1
|
McHill AW, Melanson EL, Wright KP, Depner CM. Circadian misalignment disrupts biomarkers of cardiovascular disease risk and promotes a hypercoagulable state. Eur J Neurosci 2024; 60:5450-5466. [PMID: 39053917 DOI: 10.1111/ejn.16468] [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: 03/24/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
The circadian system regulates 24-h time-of-day patterns of cardiovascular physiology, with circadian misalignment resulting in adverse cardiovascular risk. Although many proteins in the coagulation-fibrinolysis axis show 24-h time-of-day patterns, it is not understood if these temporal patterns are regulated by circadian or behavioral (e.g., sleep and food intake) cycles, or how circadian misalignment influences these patterns. Thus, we utilized a night shiftwork protocol to analyze circadian versus behavioral cycle regulation of 238 plasma proteins linked to cardiovascular physiology. Six healthy men aged 26.2 ± 5.6 years (mean ± SD) completed the protocol involving two baseline days with 8-h nighttime sleep opportunities (circadian alignment), a transition to shiftwork day, followed by 2 days of simulated night shiftwork with 8-h daytime sleep opportunities (circadian misalignment). Plasma was collected for proteomics every 4 h across 24 h during baseline and during daytime sleep and the second night shift. Cosinor analyses identified proteins with circadian or behavioral cycle-regulated 24-h time-of-day patterns. Five proteins were circadian regulated (plasminogen activator inhibitor-1, angiopoietin-2, insulin-like growth factor binding protein-4, follistatin-related protein-3, and endoplasmic reticulum resident protein-29). No cardiovascular-related proteins showed regulation by behavioral cycles. Within the coagulation pathway, circadian misalignment decreased tissue factor pathway inhibitor, increased tissue factor, and induced a 24-h time-of-day pattern in coagulation factor VII (all FDR < 0.10). Such changes in protein abundance are consistent with changes observed in hypercoagulable states. Our analyses identify circadian regulation of proteins involved in cardiovascular physiology and indicate that acute circadian misalignment could promote a hypercoagulable state, possibly contributing to elevated cardiovascular disease risk among shift workers.
Collapse
Affiliation(s)
- Andrew W McHill
- Sleep, Chronobiology, and Health Laboratory, School of Nursing, Oregon Health & Science University, Portland, Oregon, USA
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon, USA
| | - Edward L Melanson
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kenneth P Wright
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Christopher M Depner
- Department of Health and Kinesiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
2
|
An J, Wen L, Yu H, Bu Z, Feng J. Insulin-Like Growth Factor Binding Protein 2 Drives Neurodegeneration in Parkinson's Disease: Insights From In Vivo and In Vitro Studies. CNS Neurosci Ther 2024; 30:e70076. [PMID: 39412224 PMCID: PMC11480970 DOI: 10.1111/cns.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/22/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
AIMS Insulin-like growth factor binding protein 2 (IGFBP2) is implicated in various neurodegenerative diseases. However, its role in Parkinson's disease (PD) is unclear. METHODS PD rat model was established by 6-OHDA injection. After 3 weeks, mRNA-seq was conducted. Rats received rIGFBP2 via intra-MFB injection 6 h prior to 6-OHDA infusion, and the effect of IGFBP2 in PD rats was investigated by western blotting, IHC, specific kits, JC-1 staining, and TUNEL analysis. In vitro, PC12 cells were treated with 6-OHDA, and CCK-8, specific kits, Hoechst 33258 staining, Western blotting, and JC-1 staining were performed to assess the IGFBP2's role. RESULTS mRNA-seq revealed DEGs in PD, with attention to downregulated IGFBP2. rIGFBP2 treatment aggravated neurobehavioral deficits, decreased TH expression, Ψm, ATP level and SOD, GSH-Px activities but increased α-synuclein, ROS, MDA, mitochondrial cytochrome c contents, cell apoptosis in 6-OHDA-lesioned rats, which might be mediated through inactivating IGF-1R/AKT pathway. In 6-OHDA-treated PC12 cells, rIGFBP2 aggravated cell injury, demonstrated by decreased cell viability and increased apoptosis, oxidative stress, and mitochondrial dysfunction. Co-treatment with rIGFBP2 and rIGF-1 partially reversed the effect of rIGFBP2 on cell damage. CONCLUSION IGFBP2 exacerbates neurodegeneration in PD through increasing oxidative stress, mitochondrial dysfunction, and apoptosis via inhibiting IGF-1R/AKT pathway.
Collapse
Affiliation(s)
- Jing An
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Lulu Wen
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Haiyang Yu
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Zhongqi Bu
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Juan Feng
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
3
|
Dubin RF, Deo R, Ren Y, Wang J, Pico AR, Mychaleckyj JC, Kozlitina J, Arthur V, Lee H, Shah A, Feldman H, Bansal N, Zelnick L, Rao P, Sukul N, Raj DS, Mehta R, Rosas SE, Bhat Z, Weir MR, He J, Chen J, Kansal M, Kimmel PL, Ramachandran VS, Waikar SS, Segal MR, Ganz P. Incident heart failure in chronic kidney disease: proteomics informs biology and risk stratification. Eur Heart J 2024; 45:2752-2767. [PMID: 38757788 PMCID: PMC11313584 DOI: 10.1093/eurheartj/ehae288] [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: 07/30/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND AND AIMS Incident heart failure (HF) among individuals with chronic kidney disease (CKD) incurs hospitalizations that burden patients and health care systems. There are few preventative therapies, and the Pooled Cohort equations to Prevent Heart Failure (PCP-HF) perform poorly in the setting of CKD. New drug targets and better risk stratification are urgently needed. METHODS In this analysis of incident HF, SomaScan V4.0 (4638 proteins) was analysed in 2906 participants of the Chronic Renal Insufficiency Cohort (CRIC) with validation in the Atherosclerosis Risk in Communities (ARIC) study. The primary outcome was 14-year incident HF (390 events); secondary outcomes included 4-year HF (183 events), HF with reduced ejection fraction (137 events), and HF with preserved ejection fraction (165 events). Mendelian randomization and Gene Ontology were applied to examine causality and pathways. The performance of novel multi-protein risk models was compared to the PCP-HF risk score. RESULTS Over 200 proteins were associated with incident HF after adjustment for estimated glomerular filtration rate at P < 1 × 10-5. After adjustment for covariates including N-terminal pro-B-type natriuretic peptide, 17 proteins remained associated at P < 1 × 10-5. Mendelian randomization associations were found for six proteins, of which four are druggable targets: FCG2B, IGFBP3, CAH6, and ASGR1. For the primary outcome, the C-statistic (95% confidence interval [CI]) for the 48-protein model in CRIC was 0.790 (0.735, 0.844) vs. 0.703 (0.644, 0.762) for the PCP-HF model (P = .001). C-statistic (95% CI) for the protein model in ARIC was 0.747 (0.707, 0.787). CONCLUSIONS Large-scale proteomics reveal novel circulating protein biomarkers and potential mediators of HF in CKD. Proteomic risk models improve upon the PCP-HF risk score in this population.
Collapse
Affiliation(s)
- Ruth F Dubin
- Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, H5.122E, Dallas, TX 75390, USA
| | - Rajat Deo
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Ren
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jianqiao Wang
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
| | - Josyf C Mychaleckyj
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Julia Kozlitina
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Victoria Arthur
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hongzhe Lee
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amil Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Harold Feldman
- Patient-Centered Outcomes Research Institute, Washington, DC, USA
| | - Nisha Bansal
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Leila Zelnick
- Division of Nephrology, University of Washington Medical Center, Seattle, WA, USA
| | - Panduranga Rao
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Nidhi Sukul
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Dominic S Raj
- Division of Kidney Diseases and Hypertension, George Washington University School of Medicine, Washington, DC, USA
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, USA
| | - Sylvia E Rosas
- Joslin Diabetes Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zeenat Bhat
- Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jiang He
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University, New Orleans, LA, USA
| | - Mayank Kansal
- Division of Cardiology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Vasan S Ramachandran
- University of Texas School of Public Health San Antonio and the University of Texas Health Sciences Center in San Antonio, Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sushrut S Waikar
- Section of Nephrology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Mark R Segal
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Peter Ganz
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
4
|
Adasheva DA, Serebryanaya DV. IGF Signaling in the Heart in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1402-1428. [PMID: 39245453 DOI: 10.1134/s0006297924080042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/22/2024] [Indexed: 09/10/2024]
Abstract
One of the most vital processes of the body is the cardiovascular system's proper operation. Physiological processes in the heart are regulated by the balance of cardioprotective and pathological mechanisms. The insulin-like growth factor system (IGF system, IGF signaling pathway) plays a pivotal role in regulating growth and development of various cells and tissues. In myocardium, the IGF system provides cardioprotective effects as well as participates in pathological processes. This review summarizes recent data on the role of IGF signaling in cardioprotection and pathogenesis of various cardiovascular diseases, as well as analyzes severity of these effects in various scenarios.
Collapse
Affiliation(s)
- Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
5
|
Boxhammer E, Paar V, Kopp K, Gharibeh SX, Bovenkamp-Aberger E, Rezar R, Lichtenauer M, Hoppe UC, Mirna M. Insulin-like Growth Factor-Binding Protein 2 in Severe Aortic Valve Stenosis and Pulmonary Hypertension: A Gender-Based Perspective. Int J Mol Sci 2024; 25:8220. [PMID: 39125788 PMCID: PMC11312253 DOI: 10.3390/ijms25158220] [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: 06/01/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Severe aortic valve stenosis (AS) and pulmonary hypertension (PH) are life-threatening cardiovascular conditions, necessitating early detection and intervention. Recent studies have explored the role of Insulin-like Growth Factor-Binding Protein 2 (IGF-BP2) in cardiovascular pathophysiology. Understanding its involvement may offer novel insights into disease mechanisms and therapeutic targets for these conditions. A total of 102 patients (46 female, 56 male) with severe AS undergoing a transcatheter aortic valve replacement (TAVR) in a single-center study were classified using echocardiography tests to determine systolic pulmonary artery pressure (sPAP) and the presence (sPAP ≥ 40 mmHg) or absence (sPAP < 40 mmHg) of PH. Additionally, serial laboratory determinations of IGF-BP2 before, and at 24 h, 96 h, and 3 months after intervention were conducted in all study participants. Considering the entire cohort, patients with PH had significant and continuously higher serum IGF-BP2 concentrations over time than patients without PH. After subdivision by sex, it could be demonstrated that the above-mentioned results were only verifiable in males, but not in females. In the male patients, baseline IGF-BP2 levels before the TAVR was an isolated risk factor for premature death after intervention and at 1, 3, and 5 years post-intervention. The same was valid for the combination of male and echocardiographically established PH patients. The predictive role of IGF-BP2 in severe AS and concurrent PH remains unknown. A more profound comprehension of IGF-BP2 mechanisms, particularly in males, could facilitate the earlier consideration of the TAVR as a more effective and successful treatment strategy.
Collapse
Affiliation(s)
- Elke Boxhammer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Andrzejczyk K, Abou Kamar S, van Ommen AM, Canto ED, Petersen TB, Valstar G, Akkerhuis KM, Cramer MJ, Umans V, Rutten FH, Teske A, Boersma E, Menken R, van Dalen BM, Hofstra L, Verhaar M, Brugts J, Asselbergs F, den Ruijter H, Kardys I. Identifying plasma proteomic signatures from health to heart failure, across the ejection fraction spectrum. Sci Rep 2024; 14:14871. [PMID: 38937570 PMCID: PMC11211454 DOI: 10.1038/s41598-024-65667-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/23/2024] [Indexed: 06/29/2024] Open
Abstract
Circulating proteins may provide insights into the varying biological mechanisms involved in heart failure (HF) with preserved ejection fraction (HFpEF) and reduced ejection fraction (HFrEF). We aimed to identify specific proteomic patterns for HF, by comparing proteomic profiles across the ejection fraction spectrum. We investigated 4210 circulating proteins in 739 patients with normal (Stage A/Healthy) or elevated (Stage B) filling pressures, HFpEF, or ischemic HFrEF (iHFrEF). We found 2122 differentially expressed proteins between iHFrEF-Stage A/Healthy, 1462 between iHFrEF-HFpEF and 52 between HFpEF-Stage A/Healthy. Of these 52 proteins, 50 were also found in iHFrEF vs. Stage A/Healthy, leaving SLITRK6 and NELL2 expressed in lower levels only in HFpEF. Moreover, 108 proteins, linked to regulation of cell fate commitment, differed only between iHFrEF-HFpEF. Proteomics across the HF spectrum reveals overlap in differentially expressed proteins compared to stage A/Healthy. Multiple proteins are unique for distinguishing iHFrEF from HFpEF, supporting the capacity of proteomics to discern between these conditions.
Collapse
Affiliation(s)
- Karolina Andrzejczyk
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sabrina Abou Kamar
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Anne-Mar van Ommen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elisa Dal Canto
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Teun B Petersen
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gideon Valstar
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Maarten Jan Cramer
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Victor Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands
| | - Frans H Rutten
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arco Teske
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Roxana Menken
- Cardiology Centers of the Netherlands, Utrecht, The Netherlands
| | - Bas M van Dalen
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Cardiology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - Leonard Hofstra
- Cardiology Centers of the Netherlands, Utrecht, The Netherlands
| | - Marianne Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jasper Brugts
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Folkert Asselbergs
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hester den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Clinical Cardiology Department, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Hjortebjerg R, Høgdall C, Hansen KH, Høgdall E, Frystyk J. The IGF-PAPP-A-Stanniocalcin Axis in Serum and Ascites Associates with Prognosis in Patients with Ovarian Cancer. Int J Mol Sci 2024; 25:2014. [PMID: 38396692 PMCID: PMC10888379 DOI: 10.3390/ijms25042014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) and PAPP-A2 modulate insulin-like growth factor (IGF) action and are inhibited by the stanniocalcins (STC1 and STC2). We previously demonstrated increased PAPP-A and IGF activity in ascites from women with ovarian carcinomas. In this prospective, longitudinal study of 107 women with ovarian cancer and ascites accumulation, we determined corresponding serum and ascites levels of IGF-1, IGF-2, PAPP-A, PAPP-A2, STC1, and STC2 and assessed their relationship with mortality. As compared to serum, we found highly increased ascites levels of PAPP-A (51-fold) and PAPP-A2 (4-fold). Elevated levels were also observed for IGF-1 (12%), STC1 (90%) and STC2 (68%). In contrast, IGF-2 was reduced by 29% in ascites. Patients were followed for a median of 38.4 months (range: 45 days to 8.9 years), during which 73 patients (68.2%) died. Overall survival was longer for patients with high serum IGF-1 (hazard ratio (HR) per doubling in protein concentration: 0.60, 95% CI: 0.40-0.90). However, patients with high ascites levels of IGF-1 showed a poorer prognosis (HR: 2.00 (1.26-3.27)). High serum and ascites IGF-2 levels were associated with increased risk of mortality (HR: 2.01 (1.22-3.30) and HR: 1.78 (1.24-2.54), respectively). Similarly, serum PAPP-A2 was associated with mortality (HR: 1.26 (1.08-1.48)). Our findings demonstrate the presence and activity of the IGF system in the local tumor ecosystem, which is likely a characteristic feature of malignant disease and plays a role in its peritoneal dissemination. The potential clinical implications are supported by our finding that serum levels of the proteins are associated with patient prognosis.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark;
| | - Claus Høgdall
- Department of Gynecology, Juliane Marie Center, Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Kristian Horsman Hansen
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital, 5000 Odense, Denmark;
- OPEN Lab, Odense University Hospital, 5000 Odense, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev University Hospital, 2730 Herlev, Denmark;
| | - Jan Frystyk
- Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark;
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital, 5000 Odense, Denmark;
| |
Collapse
|
8
|
Wang W, Ye J, Xu L, Mo DG, Chen C, Li T, Yao HC. The effects of IGF-1 and IGFBP-2 treatments on the atherosclerosis in the aorta and the coronary arteries of the high cholesterol diet-fed rabbits. Int Immunopharmacol 2024; 127:111409. [PMID: 38118312 DOI: 10.1016/j.intimp.2023.111409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/22/2023]
Abstract
Several studies have demonstrated suppression of aortic atherosclerosis by insulin like growth factor-1 (IGF-1) in hypercholesterolemic rabbits. Though a recent study has reported that IGF-1 exerts anti-atherogenic effects in coronary arteries, the mechanisms of IGF-1 in coronary arteries need to be further verified. Studies about insulin like growth factor binding protein-2 (IGFBP-2) in atherosclerosis are rarely. The objective of this study is to examine the effects of IGF-1 and IGFBP-2 on the atherosclerosis development in the aorta and coronary arteries of the high-cholesterol diet (HCD)-fed rabbits. New Zealand white rabbits were fed either normal chow (n = 5) or a diet containing 1.0 % cholesterol (n = 18) for 12 weeks. Cholesterol-fed rabbits were given IGF-1 or IGFBP-2 or saline intravenously (each n = 6) for 10 weeks. The results revealed that IGF-1 decreased total cholesterol (TC) and low-density lipoprotein (LDL) levels (p < 0.05), whereas IGFBP-2 did not. IGF-1 significantly attenuated atherosclerotic lesions and reduced accumulated macrophages within the coronary artery plaques, whereas IGFBP-2 deteriorated these changes. Moreover, IGF-1 reduced serum platelet-activating factor acetylhydrolase levels, C reactive protein (CRP), and inhibited the protein expression levels of tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6). IGFBP-2 elevated serum 8-hydroxy-2'-deoxyguanosine levels, CRP, and promoted the protein expression levels of TNF-α and IL-6. In conclusion, IGF-1 can substantially suppress plaque formation in coronary arteries with a marked inhibition of macrophage accumulation likely via its anti-inflammatory properties, whereas IGFBP-2 plays an opposite effect on atherosclerosis. The present study highlighted a theoretical basis for pharmacological treatment of atherosclerosis.
Collapse
Affiliation(s)
- Wei Wang
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong 252000, China
| | - Jing Ye
- Department of Pathology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Li Xu
- Stem Cell and Regenerative Medicine Laboratory, Liaocheng People's Hospital, Liaocheng 252000, PR China
| | - De-Gang Mo
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong 252000, China
| | - Chen Chen
- Department of Urology, Liaocheng People's Hospital, Liaocheng 252000, PR China
| | - Tai Li
- Department of Nursing, Liaocheng Vocational & Technical College, Liaocheng 252000, China
| | - Heng-Chen Yao
- Department of Cardiology, Liaocheng People's Hospital Affiliated to Shandong First Medical University, Liaocheng, Shandong 252000, China; Department of Cardiology, Liaocheng People's Hospital, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
9
|
Lan T, Zeng Q, Fan Y, Liu T, Yao P, Liang Z, Dang X, Zhu H, Li Y, Jiang W, Lu W. Proteomics Analysis of Serum Reveals Potential Biomarkers for Heart Failure Patients with Phlegm-Blood Stasis Syndrome. J Proteome Res 2024; 23:226-237. [PMID: 38048169 DOI: 10.1021/acs.jproteome.3c00537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Heart failure (HF), a complex clinical syndrome, has become a global burden on health and economics around the world. Phlegm-blood stasis syndrome, one of the Traditional Chinese Medicine (TCM) syndrome differentiation, is the core pathogenesis dynamically throughout the occurrence, development, and prognosis of HF. Biomarkers having high sensitivity and specificity are highly demanded to facilitate the accurate differentiation of HF patients with phlegm-blood stasis syndrome. In the present study, serum samples were collected from 20 healthy controls and 40 HF patients (20 with and 20 without phlegm-blood stasis syndrome). We implemented data-independent acquisition mass spectrometry (DIA-MS) for discovery and parallel reaction monitoring (PRM) for validation of biomarkers for heart failure with phlegm-blood stasis syndrome. A total of 84 different proteins were found in the HF with phlegm-blood stasis syndrome (HF-TY) group compared with healthy controls. 37 candidate proteins were selected for the PRM assay, and five validated proteins with high sensitivity and specificity, including insulin-like growth factor-binding protein 4 (IGFBP4), β-2-microglobulin (B2M), dystroglycan (DAG1), immunoglobulin J chain (JCHAIN), and kallikrein B1 (KLKB1), were considered potential biomarkers for heart failure patients with phlegm-blood stasis syndrome. Newly identified biomarkers might provide insights into the diagnosis and treatment of HF with TCM syndrome differentiation.
Collapse
Affiliation(s)
- Taohua Lan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Qiaohuang Zeng
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Yunxiang Fan
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Tong Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Ping Yao
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Zhaoying Liang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Xiaojing Dang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Huiying Zhu
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Yanfen Li
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Wei Jiang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510020, China
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
| | - Weihui Lu
- Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment of Refractory Chronic Diseases, Guangzhou 510020, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510020, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510020, P. R. China
| |
Collapse
|
10
|
Hjortebjerg R, Kristiansen MR, Brandslund I, Aa Olsen D, Stidsen JV, Nielsen JS, Frystyk J. Associations between insulin-like growth factor binding protein-2 and insulin sensitivity, metformin, and mortality in persons with T2D. Diabetes Res Clin Pract 2023; 205:110977. [PMID: 37890435 DOI: 10.1016/j.diabres.2023.110977] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
AIMS Serum insulin-like growth factor binding protein-2 (IGFBP-2) is low in persons with type 2 diabetes mellitus (T2D) and possibly regulated by metformin. Counter-intuitively, high IGFBP-2 associates with mortality. We investigated the association between IGFBP-2, metformin-treatment, and indices of insulin sensitivity, and assessed IGFBP-2 in relation to prior comorbidity and mortality during five-year follow-up. METHODS The study included 859 treatment-naive and 558 metformin-treated persons enrolled in the Danish Centre for Strategic Research in T2D and followed for 4.9 (3.9-5.9) years through national health registries. All proteins were determined in serum collected at enrollment. RESULTS Following adjustment for age, metformin-treated and treatment-naive persons has similar IGFBP-2 levels. Low IGFBP-2 level was associated with increased BMI, fasting glucose, and C-peptide. IGFBP-2 was higher in the 437 persons who had comorbidities at enrollment than in those with T2D only (343 (213;528) vs. 242 (169;378) ng/mL). During follow-up, 87 persons died, and IGFBP-2 predicted mortality with an unadjusted HR (95% CI) per doubling in IGFBP-2 concentration of 2.62 (2.04;3.37) and a HR of 2.21 (1.61;3.01) following full adjustment. CONCLUSIONS In T2D, high IGFBP-2 associates with low glucose and insulin secretion, is unaffected by metformin treatment, and associates with risk of prior comorbidity and mortality.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Denmark; Endocrine Research Unit, Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Denmark.
| | - Maja R Kristiansen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark; Danish Centre for Strategic Research in Type 2 Diabetes (DD2), Odense, Denmark
| | - Ivan Brandslund
- Department of Biochemistry and Immunology, University Hospital of Southern Denmark, Vejle, Denmark
| | - Dorte Aa Olsen
- Department of Biochemistry and Immunology, University Hospital of Southern Denmark, Vejle, Denmark
| | - Jacob V Stidsen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark; Endocrine Research Unit, Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Denmark
| | - Jens S Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Denmark; Danish Centre for Strategic Research in Type 2 Diabetes (DD2), Odense, Denmark
| | - Jan Frystyk
- Department of Clinical Research, University of Southern Denmark, Denmark; Endocrine Research Unit, Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Denmark
| |
Collapse
|
11
|
Mohebi R, Liu Y, Hansen MK, Yavin Y, Sattar N, Pollock CA, Butler J, Jardine M, Masson S, Heerspink HJL, Januzzi JL. Insulin growth factor axis and cardio-renal risk in diabetic kidney disease: an analysis from the CREDENCE trial. Cardiovasc Diabetol 2023; 22:176. [PMID: 37438734 DOI: 10.1186/s12933-023-01916-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The insulin-like growth factors (IGF) play a crucial role in regulating cellular proliferation, apoptosis, and key metabolic pathways. The ratio of IGF-1 to IGF binding protein-3 (IGFBP-3) is an important factor in determining IGF-1 bioactivity. We sought to investigate the association of IGF-1 and IGFBP-3 with cardio-renal outcomes among persons with type 2 diabetes. METHODS Samples were available from 2627 individuals with type 2 diabetes and chronic kidney disease that were randomized to receive canagliflozin or placebo and were followed up for incident cardio-renal events. Primary outcome was defined as a composite of end-stage kidney disease, doubling of the serum creatinine level, or renal/cardiovascular death. IGF-1 and IGFBP-3 were measured at baseline, Year-1 and Year-3. Elevated IGF-1 level was defined according to age-specific cutoffs. Cox proportional hazard regression was used to investigate the association between IGF-1 level, IGFBP-3, and the ratio of IGF-1/IGFBP-3 with clinical outcomes. RESULTS Elevated IGF-1 was associated with lower glomerular filtration rate at baseline. Treatment with canagliflozin did not significantly change IGF-1 and IGFBP-3 concentrations by 3 years (p-value > 0.05). In multivariable models, elevated IGF-1 (above vs below age-specific cutoffs) was associated with the primary composite outcome (incidence rate:17.8% vs. 12.7% with a hazard ratio [HR]: 1.52; 95% confidence interval CI 1.09-2.13;P: 0.01), renal composite outcome (HR: 1.65; 95% CI 1.14-2.41; P: 0.01), and all-cause mortality (HR: 1.52; 95% CI 1.00-2.32; P; 0.05). Elevations in log IGFBP-3 did not associate with any clinical outcomes. Increase in log IGF-1/IGFBP-3 ratio was also associated with a higher risk of the primary composite outcome (HR per unit increase: 1.57; 95% CI 1.09-2.26; P; 0.01). CONCLUSIONS These results further suggest potential importance of IGF biology in the risk for cardio-renal outcomes in type 2 diabetes. SGLT2 inhibition has no impact on the biology of IGF despite its significant influence on outcomes. TRIAL REGISTRATION CREDENCE; ClinicalTrials.gov Identifier: NCT02065791.
Collapse
Affiliation(s)
- Reza Mohebi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Yuxi Liu
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Michael K Hansen
- Janssen Research Development, LLC, Spring House, Montgomery, PA, USA
| | - Yshai Yavin
- Janssen Research Development, LLC, Spring House, Montgomery, PA, USA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Carol A Pollock
- Kolling Institute, Royal North Shore Hospital University of Sydney, Sydney, NSW, Australia
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, MS, USA
- Baylor Scott & White Institute, Dallas, TX, USA
| | - Meg Jardine
- The George Institute for Global Health, UNSW Sydney, Sydney, NSW, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Concord Repatriation General Hospital, Sydney, NSW, Australia
| | - Serge Masson
- Roche Diagnostics International, Rotkreuz, Switzerland
| | - Hiddo J L Heerspink
- Department Clinical Pharmacy and Pharmacology, University of Groningen, Groningen, The Netherlands
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Heart Failure and Biomarker Trials, Baim Institute for Clinical Research, Boston, MA, USA.
| |
Collapse
|
12
|
Estrada RA, Sahay S. Insulin-like growth factor binding protein-4: The quest for the breakthrough biomarker in pulmonary arterial hypertension. Pulm Circ 2023; 13:e12252. [PMID: 37346965 PMCID: PMC10281014 DOI: 10.1002/pul2.12252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Affiliation(s)
- Rodolfo A. Estrada
- Division of Pulmonary Diseases and Critical Care MedicineUT HealthSan AntonioTexasUSA
| | - Sandeep Sahay
- Division of Pulmonary, Critical Care & Sleep MedicineHouston Methodist HospitalHoustonTexasUSA
| |
Collapse
|
13
|
Kim IK, Song BW, Lim S, Kim SW, Lee S. The Role of Epicardial Adipose Tissue-Derived MicroRNAs in the Regulation of Cardiovascular Disease: A Narrative Review. BIOLOGY 2023; 12:498. [PMID: 37106699 PMCID: PMC10135702 DOI: 10.3390/biology12040498] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Cardiovascular diseases have been leading cause of death worldwide for many decades, and obesity has been acknowledged as a risk factor for cardiovascular diseases. In the present review, human epicardial adipose tissue-derived miRNAs reported to be differentially expressed under pathologic conditions are discussed and summarized. The results of the literature review indicate that some of the epicardial adipose tissue-derived miRNAs are believed to be cardioprotective, while some others show quite the opposite effects depending on the underlying pathologic conditions. Furthermore, they suggest that that the epicardial adipose tissue-derived miRNAs have great potential as both a diagnostic and therapeutic modality. Nevertheless, mainly due to highly limited availability of human samples, it is very difficult to make any generalized claims on a given miRNA in terms of its overall impact on the cardiovascular system. Therefore, further functional investigation of a given miRNA including, but not limited to, the study of its dose effect, off-target effects, and potential toxicity is required. We hope that this review can provide novel insights to transform our current knowledge on epicardial adipose tissue-derived miRNAs into clinically viable therapeutic strategies for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Sang-Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| |
Collapse
|
14
|
Macvanin M, Gluvic Z, Radovanovic J, Essack M, Gao X, Isenovic ER. New insights on the cardiovascular effects of IGF-1. Front Endocrinol (Lausanne) 2023; 14:1142644. [PMID: 36843588 PMCID: PMC9947133 DOI: 10.3389/fendo.2023.1142644] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
INTRODUCTION Cardiovascular (CV) disorders are steadily increasing, making them the world's most prevalent health issue. New research highlights the importance of insulin-like growth factor 1 (IGF-1) for maintaining CV health. METHODS We searched PubMed and MEDLINE for English and non-English articles with English abstracts published between 1957 (when the first report on IGF-1 identification was published) and 2022. The top search terms were: IGF-1, cardiovascular disease, IGF-1 receptors, IGF-1 and microRNAs, therapeutic interventions with IGF-1, IGF-1 and diabetes, IGF-1 and cardiovascular disease. The search retrieved original peer-reviewed articles, which were further analyzed, focusing on the role of IGF-1 in pathophysiological conditions. We specifically focused on including the most recent findings published in the past five years. RESULTS IGF-1, an anabolic growth factor, regulates cell division, proliferation, and survival. In addition to its well-known growth-promoting and metabolic effects, there is mounting evidence that IGF-1 plays a specialized role in the complex activities that underpin CV function. IGF-1 promotes cardiac development and improves cardiac output, stroke volume, contractility, and ejection fraction. Furthermore, IGF-1 mediates many growth hormones (GH) actions. IGF-1 stimulates contractility and tissue remodeling in humans to improve heart function after myocardial infarction. IGF-1 also improves the lipid profile, lowers insulin levels, increases insulin sensitivity, and promotes glucose metabolism. These findings point to the intriguing medicinal potential of IGF-1. Human studies associate low serum levels of free or total IGF-1 with an increased risk of CV and cerebrovascular illness. Extensive human trials are being conducted to investigate the therapeutic efficacy and outcomes of IGF-1-related therapy. DISCUSSION We anticipate the development of novel IGF-1-related therapy with minimal side effects. This review discusses recent findings on the role of IGF-1 in the cardiovascular (CVD) system, including both normal and pathological conditions. We also discuss progress in therapeutic interventions aimed at targeting the IGF axis and provide insights into the epigenetic regulation of IGF-1 mediated by microRNAs.
Collapse
Affiliation(s)
- Mirjana Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
- *Correspondence: Mirjana Macvanin,
| | - Zoran Gluvic
- Clinic for Internal Medicine, Department of Endocrinology and Diabetes, Zemun Clinical Hospital, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Magbubah Essack
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
15
|
Jiang Y, Yan X, Yi S, Hu Z. Evaluation of Potential Biomarkers for Traditional Chinese Medicine-Based Chronic Heart Failure Syndromes; a case-control study. Eur J Integr Med 2022. [DOI: 10.1016/j.eujim.2022.102222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
16
|
Dai D, Cheng Z, Feng S, Zhu Z, Yu J, Zhang W, Lu H, Zhang R, Zhu J. Quantitative Data-Independent Acquisition Mass Spectrometry Proteomics and Weighted Correlation Network Analysis of Plasma Samples for the Discovery of Chronic Kidney Disease-Specific Atherosclerosis Risk Factors. DNA Cell Biol 2022; 41:966-980. [PMID: 36255451 DOI: 10.1089/dna.2022.0200] [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: 11/13/2022] Open
Abstract
Chronic kidney disease (CKD) accelerates atherosclerosis. The mechanism of CKD-related atherosclerosis is complex, and CKD-specific risk factors may contribute to this process in addition to traditional risk factors such as hypertension, diabetes, and hypercholesterolemia. In the present study, to discover CKD-specific atherosclerosis risk factors, a total of 62 patients with different stages of kidney function were enrolled. All patients underwent coronary angiographies and the severity of coronary atherosclerosis was defined by the SYNTAX score. Patients were divided into different groups according to their kidney function levels and coronary atherosclerosis severity. Data-independent acquisition mass spectrometry was used to identify differentially expressed proteins (DEPs) in the plasma samples, and weighted correlation network analysis (WGCNA) was employed to identify significant protein modules and hub proteins related to CKD-specific atherosclerosis. The results showed that 10 DEPs associated with atherosclerosis were found in the comparative groups with modest and severe CKD. Through WGCNA, 1768 proteins were identified and 8 protein modules were established. Enrichment analyses of protein modules revealed functional clusters mainly associated with inflammation and the complement and coagulation cascade as atherosclerosis developed under CKD conditions. The results may help to better understand the mechanisms of CKD-related atherosclerosis and guide future research on developing treatments for CKD-related atherosclerosis.
Collapse
Affiliation(s)
- Daopeng Dai
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwei Cheng
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics and Data Science, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Shuo Feng
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiwei Yu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenli Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Lu
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics and Data Science, College of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinzhou Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
CT-IGFBP-4 as a Predictive Novel Biomarker of Ischemic Cardiovascular Events and Mortality: A Systematic Review. J Interv Cardiol 2022; 2022:1816504. [PMID: 36051380 PMCID: PMC9420648 DOI: 10.1155/2022/1816504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Methods The electronic databases PubMed, medRxiv, ScienceDirect, and Google Scholar were searched for relevant literature from inception to the 10th of December, 2021. Thus, retrieved literature was screened by title and abstract, followed by full-text screening based on the eligibility criteria. The risk of bias was accessed using the quality in prognostic studies (QUIPSs) tool. The data on cardiovascular outcomes about CT-IGFBP-4 levels were studied and the results were synthesized. Results Five studies with a total of 1,417 participants were included in our study. The studies reported a low risk of bias. The mean age of the participants was 66.14 and more than 65% were males. Elevated CT-IGFBP-4 levels were associated with poor cardiovascular outcomes and increased mortality in severely ill patients. In contrast, there were no significant findings in the case of stable patients. Sandwich ELISA using lithium-heparin plasma provided a better detection limit of 0.15 ng/ml, low cross-reactivity (<2%), and generated linear results between 12 and 500 ng/ml. Conclusion CT-IGFBP-4 is an efficient biomarker for the prediction of MACE and mortality in patients with severe ischemic cardiovascular events.
Collapse
|
18
|
Frøbert AM, Brohus M, Roesen TS, Kindberg J, Fröbert O, Conover CA, Overgaard MT. Circulating insulin-like growth factor system adaptations in hibernating brown bears indicate increased tissue IGF availability. Am J Physiol Endocrinol Metab 2022; 323:E307-E318. [PMID: 35830688 DOI: 10.1152/ajpendo.00429.2021] [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] [Indexed: 11/22/2022]
Abstract
Brown bears conserve muscle and bone mass during 6 mo of inactive hibernation. The molecular mechanisms underlying hibernation physiology may have translational relevance for human therapeutics. We hypothesize that protective mechanisms involve increased tissue availability of insulin-like growth factors (IGFs). In subadult Scandinavian brown bears, we observed that mean plasma IGF-1 and IGF-2 levels during hibernation were reduced to 36 ± 10% and 56 ± 15%, respectively, compared with the active state (n = 12). Western ligand blotting identified IGF-binding protein (IGFBP)-3 as the major IGFBP in the active state, whereas IGFBP-2 was codominant during hibernation. Acid labile subunit (ALS) levels in hibernation were reduced to 41±16% compared with the active state (n = 6). Analysis of available grizzly bear RNA sequencing data revealed unaltered liver mRNA IGF-1, IGFBP-2, and IGFBP-3 levels, whereas ALS levels were significantly reduced during hibernation (n = 6). Reduced ALS synthesis and circulating levels during hibernation should prompt a shift from ternary IGF/IGFBP/ALS to smaller binary IGF/IGFBP complexes, thereby increasing IGF tissue availability. Indeed, size-exclusion chromatography of bear plasma demonstrated a shift to lower molecular weight IGF-containing complexes in the hibernating versus the active state. Furthermore, we note that the major IGF-2 mRNA isoform expressed in livers in both Scandinavian brown bears and grizzly bears was an alternative splice variant in which Ser29 is replaced with a tetrapeptide possessing a positively charged Arg residue. Homology modeling of the bear IGF-2/IGFBP-2 complex showed the tetrapeptide in proximity to the heparin-binding domain involved in bone-specific targeting of this complex. In conclusion, this study provides data which suggest that increased IGF tissue availability combined with tissue-specific targeting contribute to tissue preservation in hibernating bears.NEW & NOTEWORTHY Brown bears shift from circulating ternary IGF/IGFBP/ALS complexes in the active state to binary IGF/IGFBP complexes during hibernation, indicating increased tissue IGF-bioactivity. Furthermore, brown bears use a splice variant of IGF-2, suggesting increased bone-specific targeting of IGF anabolic signaling.
Collapse
Affiliation(s)
- Anne Mette Frøbert
- Department of Chemistry and Bioscience, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
| | - Malene Brohus
- Department of Chemistry and Bioscience, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
| | - Tinna S Roesen
- Department of Chemistry and Bioscience, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
| | - Jonas Kindberg
- Department of Wildlife, Fish and Environmental Studies, Swedish University of Agricultural Sciences, Umeå, Sweden
- Norwegian Institute for Nature Research, Trondheim, Norway
| | - Ole Fröbert
- Department of Cardiology, Faculty of Health, Örebro University, Örebro, Sweden
- Department of Clinical Medicine, Aarhus University Health, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Cheryl A Conover
- Division of Endocrinology, Metabolism, and Nutrition, Mayo Clinic, Rochester, Minnesota
| | - Michael T Overgaard
- Department of Chemistry and Bioscience, Faculty of Engineering and Science, Aalborg University, Aalborg, Denmark
| |
Collapse
|
19
|
Jin B, Bai X, Yu K, Han L, Wang N, Han W. Reduced IGFBP-2 related immunoreactivity in human serum correlates with arterial stiffness in a healthy Chinese population. Microvasc Res 2022; 144:104423. [PMID: 35995234 DOI: 10.1016/j.mvr.2022.104423] [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: 04/10/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Circulating insulin-like growth factor binding protein 2 (IGFBP-2) is associated with metabolic changes in both physiological and pathological conditions. The aim of this study was to investigate the correlation between IGFBP-2 related immunoreactivity in serum and arterial stiffness in a healthy Chinese population. METHODS In this cross-sectional study, 360 healthy participants aged 37-87 years were recruited from 1500 and were divided into three groups according to serum IGFBP-2 related immunoreactivity (Tertile I, 25.437 ng/ml-120.870 ng/ml; Tertile II, 120.871 ng/ml-161.914 ng/ml; Tertile III, 161.915 ng/ml-321.636 ng/ml). Arterial stiffness was evaluated by measuring the brachial-ankle pulse wave velocity (baPWV), ankle-brachial index (ABI), and carotid intima-media thickness (cIMT). The association between IGFBP-2 related immunoreactivity and arterial stiffness was estimated by multiple stepwise regression. RESULTS Compared with the other two groups population, the individuals in Tertile I had significantly older age (62.66 ± 13.30 years, P < 0.01), lower level of triglyceride (1.08 ± 0.70 mmol/l, P < 0.01) and E/A (peak velocity of early filling and preak velocity of atrial filling ratio) (0.90 ± 0.33, P < 0.05). IGFBP-2 related immunoreactivity was inversely related with baPWV in the total population (r = -0.171, P < 0.01) and in Tertile I (r = -0.275, P < 0.01). After adjusting for age and the other confounders, no association was found between IGFBP-2 related immunoreactivity and baPWV in the total population. However, In Tertile I, reduced IGFBP-2 related immunoreactivity in serum was an independent risk factor of baPWV acceleration in three different adjustment models: Model 1 (no adjustment, P < 0.01), Model 2 (adjusted for age, P < 0.05), and Model 3 (adjusted for all variables, P < 0.05). CONCLUSION IGFBP-2 related immunoreactivity in serum is inversely associated with baPWV in a healthy Chinese population. This association did not change after adjustment for conventional risk factors for cardiovascular diseases in the subjects with the lowest IGFBP-2 related immunoreactivity. Consequently, reduction of IGFBP-2 related immunoreactivity may be a predictor of arterial stiffness. IGFBP-2 seems to be a potential intervention target in early atherosclerosis.
Collapse
Affiliation(s)
- Bo Jin
- Department of Gerontology and Geriatrics, The Second Hospital of Dalian Medical University, Dalian, China; Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaojuan Bai
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Kai Yu
- Department of General Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Lulu Han
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Nan Wang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wen Han
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Saleh S, George J, Kott KA, Meikle PJ, Figtree GA. The Translation and Commercialisation of Biomarkers for Cardiovascular Disease—A Review. Front Cardiovasc Med 2022; 9:897106. [PMID: 35722087 PMCID: PMC9201254 DOI: 10.3389/fcvm.2022.897106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 11/19/2022] Open
Abstract
As a leading cause of mortality and morbidity worldwide, cardiovascular disease and its diagnosis, quantification, and stratification remain significant health issues. Increasingly, patients present with cardiovascular disease in the absence of known risk factors, suggesting the presence of yet unrecognized pathological processes and disease predispositions. Fortunately, a host of emerging cardiovascular biomarkers characterizing and quantifying ischaemic heart disease have shown great promise in both laboratory settings and clinical trials. These have demonstrated improved predictive value additional to widely accepted biomarkers as well as providing insight into molecular phenotypes beneath the broad umbrella of cardiovascular disease that may allow for further personalized treatment regimens. However, the process of translation into clinical practice – particularly navigating the legal and commercial landscape – poses a number of challenges. Practical and legal barriers to the biomarker translational pipeline must be further considered to develop strategies to bring novel biomarkers into the clinical sphere and apply these advances at the patient bedside. Here we review the progress of emerging biomarkers in the cardiovascular space, with particular focus on those relevant to the unmet needs in ischaemic heart disease.
Collapse
Affiliation(s)
- Soloman Saleh
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jacob George
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Katharine A. Kott
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Gemma A. Figtree
| |
Collapse
|
21
|
Topf A, Mirna M, Paar V, Motloch LJ, Bacher N, Franz M, Hoppe UC, Kretzschmar D, Lichtenauer M. Differential Diagnosis between Takotsubo Syndrome and Acute Coronary Syndrome—A Prospective Analysis of Novel Cardiovascular Biomarkers for a More Selective Triage. J Clin Med 2022; 11:jcm11112974. [PMID: 35683362 PMCID: PMC9180967 DOI: 10.3390/jcm11112974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 12/11/2022] Open
Abstract
Introduction: Takotsubo syndrome (TTS) is clinically indistinguishable from an ACS. Despite the implementation of clinical scoring systems and novel biomarkers, coronary angiography currently remains necessary for differential diagnosis. Methods: 93 patients with chest pain and the suspicion of TTS were enrolled in two study centers. Fetuin-A, IGFBP-2, Galectin-3, and TNF α were determined in serum samples, collected within 24 h after the onset of symptoms. Serum levels of biomarkers were analyzed for the differential diagnostic value between TTS and ACS. Results: Compared to TTS, patients with ACS had significantly lower serum levels of Fetuin-A and IGFBP-2. The cut-off value of Fetuin-A for the identification of TTS compared to ACS was 55.74 μg/mL (sensitivity: 100.0%, specificity: 82.6%, PPV: 63.2%, NPV: 100.0%). An optimal cut-off value for IGFBP-2 for the differential diagnosis between TTS and ACS was determined as 171.77 ng/mL (sensitivity: 76.0%, specificity: 82.6%, PPV: 76.4%, NPV 72.7%). Conclusion: Fetuin-A and IGFBP-2 might facilitate the triage between TTS and ACS and could be therefore of great benefit for the guidance of treatment.
Collapse
Affiliation(s)
- Albert Topf
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
- Correspondence: ; Tel.: +43-(0)-57-57418; Fax: +43-(0)-57255-4111
| | - Moritz Mirna
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Vera Paar
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Lukas J. Motloch
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Nina Bacher
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Marcus Franz
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, 07743 Jena, Germany; (M.F.); (D.K.)
| | - Uta C. Hoppe
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, 07743 Jena, Germany; (M.F.); (D.K.)
| | - Michael Lichtenauer
- Clinic for Internal Medicine II, University Hospital Salzburg, Paracelsus University Salzburg, Müllner Hauptstraße 48, A-5020 Salzburg, Austria; (M.M.); (V.P.); (L.J.M.); (N.B.); (U.C.H.); (M.L.)
| |
Collapse
|
22
|
Klimczak-Tomaniak D, de Bakker M, Bouwens E, Akkerhuis KM, Baart S, Rizopoulos D, Mouthaan H, van Ramshorst J, Germans T, Constantinescu A, Manintveld O, Umans V, Boersma E, Kardys I. Dynamic personalized risk prediction in chronic heart failure patients: a longitudinal, clinical investigation of 92 biomarkers (Bio-SHiFT study). Sci Rep 2022; 12:2795. [PMID: 35181700 PMCID: PMC8857321 DOI: 10.1038/s41598-022-06698-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 02/02/2022] [Indexed: 11/28/2022] Open
Abstract
The aim of our observational study was to derive a small set out of 92 repeatedly measured biomarkers with optimal predictive capacity for adverse clinical events in heart failure, which could be used for dynamic, individual risk assessment in clinical practice. In 250 chronic HFrEF (CHF) patients, we collected trimonthly blood samples during a median of 2.2 years. We selected 537 samples for repeated measurement of 92 biomarkers with the Cardiovascular Panel III (Olink Proteomics AB). We applied Least Absolute Shrinkage and Selection Operator (LASSO) penalization to select the optimal set of predictors of the primary endpoint (PE). The association between repeatedly measured levels of selected biomarkers and the PE was evaluated by multivariable joint models (mvJM) with stratified fivefold cross validation of the area under the curve (cvAUC). The PE occurred in 66(27%) patients. The optimal set of biomarkers selected by LASSO included 9 proteins: NT-proBNP, ST2, vWF, FABP4, IGFBP-1, PAI-1, PON-3, transferrin receptor protein-1, and chitotriosidase-1, that yielded a cvAUC of 0.88, outperforming the discriminative ability of models consisting of standard biomarkers (NT-proBNP, hs-TnT, eGFR clinically adjusted) − 0.82 and performing equally well as an extended literature-based set of acknowledged biomarkers (NT-proBNP, hs-TnT, hs-CRP, GDF-15, ST2, PAI-1, Galectin 3) − 0.88. Nine out of 92 serially measured circulating proteins provided a multivariable model for adverse clinical events in CHF patients with high discriminative ability. These proteins reflect wall stress, remodelling, endothelial dysfunction, iron deficiency, haemostasis/fibrinolysis and innate immunity activation. A panel containing these proteins could contribute to dynamic, personalized risk assessment. Clinical Trial Registration: 10/05/2013 https://clinicaltrials.gov/ct2/show/NCT01851538?term=nCT01851538&draw=2&rank=1.
Collapse
Affiliation(s)
- Dominika Klimczak-Tomaniak
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Cardiology, Hypertension and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Marie de Bakker
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Elke Bouwens
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - K Martijn Akkerhuis
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Sara Baart
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Jan van Ramshorst
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Tjeerd Germans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Alina Constantinescu
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Olivier Manintveld
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Victor Umans
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Room NA-316, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
23
|
Haddad F, Ataam JA, Amsallem M, Cauwenberghs N, Kuznetsova T, Rosenberg-Hasson Y, Zamanian RT, Karakikes I, Horne BD, Muhlestein JB, Kwee L, Shah S, Maecker H, Knight S, Knowlton K. Insulin Growth Factor Phenotypes in Heart Failure with Preserved Ejection Fraction, an INSPIRE Registry and CATHGEN Study: IGF axis in HFpEF. J Card Fail 2021; 28:935-946. [PMID: 34979242 DOI: 10.1016/j.cardfail.2021.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 10/04/2021] [Accepted: 12/15/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND The insulin like growth factor (IGF) axis emerged as an important pathway in heart failure with preserved ejection (HFpEF). We aimed to identify IGF phenotypes associated with HFpEF in the context high-dimensional proteomic profiling. METHODS From the Intermountain INSPIRE Registry, we identified 96 patients with HFpEF and matched controls. We performed targeted proteomics including IGF-1,2, IGF binding proteins (IGFBP) 1-7 and 111 other proteins (EMD Millipore and ELISA). We used partial least square discriminant analysis (PLS-DA) to identify a set of proteins associated with prevalent HFpEF, pulmonary hypertension (PH) and 5-year-all-cause mortality. K-mean clustering was used to identify IGF phenotypes. RESULTS Patients with HFpEF had a high prevalence of systemic hypertension (95%) and coronary artery disease (74%). Using PLS-DA, we identified a set of biomarkers including IGF1,2 and IGFBP-1,2,7 that provided a strong discrimination of HFPEF, PH and mortality with an AUC of 0.91, 0.77 and 0.83, respectively. Using K mean clustering, we identified three IGF phenotypes that were independently associated with all-cause 5-year mortality after adjustment for age, NT-proBNP and kidney disease (p=0.004). Multivariable analysis validated the prognostic value of IGFBP-1 and 2 in the CATHGEN biorepository. CONCLUSION IGF phenotypes were associated with PH and mortality in HFpEF.
Collapse
Affiliation(s)
- Francois Haddad
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Vera Moulton Wall Center at Stanford, Stanford University School of Medicine, Stanford, CA, USA.
| | - Jennifer Arthur Ataam
- Division of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Myriam Amsallem
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Vera Moulton Wall Center at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicholas Cauwenberghs
- Research Unit of Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Tatiana Kuznetsova
- Research Unit of Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Yael Rosenberg-Hasson
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Roham T Zamanian
- Vera Moulton Wall Center at Stanford, Stanford University School of Medicine, Stanford, CA, USA
| | - Ioannis Karakikes
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin D Horne
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Intermountain Medical Center, Heart Institute, Salt Lake City, UT, USA
| | | | - Lydia Kwee
- Department of Internal Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Svati Shah
- Department of Internal Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina and Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Stacey Knight
- Intermountain Medical Center, Heart Institute, Salt Lake City, UT, USA
| | - Kirk Knowlton
- Intermountain Medical Center, Heart Institute, Salt Lake City, UT, USA
| |
Collapse
|
24
|
Tantawy M, Chekka LM, Huang Y, Garrett TJ, Singh S, Shah CP, Cornell RF, Baz RC, Fradley MG, Waheed N, DeRemer DL, Yuan L, Langaee T, March K, Pepine CJ, Moreb JS, Gong Y. Lactate Dehydrogenase B and Pyruvate Oxidation Pathway Associated With Carfilzomib-Related Cardiotoxicity in Multiple Myeloma Patients: Result of a Multi-Omics Integrative Analysis. Front Cardiovasc Med 2021; 8:645122. [PMID: 33996940 PMCID: PMC8116486 DOI: 10.3389/fcvm.2021.645122] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/02/2021] [Indexed: 01/20/2023] Open
Abstract
Multiple myeloma (MM) is the second most frequent hematologic cancer in the United States. Carfilzomib (CFZ), an irreversible proteasome inhibitor being used to treat relapsed and refractory MM, has been associated with cardiotoxicity, including heart failure. We hypothesized that a multi-omics approach integrating data from different omics would provide insights into the mechanisms of CFZ-related cardiovascular adverse events (CVAEs). Plasma samples were collected from 13 MM patients treated with CFZ (including 7 with CVAEs and 6 with no CVAEs) at the University of Florida Health Cancer Center. These samples were evaluated in global metabolomic profiling, global proteomic profiling, and microRNA (miRNA) profiling. Integrative pathway analysis was performed to identify genes and pathways differentially expressed between patients with and without CVAEs. The proteomics analysis identified the up-regulation of lactate dehydrogenase B (LDHB) [fold change (FC) = 8.2, p = 0.01] in patients who experienced CVAEs. The metabolomics analysis identified lower plasma abundance of pyruvate (FC = 0.16, p = 0.0004) and higher abundance of lactate (FC = 2.4, p = 0.0001) in patients with CVAEs. Differential expression analysis of miRNAs profiling identified mir-146b to be up-regulatein (FC = 14, p = 0.046) in patients with CVAE. Pathway analysis suggested that the pyruvate fermentation to lactate pathway is associated with CFZ-CVAEs. In this pilot multi-omics integrative analysis, we observed the down-regulation of pyruvate and up-regulation of LDHB among patients who experienced CVAEs, suggesting the importance of the pyruvate oxidation pathway associated with mitochondrial dysfunction. Validation and further investigation in a larger independent cohort are warranted to better understand the mechanisms of CFZ-CVAEs.
Collapse
Affiliation(s)
- Marwa Tantawy
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Lakshmi Manasa Chekka
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Yimei Huang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Timothy J Garrett
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Sonal Singh
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Chintan P Shah
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Preston Research Building, Nashville, TN, United States
| | - Rachid C Baz
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Nida Waheed
- Department of Internal Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | | | - Lihui Yuan
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Taimour Langaee
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Keith March
- Division of Cardiovascular Medicine, Department of Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, United States
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine and Center for Regenerative Medicine, University of Florida, Gainesville, FL, United States
| | - Jan S Moreb
- Novant Health Forsyth Medical Center, Hematology, Transplantation, and Cellular Therapy Division, Winston-Salem, NC, United States
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL, United States.,UF Health Cancer Center, Gainesville, FL, United States.,Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL, United States
| |
Collapse
|
25
|
Ibrahim NE, Afilalo M, Chen-Tournoux A, Christenson RH, Gaggin HK, Hollander JE, Kastner P, Levy PD, Mang A, Masson S, Nagurney JT, Nowak RM, Pang PS, Peacock WF, Dipl-Stat VR, Walters EL, Januzzi JL. Diagnostic and Prognostic Utilities of Insulin-Like Growth Factor Binding Protein-7 in Patients With Dyspnea. JACC-HEART FAILURE 2021; 8:415-422. [PMID: 32354416 DOI: 10.1016/j.jchf.2020.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/31/2020] [Accepted: 02/19/2020] [Indexed: 12/29/2022]
Abstract
OBJECTIVES This study examined whether insulin-like growth factor binding protein-7 (IGFBP7) would aid in the diagnosis and prognosis of acute heart failure (HF) beyond N-terminal pro-B-type natriuretic peptide (NT-proBNP) concentration. BACKGROUND IGFBP7 is associated with impaired ventricular relaxation and worse prognosis. METHODS The ICON-RELOADED (International Collaborative of NT-proBNP-Re-evaluation of Acute Diagnostic Cut-Offs in the Emergency Department) study was a prospective, multicenter clinical trial that enrolled subjects presenting with dyspnea. Six-month prognosis for death or repeat hospitalization was obtained. RESULTS Among 1,449 patients, 274 (18.9%) were diagnosed with acute HF. Those with IGFBP7 concentrations in the highest quartile were older, male, had hypertension and HF, had lower estimated glomerular filtration rate (eGFR) and lowest ejection fraction (41 ± 20%; all p < 0.001). Independent predictors of IGFBP7 were age, male sex, history of diabetes, history of HF, and eGFR. Median concentrations of NT-proBNP (2,844 ng/ml vs. 99 ng/ml) and IGFBP7 (146.1 ng/ml vs. 86.1 ng/ml) were higher in those with acute HF (both; p < 0.001). Addition of IGFBP7 to NT-proBNP concentrations improved discrimination, therefore increasing the area under the receiver operating curve for diagnosis of acute HF (from 0.91 to 0.94; p < 0.001 for differences). Addition of IGFBP7 to a complete model of independent predictors of acute HF improved model calibration. IGFBP7 significantly reclassified acute HF diagnosis beyond NT-proBNP (net reclassification index: +0.25). Higher log2-IGFBP7 concentrations in patients with acute HF predicted death or rehospitalization at 6 months (hazard ratio: 1.84 per log2-SD; 95% confidence interval: 1.30 to 2.61; p = 0.001). In Kaplan-Meier analyses, supramedian concentrations of IGFBP7 were associated with shorter event-free survival (log-rank: p < 0.001). CONCLUSIONS Among patients with acute dyspnea, concentrations of IGFBP7 add to NT-proBNP for diagnosis of acute HF and provide added prognostic utility for short-term risk.
Collapse
Affiliation(s)
- Nasrien E Ibrahim
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Marc Afilalo
- Department of Emergency Medicine, McGill University and Emergency Department, Jewish General Hospital, Montreal, Quebec, Canada
| | | | - Robert H Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hanna K Gaggin
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Judd E Hollander
- Department of Emergency Medicine, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | - Phillip D Levy
- Department of Emergency Medicine, Wayne State University, Detroit, Michigan
| | | | | | - John T Nagurney
- Harvard Medical School, Boston, Massachusetts; Department of Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard M Nowak
- Department of Emergency Medicine, Wayne State University, Detroit, Michigan; Department of Emergency Medicine, University of Michigan, Ann Arbor, Michigan
| | - Peter S Pang
- Department of Emergency Medicine and Cardiology Division, Indiana University School of Medicine, Indianapolis, Indiana
| | - W Frank Peacock
- Department of Emergency Medicine, Baylor College of Medicine, Houston, Texas
| | | | - E Lea Walters
- Department of Emergency Medicine, Loma Linda University School of Medicine, Loma Linda, California
| | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Baim Institute for Clinical Research, Boston, Massachusetts.
| |
Collapse
|
26
|
Schüler R, Markova M, Osterhoff MA, Arafat A, Pivovarova O, Machann J, Hierholzer J, Hornemann S, Rohn S, Pfeiffer AFH. Similar dietary regulation of IGF-1- and IGF-binding proteins by animal and plant protein in subjects with type 2 diabetes. Eur J Nutr 2021; 60:3499-3504. [PMID: 33686453 PMCID: PMC8354897 DOI: 10.1007/s00394-021-02518-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
Increased animal but not plant protein intake has been associated with increased mortality in epidemiological studies in humans and with reduced lifespan in animal species. Protein intake increases the activity of the IGF-1 system which may provide a link to reduced lifespan. We, therefore, compared the effects of animal versus plant protein intake on circulating levels of IGF-1 and the IGF-binding proteins (IGFBP)-1 and IGFBP-2 over a 6-week period. Thirty seven participants with type 2 diabetes consumed isocaloric diets composed of either 30% energy (EN) animal or plant protein, 30% EN fat and 40% EN carbohydrates for 6 weeks. The participants were clinically phenotyped before and at the end of the study. Both diets induced similar and significant increases of IGF-1 which was unaffected by the different amino acid compositions of plant and animal protein. Despite improvements of insulin sensitivity and major reductions of liver fat, IGFBP2 decreased with both diets while IGFBP-1 was not altered. We conclude that animal and plant protein similarly increase IGF-1 bioavailability while improving metabolic parameters and may be regarded as equivalent in this regard.
Collapse
Affiliation(s)
- Rita Schüler
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke (DIfE), 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
| | - Mariya Markova
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke (DIfE), 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
| | - Martin A Osterhoff
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke (DIfE), 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany.,Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200, Berlin, Germany
| | - Ayman Arafat
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200, Berlin, Germany
| | - Olga Pivovarova
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke (DIfE), 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany.,Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200, Berlin, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.,Section of Experimental Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Johannes Hierholzer
- Diagnostic and Interventional Radiology, Klinikum Ernst von Bergmann, Academic Teaching Hospital, Charité-Universitätsmedizin Berlin, Potsdam, Germany
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke (DIfE), 14558, Nuthetal, Germany
| | - Sascha Rohn
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Hamburg, Germany
| | - Andreas F H Pfeiffer
- Department Endocrinology and Metabolism, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
27
|
Ahmed S, Ahmed A, Bouzina H, Lundgren J, Rådegran G. Elevated plasma endocan and BOC in heart failure patients decrease after heart transplantation in association with improved hemodynamics. Heart Vessels 2020; 35:1614-1628. [PMID: 32651845 PMCID: PMC7502449 DOI: 10.1007/s00380-020-01656-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The prevalence of heart failure (HF) is rising with ageing population and constitutes a major health problem globally. A common complication of HF is pulmonary hypertension (PH) which negatively impacts survival. A pathophysiological association between HF and PH with tumorigenic processes has been suggested. We aimed to identify the plasma levels of, and the association between tumour-related proteins and hemodynamic improvements in patients with HF and PH due to left heart disease (LHD) before and 1-year after heart transplantation (HT). METHODS Forty-eight tumour-related proteins were measured with proximity extension assay in plasma from 20 controls and 26 HF patients before and 1-year after HT. Patients' hemodynamics were measured with right heart catheterization. RESULTS Out of 48 proteins, specifically, plasma levels of endocan and brother of CDO (BOC) were elevated in end-stage HF patients compared to controls (p < 0.001), but decreased after HT (p < 0.01), towards controls' levels. The decrease of endocan levels after HT correlated with improved mean pulmonary arterial pressure (rs = 0.80, p < 0.0001), pulmonary arterial wedge pressure (rs = 0.63, p = 0.0012), and pulmonary vascular resistance (rs = 0.70, p < 0.001). The decrease and normalization of BOC after HT correlated with decreased mean right atrial pressure (rs = 0.61 p = 0.0015) and NT-proBNP (rs = 0.57, p = 0.0022), as well as increased cardiac index (rs = - 0.51, p = 0.0086) and left-ventricular stroke work index (rs = - 0.57, p = 0.0039). CONCLUSION Our results suggest that (i) plasma endocan in HF may reflect the state of pulmonary vascular congestion and PH-LHD, whereas (ii) plasma BOC may reflect the cardiac function and the hemodynamic overload in HF. The exact role of these proteins and their clinical applicability as biomarkers in HF and PH-LHD ought to be investigated in larger cohorts.
Collapse
Affiliation(s)
- Salaheldin Ahmed
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden.
- The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden.
| | - Abdulla Ahmed
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Habib Bouzina
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Jakob Lundgren
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| | - Göran Rådegran
- Department of Clinical Sciences Lund, Cardiology, Lund University, Lund, Sweden
- The Hemodynamic Lab, The Section for Heart Failure and Valvular Disease, VO. Heart and Lung Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
28
|
Yang J, Griffiths M, Nies MK, Brandal S, Damico R, Vaidya D, Tao X, Simpson CE, Kolb TM, Mathai SC, Pauciulo MW, Nichols WC, Ivy DD, Austin ED, Hassoun PM, Everett AD. Insulin-like growth factor binding protein-2: a new circulating indicator of pulmonary arterial hypertension severity and survival. BMC Med 2020; 18:268. [PMID: 33019943 PMCID: PMC7537100 DOI: 10.1186/s12916-020-01734-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/05/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a fatal disease that results from cardio-pulmonary dysfunction with the pathology largely unknown. Insulin-like growth factor binding protein 2 (IGFBP2) is an important member of the insulin-like growth factor family, with evidence suggesting elevation in PAH patients. We investigated the diagnostic and prognostic value of serum IGFBP2 in PAH to determine if it could discriminate PAH from healthy controls and if it was associated with disease severity and survival. METHODS Serum IGFBP2 levels, as well as IGF1/2 levels, were measured in two independent PAH cohorts, the Johns Hopkins Pulmonary Hypertension program (JHPH, N = 127), NHLBI PAHBiobank (PAHB, N = 203), and a healthy control cohort (N = 128). The protein levels in lung tissues were determined by western blot. The IGFBP2 mRNA expression levels in pulmonary artery smooth muscle cells (PASMC) and endothelial cells (PAEC) were assessed by RNA-seq, secreted protein levels by ELISA. Association of biomarkers with clinical variables was evaluated using adjusted linear or logistic regression and Kaplan-Meier analysis. RESULTS In both PAH cohorts, serum IGFBP2 levels were significantly elevated (p < 0.0001) compared to controls and discriminated PAH from controls with an AUC of 0.76 (p < 0.0001). A higher IGFBP2 level was associated with a shorter 6-min walk distance (6MWD) in both cohorts after adjustment for age and sex (coefficient - 50.235 and - 57.336 respectively). Cox multivariable analysis demonstrated that higher serum IGFBP2 was a significant independent predictor of mortality in PAHB cohort only (HR, 3.92; 95% CI, 1.37-11.21). IGF1 levels were significantly increased only in the PAHB cohort; however, neither IGF1 nor IGF2 had equivalent levels of associations with clinical variables compared with IGFBP2. Western blotting shown that IGFBP2 protein was significantly increased in the PAH vs control lung tissues. Finally, IGFBP2 mRNA expression and secreted protein levels were significantly higher in PASMC than in PAEC. CONCLUSIONS IGFBP2 protein expression was increased in the PAH lung, and secreted by PASMC. Elevated circulating IGFBP2 was associated with PAH severity and mortality and is a potentially valuable prognostic marker in PAH.
Collapse
Affiliation(s)
- Jun Yang
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University, 720 Rutland Ave. Ross RM 1143, Baltimore, MD, 21205, USA.
| | - Megan Griffiths
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University, 720 Rutland Ave. Ross RM 1143, Baltimore, MD, 21205, USA
| | - Melanie K Nies
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University, 720 Rutland Ave. Ross RM 1143, Baltimore, MD, 21205, USA
| | - Stephanie Brandal
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University, 720 Rutland Ave. Ross RM 1143, Baltimore, MD, 21205, USA
| | - Rachel Damico
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Dhananjay Vaidya
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.,Division of General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Xueting Tao
- Depart of Pediatrics, Biostatics Epidemiology and Data Management Core, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Catherine E Simpson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Todd M Kolb
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen C Mathai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - David D Ivy
- Department of Pediatric Cardiology, Children's Hospital Colorado, Denver, CO, USA
| | - Eric D Austin
- Division of Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Allen D Everett
- Division of Pediatric Cardiology, Department of Pediatrics, Johns Hopkins University, 720 Rutland Ave. Ross RM 1143, Baltimore, MD, 21205, USA
| |
Collapse
|
29
|
Ravassa S, Beaumont J, Cediel G, Lupón J, López B, Querejeta R, Díez J, Bayés-Genís A, González A. Interacción cardiorrenal y evolución de la insuficiencia cardiaca. ¿Tiene un papel la proteína de unión del factor de crecimiento de tipo insulina 2? Rev Esp Cardiol 2020. [DOI: 10.1016/j.recesp.2019.10.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Walz M, Chau L, Walz C, Sawitzky M, Ohde D, Brenmoehl J, Tuchscherer A, Langhammer M, Metzger F, Höflich C, Hoeflich A. Overlap of Peak Growth Activity and Peak IGF-1 to IGFBP Ratio: Delayed Increase of IGFBPs versus IGF-1 in Serum as a Mechanism to Speed up and down Postnatal Weight Gain in Mice. Cells 2020; 9:cells9061516. [PMID: 32580353 PMCID: PMC7348928 DOI: 10.3390/cells9061516] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/02/2020] [Accepted: 06/17/2020] [Indexed: 11/16/2022] Open
Abstract
Forced expression of insulin-like growth factor binding proteins (IGFBPs) in transgenic mice has clearly revealed inhibitory effects on somatic growth. However, by this approach, it cannot be solved if or how IGFBPs rule insulin-like growth factor (IGF)-dependent growth under normal conditions. In order to address this question, we have used growth-selected mouse models (obese and lean) and studied IGF-1 and IGFBPs in serum with respect to longitudinal growth activity in males and females compared with unselected controls. In mice of both genders, body weights were recorded and daily weight gains were calculated. Between 2 and 54 weeks of age, serum IGF-1 was determined by ELISA and intact IGFBP-2, -3 and -4 were quantified by Western ligand blotting. The molar ratio of IGF-1 to the sum of IGFBP-2 to -4 was calculated for all groups and plotted against the daily weight gain curve. Growth-selected mice are characterized by higher daily weight gains and extended periods of elevated growth activity if compared to matched unselected controls. Therefore, adult mice from the obese and lean groups can achieve more than twofold increased body weight in both genders (p < 0.001). Between 2 and 11 weeks of age, in obese and lean mice of both genders, serum IGF-1 concentrations are increased more prominently if compared to unselected controls (p < 0.001). Instead, substantial decreases of IGFBPs, particularly of IGFBP-2, are observed in males and females of all groups at the age of 2 to 4 weeks (p < 0.001). Due to the strong increase of IGF-1 but not of IGFBPs between two and four weeks of age, the ratio of IGF-1 to IGFBP-2 to -4 in serum significantly increased in all groups and genders (p < 0.05). Notably, the IGF-1 to IGFBP ratio was higher in male and female obese mice if compared to unselected controls (p < 0.05).
Collapse
Affiliation(s)
- Michael Walz
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Luong Chau
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Christina Walz
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Mandy Sawitzky
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Daniela Ohde
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Julia Brenmoehl
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
| | - Armin Tuchscherer
- Institute of Genetics and Biometry, Leibniz-Institute for Farm Animal Biology (FBN), 18197 Dummerstorf, Germany; (A.T.); (M.L.)
| | - Martina Langhammer
- Institute of Genetics and Biometry, Leibniz-Institute for Farm Animal Biology (FBN), 18197 Dummerstorf, Germany; (A.T.); (M.L.)
| | | | | | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (M.W.); (L.C.); (C.W.); (M.S.); (D.O.); (J.B.)
- Correspondence: ; Tel.: +49-(0)38208-68744; Fax: +49-(0)38208-68-702
| |
Collapse
|
31
|
Boyer MJ, Kimura Y, Akiyama T, Baggett AY, Preston KJ, Scalia R, Eguchi S, Rizzo V. Endothelial cell-derived extracellular vesicles alter vascular smooth muscle cell phenotype through high-mobility group box proteins. J Extracell Vesicles 2020; 9:1781427. [PMID: 32944170 PMCID: PMC7480479 DOI: 10.1080/20013078.2020.1781427] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The vascular endothelium and smooth muscle form adjacent cellular layers that comprise part of the vascular wall. Each cell type can regulate the other’s structure and function through a variety of paracrine effectors. Extracellular vesicles (EVs) are released from and transit between cells constituting a novel means of cell–cell communication. Here, we characterized the proteome of EVs released from each vascular cell type and examined the extent to which these vesicles participate in endothelial-vascular smooth muscle cell (VSMC) communication. EVs were collected by ultracentrifugation from media of rat aortic endothelial and smooth muscle cells cultured under serum-free conditions. Vesicle morphology, size and concentration were evaluated by transmission electron microscopy and nanoparticle tracking analysis. Western blot as well as shot gun proteomic analyses revealed sets of proteins common to both endothelial- and smooth muscle-derived EVs as well as proteins unique to each vascular cell type. Functionally, endothelial-derived EVs stimulated vascular cell adhesion molecule-1 (VCAM-1) expression and enhanced leukocyte adhesion in VSMCs while smooth muscle EVs did not elicit similar effects in endothelial cells (ECs). EVs from ECs also induced protein synthesis and senescence in VSMCs. Proteomic analysis of VSMCs following exposure to EC-derived EVs revealed upregulation of several proteins including pro-inflammatory molecules, high-mobility group box (HMGB) 1 and HMGB2. Pharmacological blockade HMGB1 and HMGB2 and siRNA depletion of HMGB1 in smooth muscle cells attenuated VCAM-1 expression and leukocyte adhesion induced by EC EVs. These data suggest that EC-derived EVs can enhance signalling pathways which influence smooth muscle cell phenotype.
Collapse
Affiliation(s)
- Michael J Boyer
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Ariele Y Baggett
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Kyle J Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
32
|
Mirna M, Topf A, Wernly B, Rezar R, Paar V, Jung C, Salmhofer H, Kopp K, Hoppe UC, Schulze PC, Kretzschmar D, Schneider MP, Schultheiss UT, Sommerer C, Paul K, Wolf G, Lichtenauer M, Busch M. Novel Biomarkers in Patients with Chronic Kidney Disease: An Analysis of Patients Enrolled in the GCKD-Study. J Clin Med 2020; 9:E886. [PMID: 32213894 PMCID: PMC7141541 DOI: 10.3390/jcm9030886] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/16/2020] [Accepted: 03/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Chronic kidney disease (CKD) and cardiovascular diseases (CVD) often occur concomitantly, and CKD is a major risk factor for cardiovascular mortality. Since some of the most commonly used biomarkers in CVD are permanently elevated in patients with CKD, novel biomarkers are warranted for clinical practice. Methods: Plasma concentrations of five cardiovascular biomarkers (soluble suppression of tumorigenicity (sST2), growth differentiation factor 15 (GDF-15), heart-type fatty acid-binding protein (H-FABP), insulin-like growth factor-binding protein 2 (IGF-BP2), and soluble urokinase plasminogen activator receptor) were analyzed by means of enzyme-linked immunosorbent assay (ELISA) in 219 patients with CKD enrolled in the German Chronic Kidney Disease (GCKD) study. Results: Except for sST2, all of the investigated biomarkers were significantly elevated in patients with CKD (2.0- to 4.4-fold increase in advanced CKD (estimated glomerular filtration rate (eGFR) < 30 mL/min/1.73 m² body surface area (BSA)) and showed a significant inverse correlation with eGFR. Moreover, all but H-FABP and sST2 were additionally elevated in patients with micro- and macro-albuminuria. Conclusions: Based on our findings, sST2 appears to be the biomarker whose diagnostic performance is least affected by decreased renal function, thus suggesting potential viability in the management of patients with CVD and concomitant CKD. The predictive potential of sST2 remains to be proven in endpoint studies.
Collapse
Affiliation(s)
- Moritz Mirna
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Albert Topf
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Bernhard Wernly
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Richard Rezar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Vera Paar
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Christian Jung
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany;
| | - Hermann Salmhofer
- Department of Internal Medicine I, Division of Nephrology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria;
| | - Kristen Kopp
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Uta C. Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - P. Christian Schulze
- Department of Internal Medicine I, Division of Cardiology, Friedrich Schiller University Jena, 07743 Jena, Germany; (P.C.S.); (D.K.)
| | - Daniel Kretzschmar
- Department of Internal Medicine I, Division of Cardiology, Friedrich Schiller University Jena, 07743 Jena, Germany; (P.C.S.); (D.K.)
| | - Markus P. Schneider
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Ulla T. Schultheiss
- Department of Medicine IV – Nephrology and Primary Care, Institute of Genetic Epidemiology, Medical Center–University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany;
| | - Claudia Sommerer
- Department of Nephrology, University of Heidelberg, 69117 Heidelberg, Germany;
| | - Katharina Paul
- Department of Internal Medicine III, Friedrich Schiller University Jena, 07743 Jena, Germany; (K.P.); (G.W.); (M.B.)
| | - Gunter Wolf
- Department of Internal Medicine III, Friedrich Schiller University Jena, 07743 Jena, Germany; (K.P.); (G.W.); (M.B.)
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University of Salzburg, 5020 Salzburg, Austria; (M.M.); (A.T.); (B.W.); (R.R.); (V.P.); (K.K.); (U.C.H.)
| | - Martin Busch
- Department of Internal Medicine III, Friedrich Schiller University Jena, 07743 Jena, Germany; (K.P.); (G.W.); (M.B.)
| |
Collapse
|
33
|
Ravassa S, Beaumont J, Cediel G, Lupón J, López B, Querejeta R, Díez J, Bayés-Genís A, González A. Cardiorenal interaction and heart failure outcomes. A role for insulin-like growth factor binding protein 2? REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2020; 73:835-843. [PMID: 31948893 DOI: 10.1016/j.rec.2019.10.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/24/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION AND OBJECTIVES Preliminary results suggest that high circulating insulin-like growth factor binding protein 2 (IGFBP2) levels are associated with mortality risk in heart failure (HF) patients. As IGFBP2 levels are increased in patients with chronic kidney disease (CKD), which is associated with a higher mortality risk in HF patients, we examined whether IGFBP2 is associated with CKD in HF patients, and whether CKD modifies the prognostic value of this protein in HF patients. METHODS HF patients (n=686, mean age 66.6 years, 32.7% women) were enrolled and followed up for a median of 3.5 (min-max range: 0.1-6) years. Patients were classified as having CKD with decreased estimated glomerular filtration rate (eGFR <60mL/min/1.73 m2) or as having CKD with nondecreased eGFR (≥ 60mL/min/1.73 m2). Serum IGFBP2 was detected by ELISA. RESULTS IGFBP2 was increased (P <.001) in CKD patients with decreased eGFR (n=290, 42.3%) compared with patients with nondecreased eGFR. IGFBP2 was directly associated with NT-proBNP (P <.001) and inversely associated with eGFR (P <.001), with both associations being independent of confounding factors. IGFBP2 was directly and independently associated with cardiovascular and all-cause death (P <.001) in the whole group of patients, but showed a stronger association with cardiovascular death in CKD patients with decreased eGFR (P for interaction <.05), improving risk prediction in these patients over clinically relevant risk factors. CONCLUSIONS Serum IGFBP2 is associated with impaired renal function and prognosticates cardiovascular death in patients with HF and CKD with decreased eGFR. Thus, there is an effect modification of CKD on circulating IGFBP2 and on its association with cardiovascular mortality in HF patients.
Collapse
Affiliation(s)
- Susana Ravassa
- Programa de Enfermedades Cardiovasculares, CIMA Universidad de Navarra, Pamplona, Navarra, Spain; Área de Enfermedades Cardiovasculares, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Instituto de Salud Carlos III, CIBERCV, Madrid, Spain.
| | - Javier Beaumont
- Programa de Enfermedades Cardiovasculares, CIMA Universidad de Navarra, Pamplona, Navarra, Spain; Área de Enfermedades Cardiovasculares, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Instituto de Salud Carlos III, CIBERCV, Madrid, Spain
| | - Germán Cediel
- Servei de Cardiologia i Unitat d'Insuficiència Cardíaca, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Josep Lupón
- Instituto de Salud Carlos III, CIBERCV, Madrid, Spain; Servei de Cardiologia i Unitat d'Insuficiència Cardíaca, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Begoña López
- Programa de Enfermedades Cardiovasculares, CIMA Universidad de Navarra, Pamplona, Navarra, Spain; Área de Enfermedades Cardiovasculares, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Instituto de Salud Carlos III, CIBERCV, Madrid, Spain
| | - Ramón Querejeta
- Servicio de Cardiología, Hospital Universitario Donostia, San Sebastián, Gipuzkoa, Spain
| | - Javier Díez
- Programa de Enfermedades Cardiovasculares, CIMA Universidad de Navarra, Pamplona, Navarra, Spain; Área de Enfermedades Cardiovasculares, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Instituto de Salud Carlos III, CIBERCV, Madrid, Spain; Departamento de Cardiología y Cirugía Cardiaca, Clínica Universidad de Navarra, Pamplona, Navarra, Spain; Departamento de Nefrología, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Antoni Bayés-Genís
- Instituto de Salud Carlos III, CIBERCV, Madrid, Spain; Servei de Cardiologia i Unitat d'Insuficiència Cardíaca, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Grupo ICREC (Insuficiència Cardíaca i Regeneració Cardíaca), Instituto de Investigación en Ciencias de la Salud Germans Trias i Pujol (IGTP), Badalona, Barcelona, Spain
| | - Arantxa González
- Programa de Enfermedades Cardiovasculares, CIMA Universidad de Navarra, Pamplona, Navarra, Spain; Área de Enfermedades Cardiovasculares, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Instituto de Salud Carlos III, CIBERCV, Madrid, Spain
| |
Collapse
|
34
|
Hjortebjerg R, Espelund U, Rasmussen TR, Folkersen B, Steiniche T, Georgsen JB, Oxvig C, Frystyk J. Pregnancy-Associated Plasma Protein-A2 Is Associated With Mortality in Patients With Lung Cancer. Front Endocrinol (Lausanne) 2020; 11:614. [PMID: 32982990 PMCID: PMC7492290 DOI: 10.3389/fendo.2020.00614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) and its homolog PAPP-A2 are enzymes that modulate the availability and mitogenic activity of insulin-like growth factor-I (IGF-I). PAPP-A has been implicated in numerous cancers but reports on PAPP-A2 in malignancy are non-existent. In a prospective observational study of 689 patients under suspicion of lung cancer, we examined levels of PAPP-A and PAPP-A2 and their relationship with mortality. Serum PAPP-A and PAPP-A2 concentrations were determined in pre-diagnostic blood samples using ELISA, and immunohistochemical staining of PAPP-A and PAPP-A2 was performed in malignant tissue from five operable patients. A total of 144 patients were diagnosed with lung cancer, whereas the diagnosis was rejected in 545 subjects, who served as a control group. PAPP-A2 concentrations were higher in patients with lung cancer [median (IQR): 0.33 (0.21-0.56) ng/mL] than in controls [0.27 (0.17-0.39) ng/mL], p < 0.001, whereas PAPP-A levels did not differ. Presence of PAPP-A and PAPP-A2 were confirmed in tumor specimens, and staining occurred in a heterogeneous pattern. Patients were observed for a median (range) of 7 (6; 8) years, during which 114 patients (79.2%) died. Patient mortality differed according to PAPP-A2 tertile (p < 0.001). PAPP-A2 was associated with mortality with an unadjusted hazard ratio (95% CI) per doubling in protein concentration of 1.30 (1.12; 1.53), p = 0.001. In a multivariable model adjusted for age, sex, and BMI, PAPP-A2 remained predictive of the endpoint with a hazard ratio per doubling in protein concentration of 1.25 (1.05; 1.48), p = 0.013. Collectively, PAPP-A2, but not PAPP-A, is elevated in patients with lung cancer and associated with mortality. This novel role of PAPP-A2 in cancer warrants further functional studies as well as validation in external cohorts.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Endocrinology (KMEB), University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- *Correspondence: Rikke Hjortebjerg
| | - Ulrick Espelund
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Birgitte Folkersen
- Department of Pulmonary Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Torben Steiniche
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan Frystyk
- Department of Molecular Endocrinology (KMEB), University of Southern Denmark, Odense, Denmark
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
35
|
Saxena S, Mathur P, Shukla V, Rani V. Differential expression of novel MicroRNAs from developing fetal heart of Gallus gallus domesticus implies a role in cardiac development. Mol Cell Biochem 2019; 462:157-165. [PMID: 31494815 DOI: 10.1007/s11010-019-03618-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/24/2019] [Indexed: 01/26/2023]
Abstract
Heart development is a complex process regulated by multi-layered genetic as well epigenetic regulators many of which are still unknown. Besides their critical role during cardiac development, these molecular regulators emerge as key modulators of cardiovascular pathologies, where fetal cardiac genes' re-expression is witnessed. MicroRNAs have recently emerged as a crucial part of signalling cascade in both development and diseases. We aimed to identify, validate, and perform functional annotation of putative novel miRNAs using chicken as a cardiac development model system. Novel miRNAs were obtained through deep sequencing of small RNAs extracted from chicken embryonic cardiac tissue of different developmental stages. After filtering out real pre-miRNAs, their expression analysis, potential target gene's prediction and functional annotations were performed. Expression analysis revealed that miRNAs were differentially expressed during different developmental stages of chicken heart. The expression of selected putative novel miRNAs was further validated by real-time PCR. Our analysis indicated the presence of novel cardiac miRNAs that might be regulating critical cardiac development events such as cardiac cell growth, differentiation, cardiac action potential generation and signal transduction.
Collapse
Affiliation(s)
- Sharad Saxena
- Transcriptome Laboratory, Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, UP, 201307, India
| | - Priyanka Mathur
- Transcriptome Laboratory, Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, UP, 201307, India
| | - Vaibhav Shukla
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vibha Rani
- Transcriptome Laboratory, Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector-62, Noida, UP, 201307, India.
| |
Collapse
|
36
|
Hjortebjerg R, Thomsen KL, Agnholt J, Frystyk J. The IGF system in patients with inflammatory bowel disease treated with prednisolone or infliximab: potential role of the stanniocalcin-2 / PAPP-A / IGFBP-4 axis. BMC Gastroenterol 2019; 19:83. [PMID: 31159802 PMCID: PMC6547608 DOI: 10.1186/s12876-019-1000-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 05/23/2019] [Indexed: 01/09/2023] Open
Abstract
Background Patients with inflammatory bowel disease (IBD) present with reduced serum insulin-like growth factor I (IGF-I). Anti-inflammatory treatment with prednisolone or infliximab ameliorates symptoms and increases circulating IGF-I, but prednisolone induces catabolism, whereas infliximab may promote protein synthesis. Recently, stanniocalcin-2 (STC2) was discovered as a novel inhibitor of the enzyme pregnancy-associated plasma protein-A (PAPP-A), which modulates IGF-I activity. PAPP-A can cleave IGF binding protein-4 (IGFBP-4), upon which IGF-I is liberated. We hypothesized that prednisolone and infliximab exert different effects on levels of STC2, PAPP-A, and IGFBP-4, thereby explaining the distinct metabolic effects of prednisolone and infliximab. Methods Thirty-eight patients with active IBD treated with either prednisolone (n = 17) or infliximab (n = 21) were examined before and after 7 days of treatment. Circulating levels of IGF-I, IGF-II, IGFBP-3, PAPP-A, and STC2 were measured by immunoassays. Intact IGFBP-4 and two IGFBP-4 fragments were determined by a novel immunoassay. Bioactive IGF was assessed by cell-based IGF receptor activation assay. Concentrations of IGFBP-4, PAPP-A, and STC2 on day 0 and 7 were compared to healthy control subjects. Results Following seven days of prednisolone treatment, total and bioactive IGF-I were increased (p < 0.001 and p < 0.05, respectively). Upon infliximab treatment, total IGF-I levels were augmented (p < 0.05), yet IGF bioactivity remained unaltered. Intact IGFBP-4 and the two IGFBP-4 fragments generated upon cleavage by PAPP-A were all decreased following treatment with either prednisolone or infliximab (all p < 0.05). PAPP-A levels were only increased by infliximab (p = 0.005), whereas the inhibitor STC2 did not respond to any of the treatments. Conclusion IGF-I and IGFBP-4 concentrations were markedly altered in patients with IBD and near-normalized with disease remission following treatment with prednisolone or infliximab. Thus, IGFBP-4 may modulate IGF bioavailability in IBD. The effect of immunosuppression did not appear to extend beyond the regulation of IGF and IGFBP-4, as neither PAPP-A nor STC2 were discernibly affected. Trial registration ClinicalTrials.gov: NCT00955123. Date of registration: August 7, 2009 (retrospectively registered).
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark. .,The Danish Diabetes Academy, Odense, Denmark.
| | - Karen L Thomsen
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Agnholt
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Clinical Research, Faculty of Health, University of Southern Denmark, Odense, Denmark.,Department of Endocrinology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
37
|
Plausible Links Between Metabolic Networks, Stem Cells, and Longevity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1201:355-388. [PMID: 31898793 DOI: 10.1007/978-3-030-31206-0_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging is an inevitable consequence of life, and all multicellular organisms undergo a decline in tissue and organ functions as they age. Several well-known risk factors, such as obesity, diabetes, and lack of physical activity that lead to the cardiovascular system, decline and impede the function of vital organs, ultimately limit overall life span. Over recent years, aging research has experienced an unparalleled growth, particularly with the discovery and recognition of genetic pathways and biochemical processes that control to some extent the rate of aging.In this chapter, we focus on several aspects of stem cell biology and aging, beginning with major cellular hallmarks of aging, endocrine regulation of aging and its impact on stem cell compartment, and mechanisms of increased longevity. We then discuss the role of epigenetic modifications associated with aging and provide an overview on a most recent search of antiaging modalities.
Collapse
|