1
|
Yuan Y, Shen Z, Teng T, Xu S, Kong C, Zeng X, A. Hofmann Bowman M, Yan L. S100a8/9 (S100 Calcium Binding Protein a8/9) Promotes Cardiac Hypertrophy Via Upregulation of FGF23 (Fibroblast Growth Factor 23) in Mice. J Am Heart Assoc 2024; 13:e028006. [PMID: 38726894 PMCID: PMC11179804 DOI: 10.1161/jaha.122.028006] [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: 08/30/2022] [Accepted: 04/04/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND S100a8/9 (S100 calcium binding protein a8/9) belongs to the S100 family and has gained a lot of interest as a critical regulator of inflammatory response. Our previous study found that S100a8/9 homolog promoted aortic valve sclerosis in mice with chronic kidney disease. However, the role of S100a8/9 in pressure overload-induced cardiac hypertrophy remains unclear. The present study was to explore the role of S100a8/9 in cardiac hypertrophy. METHODS AND RESULTS Cardiomyocyte-specific S100a9 loss or gain of function was achieved using an adeno-associated virus system, and the model of cardiac hypertrophy was established by aortic banding-induced pressure overload. The results indicate that S100a8/9 expression was increased in response to pressure overload. S100a9 deficiency alleviated pressure overload-induced hypertrophic response, whereas S100a9 overexpression accelerated cardiac hypertrophy. S100a9-overexpressed mice showed increased FGF23 (fibroblast growth factor 23) expression in the hearts after exposure to pressure overload, which activated calcineurin/NFAT (nuclear factor of activated T cells) signaling in cardiac myocytes and thus promoted hypertrophic response. A specific antibody that blocks FGFR4 (FGF receptor 4) largely abolished the prohypertrophic response of S100a9 in mice. CONCLUSIONS In conclusion, S100a8/9 promoted the development of cardiac hypertrophy in mice. Targeting S100a8/9 may be a promising therapeutic approach to treat cardiac hypertrophy.
Collapse
Affiliation(s)
- Yu‐Pei Yuan
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| | - Zhuo‐Yu Shen
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| | - Teng Teng
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| | - Si‐Chi Xu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| | - Chun‐Yan Kong
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| | - Xiao‐Feng Zeng
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| | | | - Ling Yan
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of Metabolic and Chronic DiseasesWuhanChina
| |
Collapse
|
2
|
Mazzarino M, Cetin E, Bartosova M, Marinovic I, Ipseiz N, Hughes TR, Schmitt CP, Ramji DP, Labéta MO, Raby AC. Therapeutic targeting of chronic kidney disease-associated DAMPs differentially contributing to vascular pathology. Front Immunol 2023; 14:1240679. [PMID: 37849759 PMCID: PMC10577224 DOI: 10.3389/fimmu.2023.1240679] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/08/2023] [Indexed: 10/19/2023] Open
Abstract
Chronic Kidney Disease (CKD) is associated with markedly increased cardiovascular (CV) morbidity and mortality. Chronic inflammation, a hallmark of both CKD and CV diseases (CVD), is believed to drive this association. Pro-inflammatory endogenous TLR agonists, Damage-Associated Molecular Patterns (DAMPs), have been found elevated in CKD patients' plasma and suggested to promote CVD, however, confirmation of their involvement, the underlying mechanism(s), the extent to which individual DAMPs contribute to vascular pathology in CKD and the evaluation of potential therapeutic strategies, have remained largely undescribed. A multi-TLR inhibitor, soluble TLR2, abrogated chronic vascular inflammatory responses and the increased aortic atherosclerosis-associated gene expression observed in nephropathic mice, without compromising infection clearance. Mechanistically, we confirmed elevation of 4 TLR DAMPs in CKD patients' plasma, namely Hsp70, Hyaluronic acid, HMGB-1 and Calprotectin, which displayed different abilities to promote key cellular responses associated with vascular inflammation and progression of atherosclerosis in a TLR-dependent manner. These included loss of trans-endothelial resistance, enhanced monocyte migration, increased cytokine production, and foam cell formation by macrophages, the latter via cholesterol efflux inhibition. Calprotectin and Hsp70 most consistently affected these functions. Calprotectin was further elevated in CVD-diagnosed CKD patients and strongly correlated with the predictor of CV events CRP. In nephropathic mice, Calprotectin blockade robustly reduced vascular chronic inflammatory responses and pro-atherosclerotic gene expression in the blood and aorta. Taken together, these findings demonstrated the critical extent to which the DAMP-TLR pathway contributes to vascular inflammatory and atherogenic responses in CKD, revealed the mechanistic contribution of specific DAMPs and described two alternatives therapeutic approaches to reduce chronic vascular inflammation and lower CV pathology in CKD.
Collapse
Affiliation(s)
- Morgane Mazzarino
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Esra Cetin
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Maria Bartosova
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Iva Marinovic
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Natacha Ipseiz
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Timothy R. Hughes
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
| | - Claus Peter Schmitt
- Division of Pediatric Nephrology, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Dipak P. Ramji
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Mario O. Labéta
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Anne-Catherine Raby
- Division of Infection & Immunity, Cardiff University, Cardiff, United Kingdom
- Wales Kidney Research Unit, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
3
|
Zhou Y, Zha Y, Yang Y, Ma T, Li H, Liang J. S100 proteins in cardiovascular diseases. Mol Med 2023; 29:68. [PMID: 37217870 DOI: 10.1186/s10020-023-00662-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
Cardiovascular diseases have become a serious threat to human health and life worldwide and have the highest fatality rate. Therefore, the prevention and treatment of cardiovascular diseases have become a focus for public health experts. The expression of S100 proteins is cell- and tissue-specific; they are implicated in cardiovascular, neurodegenerative, and inflammatory diseases and cancer. This review article discusses the progress in the research on the role of S100 protein family members in cardiovascular diseases. Understanding the mechanisms by which these proteins exert their biological function may provide novel concepts for preventing, treating, and predicting cardiovascular diseases.
Collapse
Affiliation(s)
- Yue Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Yiwen Zha
- Medical College, Yangzhou University, Yangzhou, China
| | - Yongqi Yang
- Medical College, Yangzhou University, Yangzhou, China
| | - Tan Ma
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Hongliang Li
- Medical College, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| | - Jingyan Liang
- Medical College, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China.
| |
Collapse
|
4
|
S100A12 is not expressed in rodents: Transgenic mouse model is needed. Mol Immunol 2022; 152:35. [DOI: 10.1016/j.molimm.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/08/2022] [Indexed: 11/21/2022]
|
5
|
Gellen B, Thorin‐Trescases N, Thorin E, Gand E, Ragot S, Montaigne D, Pucheu Y, Mohammedi K, Gatault P, Potier L, Liuu E, Hadjadj S, Saulnier P. Increased serum S100A12 levels are associated with higher risk of acute heart failure in patients with type 2 diabetes. ESC Heart Fail 2022; 9:3909-3919. [PMID: 36637406 PMCID: PMC9773733 DOI: 10.1002/ehf2.14036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/09/2022] [Accepted: 06/08/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS The hyperglycaemic stress induces the release of inflammatory proteins such as S100A12, one of the endogenous ligands of the receptors for advanced glycation end products (RAGE). Chronic activation of RAGE has multiple deleterious effects in target tissues such as the heart and the vessels by promoting oxidative stress, inflammation by the release of cytokines, macrophages infiltration, and vascular cell migration and proliferation, causing ultimately endothelial cell and cardiomyocyte dysfunction. The aim of our study was to investigate the prognostic value of circulating S100A12 beyond established cardiovascular risk factors (CVRF) for heart failure (HF) and major adverse cardiovascular events (MACE) in a cohort of patients with type 2 diabetes. METHODS AND RESULTS Serum S100A12 concentrations were measured at baseline in 1345 type 2 diabetes patients (58% men, 64 ± 11 years) recruited in the SURDIAGENE prospective cohort. Endpoints were the occurrence of acute HF requiring hospitalization (HHF) and MACE. We used a proportional hazard model adjusted for established CVRF (age, sex, duration of diabetes, estimated glomerular filtration rate, albumin/creatinine ratio, history of coronary artery disease) and serum S100A12. During the median follow-up of 84 months, 210 (16%) and 505 (38%) patients developed HHF and MACE, respectively. Baseline serum S100A12 concentrations were associated with an increased risk of HHF [hazard ratio (HR) (95% confidence interval) 1.28 (1.01-1.62)], but not MACE [1.04 (0.90-1.20)]. After adjustment for CVRF, S100A12 concentrations remained significantly associated with an increased risk of HHF [1.29 (1.01-1.65)]. In a sub-analysis, patients with high probability of pre-existing HF [N terminal pro brain natriuretic peptide (NT-proBNP) >1000 pg/mL, n = 87] were excluded. In the remaining 1258 patients, the association of serum S100A12 with the risk of HHF tended to be more pronounced [1.39 (1.06-1.83)]. When including the gold standard HF marker NT-proBNP in the model, the prognostic value of S100A12 for HHF did not reach significance. Youden method performed at 7 years for HHF prediction yielded an optimal cut-off for S100A12 concentration of 49 ng/mL (sensitivity 53.3, specificity 52.2). Compared with those with S100A12 ≤ 49 ng/mL, patients with S100A12 > 49 ng/mL had a significantly increased risk of HHF in the univariate model [HR = 1.58 (1.19-2.09), P = 0.0015] but also in the multivariate model [HR = 1.63 (1.23-2.16), P = 0.0008]. After addition of NT-proBNP to the multivariate model, S100A12 > 49 ng/mL remained associated with an increased risk of HHF [HR = 1.42 (1.07-1.90), P = 0.0160]. However, the addition of S100A12 categories on top of multivariate model enriched by NT-pro BNP did not improve the ability of the model to predict HHF (relative integrated discrimination improvement = 1.9%, P = 0.1500). CONCLUSIONS In patients with type 2 diabetes, increased serum S100A12 concentration is independently associated with risk of HHF, but not with risk of MACE. Compared with NT-proBNP, the potential clinical interest of S100A12 for the prediction of HF events remains limited. However, S100A12 could be a candidate for a multimarker approach for HF risk assessment in diabetic patients.
Collapse
Affiliation(s)
- Barnabas Gellen
- ELSAN—Polyclinique de Poitiers1 Rue de la ProvidenceF‐86000PoitiersFrance
| | | | - Eric Thorin
- Montreal Heart Institute, Research CenterMontrealQuebecCanada
- Department of Surgery, Faculty of MedicineUniversity of Montréal, Montreal Heart InstituteMontrealQuebecCanada
| | - Elise Gand
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
| | - Stephanie Ragot
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
| | - David Montaigne
- Department of Clinical Physiology—EchocardiographyCHU LilleLilleFrance
- INSERMU1011, EGID, Institut Pasteur de LilleUniversity of LilleLilleFrance
| | - Yann Pucheu
- Department of CardiologyCHU de BordeauxPessacFrance
| | - Kamel Mohammedi
- Hôpital Haut‐Lévêque, Department of Endocrinology, Diabetes and Nutrition; University of Bordeaux, Faculty of Medicine; INSERM unit 1034, Biology of Cardiovascular DiseasesBordeaux University HospitalBordeauxFrance
| | | | - Louis Potier
- Department of DiabetologyHôpital Bichat—Claude‐Bernard, APHP, Université de ParisParisFrance
- Cordeliers Research Centre, ImMeDiab team, INSERMParisFrance
| | - Evelyne Liuu
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
- Department of GeriatricsCHU de PoitiersPoitiersFrance
| | - Samy Hadjadj
- L'institut du ThoraxINSERM, CNRS, UNIV Nantes, CHU NantesNantesFrance
| | - Pierre‐Jean Saulnier
- Centre d'Investigation Clinique CIC1402Université de Poitiers, CHU de Poitiers, INSERMPoitiersFrance
| | | |
Collapse
|
6
|
Dargam V, Ng HH, Nasim S, Chaparro D, Irion CI, Seshadri SR, Barreto A, Danziger ZC, Shehadeh LA, Hutcheson JD. S2 Heart Sound Detects Aortic Valve Calcification Independent of Hemodynamic Changes in Mice. Front Cardiovasc Med 2022; 9:809301. [PMID: 35694672 PMCID: PMC9174427 DOI: 10.3389/fcvm.2022.809301] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/18/2022] [Indexed: 11/16/2022] Open
Abstract
Background Calcific aortic valve disease (CAVD) is often undiagnosed in asymptomatic patients, especially in underserved populations. Although artificial intelligence has improved murmur detection in auscultation exams, murmur manifestation depends on hemodynamic factors that can be independent of aortic valve (AoV) calcium load and function. The aim of this study was to determine if the presence of AoV calcification directly influences the S2 heart sound. Methods Adult C57BL/6J mice were assigned to the following 12-week-long diets: (1) Control group (n = 11) fed a normal chow, (2) Adenine group (n = 4) fed an adenine-supplemented diet to induce chronic kidney disease (CKD), and (3) Adenine + HP (n = 9) group fed the CKD diet for 6 weeks, then supplemented with high phosphate (HP) for another 6 weeks to induce AoV calcification. Phonocardiograms, echocardiogram-based valvular function, and AoV calcification were assessed at endpoint. Results Mice on the Adenine + HP diet had detectable AoV calcification (9.28 ± 0.74% by volume). After segmentation and dimensionality reduction, S2 sounds were labeled based on the presence of disease: Healthy, CKD, or CKD + CAVD. The dataset (2,516 S2 sounds) was split subject-wise, and an ensemble learning-based algorithm was developed to classify S2 sound features. For external validation, the areas under the receiver operating characteristic curve of the algorithm to classify mice were 0.9940 for Healthy, 0.9717 for CKD, and 0.9593 for CKD + CAVD. The algorithm had a low misclassification performance of testing set S2 sounds (1.27% false positive, 1.99% false negative). Conclusion Our ensemble learning-based algorithm demonstrated the feasibility of using the S2 sound to detect the presence of AoV calcification. The S2 sound can be used as a marker to identify AoV calcification independent of hemodynamic changes observed in echocardiography.
Collapse
Affiliation(s)
- Valentina Dargam
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Hooi Hooi Ng
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
- Department of Human and Molecular Genetics, Florida International University, Miami, FL, United States
| | - Sana Nasim
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Daniel Chaparro
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Camila Iansen Irion
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - Suhas Rathna Seshadri
- Department of Medical Education, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - Armando Barreto
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL, United States
| | - Zachary C. Danziger
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
| | - Lina A. Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Coral Gables, FL, United States
- Division of Cardiology, Department of Medicine, University of Miami Miller School of Medicine, Coral Gables, FL, United States
| | - Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL, United States
- Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| |
Collapse
|
7
|
Schwaiger JP, Kollerits B, Steinbrenner I, Weissensteiner H, Schönherr S, Forer L, Kotsis F, Lamina C, Schneider MP, Schultheiss UT, Wanner C, Köttgen A, Eckardt KU, Kronenberg F. Apolipoprotein A-IV concentrations and clinical outcomes in a large chronic kidney disease cohort: Results from the GCKD study. J Intern Med 2022; 291:622-636. [PMID: 34914850 PMCID: PMC9305919 DOI: 10.1111/joim.13437] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) represents a chronic proinflammatory state and is associated with very high cardiovascular risk. Apolipoprotein A-IV (apoA-IV) has antiatherogenic, antioxidative, anti-inflammatory and antithrombotic properties and levels increase significantly during the course of CKD. OBJECTIVES We aimed to investigate the association between apoA-IV and all-cause mortality and cardiovascular outcomes in the German Chronic Kidney Disease study. METHODS This was a prospective cohort study including 5141 Caucasian patients with available apoA-IV measurements and CKD. The majority of the patients had an estimated glomerular filtration rate (eGFR) of 30-60 ml/min/1.73m2 or an eGFR >60 ml/min/1.73m2 in the presence of overt proteinuria. Median follow-up was 6.5 years. The association of apoA-IV with comorbidities at baseline and endpoints during follow-up was modelled adjusting for major confounders. RESULTS Mean apoA-IV concentrations of the entire cohort were 28.9 ± 9.8 mg/dl. Patients in the highest apoA-IV quartile had the lowest high-sensitivity C-reactive protein values despite the highest prevalence of diabetes, albuminuria and the lowest eGFR. Each 10 mg/dl higher apoA-IV translated into lower odds of prevalent cardiovascular disease (1289 cases, odds ratio = 0.80, 95% confidence interval [CI] 0.72-0.86, p = 0.0000003). During follow-up, each 10 mg/dl higher apoA-IV was significantly associated with a lower risk for all-cause mortality (600 cases, hazard ratio [HR] = 0.81, 95% CI 0.73-0.89, p = 0.00004), incident major adverse cardiovascular events (506 cases, HR = 0.88, 95% CI 0.79-0.99, p = 0.03) and death or hospitalizations due to heart failure (346 cases, HR = 0.84, 95% CI 0.73-0.96, p = 0.01). CONCLUSIONS These data support a link between elevated apoA-IV concentrations and reduced inflammation in moderate CKD. ApoA-IV appears to be an independent risk marker for reduced all-cause mortality, cardiovascular events and heart failure in a large cohort of patients with CKD.
Collapse
Affiliation(s)
- Johannes P Schwaiger
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Internal Medicine, Landeskrankenhaus Hall i.T., Hall in Tirol, Austria
| | - Barbara Kollerits
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Hansi Weissensteiner
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Fruzsina Kotsis
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.,Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus P Schneider
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulla T Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.,Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Christoph Wanner
- Division of Nephrology, Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | -
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
8
|
Molinari P, Caldiroli L, Dozio E, Rigolini R, Giubbilini P, Romanelli MMC, Messa P, Vettoretti S. AGEs and sRAGE Variations at Different Timepoints in Patients with Chronic Kidney Disease. Antioxidants (Basel) 2021; 10:antiox10121994. [PMID: 34943097 PMCID: PMC8698924 DOI: 10.3390/antiox10121994] [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: 10/30/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 12/22/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are affected by enhanced oxidative stress and chronic inflammation, and these factors may contribute to increase advanced glycation end-products (AGEs). In this study we quantified AGEs and soluble receptors for AGE (sRAGE) isoforms and evaluated the association between their variations and eGFR at baseline and after 12 months. We evaluated 64 patients. AGEs were quantified by fluorescence intensity using a fluorescence spectrophotometer, and sRAGE by ELISA. Median age was 81 years, male patients accounted for 70%, 63% were diabetic, and eGFR was 27 ± 10 mL/min/1.73 m2. At follow up, sRAGE isoforms underwent a significant decrement (1679 [1393;2038] vs. 1442 [1117;2102], p < 0.0001), while AGEs/sRAGE ratios were increased (1.77 ± 0.92 vs. 2.24 ± 1.34, p = 0.004). Although AGEs and AGEs/sRAGE ratios were inversely related with eGFR, their basal values as well their variations did not show a significant association with eGFR changes. In a cohort of patients with a stable clinical condition at 1 year follow-up, AGEs/sRAGE was associated with renal function. The lack of association with eGFR suggests that other factors can influence its increase. In conclusion, AGEs/sRAGE can be an additional risk factor for CKD progression over a longer time, but its role as a prognostic tool needs further investigation.
Collapse
Affiliation(s)
- Paolo Molinari
- Unit of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy; (P.M.); (L.C.); (P.M.)
| | - Lara Caldiroli
- Unit of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy; (P.M.); (L.C.); (P.M.)
| | - Elena Dozio
- Laboratory of Clinical Pathology, Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
| | - Roberta Rigolini
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (R.R.); (P.G.)
| | - Paola Giubbilini
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (R.R.); (P.G.)
| | - Massimiliano M. Corsi Romanelli
- Laboratory of Clinical Pathology, Department of Biomedical Science for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (R.R.); (P.G.)
| | - Piergiorgio Messa
- Unit of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy; (P.M.); (L.C.); (P.M.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Simone Vettoretti
- Unit of Nephrology, Dialysis and Kidney Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, 20122 Milan, Italy; (P.M.); (L.C.); (P.M.)
- Correspondence: ; Tel.: +39-02-55-03-45-52
| |
Collapse
|
9
|
Soppert J, Frisch J, Wirth J, Hemmers C, Boor P, Kramann R, Vondenhoff S, Moellmann J, Lehrke M, Hohl M, van der Vorst EPC, Werner C, Speer T, Maack C, Marx N, Jankowski J, Roma LP, Noels H. A systematic review and meta-analysis of murine models of uremic cardiomyopathy. Kidney Int 2021; 101:256-273. [PMID: 34774555 DOI: 10.1016/j.kint.2021.10.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) triggers the risk of developing uremic cardiomyopathy as characterized by cardiac hypertrophy, fibrosis and functional impairment. Traditionally, animal studies are used to reveal the underlying pathological mechanism, although variable CKD models, mouse strains and readouts may reveal diverse results. Here, we systematically reviewed 88 studies and performed meta-analyses of 52 to support finding suitable animal models for future experimental studies on pathological kidney-heart crosstalk during uremic cardiomyopathy. We compared different mouse strains and the direct effect of CKD on cardiac hypertrophy, fibrosis and cardiac function in "single hit" strategies as well as cardiac effects of kidney injury combined with additional cardiovascular risk factors in "multifactorial hit" strategies. In C57BL/6 mice, CKD was associated with a mild increase in cardiac hypertrophy and fibrosis and marginal systolic dysfunction. Studies revealed high variability in results, especially regarding hypertrophy and systolic function. Cardiac hypertrophy in CKD was more consistently observed in 129/Sv mice, which express two instead of one renin gene and more consistently develop increased blood pressure upon CKD induction. Overall, "multifactorial hit" models more consistently induced cardiac hypertrophy and fibrosis compared to "single hit" kidney injury models. Thus, genetic factors and additional cardiovascular risk factors can "prime" for susceptibility to organ damage, with increased blood pressure, cardiac hypertrophy and early cardiac fibrosis more consistently observed in 129/Sv compared to C57BL/6 strains.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Janina Frisch
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Julia Wirth
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Hemmers
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Mathias Hohl
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands; Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Werner
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Würzburg, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
10
|
Rossi A, Barbieri A, Benfari G, Gaibazzi N, Erlicher A, Mureddu G, Frattini S, Faden G, Manicardi M, Beraldi M, Agostini F, Lazzarini V, Moreo A, Luigi Temporelli P, Magni G, Pressman G, Faggiano P. Heart valve calcification and cardiac hemodynamics. Echocardiography 2021; 38:525-530. [PMID: 33705585 DOI: 10.1111/echo.14994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/05/2021] [Accepted: 01/25/2021] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Heart valve calcification (VC) is associated with increased cardiovascular risk, but the hemodynamic and functional profile of patients affected by VC has not been fully explored. METHODS The study population was formed by consecutive unselected patients included in seven echocardiographic laboratories in a 2-week period. A comprehensive echocardiographic examination was performed. VC was defined by the presence of calcification on at least one valve. RESULTS Population was formed of 1098 patients (mean age 65 ± 15 years; 47% female). VC was present in 31% of the overall population. Compared with subjects without VC, VC patients were older (60 ± 14 vs 75 ± 9; P < .0001), had more hypertension (40% vs 57%; P = .0005), diabetes (11% vs 18%; P = .002), coronary artery disease (22% vs 38%; P = .04), and chronic kidney disease (4% vs 8%; P = .007). Furthermore, VC patients had lower ejection fraction (55 ± 14 vs 53 ± 25; P < .0001), worse diastolic function (E/e' 8.5 ± 4.6 vs 13.0 ± 7.1; P < .0001) and higher pulmonary artery pressure (29 ± 9 vs 37 ± 12; P < .0001). The association between VC and EF was not independent of etiology (p for VC 0.13), whereas the association with E/e' and PASP was independent in a full multivariate model (P < .0001 and P = .0002, respectively). CONCLUSION Heart valve calcification patients were characterized by a worse functional and hemodynamic profile compared to patients with normal valve. The association between VC and diastolic function and PASP were independent in comprehensive multivariate models.
Collapse
Affiliation(s)
- Andrea Rossi
- Section of Cardiology, Department of Medicine, School of Medicine, Verona, Italy
| | - Andrea Barbieri
- Department of Cardiology, Policlinico Hospital, Modena and Reggio Emilia University, Modena, Italy
| | - Giovanni Benfari
- Section of Cardiology, Department of Medicine, School of Medicine, Verona, Italy
| | | | | | | | | | - Giacomo Faden
- Cardiology Division, Pieve di Coriano, Brescia, Italy
| | - Marcella Manicardi
- Department of Cardiology, Policlinico Hospital, Modena and Reggio Emilia University, Modena, Italy
| | | | | | | | | | | | | | | | - Pompilio Faggiano
- Cardiology Division, Spedali Civili and University of Brescia, Brescia, Italy
| |
Collapse
|
11
|
Sterile inflammation in the pathogenesis of maturation failure of arteriovenous fistula. J Mol Med (Berl) 2021; 99:729-741. [PMID: 33666676 DOI: 10.1007/s00109-021-02056-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/19/2020] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
Chronic kidney disease is a widespread terminal illness that afflicts millions of people across the world. Hemodialysis is the predominant therapeutic management strategy for kidney failure and involves the external filtration of metabolic waste within the circulation. This process requires an arteriovenous fistula (AVF) for vascular access. However, AVF maturation failures are significant obstacles in establishing long-term vascular access for hemodialysis. Appropriate stimulation, activation, and proliferation of smooth muscle cells, proper endothelial cell orientation, adequate structural changes in the ECM, and the release of anti-inflammatory markers are associated with maturation. AVFs often fail to mature due to inadequate tissue repair and remodeling, leading to neointimal hyperplasia lesions. The transdifferentiation of myofibroblasts and sterile inflammation are possibly involved in AVF maturation failures; however, limited data is available in this regard. The present article critically reviews the interplay of various damage-associated molecular patterns (DAMPs) and the downstream sterile inflammatory signaling with a focus on the NLRP3 inflammasome. Improved knowledge concerning AVF maturation pathways can be unveiled by investigating the novel DAMPs and the mediators of sterile inflammation in vascular remodeling that would open improved therapeutic opportunities in the management of AVF maturation failures and its associated complications.
Collapse
|
12
|
Pawliński Ł, Tobór E, Suski M, Biela M, Polus A, Kieć-Wilk B. Proteomic biomarkers in Gaucher disease. J Clin Pathol 2020; 74:25-29. [PMID: 32409598 DOI: 10.1136/jclinpath-2020-206580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/07/2020] [Accepted: 04/13/2020] [Indexed: 12/28/2022]
Abstract
AIMS The research work was conducted to find new biomarkers and potential drug targets in Gaucher disease type 1 (GDt1) by analysing the serum proteins. METHODS This study was an observational, cross-sectional analysis of a group of 12 adult participants: six Gaucher disease (GD) patients and six healthy control. Fasting venous blood underwent proteomics analysis and molecular tests. Over 400 proteins were analysed, and in case of significantly different concentrations between the study and control group, we checked corresponding genes to confirm changes in their expression and consistency with protein alteration. RESULTS We found 31 proteins that significantly differed in concentration between GDt1 patients and a control group. These were mostly proteins involved in the regulation of the inflammatory processes and haemostasis. The levels of proteins such as alpha-1-acid glycoprotein 2, S100-A8/A9, adenyl cyclase-associated protein 1, haptoglobin or translationally controlled tumour protein related to inflammation process were significantly higher in GD patients than in control group, whereas the levels of some proteins such as heavy constant mu and gamma 4 or complement C3/C4 complex involved in humoral response like immunoglobulins were significantly decreased in GD patients. Alteration in two proteins concentration was confirmed in RNA analysis. CONCLUSIONS The work revealed few new targets for further investigation which may be useful in clinical practice for diagnosis, treatment and monitoring GDt1 patients.
Collapse
Affiliation(s)
- Łukasz Pawliński
- Metabolic Diseases and Diabetology Department, Szpital Uniwersytecki w Krakowie, Kraków, małopolskie, Poland
| | - Ewa Tobór
- Metabolic Diseases and Diabetology Department, Szpital Uniwersytecki w Krakowie, Kraków, małopolskie, Poland
| | - Maciej Suski
- Department of Pharmacology, Jagiellonian University in Krakow Medical College Faculty of Medicine, Krakow, Poland
| | - Maria Biela
- Department of Clinical Biochemistry, Jagiellonian University in Krakow Medical College Faculty of Medicine, Krakow, Poland
| | - Anna Polus
- Department of Clinical Biochemistry, Jagiellonian University in Krakow Medical College Faculty of Medicine, Krakow, Poland
| | - Beata Kieć-Wilk
- Metabolic Diseases and Diabetology Department, Szpital Uniwersytecki w Krakowie, Kraków, małopolskie, Poland
| |
Collapse
|
13
|
Steffens S, Van Linthout S, Sluijter JPG, Tocchetti CG, Thum T, Madonna R. Stimulating pro-reparative immune responses to prevent adverse cardiac remodelling: consensus document from the joint 2019 meeting of the ESC Working Groups of cellular biology of the heart and myocardial function. Cardiovasc Res 2020; 116:1850-1862. [DOI: 10.1093/cvr/cvaa137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/31/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Cardiac injury may have multiple causes, including ischaemic, non-ischaemic, autoimmune, and infectious triggers. Independent of the underlying pathophysiology, cardiac tissue damage induces an inflammatory response to initiate repair processes. Immune cells are recruited to the heart to remove dead cardiomyocytes, which is essential for cardiac healing. Insufficient clearance of dying cardiomyocytes after myocardial infarction (MI) has been shown to promote unfavourable cardiac remodelling, which may result in heart failure (HF). Although immune cells are integral key players of cardiac healing, an unbalanced or unresolved immune reaction aggravates tissue damage that triggers maladaptive remodelling and HF. Neutrophils and macrophages are involved in both, inflammatory as well as reparative processes. Stimulating the resolution of cardiac inflammation seems to be an attractive therapeutic strategy to prevent adverse remodelling. Along with numerous experimental studies, the promising outcomes from recent clinical trials testing canakinumab or colchicine in patients with MI are boosting the interest in novel therapies targeting inflammation in cardiovascular disease patients. The aim of this review is to discuss recent experimental studies that provide new insights into the signalling pathways and local regulators within the cardiac microenvironment promoting the resolution of inflammation and tissue regeneration. We will cover ischaemia- and non-ischaemic-induced as well as infection-related cardiac remodelling and address potential targets to prevent adverse cardiac remodelling.
Collapse
Affiliation(s)
- Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité, University Medicine Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Regenerative Medicine Center, Circulatory Health Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Via Paradisa, Pisa 56124, Italy
| |
Collapse
|
14
|
Kaesler N, Babler A, Floege J, Kramann R. Cardiac Remodeling in Chronic Kidney Disease. Toxins (Basel) 2020; 12:toxins12030161. [PMID: 32150864 PMCID: PMC7150902 DOI: 10.3390/toxins12030161] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
Cardiac remodeling occurs frequently in chronic kidney disease patients and affects quality of life and survival. Current treatment options are highly inadequate. As kidney function declines, numerous metabolic pathways are disturbed. Kidney and heart functions are highly connected by organ crosstalk. Among others, altered volume and pressure status, ischemia, accelerated atherosclerosis and arteriosclerosis, disturbed mineral metabolism, renal anemia, activation of the renin-angiotensin system, uremic toxins, oxidative stress and upregulation of cytokines stress the sensitive interplay between different cardiac cell types. The fatal consequences are left-ventricular hypertrophy, fibrosis and capillary rarefaction, which lead to systolic and/or diastolic left-ventricular failure. Furthermore, fibrosis triggers electric instability and sudden cardiac death. This review focuses on established and potential pathophysiological cardiorenal crosstalk mechanisms that drive uremia-induced senescence and disease progression, including potential known targets and animal models that might help us to better understand the disease and to identify novel therapeutics.
Collapse
Affiliation(s)
- Nadine Kaesler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Anne Babler
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Jürgen Floege
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
| | - Rafael Kramann
- Clinic for Renal and Hypertensive Disorders, Rheumatological and Immunological Disease, University Hospital of the RWTH Aachen, 52074 Aachen, Germany
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Correspondence:
| |
Collapse
|
15
|
S100 proteins in atherosclerosis. Clin Chim Acta 2020; 502:293-304. [DOI: 10.1016/j.cca.2019.11.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023]
|
16
|
Bian X, Su X, Wang Y, Zhao G, Zhang B, Li D. Periostin contributes to renal and cardiac dysfunction in rats with chronic kidney disease: Reduction of PPARα. Biochimie 2019; 160:172-182. [DOI: 10.1016/j.biochi.2019.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
|
17
|
Tammaro A, Florquin S, Brok M, Claessen N, Butter LM, Teske GJD, de Boer OJ, Vogl T, Leemans JC, Dessing MC. S100A8/A9 promotes parenchymal damage and renal fibrosis in obstructive nephropathy. Clin Exp Immunol 2018; 193:361-375. [PMID: 29746703 PMCID: PMC6150262 DOI: 10.1111/cei.13154] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Despite advances in our understanding of the mechanisms underlying the progression of chronic kidney disease and the development of fibrosis, only limited efficacious therapies exist. The calcium binding protein S100A8/A9 is a damage-associated molecular pattern which can activate Toll-like receptor (TLR)-4 or receptor for advanced glycation end-products (RAGE). Activation of these receptors is involved in the progression of renal fibrosis; however, the role of S100A8/A9 herein remains unknown. Therefore, we analysed S100A8/A9 expression in patients and mice with obstructive nephropathy and subjected wild-type and S100A9 knock-out mice lacking the heterodimer S100A8/A9 to unilateral ureteral obstruction (UUO). We found profound S100A8/A9 expression in granulocytes that infiltrated human and murine kidney, together with enhanced renal expression over time, following UUO. S100A9 KO mice were protected from UUO-induced renal fibrosis, independently of leucocyte infiltration and inflammation. Loss of S100A8/A9 protected tubular epithelial cells from UUO-induced apoptosis and critical epithelial-mesenchymal transition steps. In-vitro studies revealed S100A8/A9 as a novel mediator of epithelial cell injury through loss of cell polarity, cell cycle arrest and subsequent cell death. In conclusion, we demonstrate that S100A8/A9 mediates renal damage and fibrosis, presumably through loss of tubular epithelial cell contacts and irreversible damage. Suppression of S100A8/A9 could be a therapeutic strategy to halt renal fibrosis in patients with chronic kidney disease.
Collapse
Affiliation(s)
- A. Tammaro
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - S. Florquin
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - M. Brok
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - N. Claessen
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - L. M. Butter
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - G. J. D. Teske
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - O. J. de Boer
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - T. Vogl
- Institute of ImmunologyUniversity of MünsterMünsterGermany
| | - J. C. Leemans
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| | - M. C. Dessing
- Department of PathologyAmsterdam UMC, Univ(ersity) of AmsterdamAmsterdamthe Netherlands
| |
Collapse
|
18
|
Lim S, Lee ME, Jeong J, Lee J, Cho S, Seo M, Park S. sRAGE attenuates angiotensin II-induced cardiomyocyte hypertrophy by inhibiting RAGE-NFκB-NLRP3 activation. Inflamm Res 2018; 67:691-701. [DOI: 10.1007/s00011-018-1160-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 04/17/2018] [Accepted: 05/16/2018] [Indexed: 11/27/2022] Open
|
19
|
Xu X, Chen Z, Zhu X, Wang D, Liang J, Zhao C, Feng X, Wang J, Zou H, Sun L. S100A9 aggravates bleomycin-induced dermal fibrosis in mice via activation of ERK1/2 MAPK and NF-κB pathways. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:194-201. [PMID: 29456817 PMCID: PMC5811759 DOI: 10.22038/ijbms.2018.19987.5255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES This study aims to investigate the pathogenicity and possible mechanisms of S100A9 function in mice models of scleroderma. MATERIALS AND METHODS The content of S100A9 in the skin tissues of mice with scleroderma was determined. Different concentrations of bleomycin (BLM) and S100A9 were subcutaneously injected into the backs of mice simultaneously, and then pathological changes in the skin of these mice were monitored. Specifically, the levels of inflammatory cytokines and alpha smooth muscle actin (α-SMA), the activation of extracellular regulated kinase 1/2 (ERK1/2), mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways, and the expression of the receptor for advanced glycation end-product (RAGE) in the skin were determined. RESULTS The content of S100A9 in the skin tissues of mice with scleroderma was determined. Different concentrations of BLM and S100A9 were subcutaneously injected into the backs of mice simultaneously, and then pathological changes in the skin of these mice were monitored. Specifically, the levels of inflammatory cytokines and alpha smooth muscle actin (α-SMA), the activation of extracellular regulated kinase 1/2 (ERK1/2) mitogen-activated protein kinase (MAPK) and nuclear factor-kappa B (NF-κB) pathways, and the expression of the receptor for advanced glycation end-product (RAGE) in the skin were determined. CONCLUSION S100A9 aggravates dermal fibrosis in BLM-induced scleroderma (BIS) mice, and its mechanisms might be mediated by RAGE, ERK1/2, and NF-κB pathway.
Collapse
Affiliation(s)
- Xue Xu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Zhiyong Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Xiaoxia Zhu
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Cheng Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering and Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Hejian Zou
- Division of Rheumatology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| |
Collapse
|
20
|
Bansal S, Friedrichs WE, Velagapudi C, Feliers D, Khazim K, Horn D, Cornell JE, Werner SL, Fanti P. Spleen contributes significantly to increased circulating levels of fibroblast growth factor 23 in response to lipopolysaccharide-induced inflammation. Nephrol Dial Transplant 2018; 32:960-968. [PMID: 27836924 DOI: 10.1093/ndt/gfw376] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022] Open
Abstract
Background Circulating levels of fibroblast growth factor 23 (FGF23) increase progressively and correlate with systemic inflammation in chronic kidney disease (CKD). The aim of this study was to identify and characterize the causal relationship between FGF23 and inflammation in CKD. Methods Circulating FGF23 and inflammatory cytokines were correlated in healthy subjects and patients with varying levels of CKD. In addition, FGF23 expression in blood and solid organs was measured in normal mice that were exposed acutely (one time) or chronically (2-week) to low-dose lipopolysaccharide (LPS); chronic exposure being either sustained (subcutaneous pellets), intermittent (daily injections) or combined sustained plus acute (subcutaneous pellets plus acute injection on the day of sacrifice). Blood was analyzed for both terminal (cFGF23) and intact (iFGF23) FGF23 levels. Solid tissues were investigated with immunohistochemistry, enzyme-linked immunosorbent assay and reverse transcription polymerase chain reaction. Results FGF23 levels correlated significantly with neutrophil gelatinase-associated lipocalin ( r = 0.72, P < 0.001), C-reactive protein ( r = 0.38, P < 0.001), tumor necrosis factor-α ( r = 0.32, P = 0.001) and interleukin-6 ( r = 0.48, P < 0.001). Acute LPS administration increased tissue FGF23 mRNA and plasma levels of cFGF23 but not iFGF23. Neither chronic sustained nor chronic pulsatile LPS increased the tissue or circulating levels of FGF23. However, acute on chronic LPS raised tissue FGF23 mRNA and both circulating cFG23 and iFGF23. Interestingly, the spleen was the major source of FGF23. Conclusion Acute on chronic exposure to LPS stimulates FGF23 production in a normal mouse model of inflammation. We provide the first evidence that the spleen, under these conditions, contributes substantially to elevated circulating FGF23 levels.
Collapse
Affiliation(s)
- Shweta Bansal
- Division of Nephrology, Department of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA.,Renal Section, South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - William E Friedrichs
- Division of Nephrology, Department of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA
| | - Chakradhar Velagapudi
- Division of Nephrology, Department of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA.,Renal Section, South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - Denis Feliers
- Division of Nephrology, Department of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA
| | - Khaled Khazim
- Faculty of Medicine, Galilee Medical Center, Bar-Ilan University, Safed, Israel
| | - Diane Horn
- Department of Pathology, University of Texas Health Sciences Center at San Antonio and South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - John E Cornell
- Department of Epidemiology & Biostatistics, University of Texas Health Sciences Center at San Antonio, San Antonio, TX, USA
| | - Sherry L Werner
- Department of Pathology, University of Texas Health Sciences Center at San Antonio and South Texas Veterans Healthcare System, San Antonio, TX, USA
| | - Paolo Fanti
- Division of Nephrology, Department of Medicine, University of Texas Health Sciences Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
21
|
Richter B, Faul C. FGF23 Actions on Target Tissues-With and Without Klotho. Front Endocrinol (Lausanne) 2018; 9:189. [PMID: 29770125 PMCID: PMC5940753 DOI: 10.3389/fendo.2018.00189] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone whose physiologic actions on target tissues are mediated by FGF receptors (FGFR) and klotho, which functions as a co-receptor that increases the binding affinity of FGF23 for FGFRs. By stimulating FGFR/klotho complexes in the kidney and parathyroid gland, FGF23 reduces renal phosphate uptake and secretion of parathyroid hormone, respectively, thereby acting as a key regulator of phosphate metabolism. Recently, it has been shown that FGF23 can also target cell types that lack klotho. This unconventional signaling event occurs in an FGFR-dependent manner, but involves other downstream signaling pathways than in "classic" klotho-expressing target organs. It appears that klotho-independent signaling mechanisms are only activated in the presence of high FGF23 concentrations and result in pathologic cellular changes. Therefore, it has been postulated that massive elevations in circulating levels of FGF23, as found in patients with chronic kidney disease, contribute to associated pathologies by targeting cells and tissues that lack klotho. This includes the induction of cardiac hypertrophy and fibrosis, the elevation of inflammatory cytokine expression in the liver, and the inhibition of neutrophil recruitment. Here, we describe the signaling and cellular events that are caused by FGF23 in tissues lacking klotho, and we discuss FGF23's potential role as a hormone with widespread pathologic actions. Since the soluble form of klotho can function as a circulating co-receptor for FGF23, we also discuss the potential inhibitory effects of soluble klotho on FGF23-mediated signaling which might-at least partially-underlie the pleiotropic tissue-protective functions of klotho.
Collapse
|
22
|
Nazari A, Khorramdelazad H, Hassanshahi G, Day AS, Sardoo AM, Fard ET, Abedinzadeh M, Nadimi AE. S100A12 in renal and cardiovascular diseases. Life Sci 2017; 191:253-258. [PMID: 29080693 DOI: 10.1016/j.lfs.2017.10.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/12/2017] [Accepted: 10/25/2017] [Indexed: 12/21/2022]
Abstract
Expression of S100A12, a small calcium-binding protein, by neutrophils and monocytes/macrophages induces proinflammatory responses via ligation with the receptor for advanced glycation end-products (RAGE) and subsequent activation of intracellular signal transduction pathways such as the nuclear factor (NF)-κB pathway. Although S100A12 has been demonstrated to be a useful biomarker during inflammatory conditions, its precise role in the pathogenesis of renal and cardiovascular diseases has not been fully understood. Recently, several studies have employed S100A12 transgenic mice to investigate its pathological effects. Further studies using these models are required before we can translate these findings to human diseases such as renal and cardiovascular diseases.
Collapse
Affiliation(s)
- Alireza Nazari
- Department of Surgery, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Molecular Medicine Research Center, Rafsanjan University of Medical Science, Rafsanjan, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Rafsanjan University of Medical Science, Rafsanjan, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Andrew S Day
- Department of Pediatrics, University of Otago Christchurch, Christchurch, New Zealand
| | - Atlas Mashayekhi Sardoo
- School of Engineering and Design and Physical Sciences, Brunel University London, London, United Kingdom
| | | | - Mehdi Abedinzadeh
- Department of Urology, Shahid Rahnemoon Hospital, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Ali Esmaeili Nadimi
- Non Communicable Diseases Research Center, Rafsanjan University of Medical Science, Rafsanjan, Iran; Department of Cardiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
23
|
Joly P, Marshall JC, Tessier PA, Massé C, Page N, Frenette AJ, Khazoom F, Le Guillan S, Berthiaume Y, Charbonney E. S100A8/A9 and sRAGE kinetic after polytrauma; an explorative observational study. Scand J Trauma Resusc Emerg Med 2017; 25:114. [PMID: 29178941 PMCID: PMC5702249 DOI: 10.1186/s13049-017-0455-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 11/16/2017] [Indexed: 11/10/2022] Open
Abstract
Background Following tissue injury after trauma, the activation of innate immune pathways results in systemic inflammation, organ failure and an increased risk of infections. The objective of this study was to characterize the kinetics of the S100A8/S100A9 complex, a new-recognized alarmin, as well as its soluble receptor sRAGE, over time after trauma as potential early biomarkers of the risk of organ damage. Methods We collected comprehensive data from consenting patients admitted to an ICU following severe trauma. The blood samples were taken at Day 0 (admission), Day1, 3 and 5 S100A8/A9 and sRAGE were measured by ELISA. Biomarkers levels were reported as median (IQR). Results Thirty-eight patients sustaining in majority a blunt trauma (89%) with a median ISS of 39 were included. In this cohort, the S100A8/A9 complex increased significantly over time (p = 0.001), but its levels increment over time (D0 to D5) was significantly smaller in patients developing infection (7.6 vs 40.1 mcg/mL, p = 0.011). The circulating level of sRAGE circulating levels decreased over time (p < 0.0001) and was higher in patients who remained in shock on day 3 (550 vs 918 pg/mL; p = 0.02) or 5 (498 vs 644 pg/mL; p = 0.045). Admission sRAGE levels were significantly higher in non-survivors (1694 vs 745 pg/mL; p = 0.015) and was higher in patients developing renal failure (1143 vs 696 pg/mL, p = 0.011). Discussion Our findings reveal an interesting association between the biomarker S100A8/9 least increase over time and the presence of infectious complication after trauma. We describe that the sRAGE decline over time is in relation with shock and markers of ischemic injury. We also confirm the association of sRAGE levels measured at admission with mortality and the development of renal failure. Conclusions This work illustrates the importance of following the circulating level of biomarker overtime. The utilization of S1008/9 as a tool to stratify infection risk and trigger early interventions need to be validated prospectively.
Collapse
Affiliation(s)
- Philippe Joly
- Faculté de Médecine, Université de Montréal, Montréal, Canada
| | - John C Marshall
- St. Michael's Hospital and the Keenan Research Centre for Biomedical Science, University of Toronto, Toronto, Canada
| | - Philippe A Tessier
- Axe de recherche sur les maladies infectieuses et l'immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Chantal Massé
- Institut de recherches cliniques de Montréal, Université de Montréal, Montréal, Canada
| | - Nathalie Page
- Axe de recherche sur les maladies infectieuses et l'immunitaires, Centre de recherche du CHU de Québec-Université Laval, and Département de microbiologie-infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | | | | | | | - Yves Berthiaume
- Institut de recherches cliniques de Montréal, Université de Montréal, Montréal, Canada.,Département de médecine, Faculté de Médecine Université de Montréal, Montréal, Canada
| | - Emmanuel Charbonney
- Institut de recherches cliniques de Montréal, Université de Montréal, Montréal, Canada. .,Hôpital du Sacré-Coeur de Montréal, CIUSSS-NIM, Montréal, Canada. .,Département de médecine, Faculté de Médecine Université de Montréal, Montréal, Canada.
| |
Collapse
|
24
|
Jiang W, Wang Z, Hu Z, Wu H, Hu R, Hu X, Ren Z, Huang J. Blocking the ERK1/2 signal pathway can inhibit S100A12 induced human aortic smooth muscle cells damage. Cell Biol Int 2017; 41:1307-1315. [PMID: 28816402 DOI: 10.1002/cbin.10840] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/12/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Wanli Jiang
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Zhipeng Hu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Hongbing Wu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Rui Hu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Xiaoping Hu
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Zongli Ren
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| | - Jizhen Huang
- Department of Cardiovascular Surgery, Renmin Hospital; Wuhan University; Wuhan P.R. China
- Hubei Key Laboratory of Cardiology; Wuhan P.R. China
| |
Collapse
|
25
|
Abstract
Fibroblast growth factors (FGF) are mitogenic signal mediators that induce cell proliferation and survival. Although cardiac myocytes are post-mitotic, they have been shown to be able to respond to local and circulating FGFs. While precise molecular mechanisms are not well characterized, some FGF family members have been shown to induce cardiac remodeling under physiologic conditions by mediating hypertrophic growth in cardiac myocytes and by promoting angiogenesis, both events leading to increased cardiac function and output. This FGF-mediated physiologic scenario might transition into a pathologic situation involving cardiac cell death, fibrosis and inflammation, and eventually cardiac dysfunction and heart failure. As discussed here, cardiac actions of FGFs - with the majority of studies focusing on FGF2, FGF21 and FGF23 - and their specific FGF receptors (FGFR) and precise target cell types within the heart, are currently under experimental investigation. Especially cardiac effects of endocrine FGFs entered center stage over the past five years, as they might provide communication routes that couple metabolic mechanisms, such as bone-regulated phosphate homeostasis, or metabolic stress, such as hyperphosphatemia associated with kidney injury, with changes in cardiac structure and function. In this context, it has been shown that elevated serum FGF23 can directly tackle cardiac myocytes via FGFR4 thereby contributing to cardiac hypertrophy in models of chronic kidney disease, also called uremic cardiomyopathy. Precise characterization of FGFs and their origin and regulation of expression, and even more importantly, the identification of the FGFR isoforms that mediate their cardiac actions should help to develop novel pharmacological interventions for heart failure, such as FGFR4 inhibition to tackle uremic cardiomyopathy.
Collapse
Affiliation(s)
- Christian Faul
- Katz Family Drug Discovery Center, Division of Nephrology and Hypertension, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA; Department of Cell Biology and Anatomy, University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
26
|
Bagheri V, Geczy CL. Comment on “Potential Effects of Calcium Binding Protein S100A12 on Severity Evaluation and Curative Effect of Severe Acute Pancreatitis”. Inflammation 2017; 40:1811-1812. [DOI: 10.1007/s10753-017-0529-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
27
|
Takano APC, Munhoz CD, Moriscot AS, Gupta S, Barreto-Chaves MLM. S100A8/MYD88/NF-қB: a novel pathway involved in cardiomyocyte hypertrophy driven by thyroid hormone. J Mol Med (Berl) 2017; 95:671-682. [DOI: 10.1007/s00109-017-1511-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 01/02/2017] [Accepted: 01/18/2017] [Indexed: 01/25/2023]
|
28
|
Stabley JN, Towler DA. Arterial Calcification in Diabetes Mellitus: Preclinical Models and Translational Implications. Arterioscler Thromb Vasc Biol 2017; 37:205-217. [PMID: 28062508 PMCID: PMC5480317 DOI: 10.1161/atvbaha.116.306258] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus increasingly afflicts our aging and dysmetabolic population. Type 2 diabetes mellitus and the antecedent metabolic syndrome represent the vast majority of the disease burden-increasingly prevalent in children and older adults. However, type 1 diabetes mellitus is also advancing in preadolescent children. As such, a crushing wave of cardiometabolic disease burden now faces our society. Arteriosclerotic calcification is increased in metabolic syndrome, type 2 diabetes mellitus, and type 1 diabetes mellitus-impairing conduit vessel compliance and function, thereby increasing the risk for dementia, stroke, heart attack, limb ischemia, renal insufficiency, and lower extremity amputation. Preclinical models of these dysmetabolic settings have provided insights into the pathobiology of arterial calcification. Osteochondrogenic morphogens in the BMP-Wnt signaling relay and transcriptional regulatory programs driven by Msx and Runx gene families are entrained to innate immune responses-responses activated by the dysmetabolic state-to direct arterial matrix deposition and mineralization. Recent studies implicate the endothelial-mesenchymal transition in contributing to the phenotypic drift of mineralizing vascular progenitors. In this brief overview, we discuss preclinical disease models that provide mechanistic insights-and point to challenges and opportunities to translate these insights into new therapeutic strategies for our patients afflicted with diabetes mellitus and its arteriosclerotic complications.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Arteries/metabolism
- Arteries/pathology
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Angiopathies/etiology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Diet, High-Fat
- Disease Models, Animal
- Female
- Genetic Predisposition to Disease
- Humans
- Hyperlipidemias/complications
- Hyperlipidemias/genetics
- Male
- Phenotype
- Plaque, Atherosclerotic
- Rats
- Signal Transduction
- Translational Research, Biomedical
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- John N Stabley
- From the Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Dwight A Towler
- From the Division of Endocrinology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
29
|
Liu Y, Yu M, Zhang Z, Yu Y, Chen Q, Zhang W, Zhao X. Blockade of receptor for advanced glycation end products protects against systolic overload-induced heart failure after transverse aortic constriction in mice. Eur J Pharmacol 2016; 791:535-543. [PMID: 27393458 DOI: 10.1016/j.ejphar.2016.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 07/02/2016] [Accepted: 07/05/2016] [Indexed: 11/15/2022]
Abstract
Heart failure is the consequence of sustained, abnormal neurohormonal and mechanical stress and remains a leading cause of death worldwide. The aim of this work was to identify whether blockade of receptor for advanced glycation end products (RAGE) protected against systolic overload-induced heart failure and investigate the possible underlying mechanism. It was found that RAGE mRNA and protein expression was up-regulated in cardiac tissues from mice subjected to pressure overload by transverse aortic constriction (TAC). Importantly, inhibition of RAGE by treatment with soluble RAGE (sRAGE) or FPS-ZM1 (a high-affinity RAGE-specific inhibitor) for 8 weeks attenuated cardiac remodeling (including cardiac hypertrophy and fibrosis), and dysfunction in mice exposed to TAC. Furthermore, treatment of TAC mice with sRAGE or FPS-ZM1 enhanced phosphorylation of AMPK and reduced phosphorylation of mTOR and protein expression of NFκB p65 in cardiac tissues. In addition, treatment of TAC mice with sRAGE or FPS-ZM1 abated oxidative stress, attenuated endoplasmic reticulum stress, and suppressed inflammation in cardiac tissues. These data demonstrated the benefits of blocking RAGE on the progression of systolic overload-induced heart failure in mice, which was possibly through modulating AMPK/mTOR and NFκB pathways.
Collapse
Affiliation(s)
- Yu Liu
- Department of Cardiology, Jinling Hospital, Nanjing University, Zhongshan East Road 305, Nanjing 210002, China; Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Manli Yu
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhigang Zhang
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yunhua Yu
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qi Chen
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wei Zhang
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xianxian Zhao
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
30
|
Lin H, Shen L, Zhang X, Xie J, Hao H, Zhang Y, Chen Z, Yamamoto H, Liao W, Bin J, Cao S, Huang X, Liao Y. HMGB1-RAGE Axis Makes No Contribution to Cardiac Remodeling Induced by Pressure-Overload. PLoS One 2016; 11:e0158514. [PMID: 27355349 PMCID: PMC4927190 DOI: 10.1371/journal.pone.0158514] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/16/2016] [Indexed: 12/16/2022] Open
Abstract
High-mobility group box1 (HMGB1) exerts effects on inflammation by binding to receptor for advanced glycation end products (RAGE) or Toll-like receptor 4. Considering that inflammation is involved in pressure overload-induced cardiac hypertrophy, we herein attempted to investigate whether HMGB1 plays a role in myocardial hypertrophy in RAGE knockout mice as well as in the growth and apoptosis of cardiomyocytes. The myocardial expression of RAGE was not significantly changed while TLR4 mRNA was upregulated in response to transverse aortic constriction (TAC) for 1 week. The myocardial expression of HMGB1 protein was markedly increased in TAC group when compared to the sham group. Heart weight to body weight ratio (HW/BW) and lung weight to body weight ratio (LW/BW) were evaluated in RAGE knockout (KO) and wild-type (WT) mice 1 week after TAC. Significant larger HW/BW and LW/BW ratios were found in TAC groups than the corresponding sham groups, but no significant difference was found between KO and WT TAC mice. Similar results were also found when TAC duration was extended to 4 weeks. Cultured neonatal rat cardiomyocytes were treated with different concentrations of recombinant HMGB1, then cell viability was determined using MTT and CCK8 assays and cell apoptosis was determined by Hoechst staining and TUNEL assay. The results came out that HMGB1 exerted no influence on viability or apoptosis of cardiomyocytes. Besides, the protein expression levels of Bax and Bcl2 in response to different concentrations of HMGB1 were similar. These findings indicate that HMGB1 neither exerts influence on cardiac remodeling by binding to RAGE nor induces apoptosis of cardiomyocytes under physiological condition.
Collapse
Affiliation(s)
- Hairuo Lin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Liang Shen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Cardiology, first affiliated hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiajun Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiahe Xie
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huixin Hao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yingxue Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhenhuan Chen
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hiroshi Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shiping Cao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaobo Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- * E-mail:
| |
Collapse
|
31
|
Pham DL, Yoon MG, Ban GY, Kim SH, Kim MA, Ye YM, Shin YS, Park HS. Serum S100A8 and S100A9 Enhance Innate Immune Responses in the Pathogenesis of Baker's Asthma. Int Arch Allergy Immunol 2016; 168:138-46. [PMID: 26745257 DOI: 10.1159/000441678] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/12/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND S100A8 and S100A9 can be produced by lipopolysaccharide-stimulated granulocytes and provoke an innate immune-mediated airway inflammation. Involvement of S100A8 and S100A9 has been implicated in asthma. To further understand the role of S100A8 and S100A9 during innate immune responses in baker's asthma, we investigated the associations of serum S100A8 and S100A9 with exposure to bakery allergens and polymorphisms of the Toll-like receptor 4 (TLR4) gene. METHODS Totally, 381 bakery workers and 100 unexposed healthy controls were recruited. Skin prick tests for bakery allergens were performed. Serum levels of S100A8, S100A9, myeloperoxidase (MPO), tumor necrosis factor (TNF)-α, and interleukin (IL)-8 were measured using ELISA. Predictive values of serum S100A8 and S100A9 in bakery workers were evaluated by receiver-operating characteristic (ROC) curves. Polymorphisms of TLR4 -2027Ax2192;G and -1608Tx2192;C were genotyped. RESULTS Higher serum levels of S100A8 and S100A9 were noted in bakery workers compared to the normal controls (p < 0.001); however, no significant differences were noted according to work-related symptoms. The area under the ROC curve of serum S100A8 was 0.886 for occupational exposure (p < 0.001). The TLR4 -1608CC genotype was significantly associated with a higher serum S100A8 level (p = 0.025). Serum S100A8 and S100A9 levels were correlated with serum levels of MPO (r = 0.396 and 0.189, respectively), TNF-α (r = 0.536 and 0.280, respectively), and IL-8 (r = 0.540 and 0.205, respectively; p < 0.001 for all). CONCLUSION S100A8 and S100A9 are involved in innate immune responses under the regulation of TLR4 polymorphisms in baker's asthma pathogenesis. Serum S100A8 could be a potential biomarker for predicting occupational exposure to wheat flour in bakery workers.
Collapse
Affiliation(s)
- Duy Le Pham
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Pulskens WP, Verkaik M, Sheedfar F, van Loon EP, van de Sluis B, Vervloet MG, Hoenderop JG, Bindels RJ. Deregulated Renal Calcium and Phosphate Transport during Experimental Kidney Failure. PLoS One 2015; 10:e0142510. [PMID: 26566277 PMCID: PMC4643984 DOI: 10.1371/journal.pone.0142510] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/22/2015] [Indexed: 12/17/2022] Open
Abstract
Impaired mineral homeostasis and inflammation are hallmarks of chronic kidney disease (CKD), yet the underlying mechanisms of electrolyte regulation during CKD are still unclear. Here, we applied two different murine models, partial nephrectomy and adenine-enriched dietary intervention, to induce kidney failure and to investigate the subsequent impact on systemic and local renal factors involved in Ca(2+) and Pi regulation. Our results demonstrated that both experimental models induce features of CKD, as reflected by uremia, and elevated renal neutrophil gelatinase-associated lipocalin (NGAL) expression. In our model kidney failure was associated with polyuria, hypercalcemia and elevated urinary Ca(2+) excretion. In accordance, CKD augmented systemic PTH and affected the FGF23-αklotho-vitamin-D axis by elevating circulatory FGF23 levels and reducing renal αklotho expression. Interestingly, renal FGF23 expression was also induced by inflammatory stimuli directly. Renal expression of Cyp27b1, but not Cyp24a1, and blood levels of 1,25-dihydroxy vitamin D3 were significantly elevated in both models. Furthermore, kidney failure was characterized by enhanced renal expression of the transient receptor potential cation channel subfamily V member 5 (TRPV5), calbindin-D28k, and sodium-dependent Pi transporter type 2b (NaPi2b), whereas the renal expression of sodium-dependent Pi transporter type 2a (NaPi2a) and type 3 (PIT2) were reduced. Together, our data indicates two different models of experimental kidney failure comparably associate with disturbed FGF23-αklotho-vitamin-D signalling and a deregulated electrolyte homeostasis. Moreover, this study identifies local tubular, possibly inflammation- or PTH- and/or FGF23-associated, adaptive mechanisms, impacting on Ca(2+)/Pi homeostasis, hence enabling new opportunities to target electrolyte disturbances that emerge as a consequence of CKD development.
Collapse
Affiliation(s)
- Wilco P. Pulskens
- Dept. of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Dept. of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Melissa Verkaik
- Dept. of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Fareeba Sheedfar
- Dept. of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ellen P. van Loon
- Dept. of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bart van de Sluis
- Dept. of Pediatrics, Molecular Genetics Section, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark G. Vervloet
- Dept. of Nephrology, VU University Medical Center, Amsterdam, The Netherlands
| | - Joost G. Hoenderop
- Dept. of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - René J. Bindels
- Dept. of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | |
Collapse
|
33
|
Oesterle A, Bowman MAH. S100A12 and the S100/Calgranulins: Emerging Biomarkers for Atherosclerosis and Possibly Therapeutic Targets. Arterioscler Thromb Vasc Biol 2015; 35:2496-507. [PMID: 26515415 DOI: 10.1161/atvbaha.115.302072] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/14/2015] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is mediated by local and systematic inflammation. The multiligand receptor for advanced glycation end products (RAGE) has been studied in animals and humans and is an important mediator of inflammation and atherosclerosis. This review focuses on S100/calgranulin proteins (S100A8, S100A9, and S100A12) and their receptor RAGE in mediating vascular inflammation. Mice lack the gene for S100A12, which in humans is located on chromosome 3 between S100A8 and S100A9. Transgenic mice with smooth muscle cell-targeted expression of S100A12 demonstrate increased coronary and aortic calcification, as well as increased plaque vulnerability. Serum S100A12 has recently been shown to predict future cardiovascular events in a longitudinal population study, underscoring a role for S100A12 as a potential biomarker for coronary artery disease. Genetic ablation of S100A9 or RAGE in atherosclerosis-susceptible apolipoprotein E null mice results in reduced atherosclerosis. Importantly, S100A12 and the RAGE axis can be modified pharmacologically. For example, soluble RAGE reduces murine atherosclerosis and vascular inflammation. Additionally, a class of compounds currently in phase III clinical trials for multiple sclerosis and rheumatologic conditions, the quinoline-3-carboxamides, reduce atherosclerotic plaque burden and complexity in transgenic S100A12 apolipoprotein E null mice, but have not been tested with regards to human atherosclerosis. The RAGE axis is an important mediator for inflammation-induced atherosclerosis, and S100A12 has emerged as biomarker for human atherosclerosis. Decreasing inflammation by inhibiting S100/calgranulin-mediated activation of RAGE attenuates murine atherosclerosis, and future studies in patients with coronary artery disease are warranted to confirm S100/RAGE as therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Adam Oesterle
- From the Department of Medicine, The University of Chicago, IL
| | | |
Collapse
|
34
|
Maharaj A, Slusher AL, Zourdos MC, Whitehurst M, Fico BG, Huang CJ. Association of calprotectin with leukocyte chemotactic and inflammatory mediators following acute aerobic exercise. Appl Physiol Nutr Metab 2015; 41:83-7. [PMID: 26673245 DOI: 10.1139/apnm-2015-0385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The objective of this study was to examine whether acute aerobic exercise-mediated calprotectin in plasma would be associated with monocyte chemotactic protein-1 (MCP-1), myeloperoxidase (MPO), and interleukin-6 (IL-6) in healthy individuals. Eleven healthy participants, aged 18 to 30 years, were recruited to perform a 30-min bout of aerobic exercise at 75% maximal oxygen uptake. Acute aerobic exercise elicited a significant elevation across time in plasma calprotectin, MCP-1, MPO, and IL-6. Body mass index (BMI) was positively correlated with calprotectin area-under-the-curve with "respect to increase" (AUCi) and IL-6 AUCi. Furthermore, calprotectin AUCi was positively correlated with IL-6 AUCi and MPO AUCi, even after controlling for BMI. Although MPO AUCi was positively correlated with IL-6 AUCi, this relationship no longer existed after controlling for BMI. These results suggest that acute aerobic exercise could mediate innate immune response associated with calprotectin and its related leukocyte chemotactic and inflammatory mediators, especially in individuals with elevated BMI.
Collapse
Affiliation(s)
- Arun Maharaj
- a Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Aaron L Slusher
- a Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA.,b Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Michael C Zourdos
- a Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Michael Whitehurst
- a Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Brandon G Fico
- a Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Chun-Jung Huang
- a Exercise Biochemistry Laboratory, Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
35
|
Victorino VJ, Mencalha AL, Panis C. Post-translational modifications disclose a dual role for redox stress in cardiovascular pathophysiology. Life Sci 2015; 129:42-7. [DOI: 10.1016/j.lfs.2014.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/03/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023]
|
36
|
Deb A, Wang Y. Hypertrophic preconditioning: short-term tricks for long-term gain. Circulation 2015; 131:1468-70. [PMID: 25820337 DOI: 10.1161/circulationaha.115.016330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Arjun Deb
- From David Geffen School of Medicine, University of California, Los Angeles
| | - Yibin Wang
- From David Geffen School of Medicine, University of California, Los Angeles.
| |
Collapse
|
37
|
He YY, Yan W, Liu CL, Li X, Li RJ, Mu Y, Jia Q, Wu FF, Wang LL, He KL. Usefulness of S100A12 as a prognostic biomarker for adverse events in patients with heart failure. Clin Biochem 2015; 48:329-33. [DOI: 10.1016/j.clinbiochem.2014.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/13/2014] [Accepted: 11/18/2014] [Indexed: 01/04/2023]
|
38
|
Ligthart S, Sedaghat S, Ikram MA, Hofman A, Franco OH, Dehghan A. EN-RAGE. Arterioscler Thromb Vasc Biol 2014; 34:2695-9. [DOI: 10.1161/atvbaha.114.304306] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Symen Ligthart
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sanaz Sedaghat
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M. Arfan Ikram
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Oscar H. Franco
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Abbas Dehghan
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
39
|
Yan L, Bowman MAH. Chronic sustained inflammation links to left ventricular hypertrophy and aortic valve sclerosis: a new link between S100/RAGE and FGF23. INFLAMMATION AND CELL SIGNALING 2014; 1. [PMID: 26082935 DOI: 10.14800/ics.279] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cardiovascular disease including left ventricular hypertrophy, diastolic dysfunction and ectopic valvular calcification are common in patients with chronic kidney disease (CKD). Both S100A12 and fibroblast growth factor 23 (FGF23) have been identified as biomarkers of cardiovascular morbidity and mortality in patients with CKD. We tested the hypothesis that human S100/calgranulin would accelerate cardiovascular disease in mice subjected to CKD. METHODS This review paper focuses on S100 proteins and their receptor for advanced glycation end products (RAGE) and summarizes recent findings obtained in novel developed transgenic hBAC-S100 mice that express S100A12 and S100A8/9 proteins. A bacterial artificial chromosome of the human S100/calgranulin gene cluster containing the genes and regulatory elements for S100A8, S100A9 and S100A12 was expressed in C57BL/6J mice (hBAC-S100). CKD was induced by ureteral ligation, and hBAC-S100 mice and WT mice were studied after 10 weeks of chronic uremia. RESULTS hBAC-S100 mice with CKD showed increased FGF23 in the heart, left ventricular hypertrophy (LVH), diastolic dysfunction, focal cartilaginous metaplasia and calcification of the mitral and aortic valve annulus together with aortic valve sclerosis. This phenotype was not observed in WT mice with CKD or in hBAC-S100 mice lacking RAGE with CKD, suggesting that the inflammatory milieu mediated by S100/RAGE promotes pathological cardiac hypertrophy in CKD. In vitro, inflammatory stimuli including IL-6, TNFα, LPS, or serum from hBAC-S100 mice up regulated FGF23 mRNA and protein in primary murine neonatal and adult cardiac fibroblasts. CONCLUSIONS Taken together, our study shows that myeloid-derived human S100/calgranulin is associated with the development of cardiac hypertrophy and ectopic cardiac calcification in a RAGE dependent manner in a mouse model of CKD. We speculate that FGF23 produced by cardiac fibroblasts in response to cytokines may act in a paracrine manner to accelerate LVH and diastolic dysfunction in hBAC-S100 mice with CKD. We suggest that S100/RAGE-mediated chronic sustained systemic inflammation is linked to pathological cardiac remodeling via direct up regulation of FGF23 in cardiac fibroblasts, thereby providing a new mechanistic understanding for the common association between CKD, diabetes, metabolic syndrome, or hypertension with left ventricular hypertrophy with diastolic dysfunction.
Collapse
Affiliation(s)
- Ling Yan
- Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL 60637 USA
| | - Marion A Hofmann Bowman
- Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL 60637 USA
| |
Collapse
|
40
|
Local and systemic RAGE axis changes in pulmonary hypertension: CTEPH and iPAH. PLoS One 2014; 9:e106440. [PMID: 25188497 PMCID: PMC4154707 DOI: 10.1371/journal.pone.0106440] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/29/2014] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The molecular determinants of chronic thromboembolic pulmonary hypertension (CTEPH) and idiopathic pulmonary arterial hypertension (iPAH) remain poorly understood. The receptor for advanced glycation endproducts (RAGE) and its ligands: HMGB1 and S100A9 are involved in inflammatory disorders. We sought to investigate the role of the RAGE axis in patients with CTEPH undergoing pulmonary endarterectomy (PEA), iPAH undergoing lung transplantation (LuTX). The high pulmonary vascular resistance in CTEPH/iPAH results in pressure overload of the right ventricle. We compared sRAGE measurements to that of patients with aortic valve stenosis (AVS) - pressure overload of the left ventricle. METHODS We enrolled patients with CTEPH(26), iPAH(15), AVS(15) and volunteers(33). Immunohistochemistry with antibodies to RAGE and HMGB1 was performed on PEA specimens and lung tissues. We employed enzyme-linked immunosorbent assays to determine the concentrations of sRAGE, esRAGE, HMGB1 and S100A9 in serum of volunteers and patients with CTEPH, iPAH, AVS before and after PEA, LuTX and aortic valve replacement (AVR). RESULTS In endarterectomised tissues from patients with CTEPH RAGE and HMGB1 were identified in myofibroblasts (α-SMA+vimentin+CD34-), recanalizing vessel-like structures of distal myofibrotic tissues and endothelium of neointima. RAGE was differentially expressed in prototypical Heath Edwards lesions in iPAH. We found significantly increased serum concentrations of sRAGE, esRAGE and HMGB1 in CTEPH. In iPAH, sRAGE and esRAGE were significantly higher than in controls. Serum concentrations of sRAGE were significantly elevated in iPAH(p<0.001) and CTEPH(p = 0.001) compared to AVS. Serum sRAGE was significantly higher in iPAH compared to CTEPH(p = 0.042) and significantly reduced in AVS compared to controls(p = 0.001). There were no significant differences in sRAGE serum concentrations before and after surgical therapy for CTEPH, iPAH or AVS. CONCLUSIONS Our data suggest a role for the RAGE pathway in the pathophysiology of CTEPH and iPAH. PEA improves the local control of disease but may not influence the systemic inflammatory mechanisms in CTEPH patients through the RAGE pathway.
Collapse
|