan
- From the, Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - G Marko-Varga
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - D Erlinge
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - S James
- Uppsala Clinical Research Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - B Lindahl
- Uppsala Clinical Research Center, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - T Jernberg
- From the, Department of Clinical Sciences, Division of Cardiovascular Medicine, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Association between nonsteroidal anti-inflammatory drug use and major adverse cardiovascular events in patients with end-stage renal disease: a population-based cohort study. J Nephrol 2020; 34:441-449. [PMID: 32840754 DOI: 10.1007/s40620-020-00830-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/08/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used for patients with end-stage renal disease (ESRD) despite clinical guideline recommendations that the use of NSAIDs be avoided in this population. However, the relationship between NSAID use and adverse cardiovascular events remains unclear. Thus, this study investigated the association between NSAID use and major adverse cardiovascular events (MACEs) in patients with ESRD. METHODS We used the Taiwan National Health Insurance Research Database to conduct this population-based cohort study of patients with newly diagnosed ESRD requiring long-term dialysis between 1998 and 2012. Clinical outcomes were evaluated until the end of 2013. Time-dependent Cox regression models were used to investigate the association between NSAID use and MACEs in patients with ESRD. RESULTS Among 2349 patients with ESRD receiving dialysis, 1923 (82%) patients used NSAIDs during the follow-up period. Multivariable analysis revealed that compared with nonusers, NSAID users exhibited an increased risk of MACEs with an adjusted hazard ratio (HR) of 1.70 (95% confidence interval [CI] 1.22-2.36). Further analysis demonstrated a significant dose-response relationship between the cumulative use of NSAIDs and MACEs. Adjusted HRs for MACEs were 1.63 (95% CI 1.16-2.30), 1.86 (95% CI 1.22-2.83), and 1.99 (95% CI 1.24-3.20) for cumulative NSAID use of 1-30 defined daily doses (DDDs), 31-90 DDDs, and > 90 DDDs, respectively. CONCLUSIONS The results of this study suggest that NSAID use may increase the risk of MACEs in patients with ESRD. Clinicians and patients with ESRD should be aware of the potential cardiovascular risks associated with NSAIDs.
Collapse
|
38
|
Feldreich T, Nowak C, Carlsson AC, Östgren CJ, Nyström FH, Sundström J, Carrero-Roig JJ, Leppert J, Hedberg P, Giedraitis V, Lind L, Cordeiro A, Ärnlöv J. The association between plasma proteomics and incident cardiovascular disease identifies MMP-12 as a promising cardiovascular risk marker in patients with chronic kidney disease. Atherosclerosis 2020; 307:11-15. [PMID: 32702535 DOI: 10.1016/j.atherosclerosis.2020.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/19/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Previous proteomics efforts in patients with chronic kidney disease (CKD) have predominantly evaluated urinary protein levels. Therefore, our aim was to investigate the association between plasma levels of 80 cardiovascular disease-related proteins and the risk of major adverse cardiovascular events (MACE) in patients with CKD. METHODS Individuals with CKD stages 3-5 (eGFR below 60 ml min-1 [1.73 m]-2) from three community-based cohorts (PIVUS, ULSAM, SAVA), one diabetes cohort (CARDIPP) and one cohort with peripheral artery disease patients (PADVA) with information on 80 plasma protein biomarkers, assessed with a proximity extension assay, and follow-up data on incident MACE, were used as discovery sample. To validate findings and to asses generalizability to patients with CKD in clinical practice, an outpatient CKD-cohort (Malnutrition, Inflammation and Vascular Calcification (MIVC)) was used as replication sample. RESULTS In the discovery sample (total n = 1316), 249 individuals experienced MACE during 7.0 ± 2.9 years (range 0.005-12.9) of follow-up, and in the replication sample, 71 MACE events in 283 individuals over a mean ± SD change of 2.9 ± 1.2 years (range 0.1-4.0) were documented. Applying Bonferroni correction, 18 proteins were significantly associated with risk of MACE in the discovery cohort, adjusting for age and sex in order of significance, GDF-15, FGF-23, REN, FABP4, IL6, TNF-R1, AGRP, MMP-12, AM, KIM-1, TRAILR2, TNFR2, CTSL1, CSF1, PlGF, CA-125, CCL20 and PAR-1 (p < 0.000625 for all). Only matrix metalloproteinase 12 (MMP-12) was significantly associated with an increased risk of MACE in the replication sample (hazard ratio (HR) per SD increase, 1.36, 95% CI (1.07-1.75), p = 0.013). CONCLUSIONS Our proteomics analyses identified plasma MMP-12 as a promising cardiovascular risk marker in patients with CKD.
Collapse
Affiliation(s)
- Tobias Feldreich
- Education, Health and Social Studies, Dalarna University, Falun, Sweden.
| | - Christoph Nowak
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Alfred Nobels Allé 23, SE 14183, Huddinge, Sweden
| | - Axel C Carlsson
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Alfred Nobels Allé 23, SE 14183, Huddinge, Sweden
| | - Carl-Johan Östgren
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Fredrik H Nyström
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Juan-Jesus Carrero-Roig
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jerzy Leppert
- Centre for Clinical Research, Uppsala University, Västerås, Sweden
| | - Pär Hedberg
- Centre for Clinical Research, Uppsala University, Västerås, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Antonio Cordeiro
- Department of Hypertension and Nephrology, Dante Pazzanese Institute of Cardiology, São Paulo, Brazil
| | - Johan Ärnlöv
- Education, Health and Social Studies, Dalarna University, Falun, Sweden; Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Alfred Nobels Allé 23, SE 14183, Huddinge, Sweden
| |
Collapse
|
39
|
Raffield LM, Dang H, Pratte KA, Jacobson S, Gillenwater L, Ampleford E, Barjaktarevic I, Basta P, Clish CB, Comellas AP, Cornell E, Curtis JL, Doerschuk C, Durda P, Emson C, Freeman C, Guo X, Hastie AT, Hawkins GA, Herrera J, Johnson WC, Labaki WW, Liu Y, Masters B, Miller M, Ortega VE, Papanicolaou G, Peters S, Taylor KD, Rich SS, Rotter JI, Auer P, Reiner AP, Tracy RP, Ngo D, Gerszten RE, O’Neal WK, Bowler RP. Comparison of Proteomic Assessment Methods in Multiple Cohort Studies. Proteomics 2020; 20:e1900278. [PMID: 32386347 PMCID: PMC7425176 DOI: 10.1002/pmic.201900278] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 04/30/2020] [Indexed: 11/09/2022]
Abstract
Novel proteomics platforms, such as the aptamer-based SOMAscan platform, can quantify large numbers of proteins efficiently and cost-effectively and are rapidly growing in popularity. However, comparisons to conventional immunoassays remain underexplored, leaving investigators unsure when cross-assay comparisons are appropriate. The correlation of results from immunoassays with relative protein quantification is explored by SOMAscan. For 63 proteins assessed in two chronic obstructive pulmonary disease (COPD) cohorts, subpopulations and intermediate outcome measures in COPD Study (SPIROMICS), and COPDGene, using myriad rules based medicine multiplex immunoassays and SOMAscan, Spearman correlation coefficients range from -0.13 to 0.97, with a median correlation coefficient of ≈0.5 and consistent results across cohorts. A similar range is observed for immunoassays in the population-based Multi-Ethnic Study of Atherosclerosis and for other assays in COPDGene and SPIROMICS. Comparisons of relative quantification from the antibody-based Olink platform and SOMAscan in a small cohort of myocardial infarction patients also show a wide correlation range. Finally, cis pQTL data, mass spectrometry aptamer confirmation, and other publicly available data are integrated to assess relationships with observed correlations. Correlation between proteomics assays shows a wide range and should be carefully considered when comparing and meta-analyzing proteomics data across assays and studies.
Collapse
Affiliation(s)
- Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Katherine A. Pratte
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | - Sean Jacobson
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | - Lucas Gillenwater
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | - Elizabeth Ampleford
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Igor Barjaktarevic
- UCLA Division of Pulmonary and Critical Care, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Patricia Basta
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Clary B. Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Elaine Cornell
- Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Jeffrey L. Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI
- VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Claire Doerschuk
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Peter Durda
- Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Claire Emson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZenenca, Gaithersburg, MD
| | - Christine Freeman
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI
- VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Annette T. Hastie
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Gregory A. Hawkins
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI
| | - Yongmei Liu
- Department of Epidemiology & Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | | | | | - Victor E. Ortega
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - George Papanicolaou
- Epidemiology Branch, Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD
| | - Stephen Peters
- Department of Internal Medicine, Section for Pulmonary, Critical Care, Allergy & Immunology, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Paul Auer
- Zilber School of Public Health, University of Wisconsin Milwaukee, Milwaukee, WI
| | - Alex P. Reiner
- Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT
- Department of Biochemistry, Larner College of Medicine at the University of Vermont, Burlington, VT
| | - Debby Ngo
- Division of Pulmonary, Sleep and Critical Care Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Wanda Kay O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Russell P. Bowler
- Division of Pulmonary Medicine, Department of Medicine, National Jewish Health (NJH), Denver, CO
| | | |
Collapse
|
40
|
Thomford NE, Bope CD, Agamah FE, Dzobo K, Owusu Ateko R, Chimusa E, Mazandu GK, Ntumba SB, Dandara C, Wonkam A. Implementing Artificial Intelligence and Digital Health in Resource-Limited Settings? Top 10 Lessons We Learned in Congenital Heart Defects and Cardiology. ACTA ACUST UNITED AC 2020; 24:264-277. [DOI: 10.1089/omi.2019.0142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Nicholas Ekow Thomford
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- School of Medical Sciences, Department of Medical Biochemistry, University of Cape Coast, Cape Coast, Ghana
| | - Christian Domilongo Bope
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- School of Medical Sciences, Department of Medical Biochemistry, University of Cape Coast, Cape Coast, Ghana
- Department of Mathematics and Computer Sciences, Faculty of Sciences, University of Kinshasa, Kinshasa, D.R. Congo
| | - Francis Edem Agamah
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Kevin Dzobo
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Richmond Owusu Ateko
- University of Ghana Medical School, Department of Chemical Pathology, University of Ghana, Accra, Ghana
| | - Emile Chimusa
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gaston Kuzamunu Mazandu
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Simon Badibanga Ntumba
- Department of Mathematics and Computer Sciences, Faculty of Sciences, University of Kinshasa, Kinshasa, D.R. Congo
| | - Collet Dandara
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ambroise Wonkam
- Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Institute for Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
41
|
Lekva T, Sugulle M, Moe K, Redman C, Dechend R, Staff AC. Multiplex Analysis of Circulating Maternal Cardiovascular Biomarkers Comparing Preeclampsia Subtypes. Hypertension 2020; 75:1513-1522. [PMID: 32336238 DOI: 10.1161/hypertensionaha.119.14580] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Preeclampsia, a hypertensive pregnancy disorder, links to increased long-term maternal cardiovascular disease (CVD). The risk is further increased with early-onset preeclampsia (EPE) and delivery of a growth-restricted child. We hypothesized that circulating biomarkers associated with CVD risk differed between preeclampsia subtypes and controls. We compared EPE; n=37, delivery <week 34, late-onset preeclampsia (LPE); n=29, delivery ≥week 34, and normotensive controls (n=49) using Olink Proseek multiplex CVD I assay (targeting 92 biomarkers). We stratified analysis to uteroplacental spiral artery acute atherosis presence in preeclampsia patients, sharing morphological similarities with atherosclerosis. We found 47 CVD-related biomarkers differing between the groups, 42 markers between normotensive controls and EPE, 28 markers between normotensive controls and LPE, and 9 markers between EPE and LPE. Among these 9 markers, ST2 (ST2 protein), MMP (matrix metalloproteinase) 1, MMP3, and fractalkine (CX3CL1) were uniquely dysregulated in EPE. Principal component (PC) analysis of the differing markers identified 4 clusters (named PC1-PC4) that largely separated the preeclampsia and control groups as well as pregnancies with low and high circulating PlGF (placental growth factor). The combination of the single markers PlGF, ST2, MMP1, MMP3, and CX3CL1 had a high discriminatory property to differentiate between EPE and LPE. Preeclampsia with acute atherosis or with fetal growth restriction could be differentiated by Olink biomarkers as compared with preeclampsia without these features. We identified specific CVD-related biomarkers in pregnancy depending on preeclampsia subtypes and uteroplacental acute atherosis. Assessment of these pregnancy measured biomarkers' relation to long-term cardiovascular dysfunction and hard end points is warranted.
Collapse
Affiliation(s)
- Tove Lekva
- From the Department of Obstetrics and Gynecology, Oslo University Hospital, Ullevål, Norway (T.L., M.S., K.M., A.C.S.).,Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Norway (T.L.)
| | - Meryam Sugulle
- From the Department of Obstetrics and Gynecology, Oslo University Hospital, Ullevål, Norway (T.L., M.S., K.M., A.C.S.).,Faculty of Medicine, University of Oslo, Norway (M.S., K.M., A.C.S.)
| | - Kjartan Moe
- From the Department of Obstetrics and Gynecology, Oslo University Hospital, Ullevål, Norway (T.L., M.S., K.M., A.C.S.).,Faculty of Medicine, University of Oslo, Norway (M.S., K.M., A.C.S.).,Department of Gynecology and Obstetrics, Bærum Hospital, Vestre Viken HF, Norway (K.M.)
| | | | - Ralf Dechend
- Experimental and Clinical Research Center, a cooperation of Charité-Universitätsmedizin Berlin and Max-Delbruck Center for Molecular Medicine, Germany (R.D.).,Department of Cardiology and Nephrology, HELIOS-Klinikum, Berlin, Germany (R.D.)
| | - Anne Cathrine Staff
- From the Department of Obstetrics and Gynecology, Oslo University Hospital, Ullevål, Norway (T.L., M.S., K.M., A.C.S.).,Faculty of Medicine, University of Oslo, Norway (M.S., K.M., A.C.S.)
| |
Collapse
|
42
|
Lee S, Oh J, Kim H, Ha J, Chun KH, Lee CJ, Park S, Lee SH, Kang SM. Sacubitril/valsartan in patients with heart failure with reduced ejection fraction with end-stage of renal disease. ESC Heart Fail 2020; 7:1125-1129. [PMID: 32153122 PMCID: PMC7261577 DOI: 10.1002/ehf2.12659] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/15/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Aims Sacubitril/valsartan (SV) reduced heart failure hospitalization and cardiovascular mortality compared with enalapril in the Prospective Comparison of ARNI with ACE‐I to Determine Impact on Global Mortality and Morbidity in Heart Failure trial. However, this trial excluded patients with end stage of renal disease (ESRD); thus, the efficacy and safety of SV in heart failure with reduced ejection fraction (HFrEF) with ESRD remains uncertain. Methods and results We retrospectively analysed the clinical and laboratory data of 501 HFrEF patients who administered with SV from March 2017 to April 2019 in a single tertiary university hospital. A total of 23 HFrEF patients with ESRD on dialysis [58.3% non‐ischaemic heart failure; left ventricular ejection fraction (LVEF): 29.7 ± 4.4%] were included in this study. At baseline and follow‐up visit, we evaluated cardiovascular biomarkers such as high‐sensitive troponin T (hsTnT), soluble ST2 (sST2), echocardiographic parameters, and clinical and adverse events. The mean dose of SV was 90 ± 43 mg/day at baseline and 123 ± 62 mg/day at last follow‐up (follow‐up duration: median 132 days). The level of hsTnT was significantly reduced from 236.2 ± 355.3 to 97.0 ± 14.0 pg/mL (P = 0.002), and the sST2 level was significantly reduced from 40.4 ± 44.0 to 19.6 ± 14.1 ng/mL (P = 0.005). LVEF was significantly improved from 29.7 ± 4.4% to 40.8 ± 10.4% (P = 0.002). During the follow‐up, up‐titration, down‐titration, and maintenance of SV dosing were observed in 7 (30%), 5 (21.7%), and 11 patients (47.8%), respectively. SV down‐titration group had adverse events including symptomatic hypotension (systolic blood pressure <100 mmHg) (n = 4) and dizziness (n = 1), but they did not discontinue SV therapy. Conclusions We found that SV could safely reduce the hsTnT and sST2 levels and improve LVEF in HFrEF patients with ESRD. As far as we know, this is the first study to show the efficacy and safety of SV in HFrEF with ESRD on dialysis. Larger prospective, long‐term follow‐up study should be warranted.
Collapse
Affiliation(s)
- Seonhwa Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jaewon Oh
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Hyoeun Kim
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jaehyung Ha
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Kyeong-Hyeon Chun
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Chan Joo Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Sungha Park
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Sang-Hak Lee
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Seok-Min Kang
- Division of Cardiology, Severance Cardiovascular Hospital, Cardiovascular Research Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-gu, Seoul, 03722, Korea
| |
Collapse
|
43
|
Semba RD, Tian Q, Carlson MC, Xue QL, Ferrucci L. Motoric cognitive risk syndrome: Integration of two early harbingers of dementia in older adults. Ageing Res Rev 2020; 58:101022. [PMID: 31996326 PMCID: PMC7697173 DOI: 10.1016/j.arr.2020.101022] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 12/17/2022]
Abstract
Dementia is characterized by a long preclinical phase that may last years to decades before the onset of mild cognitive impairment. Slow gait speed and subjective memory complaint commonly co-occur during this preclinical phase, and each is a strong independent predictor of cognitive decline and dementia. Motoric cognitive risk (MCR) syndrome is a pre-dementia syndrome that combines these two early harbingers of dementia. The risk of cognitive decline or dementia is stronger for MCR than for either slow gait speed or subjective memory complaint alone. Slow gait speed and subjective memory complaint have several common risk factors: cardiovascular disease, diabetes mellitus, abnormal cortisol profiles, low vitamin D levels, brain atrophy with decreased hippocampal volume, and increased deposition of beta-amyloid in the brain. The underlying pathogenesis of MCR remains poorly understood. Metabolomics and proteomics have great potential to provide new insights into biological pathways involved in MCR during the long preclinical phase preceding dementia.
Collapse
Affiliation(s)
- Richard D Semba
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Qu Tian
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michelle C Carlson
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Qian-Li Xue
- Departments of Medicine, Biostatistics, and Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
44
|
Hultberg J, Ernerudh J, Larsson M, Nilsdotter-Augustinsson Å, Nyström S. Plasma protein profiling reflects T H1-driven immune dysregulation in common variable immunodeficiency. J Allergy Clin Immunol 2020; 146:417-428. [PMID: 32057767 DOI: 10.1016/j.jaci.2020.01.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/14/2020] [Accepted: 01/17/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is a disorder characterized by antibody deficiency. A significant fraction of the patients suffer from immune dysregulation, which leads to increased morbidity and mortality. The pathogenesis of this condition is poorly understood. OBJECTIVE Our aim was to find out whether the plasma protein signature in CVID is associated with clinical characteristics and lymphocyte aberrations. METHODS A highly sensitive proximity extension assay was used for targeted profiling of 145 plasma proteins in 29 patients with CVID. Phenotyping of peripheral lymphocytes was done by flow cytometry. The findings were correlated with the burden of immune dysregulation. RESULTS Unsupervised clustering of plasma protein profiles identified 2 distinct groups of patients with CVID that differed significantly in terms of the degree of complications due to immune dysregulation and in terms of the frequency of activated B- and T-cell subpopulations. Pathway analysis identified IFN-γ and IL-1β as the top enriched upstream regulators associated with higher grade of immune dysregulation. In addition, CVID was found to be associated with increased plasma levels of the B-cell-attracting chemokine CXCL13. CONCLUSION Clustering based on plasma protein profiles delineated a subgroup of patients with CVID with activated T cells and clinical complications due to immune dysregulation. Thus, data indicate that CVID-associated immune dysregulation is a TH1-mediated inflammatory process driven by the IFN-γ pathway.
Collapse
Affiliation(s)
- Jonas Hultberg
- Division of Molecular Virology, Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Clinical Immunology and Transfusions Medicine, and Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusions Medicine, and Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Marie Larsson
- Division of Molecular Virology, Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Department of Infectious Diseases, and Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Division of Molecular Virology, Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Clinical Immunology and Transfusions Medicine, and Department of Biomedicine and Clinical Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
45
|
Carlsson AC, Nowak C, Lind L, Östgren CJ, Nyström FH, Sundström J, Carrero JJ, Riserus U, Ingelsson E, Fall T, Ärnlöv J. Growth differentiation factor 15 (GDF-15) is a potential biomarker of both diabetic kidney disease and future cardiovascular events in cohorts of individuals with type 2 diabetes: a proteomics approach. Ups J Med Sci 2020; 125:37-43. [PMID: 31805809 PMCID: PMC7054929 DOI: 10.1080/03009734.2019.1696430] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Diabetic kidney disease (DKD) is a leading risk factor for end-stage renal disease and is one of the most important risk factors for cardiovascular disease in patients with diabetes. It is possible that novel markers portraying the pathophysiological underpinning processes may be useful.Aim: To investigate the associations between 80 circulating proteins, measured by a proximity extension assay, and prevalent DKD and major adverse cardiovascular events (MACE) in type 2 diabetes.Methods: We randomly divided individuals with type 2 diabetes from three cohorts into a two-thirds discovery and one-third replication set (total n = 813, of whom 231 had DKD defined by estimated glomerular filtration rate <60 mg/mL/1.73 m2 and/or urinary albumin-creatinine ratio ≥3 g/mol). Proteins associated with DKD were also assessed as predictors for incident major adverse cardiovascular events (MACE) in persons with DKD at baseline.Results: Four proteins were positively associated with DKD in models adjusted for age, sex, cardiovascular risk factors, glucose control, and diabetes medication: kidney injury molecule-1 (KIM-1, odds ratio [OR] per standard deviation increment, 1.65, 95% confidence interval [CI] 1.27-2.14); growth differentiation factor 15 (GDF-15, OR 1.40, 95% CI 1.16-1.69); myoglobin (OR 1.57, 95% CI 1.30-1.91), and matrix metalloproteinase 10 (MMP-10, OR 1.43, 95% CI 1.17-1.74). In patients with DKD, GDF-15 was significantly associated with increased risk of MACE after adjustments for baseline age, sex, microalbuminuria, and kidney function and (59 MACE events during 7 years follow-up, hazard ratio per standard deviation increase 1.43 [95% CI 1.03-1.98]) but not after further adjustments for cardiovascular risk factors.Conclusion: Our proteomics approach confirms and extends previous associations of higher circulating levels of GDF-15 with both micro- and macrovascular disease in patients with type 2 diabetes. Our data encourage additional studies evaluating the clinical utility of our findings.
Collapse
Affiliation(s)
- Axel C. Carlsson
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
- CONTACT Axel C. Carlsson Department of Neurobiology Care Sciences and Society, Division for Family Medicine and Primary Care, 141 83 Huddinge, Sweden
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl Johan Östgren
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Fredrik H. Nyström
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Juan Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Riserus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
- Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| |
Collapse
|
46
|
Santagati G, Cataldo E, Columbano V, Chatrenet A, Penna D, Pelosi E, Hachemi M, Gendrot L, Nielsen L, Cinquantini F, Saulnier P, Arena V, Boursot C, Piccoli GB. Positron Emission Tomography Can Support the Diagnosis of Dialysis-Related Amyloidosis. J Clin Med 2019; 8:jcm8091494. [PMID: 31546847 PMCID: PMC6781261 DOI: 10.3390/jcm8091494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/31/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The improvements in dialysis have not eliminated long-term problems, including dialysis-related amyloidosis (DRA), caused by Beta-2 microglobulin deposition. Several types of scintigraphy have been tested to detect DRA, none entered the clinical practice. Aim of the study was to assess the potential of PET-FDG scan in the diagnosis of DRA. METHODS Forty-six dialysis patients with at least one PET scan (72 scans) were selected out 162 patients treated in 2016-2018. Subjective global assessment (SGA), malnutrition inflammation score (A), Charlson Comorbidity Index (CCI), were assessed at time of scan; 218 age-matched cases with normal kidney function were selected as controls. PET scans were read in duplicate. Carpal tunnel syndrome was considered a proxy for DRA. A composite "amyloid score" score considered each dialysis year = 1 point; carpal tunnel-DRA = 5 points per site. Logistic regression, ROC curves and a prediction model were built. RESULTS The prevalence of positive PET was 43.5% in dialysis, 5% in controls (p < 0.0001). PET was positive in 14/15 (93.3%) scans in patients with carpal tunnel. PET sensitivity for detecting DRA was 95% (specificity 64%). Carpal tunnel was related to dialysis vintage and MIS. A positive PET scan was significantly associated with dialysis vintage, MIS and amyloid score. A prediction model to explain PET positivity combined clinical score and MIS, allowing for an AUC of 0.906 (CI: 0.813-0.962; p < 0.001). CONCLUSIONS PET-FDG may identify DRA, and may be useful in detecting cases in which inflammation favours B2M deposition. This finding, needing large-scale confirmation, could open new perspectives in the study of DRA.
Collapse
Affiliation(s)
| | | | | | | | - Daniele Penna
- Affidea IRMET, PET CENTER, Torino via Onorato Vigliani 89, 10135 Torino, Italy.
| | - Ettore Pelosi
- Affidea IRMET, PET CENTER, Torino via Onorato Vigliani 89, 10135 Torino, Italy.
| | - Mammar Hachemi
- Medecine Nucleaire, Centre Hospitalier du Mans, 72037 Le Mans, France.
| | | | - Louise Nielsen
- Néphrologie, Centre Hospitalier du Mans, 72037 Le Mans, France.
| | | | | | - Vincenzo Arena
- Affidea IRMET, PET CENTER, Torino via Onorato Vigliani 89, 10135 Torino, Italy.
| | - Charles Boursot
- Medecine Nucleaire, Centre Hospitalier du Mans, 72037 Le Mans, France.
| | - Giorgina Barbara Piccoli
- Néphrologie, Centre Hospitalier du Mans, 72037 Le Mans, France.
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino, 10100 Torino, Italy.
| |
Collapse
|
47
|
Arici M. Kidney injury molecule-1: a successful quest for a predictive kidney disease marker? Nephrol Dial Transplant 2019; 35:194-197. [DOI: 10.1093/ndt/gfz074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 03/23/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mustafa Arici
- Department of Nephrology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|