1
|
Javier AJS, Kennedy FM, Yi X, Wehling-Henricks M, Tidball JG, White KE, Witczak CA, Kuro-O M, Welc SS. Klotho Is Cardioprotective in the mdx Mouse Model of Duchenne Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:923-940. [PMID: 39889824 PMCID: PMC12016860 DOI: 10.1016/j.ajpath.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, progressive skeletal and cardiac myopathy. Cardiomyopathy is the leading cause of death in patients with DMD, but the molecular basis for heart failure is incompletely understood. As with humans, in the mdx mouse model of DMD, cardiac function is impaired after the onset of skeletal muscle pathology. Dysregulation of Klotho gene regulation in dystrophic skeletal muscles occurs at disease onset, affecting pathogenesis. Whether Klotho is protective against dystrophin-deficient cardiomyopathy is unknown. This study found that expression of a Klotho transgene prevented deficits in left ventricular ejection fraction and fractional shortening in mdx mice. Improvements in cardiac performance were associated with reductions in adverse cardiac remodeling, cardiac myocyte hypertrophy, and fibrosis. In addition, mdx mice expressed high concentrations of plasma fibroblast growth factor 23 (FGF23), and expression was increased locally in hearts. The cardioprotective effects of Klotho were not associated with differences in renal function or serum biochemistries, but transgene expression prevented increased expression of plasma FGF23 and cardiac Fgf23 mRNA expression. Cardiac reactive oxygen species, oxidative damage, mitochondrial damage, and apoptosis were reduced in transgenic hearts. FGF23 stimulated hypertrophic growth in dystrophic neonatal mouse ventricular myocytes in vitro, which was inhibited by co-stimulation with soluble Klotho. Taken together, these results show that Klotho prevented dystrophic cardiac remodeling and improved function.
Collapse
MESH Headings
- Animals
- Klotho Proteins
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/physiopathology
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/genetics
- Glucuronidase/metabolism
- Glucuronidase/genetics
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/blood
- Fibroblast Growth Factors/genetics
- Mice, Inbred mdx
- Disease Models, Animal
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Mice, Transgenic
- Mice, Inbred C57BL
- Male
- Apoptosis
- Reactive Oxygen Species/metabolism
- Fibrosis
- Humans
- Myocardium/pathology
- Myocardium/metabolism
Collapse
Affiliation(s)
- Areli Jannes S Javier
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - Felicia M Kennedy
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xin Yi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Department of Pathology and Laboratory Medicine, University of California, Los Angeles, California
| | - Kenneth E White
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carol A Witczak
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Makoto Kuro-O
- Division of Anti-Aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Steven S Welc
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana; Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
2
|
Liu D, Yu S, Zhang Y, Li Q, Kang P, Wang L, Han R, Cheng D, Chen A, Hou X, Wu L, Zang S, Fang Q, Jia W, Li H. Fibroblast growth factor 23 predicts incident diabetic kidney disease: A 4.6-year prospective study. Diabetes Obes Metab 2025; 27:2232-2241. [PMID: 39895483 PMCID: PMC11885106 DOI: 10.1111/dom.16224] [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: 11/11/2024] [Revised: 01/11/2025] [Accepted: 01/19/2025] [Indexed: 02/04/2025]
Abstract
AIMS Fibroblast growth factor (FGF) 23 is a bone-derived phosphaturic hormone that participates in the regulation of mineral metabolism and the development of chronic kidney disease. This study aimed to investigate the association between FGF23 and diabetic kidney disease (DKD) in a community-based prospective cohort. MATERIALS AND METHODS Of 7230 individuals who completed a 4.6-year follow-up survey, 1614 individuals with diabetes at baseline were included in this study. Baseline serum FGF23 levels were measured by enzyme-linked immunosorbent assay. Multiple and ordinal logistic regression analyses were used to examine the predictive performance of baseline FGF23 for incident DKD. RESULTS Baseline serum FGF23 levels exhibited an earlier elevation in the course of DKD and a gradual increase with the progressive stages of DKD (p < 0.05), while no statistical changes were observed in serum calcium and phosphorus levels. Over a 4.6-year follow-up, 198 individuals with diabetes developed incident DKD. Baseline FGF23 was significantly associated with the incidence of DKD (odds ratio 1.290 [95% CI 1.063, 1.565]) after adjusting for conventional DKD risk factors, especially in individuals with lower body mass index (<24 kg/m2), worse glycaemic control (HbA1c ≥7%), and shorter duration of diabetes (<5 years). Moreover, FGF23 models exhibited great performances in DKD risk prediction and yielded increments compared to traditional DKD risk factors (p < 0.05). CONCLUSIONS Serum FGF23 level increased at early stages of DKD, and it was an independent predictor of incident DKD, suggesting its potential for early identification of individuals at risk.
Collapse
Affiliation(s)
- Dan Liu
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shujie Yu
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ying Zhang
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qian Li
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
| | - Piao Kang
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lihong Wang
- Department of Endocrinology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
| | - Rui Han
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Di Cheng
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Anran Chen
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
- Department of MedicineShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xuhong Hou
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
| | - Liang Wu
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
| | - Shufei Zang
- Department of Endocrinology, Shanghai Fifth People's HospitalFudan UniversityShanghaiChina
| | - Qichen Fang
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Institute for Proactive HealthcareShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
3
|
Adeoye M, Hamdallah H, Adeoye AM. Homocysteine levels and cardiovascular disease risk factors in chronic kidney disease (CKD), hypertensive and healthy Nigerian adults: a comparative retrospective study. BMJ Open 2025; 15:e089644. [PMID: 40074258 PMCID: PMC11904321 DOI: 10.1136/bmjopen-2024-089644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVES To investigate homocysteine (Hcy) levels in individuals with chronic kidney disease (CKD), hypertension and a healthy Nigerian population, and to assess their association with cardiovascular disease (CVD) risk. SETTING The study was conducted using data from the Ibadan CRECKID (Cardiovascular and Renal Event in People with Chronic Kidney Disease) study in Nigeria. PARTICIPANTS A total of 420 adults (aged 18+) categorised into three groups: individuals with stage 2 CKD or higher, hypertensive non-CKD individuals and normotensive individuals. OUTCOMES The primary outcome was the difference in serum Hcy levels across the groups; secondary outcomes included the prevalence of hyperhomocysteinaemia (HHcy) and correlation with fibroblast growth factor (FGF). RESULTS No significant difference in mean serum Hcy levels among the CKD, hypertensive and healthy groups (p=0.39) was observed. However, HHcy (≥15 µmol/L) prevalence was significantly higher in the hypertensive group (p<0.05). A strong positive correlation between Hcy levels and FGF was identified across all groups (p<0.001). CONCLUSIONS The present study indicates that Hcy levels may not serve as a reliable predictor of CVD outcomes across populations with varying kidney function and CVD risk profiles.
Collapse
Affiliation(s)
- Marvellous Adeoye
- School of Medicine, University of Chester, Chester, UK
- Institute of Public Health and Wellbeing, University of Essex-Colchester Campus, Colchester, UK
| | - Hanady Hamdallah
- Primary Care and Population Health, University College London, London, UK
| | | |
Collapse
|
4
|
Oliveira-Sousa J, Fragão-Marques M, Duarte-Gamas L, Ribeiro H, Rocha-Neves J. FGF-23 as a Biomarker for Carotid Plaque Vulnerability: A Systematic Review. Med Sci (Basel) 2025; 13:27. [PMID: 40137447 PMCID: PMC11943977 DOI: 10.3390/medsci13010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Carotid artery disease is a condition affecting 3% of the general population which significantly contributes to the development of cerebrovascular events. Fibroblast Growth Factor-23 (FGF-23) is a hormone that has been linked to atherosclerosis and increased cardiovascular risk, including stroke and myocardial infarction. This review explores the association of FGF-23 with carotid artery disease progression in an endarterectomy clinical context. METHODS Based on Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA), a search was performed relying on MEDLINE, Scopus and Web of Science, identifying publications focused on the correlation between serum FGF-23 and carotid artery disease. Assessment of study quality was made using National Heart, Lung and Blood Institute Study Quality Assessment Tool (NHLBI). RESULTS Three observational studies, comprising 1039 participants, were included. There was considerable heterogeneity among the populations from the different studies. Elevated FGF-23 levels were consistently associated with unstable plaque features, including intraplaque neovascularization, as identified through Superb Microvascular Imaging (SMI). Plasma levels of inflammatory mediators, such as Interleukin-6 (Il-6), Monocyte Chemoattractant Protein-1 (MCP-1), and Osteoprotegerin (OPG), positively correlated with carotid artery disease, but their link to unstable plaques is conflicting. None of the studies investigated clinical complications following carotid endarterectomy. CONCLUSIONS FGF-23 is a potential biomarker for plaque vulnerability in carotid disease. Despite promising findings, limitations such as small sample sizes and lack of longitudinal data suggest the need for larger and more diverse studies to improve risk stratification and inform personalized treatment strategies for carotid atherosclerosis.
Collapse
Affiliation(s)
- Joana Oliveira-Sousa
- RISE-Health, Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (M.F.-M.); (H.R.); (J.R.-N.)
| | - Mariana Fragão-Marques
- RISE-Health, Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (M.F.-M.); (H.R.); (J.R.-N.)
| | - Luís Duarte-Gamas
- Department of Angiology and Vascular Surgery, Local Health Unit Tâmega e Sousa, 4560-136 Penafiel, Portugal;
| | - Hugo Ribeiro
- RISE-Health, Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (M.F.-M.); (H.R.); (J.R.-N.)
- Faculty of Medicine, University of Coimbra, 3004-528 Coimbra, Portugal
- Community Palliative Care Team Gaia–Local Health Unit Gaia and Espinho, 4434-502 Vila Nova de Gaia, Portugal
- Coimbra Institute for Clinical and Biomedical Research, 3000-548 Coimbra, Portugal
| | - João Rocha-Neves
- RISE-Health, Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal; (M.F.-M.); (H.R.); (J.R.-N.)
| |
Collapse
|
5
|
Wu VC, Peng KY, Chen TI, Sun CY, Liao HW, Chan CK, Lin YH, Liou HH, Chueh JS. C-terminal FGF-23 production coupling with aldosterone via FAM20C and predicting cardiovascular events in primary aldosteronism. JCI Insight 2025; 10:e166461. [PMID: 39989455 PMCID: PMC11949054 DOI: 10.1172/jci.insight.166461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/08/2025] [Indexed: 02/25/2025] Open
Abstract
This study examined the involvement of fibroblast growth factor-23 (FGF-23) in primary aldosteronism (PA), a condition characterized by elevated aldosterone levels and hypertension. We recruited patients with unilateral PA (uPA) and observed increased levels of C-terminal FGF-23 (cFGF-23) and C-terminal to intact FGF-23 (iFGF-23) in patients with uPA compared with essential hypertension control participants. Elevated preoperative cFGF-23 levels were associated with adverse outcomes, including mortality and cardiovascular or kidney events. Plasma cFGF-23 levels demonstrated a nonlinear rise with aldosterone, but iFGF-23 levels were not correlated with plasma aldosterone concentration. Higher cFGF-23 levels independently predicted hypertension remission after adrenalectomy for patients with uPA. Patients with uPA, who exhibited elevated cFGF-23 levels, had decreased levels after adrenalectomy. In cell cultures, aldosterone enhanced cleavage of iFGF-23, leading to increased levels of cFGF-23 fragments, an effect mitigated by silencing of family with sequence similarity 20, member C (FAM20C). However, the enhancement of cFGF-23 levels remained unaffected by the furin inhibitor. The study suggests that aldosterone influences FGF-23 phosphorylation by interacting with FAM20C, with docking experiments indicating aldosterone's binding to FAM20C. This work highlights that patients with uPA with elevated cFGF-23 levels are associated with cardiovascular risks, and adrenalectomy reduces cFGF-23. Aldosterone likely promotes cFGF-23 production through FAM20C-mediated phosphorylation of iFGF-23.
Collapse
Affiliation(s)
| | | | - Tsu-I Chen
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiao-Yin Sun
- Division of Nephrology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Hung-Wei Liao
- Department of Internal Medicine, Wan-Fang Hospital, Taipei, Taiwan
| | - Chieh-Kai Chan
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Hsinchu City, Taiwan
| | | | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, Hsin-Jen Hospital, New Taipei City, Taiwan
| | - Jeff S. Chueh
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Fajkić A, Jahić R, Ejubović M, Đešević M, Ejubović AJ, Lepara O. The Trend of Changes in Adiponectin, Resistin, and Adiponectin-Resistin Index Values in Type 2 Diabetic Patients with the Development of Metabolic Syndrome. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1795. [PMID: 39596980 PMCID: PMC11596469 DOI: 10.3390/medicina60111795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: This study aimed to investigate the novel adiponectin-resistin (AR) index as a predictor of the development of metabolic syndrome (MetS) in individuals with type 2 diabetes mellitus (T2DM). MetS is common in T2DM and increases cardiovascular risk. Adiponectin and resistin, adipokines with opposing effects on insulin sensitivity and inflammation, make the AR index a potential marker for metabolic risk. Materials and Methods: This prospective observational study included 80 T2DM participants (ages 30-60) from Sarajevo, Bosnia and Herzegovina, over 24 months. The participants were divided into two groups: T2DM with MetS (n = 48) and T2DM without MetS (n = 32). Anthropometric data, biochemical analyses, and serum levels of adiponectin and resistin were measured at baseline and every six months. The AR index was calculated using the formula AR = 1 + log10(R) - 1 + log10(A), where R and A represent resistin and adiponectin concentrations. Logistic regression identified predictors of MetS. Results: T2DM patients who developed MetS showed a significant decline in adiponectin levels (40.19 to 32.49 ng/mL, p = 0.02) and a rise in resistin levels (284.50 to 315.21 pg/mL, p = 0.001). The AR index increased from 2.85 to 2.98 (p = 0.001). The AR index and resistin were independent predictors of MetS after 18 months, with the AR index showing a stronger predictive value (p = 0.007; EXP(B) = 1.265). Conclusions: The AR index is a practical marker for predicting MetS development in T2DM participants, improving metabolic risk stratification. Incorporating it into clinical assessments may enhance early detection and treatment strategies.
Collapse
Affiliation(s)
- Almir Fajkić
- Department of Pathophysiology, Faculty of Medicine, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina
| | - Rijad Jahić
- University Clinical Center Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Malik Ejubović
- Department of Internal Medicine, Cantonal Hospital Zenica, 72000 Zenica, Bosnia and Herzegovina; (M.E.); (A.J.E.)
| | - Miralem Đešević
- Department of Cardiology, Polyclinic Eurofarm Center Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Amira Jagodić Ejubović
- Department of Internal Medicine, Cantonal Hospital Zenica, 72000 Zenica, Bosnia and Herzegovina; (M.E.); (A.J.E.)
| | - Orhan Lepara
- Department of Human Physiology, Faculty of Medicine, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| |
Collapse
|
7
|
Métivier L, Vivien D, Goy R, Agin V, Bui E, Benbrika S. Plasminogen Activator Inhibitor-1 in the Pathophysiology of Late Life Depression. Int J Geriatr Psychiatry 2024; 39:e70015. [PMID: 39578639 DOI: 10.1002/gps.70015] [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: 05/16/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Abstract
INTRODUCTION Late life depression (LLD) is characterized by specific clinical features including a high frequency of vascular form and frequent antidepressant treatment resistance. The expression and functions of the serine protease inhibitor, Plasminogen Activator Inhibitor-1 (PAI-1) is known to be altered by aging, vascular damage, insulin levels associated with a sedentary lifestyle, chronic stress leading to hypercortisolemia, and inflammatory changes linked to stress responses. These phenomena would be implicated in LLD like vascular depression. This article thus aims to review the existing literature regarding the association between LLD and plasmatic levels of PAI-1, a marker of hypofibrinolysis. We hypothesize that increased age would be associated with changes in PAI-1 plasma level and function which influence LLD pathogenesis and its treatment. RESULTS Although a large number of studies on PAI-1 changes in the elderly exist, studies about its implications in LLD are sparse. Despite heterogeneous findings regarding the direction of variation in plasmatic PAI-1 levels among elderly participants with LLD, all studies demonstrated an association between PAI-1 levels and current or remitted depressive symptoms. Moreover, disruptions in the concentrations of other biological markers influencing PAI-1 expression, such as cytokines or adipokines, were also observed, notably an increase in the levels of interleukins 6 and 8. DISCUSSION LLD genesis appears to be influenced by PAI-1 regulatory loops which are implicated in senescence or cell death. The resistance to antidepressant treatment appears to be linked to distinct biological profiles involving inflammatory and fibrinolytic factors. Taken together these data suggest that PAI-1 pathway may be a promising target of treatment development efforts for LLD, and depression in general.
Collapse
Affiliation(s)
- L Métivier
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
| | - D Vivien
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - R Goy
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
| | - V Agin
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| | - E Bui
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
- CHU de CAEN Normandie, Service de Psychiatrie, Centre Esquirol, Caen, France
| | - S Benbrika
- INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Normandie University, UNICAEN, Caen, France
- Fédération Hospitalo-Universitaire (FHU A2M2P), GIP Cyceron, Normandie Univ, UNICAEN, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
- CHU de CAEN Normandie, Service de Psychiatrie, Centre Esquirol, Caen, France
| |
Collapse
|
8
|
Souissi A, Dergaa I, Hajri SE, Chamari K, Saad HB. A new perspective on cardiovascular function and dysfunction during endurance exercise: identifying the primary cause of cardiovascular risk. Biol Sport 2024; 41:131-144. [PMID: 39416509 PMCID: PMC11474989 DOI: 10.5114/biolsport.2024.134757] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/12/2023] [Accepted: 01/04/2024] [Indexed: 10/19/2024] Open
Abstract
Exercise mechanical efficiency typically falls within the range of approximately 20 to 25%. This means that a great part of the metabolic energy converted to generate movement is released as heat. Therefore, the rise in core temperature during endurance exercise in humans is proportional to generated work. Cutaneous vasodilation occurs when the core temperature threshold is reached. The rise in heart rate in response to thermal stress is a cardiovascular response that increases cardiac output and skin blood flow. The cardiovascular response during endurance exercise is a complex phenomenon potentially influenced by the involvement of nitric oxide in active thermoregulatory vasodilation. Excessive exercise can create high oxidative stress by disrupting the balance between free radicals' production and scavenging, resulting in impaired cardiovascular function. The above considerations are related to the severity and duration of endurance exercise. The first focus of this narrative review is to provide an updated understanding of cardiovascular function during endurance exercise. We aim to explore the potential role of oxidative stress in causing cardiovascular dysfunction during endurance exercise from a fresh perspective. Additionally, we aim to identify the primary factors contributing to cardiovascular risk during strenuous prolonged exercise by highlighting recent progress in this area, which may shed light on previously unexplained physiological responses. To ascertain the effect of endurance exercise on cardiovascular function and dysfunction, a narrative review of the literature was undertaken using PubMed, ScienceDirect, Medline, Google Scholar, and Scopus. The review highlighted that high oxidative stress (due to high levels of catecholamines, shear stress, immune system activation, and renal dysfunction) leads to a rise in platelet aggregation during endurance exercise. Importantly, we clearly revealed for the first time that endothelial damage, vasoconstriction, and blood coagulation (inducing thrombosis) are potentially the primary factors of cardiovascular dysfunction and myocardial infarction during and/or following endurance exercise.
Collapse
Affiliation(s)
- Amine Souissi
- Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie
| | - Ismail Dergaa
- Primary Health Care Corporation (PHCC), Doha, P.O. Box 26555, Qatar
| | - Samia Ernez Hajri
- Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie
| | - Karim Chamari
- High Institute of Sport and Physical Education of Ksar-Said, University of La Manouba, Tunis, Tunisia
- Naufar Wellness & Recovery Center, Doha, Qatar
| | - Helmi Ben Saad
- Université de Sousse, Faculté de Médecine de Sousse, Hôpital Farhat HACHED, Laboratoire de Recherche (Insuffisance Cardiaque, LR12SP09), Sousse, Tunisie
| |
Collapse
|
9
|
Mitterer W, Odri Komazec I, Huber E, Schaefer B, Posod A, Kiechl-Kohlendorfer U. Young hearts, early risks: novel cardiovascular biomarkers in former very preterm infants at kindergarten age. Pediatr Res 2024; 96:999-1005. [PMID: 38658663 PMCID: PMC11502516 DOI: 10.1038/s41390-024-03210-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/01/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Preterm birth is associated with long-term cardiovascular morbidity and mortality. In adults, fibroblast growth factor-23 (FGF-23), α-Klotho, and secretoneurin have all garnered attention as cardiovascular biomarkers, but their utility in pediatric populations has not yet been ascertained. The aim of this pilot study was to evaluate these novel cardiovascular biomarkers and their association with indicators of cardiovascular impairment in the highly vulnerable population of former very preterm infants. METHODS Five- to seven-year-old children born at < 32 weeks' gestation were eligible for the study. Healthy same-aged children born at term served as controls. Biomarkers were quantified in fasting blood samples, and echocardiographic measurements including assessment of aortic elastic properties were obtained. RESULTS We included 26 former very preterm infants and 21 term-born children in the study. At kindergarten age, former very preterm infants exhibited significantly higher plasma concentrations of biologically active intact FGF-23 (iFGF-23; mean 43.2 pg/mL vs. 29.1 pg/mL, p = 0.003) and secretoneurin (median 93.8 pmol/L vs. 70.5 pmol/L, p = 0.046). iFGF-23 inversely correlated with distensibility of the descending aorta. CONCLUSION In preterm-born children, iFGF-23 and secretoneurin both offer prospects as valuable cardiovascular biomarkers, potentially allowing for risk stratification and timely implementation of preventive measures. IMPACT Former very preterm infants have increased plasma concentrations of the novel cardiovascular biomarkers intact fibroblast growth factor-23 (iFGF-23) and secretoneurin at kindergarten age. Increases in iFGF-23 concentrations are associated with decreased distensibility of the descending aorta even at this early age. Monitoring of cardiovascular risk factors is essential in individuals with a history of preterm birth. Both iFGF-23 and secretoneurin hold promise as clinically valuable biomarkers for risk stratification, enabling the implementation of early preventive measures.
Collapse
Affiliation(s)
- Wolfgang Mitterer
- Department of Pediatrics II, Medical University of Innsbruck, Innsbruck, Austria
- VASCage GmbH, Research Centre on Vascular Ageing and Stroke, Innsbruck, Austria
| | - Irena Odri Komazec
- Department of Pediatrics III, Medical University of Innsbruck, Innsbruck, Austria
| | - Eva Huber
- Department of Pediatrics II, Medical University of Innsbruck, Innsbruck, Austria
| | - Benedikt Schaefer
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
| | - Anna Posod
- Department of Pediatrics II, Medical University of Innsbruck, Innsbruck, Austria.
| | | |
Collapse
|
10
|
Fang L, Zhang G, Wu Y, Li H, Li Z, Yu B, Wang B, Zhou L. Fibroblast growth factor 23 inhibition attenuates steroid-induced osteonecrosis of the femoral head through pyroptosis. Sci Rep 2024; 14:16270. [PMID: 39009650 PMCID: PMC11251279 DOI: 10.1038/s41598-024-66799-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024] Open
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is the predominant cause of non-traumatic osteonecrosis of the femoral head (ONFH). Impaired blood supply and reduced osteogenic activity of the femoral head are the key pathogenic mechanisms of SONFH. Fibroblast growth factor 23 (FGF23) levels are not only a biomarker for early vascular lesions caused by abnormal mineral metabolism, but can also act directly on the peripheral vascular system, leading to vascular pathology. The aim of this study was to observe the role of FGF23 on bone microarchitecture and vascular endothelium, and to investigate activation of pyroptosis in SONFH. Lipopolysaccharide (LPS) combined with methylprednisolone (MPS) was applied for SONFH mouse models, and adenovirus was used to increase or decrease the level of FGF23. Micro-CT and histopathological staining were used to observe the structure of the femoral head, and immunohistochemical staining was used to observe the vascular density. The cells were further cultured in vitro and placed in a hypoxic environment for 12 h to simulate the microenvironment of vascular injury during SONFH. The effect of FGF23 on osteogenic differentiation was evaluated using alkaline phosphatase staining, alizarin red S staining and expression of bone formation-related proteins. Matrigel tube formation assay in vitro and immunofluorescence were used to detect the ability of FGF23 to affect endothelial cell angiogenesis. Steroids activated the pyroptosis signaling pathway, promoted the secretion of inflammatory factors in SONFH models, led to vascular endothelial dysfunction and damaged the femoral head structure. In addition, FGF23 inhibited the HUVECs angiogenesis and BMSCs osteogenic differentiation. FGF23 silencing attenuated steroid-induced osteonecrosis of the femoral head by inhibiting the pyroptosis signaling pathway, and promoting osteogenic differentiation of BMSCs and angiogenesis of HUVECs in vitro.
Collapse
Affiliation(s)
- Lun Fang
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
- Medical School of Nanjing University, Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Gang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Shandong First Medical University, Taian, 271000, Shandong, People's Republic of China
| | - Yadi Wu
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Hao Li
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Zhongzhe Li
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Beilei Yu
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Bin Wang
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China
| | - Lu Zhou
- Institute of Sports Medicine, College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy Medical Sciences, 619 Changcheng Road, Taian, 271016, Shandong, People's Republic of China.
| |
Collapse
|
11
|
Chen J, Liu C, Sun C, Zeng J, Chi J, Che K, Wang Y. Association between Serum Phosphorus Levels and Diabetic Retinopathy: A Cross-Sectional Study. Int J Endocrinol 2024; 2024:3830246. [PMID: 38904033 PMCID: PMC11187971 DOI: 10.1155/2024/3830246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/22/2024] Open
Abstract
Background and Aims The aim of this study was to investigate the association between serum phosphate levels and diabetic retinopathy (DR) in patients with type 2 diabetes mellitus (T2DM). Methods and Results The study sample consisted of 1657 T2DM patients hospitalized between 2017 and 2019. Patients were categorized into quartiles based on their serum phosphate levels (Q1-Q4). An increasing trend in the prevalence of DR was observed across these quartiles. Subsequently, logistic regression analysis was employed to adjust for potential confounders, such as gender, age, BMI, and duration of diabetes, and to evaluate the odds ratios (ORs) associated with these quartiles. The prevalence of DR showed an increasing trend with elevated serum phosphate levels. Logistic regression further confirmed that serum phosphate levels remain an independent risk factor for DR. Conclusion Elevated serum phosphate levels are closely associated with the prevalence of DR in hospitalized T2DM patients. Further studies are needed to establish causality. This trial is registered with chiCTR2000032374.
Collapse
Affiliation(s)
- Jintao Chen
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chuanfeng Liu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cunwei Sun
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Zeng
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingwei Chi
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kui Che
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yangang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Pellicano C, Colalillo A, De Marco O, Carnazzo V, Basile U, Gigante A, Cianci R, Rosato E. Iloprost infusion reduces serological cytokines and hormones of hypoxia and inflammation in systemic sclerosis patients. Clin Exp Med 2024; 24:109. [PMID: 38777916 PMCID: PMC11111538 DOI: 10.1007/s10238-024-01374-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is characterized by microvascular damage of skin and internal organs with chronic hypoxia and release of cytokines and hormones such as neutrophil gelatinase-associated lipocalin (NGAL), fibroblast growth factor-23 (FGF-23) and Klotho. Aim of the study was to evaluate FGF-23, Klotho and NGAL serum levels in SSc patients and healthy controls (HC) and to evaluate serum levels changes of FGF-23, Klotho and NGAL after Iloprost. METHODS Twenty-one SSc patients and 20 HC were enrolled. In SSc patients, peripheral venous blood samples were collected at the first day before the autumn Iloprost infusion (t0), 60 min (t1) and 14 days after Iloprost infusion (t2). RESULTS SSc patients had higher serum level of FGF-23 [18.7 ± 6.4 pg/ml versus 3.6 ± 2.2 pg/ml, p < 0.001], Klotho [5.1 ± 0.8 pg/ml versus 2.3 ± 0.6 pg/ml, p < 0.001] and NGAL [20.9 ± 2.6 pg/ml versus 14.5 ± 1.7 pg/ml, p < 0.001] than HC. Iloprost infusion reduces serum level of FGF-23 (18.7 ± 6.4 pg/ml versus 10.4 ± 5.5 pg/ml, p < 0.001), Klotho (5.1 ± 0.8 pg/ml versus 2.5 ± 0.6 pg/ml, p < 0.001) and NGAL (20.9 ± 2.6 pg/ml versus 15.1 ± 2.3 pg/ml, p < 0.001) between t0 and t1. The Iloprost infusion reduces serum level of FGF-23 (18.7 ± 6.4 pg/ml versus 6.6 ± 5.1 pg/ml), Klotho (5.1 ± 0.8 pg/ml versus 2.3 ± 0.4 pg/ml) and NGAL (20.9 ± 2.6 pg/ml versus 15.5 ± 1.9 pg/ml) between t0 and t2. CONCLUSIONS SSc patients had higher FGF-23, Klotho and NGAL than HC. Iloprost reduces serum levels of FGF-23, Klotho and NGAL.
Collapse
Affiliation(s)
- Chiara Pellicano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Amalia Colalillo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Oriana De Marco
- Department of Public Health, Nephrology Unit, University Federico II, 80138, Naples, Italy
| | - Valeria Carnazzo
- UOC of Clinical Pathology DEA II Level, Hospital Santa Maria Goretti-ASL Latina, 04100, Latina, Italy
| | - Umberto Basile
- UOC of Clinical Pathology DEA II Level, Hospital Santa Maria Goretti-ASL Latina, 04100, Latina, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Rosario Cianci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale Dell'Università 37, 00185, Rome, Italy.
| |
Collapse
|
13
|
Curaj A, Vanholder R, Loscalzo J, Quach K, Wu Z, Jankowski V, Jankowski J. Cardiovascular Consequences of Uremic Metabolites: an Overview of the Involved Signaling Pathways. Circ Res 2024; 134:592-613. [PMID: 38422175 DOI: 10.1161/circresaha.123.324001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
The crosstalk of the heart with distant organs such as the lung, liver, gut, and kidney has been intensively approached lately. The kidney is involved in (1) the production of systemic relevant products, such as renin, as part of the most essential vasoregulatory system of the human body, and (2) in the clearance of metabolites with systemic and organ effects. Metabolic residue accumulation during kidney dysfunction is known to determine cardiovascular pathologies such as endothelial activation/dysfunction, atherosclerosis, cardiomyocyte apoptosis, cardiac fibrosis, and vascular and valvular calcification, leading to hypertension, arrhythmias, myocardial infarction, and cardiomyopathies. However, this review offers an overview of the uremic metabolites and details their signaling pathways involved in cardiorenal syndrome and the development of heart failure. A holistic view of the metabolites, but more importantly, an exhaustive crosstalk of their known signaling pathways, is important for depicting new therapeutic strategies in the cardiovascular field.
Collapse
Affiliation(s)
- Adelina Curaj
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Raymond Vanholder
- Department of Internal Medicine and Pediatrics, Nephrology Section, University Hospital, Ghent, Belgium (R.V.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Kaiseng Quach
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Zhuojun Wu
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Vera Jankowski
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, RWTH Aachen University, Germany (A.C., K.Q., Z.W., V.J., J.J.)
- Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands (J.J.)
- Aachen-Maastricht Institute for Cardiorenal Disease, RWTH Aachen University, Aachen, Germany (J.J.)
| |
Collapse
|
14
|
Huang HJ, Hsu BG, Wang CH, Tsai JP, Chen YH, Hung SC, Lin YL. Diabetes mellitus modifies the association between chronic kidney disease-mineral and bone disorder biomarkers and aortic stiffness in peritoneal dialysis patients. Sci Rep 2024; 14:4554. [PMID: 38402283 PMCID: PMC10894213 DOI: 10.1038/s41598-024-55364-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 02/22/2024] [Indexed: 02/26/2024] Open
Abstract
This study aimed to investigate the relationship of four chronic kidney disease-mineral and bone disorder (CKD-MBD) biomarkers, including intact parathyroid hormone (PTH), fibroblast growth factor 23 (FGF23), soluble klotho, and fetuin-A, with aortic stiffness in peritoneal dialysis (PD) patients, comparing those with and without diabetes mellitus (DM). A total of 213 patients (mean age 58 ± 14 years; 81 (38.0%) patients with DM) were enrolled. Their aortic pulse wave velocity (PWV) was measured using pressure applanation tonometry, while serum intact PTH, FGF23, α-klotho, and fetuin-A levels were measured using enzyme-linked immunosorbent assay. Overall, patients with DM had higher aortic PWV than those without (9.9 ± 1.8 vs. 8.6 ± 1.4 m/s, p < 0.001). Among the four CKD-MBD biomarkers, FGF23 levels were significantly lower in DM group (462 [127-1790] vs. 1237 [251-3120] pg/mL, p = 0.028) and log-FGF23 independently predicted aortic PWV in DM group (β: 0.61, 95% confidence interval: 0.06-1.16, p = 0.029 in DM group; β: 0.10, 95% confidence interval: - 0.24-0.45, p = 0.546 in nonDM group; interaction p = 0.016). In conclusion, the association between FGF23 and aortic PWV was significantly modified by DM status in PD patients.
Collapse
Affiliation(s)
- Hsiang-Jung Huang
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
| | - Bang-Gee Hsu
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
| | - Chih-Hsien Wang
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
| | - Yi-Hsin Chen
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 40201, Taiwan
| | - Szu-Chun Hung
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taipei, 23142, Taiwan
| | - Yu-Li Lin
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan.
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan.
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan.
| |
Collapse
|
15
|
Kirkman DL, Chavez DA. Exercise for chronic kidney disease: effects on vascular and cardiopulmonary function. Am J Physiol Heart Circ Physiol 2024; 326:H138-H147. [PMID: 37975707 DOI: 10.1152/ajpheart.00400.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Patients with chronic kidney disease (CKD) have an exacerbated prevalence of cardiovascular disease (CVD). Vascular dysfunction, characterized by impaired endothelial function and arterial stiffness, and markedly low cardiorespiratory fitness levels are hallmark manifestations of the disease that contribute to the CVD burden. Despite advancements in blood pressure and lipid lowering pharmacological therapies, CVD remains markedly prevalent across the spectrum of CKD. This highlights a stagnation in effective clinical strategies to improve cardiovascular health and reinforces the critical need for adjuvant lifestyle strategies such as physical activity and exercise training to be incorporated into routine clinical care. This narrative review provides an overview of the known effects of exercise on vascular and cardiopulmonary function across the spectrum of CKD. The physiological mechanisms of vascular dysfunction that serve as exercise-specific therapeutic targets are highlighted and future perspectives are discussed.
Collapse
Affiliation(s)
- Danielle L Kirkman
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, United States
| | - Domenico A Chavez
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, United States
| |
Collapse
|
16
|
Edmonston D, Grabner A, Wolf M. FGF23 and klotho at the intersection of kidney and cardiovascular disease. Nat Rev Cardiol 2024; 21:11-24. [PMID: 37443358 DOI: 10.1038/s41569-023-00903-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Cardiovascular disease is the leading cause of death in patients with chronic kidney disease (CKD). As CKD progresses, CKD-specific risk factors, such as disordered mineral homeostasis, amplify traditional cardiovascular risk factors. Fibroblast growth factor 23 (FGF23) regulates mineral homeostasis by activating complexes of FGF receptors and transmembrane klotho co-receptors. A soluble form of klotho also acts as a 'portable' FGF23 co-receptor in tissues that do not express klotho. In progressive CKD, rising circulating FGF23 levels in combination with decreasing kidney expression of klotho results in klotho-independent effects of FGF23 on the heart that promote left ventricular hypertrophy, heart failure, atrial fibrillation and death. Emerging data suggest that soluble klotho might mitigate some of these effects via several candidate mechanisms. More research is needed to investigate FGF23 excess and klotho deficiency in specific cardiovascular complications of CKD, but the pathophysiological primacy of FGF23 excess versus klotho deficiency might never be precisely resolved, given the entangled feedback loops that they share. Therefore, randomized trials should prioritize clinical practicality over scientific certainty by targeting disordered mineral homeostasis holistically in an effort to improve cardiovascular outcomes in patients with CKD.
Collapse
Affiliation(s)
- Daniel Edmonston
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander Grabner
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Myles Wolf
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
17
|
Khreba N, Khedr D, Abdel-Baky A, Kannishy GE, Samaan E. Nephron index rather than serum FGF 23 predicts endothelial dysfunction in early but not advanced chronic kidney disease patients. Int Urol Nephrol 2023; 55:3159-3165. [PMID: 37043155 PMCID: PMC10611818 DOI: 10.1007/s11255-023-03589-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/03/2023] [Indexed: 04/13/2023]
Abstract
BACKGROUND Endothelial dysfunction is the primary step for the development of CKD-related cardiovascular disease. Early prediction and management can influence patient survival. Serum testing of FGF 23 hormone and urinary phosphate excretion were studied as predictors of all-cause cardiovascular morbidity in CKD patients; however, their relation to endothelial dysfunction is controversial. A combination of both in one index is hypothesized to increase their sensitivity in detecting endothelial dysfunction, especially in the early stages of CKD before the dominance of hyperphosphatemia, the original risk. METHODS A cross-sectional comparative analysis between thirty CKD stage 3 patients and sixty stage 4-5 CKD patients was conducted. All patients were tested for markers of mineral bone disorders including serum FGF 23 and 24-h urinary phosphate excretion. A combination of both in one index (nephron index) is calculated and hypothesized to correlate with nephron number. Endothelial dysfunction was assessed by measuring the post-occlusion brachial flow-mediated dilatation (FMD). RESULTS In univariate and multivariate regression analyses, the nephron index was the only predictor of endothelial dysfunction in individuals with stage 3 CKD (r = 0.74, P 0.01). This was not applied to stage 4-5 CKD patients where serum phosphorus (r = - 0.53, P 0.001), intact PTH (r = - 0.53, P 0.001), uric acid (r = - 0.5, P 0.001), and measured GFR (r = 0.59, P 0.001) were the highest correlates to FMD; the Nephron index had the weakest correlation (r = 0.28, P = 0.02) and is not predictive of endothelial dysfunction. CONCLUSION Nephron index calculation showed better correlation with endothelial dysfunction than using any of its determinants alone in early stages of CKD when FGF 23 levels are just beginning to rise. In advanced CKD patients, hyperphosphatemia, hyperparathyroidism, hyperuricemia, and measured GFR are more reliable than nephron index.
Collapse
Affiliation(s)
- Nora Khreba
- Mansoura Nephrology and Dialysis Unit, Mansoura Faculty of Medicine, Internal Medicine Depament, Mansoura University, El Gomhoria St., Mansoura, 35516, Egypt
| | - Doaa Khedr
- Diagnostic and Interventional Radiology Department, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Azza Abdel-Baky
- Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura, Egypt
| | - Ghada El Kannishy
- Mansoura Nephrology and Dialysis Unit, Mansoura Faculty of Medicine, Internal Medicine Depament, Mansoura University, El Gomhoria St., Mansoura, 35516, Egypt
| | - Emad Samaan
- Mansoura Nephrology and Dialysis Unit, Mansoura Faculty of Medicine, Internal Medicine Depament, Mansoura University, El Gomhoria St., Mansoura, 35516, Egypt.
| |
Collapse
|
18
|
Natsuki Y, Morioka T, Kakutani Y, Yamazaki Y, Ochi A, Kurajoh M, Mori K, Imanishi Y, Shoji T, Inaba M, Emoto M. Serum Fibroblast Growth Factor 23 Levels are Associated with Vascular Smooth Muscle Dysfunction in Type 2 Diabetes. J Atheroscler Thromb 2023; 30:1838-1848. [PMID: 37225519 DOI: 10.5551/jat.64000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023] Open
Abstract
AIM Increased level of serum fibroblast growth factor 23 (FGF23) is a hallmark of abnormal phosphate metabolism in patients with chronic kidney disease (CKD) and is recently shown to be associated with the risk of cardiovascular disease even in those without CKD. This study investigated the association between serum FGF23 levels and vascular function in patients with type 2 diabetes. METHODS This was a cross-sectional study involving 283 Japanese patients with type 2 diabetes. Flow-mediated dilatation (FMD) and nitroglycerin-mediated dilatation (NMD) of the brachial artery were measured via ultrasonography to evaluate vascular endothelial and smooth muscle functions, respectively. Serum intact FGF23 levels were determined via a sandwich enzyme-linked immunosorbent assay. RESULTS The median values of FMD, NMD, and serum FGF23 were 6.0%, 14.0%, and 27.3 pg/mL, respectively. The serum FGF23 levels were inversely associated with NMD but not with FMD, and the association was independent of atherosclerotic risk factors, estimated glomerular filtration rate (eGFR), and serum phosphate levels. Furthermore, the relationship between serum FGF23 levels and NMD was modified by kidney function, which was pronounced in subjects with normal kidney function (eGFR ≥ 60 mL/min/1.73 m2). CONCLUSION Serum FGF23 levels are independently and inversely associated with NMD in patients with type 2 diabetes, particularly in those with normal kidney function. Our results indicate that FGF23 is involved in vascular smooth muscle dysfunction and that increased serum levels of FGF23 may serve as a novel biomarker for vascular smooth muscle dysfunction in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Yuka Natsuki
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Yoshinori Kakutani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Yuko Yamazaki
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Akinobu Ochi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Masafumi Kurajoh
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Katsuhito Mori
- Department of Nephrology, Osaka Metropolitan University Graduate School of Medicine
| | - Yasuo Imanishi
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka Metropolitan University Graduate School of Medicine
- Vascular Science Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine
| | - Masaaki Inaba
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
| | - Masanori Emoto
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine
- Vascular Science Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine
| |
Collapse
|
19
|
Wu KL, Chen CL, Thi Nguyen MH, Tsai JC, Wang SC, Chiang WF, Hsiao PJ, Chan JS, Hou JJ, Ma N. MicroRNA regulators of vascular pathophysiology in chronic kidney disease. Clin Chim Acta 2023; 551:117610. [PMID: 37863246 DOI: 10.1016/j.cca.2023.117610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Coronary artery disease (CAD) is a severe comorbidity in chronic kidney disease (CKD) due to heavy calcification in the medial layer and inflamed plaques. Chronic inflammation, endothelial dysfunction and vascular calcification are major contributors that lead to artherosclerosis in CKD. The lack of specific symptoms and signs of CAD and decreased accuracy of noninvasive diagnostic tools result in delayed diagnosis leading to increased mortality. MicroRNAs (miRNAs) are post-transcriptional regulators present in various biofluids throughout the body. In the circulation, miRNAs have been reported to be encapsulated in extracellular vesicles and serve as stable messengers for crosstalk among cells. miRNAs are involved in pathophysiologic mechanisms including CAD and can potentially be extended from basic research to clinical translational practice.
Collapse
Affiliation(s)
- Kun-Lin Wu
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Lung Chen
- Division of Nephrology, Department of Medicine, Landseed International Hospital, Taoyuan, Taiwan
| | - Mai-Huong Thi Nguyen
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Jen-Chieh Tsai
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan; Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Taiwan
| | - Sun-Chong Wang
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan
| | - Wen-Fang Chiang
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Po-Jen Hsiao
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jenq-Shyong Chan
- Division of Nephrology, Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ju Jung Hou
- Kaohsiung Medical University Hospital, Department of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Nianhan Ma
- Department of Biomedical Sciences and Engineering, Institute of Systems Biology and Bioinformatics, National Central University, Taoyuan, Taiwan.
| |
Collapse
|
20
|
Rabkin SW. Collagen type IV as the link between arterial stiffness and dementia. Am J Transl Res 2023; 15:5961-5971. [PMID: 37969177 PMCID: PMC10641358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 03/14/2023] [Indexed: 11/17/2023]
Abstract
Arterial stiffness has been linked to impaired cognitive function and dementia but the reason for the association is uncertain. This review proposes that collagen type IV is a critical factor linking arterial stiffness and dementia. Several genome wide association studies have related arterial stiffness to Collagen type IVα. Proteomic studies of arteries, demonstrated higher levels of collagen IVα1 in persons with high arterial stiffness. Collagen type IV defects are associated genetic causes of dementia as well as dementia of a variety of other causes. There are plausible causal roles for collagen type IV in dementia. Disorders of Collagen type IV can produce (I) fibro-hyalinosis and elastosis of small arterioles leading to cerebral ischemia and infarction; (II) dysfunction of the blood brain barrier leading to cerebral hemorrhage; (III) carotid artery stiffness with increase pulse pressure induces cerebral blood vessel damage leading to cerebral atrophy. The mechanisms by which Collagen type IV can lead to vascular stiffness include its degradation by matrix metalloprotease type 2 that (a) stimulates vascular smooth muscle cells to produce more extracellular matrix or (b) liberates peptides that damage the subendothelial space. Factors, such as TGF-β1, and LDL cholesterol especially oxidized LDL can increase collagen type IV and produce vascular stiffness and dementia. Fibroblast growth factor 23, and abnormal NO signaling have been linked to collagen type IV or increased vascular stiffness and an increased risk of dementia. Recognition of the central role of collagen type IV in arterial stiffness and dementia will inspire new research focused on determining whether its modification can benefit arterial and brain health.
Collapse
Affiliation(s)
- Simon W Rabkin
- Department of Medicine, University of British Columbia Vancouver, B.C., Canada
| |
Collapse
|
21
|
Mattinzoli D, Turolo S, Ikehata M, Vettoretti S, Montini G, Agostoni C, Conti C, Benedetti M, Messa P, Alfieri CM, Castellano G. MCP1 Inverts the Correlation between FGF23 and Omega 6/3 Ratio: Is It Also True in Renal Transplantation? J Clin Med 2023; 12:5928. [PMID: 37762869 PMCID: PMC10532002 DOI: 10.3390/jcm12185928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
During chronic kidney disease (CKD) progression, an increase in fibroblast growth factor (FGF23) is present. In stage 5, a positive correlation between FGF23 and omega-6 (n-6) polyunsaturated fatty acids (PUFAs) emerges. Hypothesizing that the rising positive correlation between monocyte chemoattractant protein 1 (MCP1) and n-6 in stage 4 could be the cause, we previously explored FGF23 and MCP1's roles in dyslipidemia and cardiovascular risk in CKD. In the present paper, we retraced the study evaluating 40 kidney transplant patients (KTx), a cohort where several factors might modify the previous relationships found. An ELISA and gas chromatography assessed the MCP1, FGF23, and PUFA levels. Despite the FGF23 increase (p < 0.0001), low MCP1 levels were found. A decrease in the n-6/n-3 ratio (p = 0.042 CKD stage 4 vs. 5) lowered by the increase in both n-3 αlinolenic (p = 0.012) and docosapentaenoic acid (p = 0.049) was observed. A negative correlation between FGF23 and the n-6/n-3 ratio in CKD stage 4 (r2 -0.3 p = 0.043) and none with MCP1 appeared. According to our findings, different mechanisms in the relationship between FGF23, PUFAs, and MCP1 in CKD and KTx patients might be present, which is possibly related to the immunosuppressive status of the last. Future research will further clarify our hypothesis.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefano Turolo
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Masami Ikehata
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Simone Vettoretti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo Agostoni
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Pediatric-Immunorheumatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Costanza Conti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, 20157 Milan, Italy
| | - Matteo Benedetti
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Post-Graduate School of Specialization in Nephrology, University of Milan, 20157 Milan, Italy
| | - Piergiorgio Messa
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Carlo Maria Alfieri
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Giuseppe Castellano
- Department of Nephrology, Dialysis and Renal Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
22
|
Wu C, Yu C, Yang Y, Jin H. Heart failure in erythrodermic psoriasis: a retrospective study of 225 patients. Front Cardiovasc Med 2023; 10:1169474. [PMID: 37593148 PMCID: PMC10427504 DOI: 10.3389/fcvm.2023.1169474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023] Open
Abstract
Purpose Erythrodermic psoriasis (EP) is a severe form of psoriasis that affects multiple organs, including the cardiovascular system. However, few studies have focused on this condition.This study is aimed to assess the prevalence and factors associated with heart failure in EP patient, and to the measure the serum concentrations of fibroblast growth factor 23 (FGF23), a potential predictor of chronic heart failure. Methods We retrospectively studied patients with EP hospitalized at Peking Union Medical College Hospital between January 2005 to October 2021. The prevalence of heart failure and associated factors was measured. In addition, peripheral blood samples were collected from 17 patients and matched with samples from eight healthy controls, and their serum concentrations of FGF23 were measured by ELISA. Results We studied 225 patients with EP, with a male: female ratio of 2.7:1 and a mean age of 47.6 ± 16.7 years. Twenty-five (11.1%) participants were diagnosed with heart failure during their hospital stay. The patients with EP and heart failure were older (58.2 years vs. 46.2 years, p = 0.001); had a higher prevalence of a history of coronary heart disease (32.0% vs. 21.5%, p < 0.001), fever (48.0% vs. 23.0%, p = 0.007), infection (56.0% vs. 35.5%, p = 0.046); higher hsCRP concentration (43.2 mg/L vs. 8.2 mg/L, p = 0.005); and higher prevalence of anemia (60.0% vs. 22.0%, p < 0.001), hypoalbuminemia (64.0% vs. 42.0%, p = 0.037), and hyperlipidemia (40.0% vs. 20.0%, p = 0.023) than those without heart failure. The serum FGF23 concentration was significantly higher in patients with EP than controls (493.1 pg/ml vs. 277.8 pg/ml, p = 0.027), and was significantly lower after treatment (395.7 pg/ml vs. 463.1 pg/ml, p = 0.022). Conclusions Clinicians should be aware of the risk of heart failure in patients with EP, and especially those of advanced age and with a history of coronary heart disease, severe systemic symptoms, high concentrations of inflammatory biomarkers, and poor nutritional status.
Collapse
Affiliation(s)
| | | | | | - Hongzhong Jin
- Department of Dermatology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
23
|
Chen G, Ren C, Xiao Y, Wang Y, Yao R, Wang Q, You G, Lu M, Yan S, Zhang X, Zhang J, Yao Y, Zhou H. Time-resolved single-cell transcriptomics reveals the landscape and dynamics of hepatic cells in sepsis-induced acute liver dysfunction. JHEP Rep 2023; 5:100718. [PMID: 37122356 PMCID: PMC10130477 DOI: 10.1016/j.jhepr.2023.100718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND & AIMS Sepsis-induced acute liver dysfunction often occurs early in sepsis and can exacerbate the pathology by triggering multiple organ dysfunction and increasing lethality. Nevertheless, our understanding of the cellular heterogeneity and dynamic regulation of major nonparenchymal cell lineages remains unclear. METHODS Here, single-cell RNA sequencing was used to profile multiple nonparenchymal cell subsets and dissect their crosstalk during sepsis-induced acute liver dysfunction in a clinically relevant polymicrobial sepsis model. The transcriptomes of major liver nonparenchymal cells from control and sepsis mice were analysed. The alterations in the endothelial cell and neutrophil subsets that were closely associated with acute liver dysfunction were validated using multiplex immunofluorescence staining. In addition, the therapeutic efficacy of inhibiting activating transcription factor 4 (ATF4) in sepsis and sepsis-induced acute liver dysfunction was explored. RESULTS Our results present the dynamic transcriptomic landscape of major nonparenchymal cells at single-cell resolution. We observed significant alterations and heterogeneity in major hepatic nonparenchymal cell subsets during sepsis. Importantly, we identified endothelial cell (CD31+Sele+Glut1+) and neutrophil (Ly6G+Lta4h+Sort1+) subsets that were closely associated with acute liver dysfunction during sepsis progression. Furthermore, we found that ATF4 inhibition alleviated sepsis-induced acute liver dysfunction, prolonging the survival of septic mice. CONCLUSIONS These results elucidate the potential mechanisms and subsequent therapeutic targets for the prevention and treatment of sepsis-induced acute liver dysfunction and other liver-related diseases. IMPACT AND IMPLICATIONS Sepsis-induced acute liver dysfunction often occurs early in sepsis and can lead to the death of the patient. Nevertheless, the pathogenesis of sepsis-induced acute liver dysfunction is not yet clear. We identified the major cell types associated with acute liver dysfunction and explored their interactions during sepsis. In addition, we also found that ATF-4 inhibition could be invoked as a potential therapeutic for sepsis-induced acute liver dysfunction.
Collapse
Affiliation(s)
- Gan Chen
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Chao Ren
- Translational Medicine Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yao Xiao
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Yujing Wang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Renqi Yao
- Translational Medicine Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, China
| | - Quan Wang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Mingzi Lu
- Beijing Science and Technology Innovation Research Center, Beijing, China
| | - Shaoduo Yan
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Xiaoyong Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Jun Zhang
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| | - Yongming Yao
- Translational Medicine Research Center, Fourth Medical Center and Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, China
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Dai Z, Zhang X. Pathophysiology and Clinical Impacts of Chronic Kidney Disease on Coronary Artery Calcification. J Cardiovasc Dev Dis 2023; 10:jcdd10050207. [PMID: 37233174 DOI: 10.3390/jcdd10050207] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
The global prevalence of chronic kidney disease (CKD) has increased in recent years. Adverse cardiovascular events have become the main cause of life-threatening events in patients with CKD, and vascular calcification is a risk factor for cardiovascular disease. Vascular calcification, especially coronary artery calcification, is more prevalent, severe, rapidly progressive, and harmful in patients with CKD. Some features and risk factors are unique to vascular calcification in patients with CKD; the formation of vascular calcification is not only influenced by the phenotypic transformation of vascular smooth muscle cells, but also by electrolyte and endocrine dysfunction, uremic toxin accumulation, and other novel factors. The study on the mechanism of vascular calcification in patients with renal insufficiency can provide a basis and new target for the prevention and treatment of this disease. This review aims to illustrate the impact of CKD on vascular calcification and to discuss the recent research data on the pathogenesis and factors involved in vascular calcification, mainly focusing on coronary artery calcification, in patients with CKD.
Collapse
Affiliation(s)
- Zhuoming Dai
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| |
Collapse
|
25
|
Wu Q, Ye Z, Zhou C, Liu M, Zhang Y, Zhang Z, He P, Li R, Li H, Yang S, Zhang Y, Jiang J, Nie J, Liu C, Qin X. A U-shaped association between dietary phosphorus intake and new-onset hypertension: a nationwide cohort study in China. Am Heart J 2023; 259:21-29. [PMID: 36690241 DOI: 10.1016/j.ahj.2023.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/30/2022] [Accepted: 01/16/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND The association between dietary phosphorus intake and the risk of hypertension remains uncertain. We aimed to investigate the relation of dietary phosphorus intake with new-onset hypertension among Chinese adults. METHODS A total of 12,177 participants who were free of hypertension at baseline from the China Health and Nutrition Survey (CHNS) were included. Dietary intake was measured by 3 consecutive 24-hour dietary recalls combined with a household food inventory. New-onset hypertension was defined as systolic blood pressure ≥140 mm Hg or diastolic blood pressure ≥90 mm Hg or diagnosed by a physician or under antihypertensive treatment during the follow-up. RESULTS During a median follow-up of 6.1 years, 4,269 participants developed new-onset hypertension. Overall, the association between dietary phosphorus intake and new-onset hypertension followed a U-shape (P for nonlinearity<.001). Consistently, when dietary phosphorus intake was assessed as quintiles, compared with those in the 3rd to 4th quintiles (912.0-<1089.5 mg/d), a significantly higher risk of new-onset hypertension was found in participants in the 1st to 2nd quintiles (<912.0 mg/d: HR, 1.23; 95% CI, 1.14-1.33), and the fifth quintile (≥1089.5 mg/d: HR, 1.21; 95% CI, 1.10-1.33). CONCLUSION There was a U-shaped association between dietary phosphorus intake and new-onset hypertension in general Chinese adults.
Collapse
Affiliation(s)
- Qimeng Wu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Ziliang Ye
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Chun Zhou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Mengyi Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Yuanyuan Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Zhuxian Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Panpan He
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Rui Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Institute of Biomedicine, Anhui Medical University, Hefei, 230032, China
| | - Huan Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Sisi Yang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Yanjun Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Jianping Jiang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Jing Nie
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China
| | - Chengzhang Liu
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Institute of Biomedicine, Anhui Medical University, Hefei, 230032, China
| | - Xianhui Qin
- Division of Nephrology, Nanfang Hospital, Southern Medical University, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Guangdong Provincial Institute of Nephrology, Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China; Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Institute of Biomedicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
26
|
Guo LW, Wang YK, Li SJ, Yin GT, Li D. Elevated Fibroblast Growth Factor 23 Impairs Endothelial Function through the NF-κB Signaling Pathway. J Atheroscler Thromb 2023; 30:138-149. [PMID: 35314565 PMCID: PMC9925204 DOI: 10.5551/jat.63460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIM Roles of fibroblast growth factor 23 (FGF23) in endothelial dysfunction remain controversial, and evidence from population-based studies is lacking. The present study aimed to explore the effects of FGF23 on endothelial dysfunction on the basis of both clinical data of patients with coronary artery disease (CAD) and the in vitro research in human umbilical vein endothelial cells (HUVECs). METHODS A total of 321 CAD patients were enrolled after coronary angiography, brachial artery flow-mediated dilation (FMD) was assessed using ultrasound equipment. Serum FGF23, nitric oxide (NO), and endothelin-1 (ET-1) were detected via enzyme-linked immunosorbent assay. Apoptosis was determined using the annexin V-fluorescein isothiocyanate/propidium lodide apoptosis detection kit. Cell migration was evaluated by wound healing and transwell migration assays. Reactive oxide species levels were determined using fluorescent probes, and NF-κB p65 nuclear translocation was assessed via immunofluorescence. RESULTS Serum FGF23 was significantly increased in CAD patients combined with severe endothelial dysfunction (FMD <2%) compared to those with FMD ≥ 2% (P<0.001). Furthermore, the levels of FGF23 were negatively correlated with NO, whereas positively correlated with ET-1 both in unadjusted analysis and multivariate-adjusted analysis. In HUVECs, FGF23 interfered with the bioavailability of NO via increased oxidative stress. Moreover, FGF23 directly impaired the endothelium by promoting HUVECs apoptosis and attenuating the migration of HUVECs. Additional experiments showed that FGF23 induced endothelial injury through activation of the NF-κB signaling pathway. CONCLUSIONS Elevated FGF23 is clinically associated with endothelial dysfunction in CAD patients, and FGF23 impairs endothelial function through activation of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Li-wei Guo
- School of Forensic Medicine, Xinxiang Medical University, Henan, China,Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Henan, China
| | - Yi-kai Wang
- School of Forensic Medicine, Xinxiang Medical University, Henan, China,Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Henan, China
| | - Shi-jie Li
- School of Forensic Medicine, Xinxiang Medical University, Henan, China,Xinxiang Key Laboratory of Metabolism and Integrative Physiology, Henan, China
| | - Guo-tian Yin
- The Third Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Duan Li
- School of Forensic Medicine, Xinxiang Medical University, Henan, China
| |
Collapse
|
27
|
Negah SS, Forouzanfar F. Dual Role of Fibroblast Growth Factor Pathways in Sleep Regulation. Endocr Metab Immune Disord Drug Targets 2023; 23:63-69. [PMID: 35927892 DOI: 10.2174/1871530322666220802161031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/11/2022] [Accepted: 05/20/2022] [Indexed: 11/22/2022]
Abstract
Sleep plays an important function in neuro-immuno-endocrine homeostasis. Sleep disorders have been associated with an increased risk of metabolic and cognitive impairments. Among different factors that have an effect on sleep metabolism, a growing body of literature has investigated growth factors in the course of sleep quality and disorders. A good example of growth factors is fibroblast growth factors (FGFs), which are a large family of polypeptide growth factors. Evidence has shown that FGFs are involved in the modulation of sleep-wake behavior by their receptor subtypes and ligands, e.g., FFG1 plays an important role in the quality of sleep through somnogenic effects, while the high level of FGF23 is associated with secondary disorders in shift workers. Therefore, a controversial effect of FGFs can be seen in the course of sleep in physiologic and pathologic conditions. Further investigation on this topic would help us to understand the role of FGFs in sleep disorders as a therapeutic option and biomarker.
Collapse
Affiliation(s)
- Sajad Sahab Negah
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.,Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
28
|
Miceli G, Basso MG, Rizzo G, Pintus C, Tuttolomondo A. The Role of the Coagulation System in Peripheral Arterial Disease: Interactions with the Arterial Wall and Its Vascular Microenvironment and Implications for Rational Therapies. Int J Mol Sci 2022; 23:14914. [PMID: 36499242 PMCID: PMC9739112 DOI: 10.3390/ijms232314914] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Peripheral artery disease (PAD) is a clinical manifestation of atherosclerotic disease with a large-scale impact on the economy and global health. Despite the role played by platelets in the process of atherogenesis being well recognized, evidence has been increasing on the contribution of the coagulation system to the atherosclerosis formation and PAD development, with important repercussions for the therapeutic approach. Histopathological analysis and some clinical studies conducted on atherosclerotic plaques testify to the existence of different types of plaques. Likely, the role of coagulation in each specific type of plaque can be an important determinant in the histopathological composition of atherosclerosis and in its future stability. In this review, we analyze the molecular contribution of inflammation and the coagulation system on PAD pathogenesis, focusing on molecular similarities and differences between atherogenesis in PAD and coronary artery disease (CAD) and discussing the possible implications for current therapeutic strategies and future perspectives accounting for molecular inflammatory and coagulation targets. Understanding the role of cross-talking between coagulation and inflammation in atherosclerosis genesis and progression could help in choosing the right patients for future dual pathway inhibition strategies, where an antiplatelet agent is combined with an anticoagulant, whose role, despite pathophysiological premises and trials' results, is still under debate.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| |
Collapse
|
29
|
Liu Y, Chen Q, Li Y, Bi L, He Z, Shao C, Jin L, Peng R, Zhang X. Advances in FGFs for diabetes care applications. Life Sci 2022; 310:121015. [PMID: 36179818 DOI: 10.1016/j.lfs.2022.121015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Diabetes mellitus (DM) is an endocrine and metabolic disease caused by a variety of pathogenic factors, including genetic factors, environmental factors and behavior. In recent decades, the number of cases and the prevalence of diabetes have steadily increased, and it has become one of the most threatening diseases to human health in the world. Currently, insulin is the most effective and direct way to control hyperglycemia for diabetes treatment at a low cost. However, hypoglycemia is often a common complication of insulin treatment. Moreover, with the extension of treatment time, insulin resistance, considered the typical adverse symptom, can appear. Therefore, it is urgent to develop new targets and more effective and safer drugs for diabetes treatment to avoid adverse reactions and the insulin tolerance of traditional hypoglycemic drugs. SCOPE OF REVIEW In recent years, it has been found that some fibroblast growth factors (FGFs), including FGF1, FGF19 and FGF21, can safely and effectively reduce hyperglycemia and have the potential to be developed as new drugs for the treatment of diabetes. FGF23 is also closely related to diabetes and its complications, which provides a new approach for regulating blood glucose and solving the problem of insulin tolerance. MAJOR CONCLUSIONS This article reviews the research progress on the physiology and pharmacology of fibroblast growth factor in the treatment of diabetes. We focus on the application of FGFs in diabetes care and prevention.
Collapse
Affiliation(s)
- Yinai Liu
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Qianqian Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yaoqi Li
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Liuliu Bi
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Zhiying He
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chuxiao Shao
- Department of Hepatopancreatobiliary Surgery, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Hospital of Zhejiang University, Lishui 323000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
30
|
Buyukdemirci S, Oguz EG, Cimen SG, Sahin H, Cimen S, Ayli MD. Vitamin D deficiency may predispose patients to increased risk of kidney transplant rejection. World J Transplant 2022; 12:299-309. [PMID: 36187881 PMCID: PMC9516489 DOI: 10.5500/wjt.v12.i9.299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/01/2022] [Accepted: 09/07/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Vitamin D deficiency occurs in more than 80% of kidney transplant recipients. Its immunomodulatory effects can predispose transplant recipients to rejection and chronic allograft nephropathy (CAN). This study determined the association between serum 25 (OH) vitamin D, biopsy-proven allograft rejection, and CAN rates.
AIM To determine the relationship between serum 25 (OH) vitamin D level and biopsy-proven allograft rejection and CAN rate in renal transplant recipients.
METHODS Adult renal transplant recipients followed at the clinic between January 2013 and 2018 were included. Recipients requiring graft biopsy due to declined function, hematuria, and proteinuria were reviewed. The two groups were compared regarding collected data, including the biopsy results, immunologic parameters, vitamin D, parathyroid hormone (PTH), phosphorus, albumin levels, and graft function tests.
RESULTS Fifty-two recipients who underwent graft biopsy met the inclusion criteria. In all, 14 recipients had a vitamin D level > 15 ng/mL (group 1) vs ≤ 15 ng/mL (group 2) in 38. In total, 27 patients had biopsy-proven rejection, and 19 had CAN. There was only 1 recipient with biopsy-proven rejection in group 1, whereas there were 24 patients with rejection in group 2. The rejection rate was significantly higher in group 2 than in group 1 (P < 0.001). Four patients were diagnosed with CAN in group 1 vs fifteen in group 2. There was no significant difference in the CAN rate between the two groups. PTH was higher at the time of graft biopsy (P = 0.009, P = 0.022) in group 1 with a mean of 268 pg/mL. Donor-specific antibodies were detected in 14 (56.0%) of the recipients with rejection. Vitamin D level was 9.7 ± 3.4 ng/mL in the rejection group vs 14.7 ± 7.2 in the non-rejection group; this difference was statistically significant (P = 0.003). The albumin levels were significantly lower in patients with rejection than in those without rejection (P = 0.001). In univariate regression analysis of risk factors affecting rejection, sex, serum vitamin D, phosphorus and albumin were found to have an impact (P = 0.027, P = 0.007, P = 0.023, P = 0.008). In multivariate regression analysis, the same factors did not affect rejection.
CONCLUSION The serum 25 (OH) vitamin D level in kidney transplant recipients remained low. Although low serum vitamin D level emerged as a risk factor for rejection in univariate analysis, this finding was not confirmed by multivariate analysis. Prospective studies are required to determine the effect of serum vitamin D levels on allograft rejection.
Collapse
Affiliation(s)
- Semih Buyukdemirci
- Department of Internal Medicine, Sağlık Bilimleri Universitesi, Ankara 65000, Altındağ, Turkey
| | - Ebru Gok Oguz
- Department of Nephrology, Sağlık Bilimleri Universitesi, Ankara 65000, Altındağ, Turkey
| | - Sanem Guler Cimen
- Department of General Surgery, Sağlık Bilimleri Universitesi, Ankara 65000, Altındağ, Turkey
| | - Hatice Sahin
- Department of Nephrology, Sağlık Bilimleri Universitesi, Ankara 65000, Altındağ, Turkey
| | - Sertac Cimen
- Department of Urology, Sağlık Bilimleri Universitesi, Ankara 65000, Altındağ, Turkey
| | - Mehmet Deniz Ayli
- Department of Nephrology, Sağlık Bilimleri Universitesi, Ankara 65000, Altındağ, Turkey
| |
Collapse
|
31
|
Vergara N, de Mier MVPR, Rodelo-Haad C, Revilla-González G, Membrives C, Díaz-Tocados JM, Martínez-Moreno JM, Torralbo AI, Herencia C, Rodríguez-Ortiz ME, López-Baltanás R, Richards WG, Felsenfeld A, Almadén Y, Martin-Malo A, Ureña J, Santamaría R, Soriano S, Rodríguez M, Muñoz-Castañeda JR. The direct effect of fibroblast growth factor 23 on vascular smooth muscle cell phenotype and function. Nephrol Dial Transplant 2022; 38:322-343. [PMID: 35867864 PMCID: PMC9923714 DOI: 10.1093/ndt/gfac220] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In chronic kidney disease (CKD) patients, increased levels of fibroblast growth factor 23 (FGF23) are associated with cardiovascular mortality. The relationship between FGF23 and heart hypertrophy has been documented, however, it is not known whether FGF23 has an effect on vasculature. Vascular smooth muscle cells VSMCs may exhibit different phenotypes; our hypothesis is that FGF23 favours a switch from a contractile to synthetic phenotype that may cause vascular dysfunction. Our objective was to determine whether FGF23 may directly control a change in VSMC phenotype. METHODS This study includes in vitro, in vivo and ex vivo experiments and evaluation of patients with CKD stages 2-3 studying a relationship between FGF23 and vascular dysfunction. RESULTS In vitro studies show that high levels of FGF23, by acting on its specific receptor FGFR1 and Erk1/2, causes a change in the phenotype of VSMCs from contractile to synthetic. This change is mediated by a downregulation of miR-221/222, which augments the expression of MAP3K2 and PAK1. miR-221/222 transfections recovered the contractile phenotype of VSMCs. Infusion of recombinant FGF23 to rats increased vascular wall thickness, with VSMCs showing a synthetic phenotype with a reduction of miR-221 expression. Ex-vivo studies on aortic rings demonstrate also that high FGF23 increases arterial stiffening. In CKD 2-3 patients, elevation of FGF23 was associated with increased pulse wave velocity and reduced plasma levels of miR-221/222. CONCLUSION In VSMCs, high levels of FGF23, through the downregulation of miR-221/222, causes a change to a synthetic phenotype. This change in VSMCs increases arterial stiffening and impairs vascular function, which might ultimately worsen cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | - Gonzalo Revilla-González
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Departemento de Fisiología Médica y Biofísica, Sevilla, Spain
| | - Cristina Membrives
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Juan M Díaz-Tocados
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Julio M Martínez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Ana I Torralbo
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | - Carmen Herencia
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | | | - Rodrigo López-Baltanás
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain
| | | | - Arnold Felsenfeld
- Department of Medicine, Veterans Affairs Greater Los Angeles Healthcare System and the David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yolanda Almadén
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,Internal Medicine Service, Reina Sofia University Hospital, Cordoba, Spain,Spanish Biomedical Research Networking Centre consortium for the area of Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Alejandro Martin-Malo
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain,Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain,Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, Madrid, Spain, and the European Uremic Toxins group
| | - Juan Ureña
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Departemento de Fisiología Médica y Biofísica, Sevilla, Spain
| | | | - Sagrario Soriano
- Maimonides Institute for Biomedical Research of Cordoba, Cordoba, Spain,University of Cordoba, Spain,Nephrology Service, Reina Sofia University Hospital, Cordoba, Spain,Spanish Renal Research Network (REDinREN), Institute of Health Carlos III, Madrid, Spain, and the European Uremic Toxins group
| | | | | |
Collapse
|
32
|
Lee S, Kim SJ. Effects of Normal Reference Range of Phosphorus and Corresponding PTH on Endothelial Function in CKD Patients. Front Med (Lausanne) 2022; 9:935977. [PMID: 35903313 PMCID: PMC9314747 DOI: 10.3389/fmed.2022.935977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionEndothelial dysfunction commonly occurs in chronic kidney disease (CKD) patients and increases the risk for cardiovascular disease. Among CKD patients, biomarkers involved in the pathogenesis of CKD-mineral bone disorder (CKD-MBD), such as phosphorus, parathyroid hormone, and fibroblast growth factor 23, are associated with endothelial dysfunction. We investigated whether these biomarkers induce endothelial dysfunction in CKD patients with normal phosphorus levels.MethodsThis cross-sectional study examined CKD patients with normal phosphorus levels; patients with an estimated glomerular filtration rate (eGFR) <15 or who were under dialysis were excluded. Iontophoresis with laser doppler flowmetry (ILDF) and peripheral arterial tonometry were performed to assess endothelial function in 85 patients. Pearson's correlation coefficient, multiple regression, and mediation analyses were performed to examine the association between CKD-MBD biomarkers and endothelial dysfunction.ResultsEndothelial dysfunction was observed in all subjects with a low response to ILDF and 27% of subjects according to peripheral arterial tonometry. Acetylcholine (Ach)-induced ILDF was significantly associated with eGFR (r = 0.22, P = 0.04), intact parathyroid hormone (iPTH; r = −0.46, P < 0.01), and VCAM-1 (r = −0.36, P < 0.01). The reactive hyperemia index (RHI) was significantly related to phosphorus levels (r = 0.32, P < 0.01) and iPTH (r = −0.39, P = 0.02). After adjusting for eGFR, iPTH and VCAM-1 remained independent factors for predicting endothelial dysfunction measured using Ach-induced ILDF. In addition, iPTH and phosphorus levels were independent predictors for endothelial dysfunction measured using RHI in the eGFR-adjusted model. Mediation analyses showed that the individual indirect effects of iPTH were significantly affected ILDF and RHI.ConclusionSerum levels of phosphorus and iPTH are associated with endothelial dysfunction, even in CKD patients with normal phosphorus levels.
Collapse
|
33
|
Quiroga B, Ortiz A, Navarro-González JF, Santamaría R, de Sequera P, Díez J. From cardiorenal syndromes to cardionephrology: a reflection by nephrologists on renocardiac syndromes. Clin Kidney J 2022; 16:19-29. [PMID: 36726435 PMCID: PMC9871856 DOI: 10.1093/ckj/sfac113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Cardiorenal syndromes (CRS) are broadly defined as disorders of the heart and kidneys whereby acute or chronic dysfunction in one organ may induce acute or chronic dysfunction of the other. CRS are currently classified into five categories, mostly based on disease-initiating events and their acuity or chronicity. CRS types 3 and 4 (also called renocardiac syndromes) refer to acute and chronic kidney dysfunction resulting in acute and chronic heart dysfunction, respectively. The notion of renocardiac syndromes has broadened interest in kidney-heart interactions but uncertainty remains in the nephrological community's understanding of the clinical diversity, pathophysiological mechanisms and optimal management approaches of these syndromes. This triple challenge that renocardiac syndromes (and likely other cardiorenal syndromes) pose to the nephrologist can only be faced through a specific and demanding training plan to enhance his/her cardiological scientific knowledge and through an appropriate clinical environment to develop his/her cardiological clinical skills. The first must be the objective of the subspecialty of cardionephrology (or nephrocardiology) and the second must be the result of collaboration with cardiologists (and other specialists) in cardiorenal care units. This review will first consider various aspects of the challenges that renocardiac syndromes pose to nephrologists and, then, will discuss those aspects of cardionephrology and cardiorenal units that can facilitate an effective response to the challenges.
Collapse
Affiliation(s)
| | | | - Juan F Navarro-González
- RICORS2040, Carlos III Institute of Health, Madrid, Spain,Division of Nephrology and Research Unit, University Hospital Nuestra Señora de Candelaria, and University Institute of Biomedical Technologies, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Rafael Santamaría
- RICORS2040, Carlos III Institute of Health, Madrid, Spain,Division of Nephrology, University Hospital Reina Sofia, Cordoba, Spain,Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Cordoba, Spain
| | - Patricia de Sequera
- Department of Nephrology, University Hospital Infanta Leonor, University Complutense of Madrid, Madrid, Spain
| | | |
Collapse
|
34
|
Volk C, Schmidt B, Brandsch C, Kurze T, Schlegelmilch U, Grosse I, Ulrich C, Girndt M, Stangl GI. Acute Effects of an Inorganic Phosphorus Additive on Mineral Metabolism and Cardiometabolic Risk Factors in Healthy Subjects. J Clin Endocrinol Metab 2022; 107:e852-e864. [PMID: 34448875 DOI: 10.1210/clinem/dgab635] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Hyperphosphatemia and high levels of fibroblast growth factor 23 (FGF23) are risk factors for cardiovascular events in patients with chronic kidney diseases. However, the impact of an inorganic phosphorus additive in healthy people is largely unknown. OBJECTIVE We aimed to investigate the acute effect of excessive dietary phosphorus administered as sodium dihydrogen phosphate on the postprandial levels of Pi and FGF23 and the response to food. METHODS This study was a double-blind placebo-controlled crossover study with 29 healthy male and female participants from the general community who were administered a single dose of either 700 mg phosphorus (NaH2PO4) or a sodium-adjusted placebo in combination with a test meal. Postprandial plasma levels of Pi and FGF23 were measured. RESULTS Compared with placebo, oral phosphorus increased the plasma Pi level, which remained elevated during the ensuing 8 hours (at 480 minutes: 1.31 vs 1.16 mmol/l; P < 0.001), increased urinary Pi (iAUC0-480 789 vs 95 mmol/mmol; P < 0.001), reduced tubular Pi reabsorption (iAUC0-480 -31.5 vs -6.2; P < 0.001), decreased urinary calcium (iAUC0-240 30.6 vs 53.0 mmol/mmol; P = 0.009), and stimulated the release of parathyroid hormone (iAUC0-480 2212 vs 768 ng/l; P < 0.001). However, the FGF23 levels did not change. Postprandial levels of glucose, insulin, and lipids were not substantially affected by phosphorus vs placebo. CONCLUSION An oral phosphorus load can induce elevated postprandial levels of circulating Pi for hours in healthy subjects, despite rapid homeostatic counterreactions. FGF23 levels and the postprandial response to food were not affected.
Collapse
Affiliation(s)
- Christin Volk
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Benjamin Schmidt
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Corinna Brandsch
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Tabea Kurze
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Ulf Schlegelmilch
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Ivo Grosse
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Competence Cluster of Cardiovascular Health and Nutrition (nutriCARD), Halle-Jena-Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Germany
| | - Christof Ulrich
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Gabriele I Stangl
- Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Institute of Computer Science, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| |
Collapse
|
35
|
Eitner F, Richter B, Schwänen S, Szaroszyk M, Vogt I, Grund A, Thum T, Heineke J, Haffner D, Leifheit-Nestler M. Comprehensive Expression Analysis of Cardiac Fibroblast Growth Factor 23 in Health and Pressure-induced Cardiac Hypertrophy. Front Cell Dev Biol 2022; 9:791479. [PMID: 35118076 PMCID: PMC8804498 DOI: 10.3389/fcell.2021.791479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022] Open
Abstract
Enhanced fibroblast growth factor 23 (FGF23) is associated with left ventricular hypertrophy (LVH) in patients with chronic kidney and heart disease. Experimentally, FGF23 directly induces cardiac hypertrophy and vice versa cardiac hypertrophy stimulates FGF23. Besides the bone, FGF23 is expressed by cardiac myocytes, whereas its synthesis in other cardiac cell types and its paracrine role in the heart in health and disease is unknown. By co-immunofluorescence staining of heart tissue of wild-type mice, we show that Fgf23 is expressed by cardiac myocytes, fibroblasts and endothelial cells. Cardiac Fgf23 mRNA and protein level increases from neonatal to six months of age, whereas no age-related changes in bone Fgf23 mRNA expression were noted. Cardiac myocyte-specific disruption of Fgf23 using Cre-LoxP system (Fgf23fl/fl/cre+) caused enhanced mortality, but no differences in cardiac function or structure. Although pressure overload-induced cardiac hypertrophy induced by transverse aortic constriction (TAC) resulted in a slightly worse phenotype with a more severe reduced ejection fraction, higher end-systolic volume and more enlarged systolic LV diameter in Fgf23fl/fl/cre+ mice compared to controls, this was not translated to any worse cellular hypertrophy, fibrosis or chamber remodeling. TAC induced Fgf23 mRNA expression in whole cardiac tissue in both genotypes. Interestingly, co-immunofluorescence staining revealed enhanced Fgf23 synthesis in cardiac fibroblasts and endothelial cells but not in cardiac myocytes. RNA sequencing of isolated adult cardiac myocytes, cardiac fibroblasts and endothelial cells confirmed significantly higher Fgf23 transcription in cardiac fibroblasts and endothelial cells after TAC. Our data indicate that Fgf23 is physiologically expressed in various cardiac cell types and that cardiac fibroblasts and endothelial cells might be an important source of FGF23 in pathological conditions. In addition, investigations in Fgf23fl/fl/cre+ mice suggest that cardiac myocyte-derived FGF23 is needed to maintain cardiac function during pressure overload.
Collapse
Affiliation(s)
- Fiona Eitner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Beatrice Richter
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Saskia Schwänen
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Malgorzata Szaroszyk
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Isabel Vogt
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Andrea Grund
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Joerg Heineke
- Department for Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
| | - Maren Leifheit-Nestler
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Pediatric Research Center, Hannover Medical School, Hannover, Germany
- *Correspondence: Maren Leifheit-Nestler,
| |
Collapse
|
36
|
Impact of Uremic Toxins on Endothelial Dysfunction in Chronic Kidney Disease: A Systematic Review. Int J Mol Sci 2022; 23:ijms23010531. [PMID: 35008960 PMCID: PMC8745705 DOI: 10.3390/ijms23010531] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at a highly increased risk of cardiovascular complications, with increased vascular inflammation, accelerated atherogenesis and enhanced thrombotic risk. Considering the central role of the endothelium in protecting from atherogenesis and thrombosis, as well as its cardioprotective role in regulating vasorelaxation, this study aimed to systematically integrate literature on CKD-associated endothelial dysfunction, including the underlying molecular mechanisms, into a comprehensive overview. Therefore, we conducted a systematic review of literature describing uremic serum or uremic toxin-induced vascular dysfunction with a special focus on the endothelium. This revealed 39 studies analyzing the effects of uremic serum or the uremic toxins indoxyl sulfate, cyanate, modified LDL, the advanced glycation end products N-carboxymethyl-lysine and N-carboxyethyl-lysine, p-cresol and p-cresyl sulfate, phosphate, uric acid and asymmetric dimethylarginine. Most studies described an increase in inflammation, oxidative stress, leukocyte migration and adhesion, cell death and a thrombotic phenotype upon uremic conditions or uremic toxin treatment of endothelial cells. Cellular signaling pathways that were frequently activated included the ROS, MAPK/NF-κB, the Aryl-Hydrocarbon-Receptor and RAGE pathways. Overall, this review provides detailed insights into pathophysiological and molecular mechanisms underlying endothelial dysfunction in CKD. Targeting these pathways may provide new therapeutic strategies reducing increased the cardiovascular risk in CKD.
Collapse
|
37
|
Chen HH, Pan JY, Lu WH, Wu CJ, Tseng CJ. Prazosin improves neurogenic acute heart failure through downregulation of fibroblast growth factor 23 in rat hearts. CHINESE J PHYSIOL 2022; 65:179-186. [DOI: 10.4103/cjp.cjp_9_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
38
|
Nashawi M, Ahmed MS, Amin T, Abualfoul M, Chilton R. Cardiovascular benefits from SGLT2 inhibition in type 2 diabetes mellitus patients is not impaired with phosphate flux related to pharmacotherapy. World J Cardiol 2021; 13:676-694. [PMID: 35070111 PMCID: PMC8716977 DOI: 10.4330/wjc.v13.i12.676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
The beneficial cardiorenal outcomes of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in patients with type 2 diabetes mellitus (T2DM) have been substantiated by multiple clinical trials, resulting in increased interest in the multifarious pathways by which their mechanisms act. The principal effect of SGLT2i (-flozin drugs) can be appreciated in their ability to block the SGLT2 protein within the kidneys, inhibiting glucose reabsorption, and causing an associated osmotic diuresis. This ameliorates plasma glucose elevations and the negative cardiorenal sequelae associated with the latter. These include aberrant mitochondrial metabolism and oxidative stress burden, endothelial cell dysfunction, pernicious neurohormonal activation, and the development of inimical hemodynamics. Positive outcomes within these domains have been validated with SGLT2i administration. However, by modulating the sodium-glucose cotransporter in the proximal tubule (PT), SGLT2i consequently promotes sodium-phosphate cotransporter activity with phosphate retention. Phosphatemia, even at physiologic levels, poses a risk in cardiovascular disease burden, more so in patients with type 2 diabetes mellitus (T2DM). There also exists an association between phosphatemia and renal impairment, the latter hampering cardiovascular function through an array of physiologic roles, such as fluid regulation, hormonal tone, and neuromodulation. Moreover, increased phosphate flux is associated with an associated increase in fibroblast growth factor 23 levels, also detrimental to homeostatic cardiometabolic function. A contemporary commentary concerning this notion unifying cardiovascular outcome trial data with the translational biology of phosphate is scant within the literature. Given the apparent beneficial outcomes associated with SGLT2i administration notwithstanding negative effects of phosphatemia, we discuss in this review the effects of phosphate on the cardiometabolic status in patients with T2DM and cardiorenal disease, as well as the mechanisms by which SGLT2i counteract or overcome them to achieve their net effects. Content drawn to develop this conversation begins with proceedings in the basic sciences and works towards clinical trial data.
Collapse
Affiliation(s)
- Mouhamed Nashawi
- Department of Internal Medicine, Baylor Scott and White All Saints Medical Center, Fort Worth, TX 76132, United States.
| | - Mahmoud S Ahmed
- Division of Medicine-Cardiology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Toka Amin
- Division of Medicine-Cardiology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Mujahed Abualfoul
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Dallas, TX 75203, United States
| | - Robert Chilton
- Department of Internal Medicine, Methodist Dallas Medical Center, Dallas, TX 75203, United States
| |
Collapse
|
39
|
Deng J, Liu Y, Liu Y, Li W, Nie X. The Multiple Roles of Fibroblast Growth Factor in Diabetic Nephropathy. J Inflamm Res 2021; 14:5273-5290. [PMID: 34703268 PMCID: PMC8524061 DOI: 10.2147/jir.s334996] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022] Open
Abstract
Diabetic nephropathy (DN) is a common microvascular complication in the late stages of diabetes. Currently, the etiology and pathogenesis of DN are not well understood. Even so, available evidence shows its development is associated with metabolism, oxidative stress, cytokine interaction, genetic factors, and renal microvascular disease. Diabetic nephropathy can lead to proteinuria, edema and hypertension, among other complications. In severe cases, it can cause life-threatening complications such as renal failure. Patients with type 1 diabetes, hypertension, high protein intake, and smokers have a higher risk of developing DN. Fibroblast growth factor (FGF) regulates several human processes essential for normal development. Even though FGF has been implicated in the pathological development of DN, the underlying mechanisms are not well understood. This review summarizes the role of FGF in the development of DN. Moreover, the association of FGF with metabolism, inflammation, oxidative stress and fibrosis in the context of DN is discussed. Findings of this review are expected to deepen our understanding of DN and generate ideas for developing effective prevention and treatments for the disease.
Collapse
Affiliation(s)
- Junyu Deng
- College of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Ye Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Yiqiu Liu
- College of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Wei Li
- College of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Joint International Research Laboratory of Ethnomedicine of Chinese Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Key Laboratory of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi, 563000, People’s Republic of China
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, People’s Republic of China
| |
Collapse
|
40
|
Alderson HV, Chinnadurai R, Ibrahim ST, Asar O, Ritchie JP, Middleton R, Larsson A, Diggle PJ, Larsson TE, Kalra PA. Longitudinal change in c-terminal fibroblast growth factor 23 and outcomes in patients with advanced chronic kidney disease. BMC Nephrol 2021; 22:329. [PMID: 34600515 PMCID: PMC8487581 DOI: 10.1186/s12882-021-02528-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 09/10/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fibroblast growth factor23 (FGF23) is elevated in CKD and has been associated with outcomes such as death, cardiovascular (CV) events and progression to Renal Replacement therapy (RRT). The majority of studies have been unable to account for change in FGF23 over time and those which have demonstrate conflicting results. We performed a survival analysis looking at change in c-terminal FGF23 (cFGF23) over time to assess the relative contribution of cFGF23 to these outcomes. METHODS We measured cFGF23 on plasma samples from 388 patients with CKD 3-5 who had serial measurements of cFGF23, with a mean of 4.2 samples per individual. We used linear regression analysis to assess the annual rate of change in cFGF23 and assessed the relationship between time-varying cFGF23 and the outcomes in a cox-regression analysis. RESULTS Across our population, median baseline eGFR was 32.3mls/min/1.73m2, median baseline cFGF23 was 162 relative units/ml (RU/ml) (IQR 101-244 RU/mL). Over 70 months (IQR 53-97) median follow-up, 76 (19.6%) patients progressed to RRT, 86 (22.2%) died, and 52 (13.4%) suffered a major non-fatal CV event. On multivariate analysis, longitudinal change in cFGF23 was significantly associated with risk for death and progression to RRT but not non-fatal cardiovascular events. CONCLUSION In our study, increasing cFGF23 was significantly associated with risk for death and RRT.
Collapse
Affiliation(s)
- Helen V Alderson
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK. .,Department of Renal Medicine, Salford Royal Hospital, Level 2, Hope Building, Stott Lane, Salford, M6 8HD, UK.
| | - Rajkumar Chinnadurai
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| | - Sara T Ibrahim
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK.,Department of Internal Medicine and Nephrology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Ozgur Asar
- Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - James P Ritchie
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| | - Rachel Middleton
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| | - Anders Larsson
- Section of Clinical Chemistry, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter J Diggle
- CHICAS, Lancaster Medical School, Lancaster University, Lancaster, UK
| | - Tobias E Larsson
- Department of Clinical Science, Intervention and Technology, Renal Unit, Karolinska Institute, Stockholm, Sweden
| | - Philip A Kalra
- Vascular Research Group, Manchester Academic Health Sciences Centre, University of Manchester, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|
41
|
Zhang R, Wang SY, Yang F, Ma S, Lu X, Kan C, Zhang JB. Crosstalk of fibroblast growth factor 23 and anemia-related factors during the development and progression of CKD (Review). Exp Ther Med 2021; 22:1159. [PMID: 34504604 PMCID: PMC8393509 DOI: 10.3892/etm.2021.10593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 03/08/2021] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) plays an important role in the development of chronic kidney disease-mineral bone disorder (CKD-MBD). Abnormally elevated levels of 1,25-dihydroxyvitamin D cause osteocytes to secrete FGF23, which subsequently induces phosphaturia. Recent studies have reported that iron deficiency, erythropoietin (EPO) and hypoxia regulate the pathways responsible for FGF23 production. However, the molecular mechanisms underlying the interactions between FGF23 and anemia-related factors are not yet fully understood. The present review discusses the associations between FGF23, iron, EPO and hypoxia-inducible factors (HIFs), and their impact on FGF23 bioactivity, focusing on recent studies. Collectively, these findings propose interactions between FGF23 gene expression and anemia-related factors, including iron deficiency, EPO and HIFs. Taken together, these results suggest that FGF23 bioactivity is closely associated with the occurrence of CKD-related anemia and CKD-MBD.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Song-Yan Wang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Fan Yang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Shuang Ma
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| | - Xu Lu
- Department of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Chao Kan
- Department of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130000, P.R. China
| | - Jing-Bin Zhang
- Department of Nephrology, Jilin Province People's Hospital, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
42
|
Kalantar-Zadeh K, Jafar TH, Nitsch D, Neuen BL, Perkovic V. Chronic kidney disease. Lancet 2021; 398:786-802. [PMID: 34175022 DOI: 10.1016/s0140-6736(21)00519-5] [Citation(s) in RCA: 691] [Impact Index Per Article: 172.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease is a progressive disease with no cure and high morbidity and mortality that occurs commonly in the general adult population, especially in people with diabetes and hypertension. Preservation of kidney function can improve outcomes and can be achieved through non-pharmacological strategies (eg, dietary and lifestyle adjustments) and chronic kidney disease-targeted and kidney disease-specific pharmacological interventions. A plant-dominant, low-protein, and low-salt diet might help to mitigate glomerular hyperfiltration and preserve renal function for longer, possibly while also leading to favourable alterations in acid-base homoeostasis and in the gut microbiome. Pharmacotherapies that alter intrarenal haemodynamics (eg, renin-angiotensin-aldosterone pathway modulators and SGLT2 [SLC5A2] inhibitors) can preserve kidney function by reducing intraglomerular pressure independently of blood pressure and glucose control, whereas other novel agents (eg, non-steroidal mineralocorticoid receptor antagonists) might protect the kidney through anti-inflammatory or antifibrotic mechanisms. Some glomerular and cystic kidney diseases might benefit from disease-specific therapies. Managing chronic kidney disease-associated cardiovascular risk, minimising the risk of infection, and preventing acute kidney injury are crucial interventions for these patients, given the high burden of complications, associated morbidity and mortality, and the role of non-conventional risk factors in chronic kidney disease. When renal replacement therapy becomes inevitable, an incremental transition to dialysis can be considered and has been proposed to possibly preserve residual kidney function longer. There are similarities and distinctions between kidney-preserving care and supportive care. Additional studies of dietary and pharmacological interventions and development of innovative strategies are necessary to ensure optimal kidney-preserving care and to achieve greater longevity and better health-related quality of life for these patients.
Collapse
Affiliation(s)
- Kamyar Kalantar-Zadeh
- Division of Nephrology, Hypertension and Kidney Transplantation, University of California Irvine, Orange, CA, USA; Tibor Rubin Veterans Affairs Medical Center, Long Beach, CA, USA.
| | - Tazeen H Jafar
- Duke-NUS Graduate Medical School, Singapore; Department of Renal Medicine, Singapore General Hospital, Singapore; Duke Global Health Institute, Durham, NC, USA
| | - Dorothea Nitsch
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK; United Kingdom Renal Registry, Bristol, UK; Department of Nephrology, Royal Free London NHS Foundation Trust, London, UK
| | - Brendon L Neuen
- The George Institute for Global Health, University of New South Wales Sydney, Sydney, NSW, Australia
| | - Vlado Perkovic
- Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
Kusmic C, Vizzoca A, Taranta M, Tedeschi L, Gherardini L, Pelosi G, Giannetti A, Tombelli S, Grimaldi S, Baldini F, Domenici C, Trivella MG, Cinti C. Silencing Survivin: a Key Therapeutic Strategy for Cardiac Hypertrophy. J Cardiovasc Transl Res 2021; 15:391-407. [PMID: 34409583 DOI: 10.1007/s12265-021-10165-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/05/2021] [Indexed: 11/29/2022]
Abstract
Cardiac hypertrophy, in its aspects of localized thickening of the interventricular septum and concentric increase of the left ventricle, constitutes a risk factor of heart failure. Myocardial hypertrophy, in the presence of different degree of myocardial fibrosis, is paralleled by significant molecular, cellular, and histological changes inducing alteration of cardiac extracellular matrix composition as well as sarcomeres and cytoskeleton remodeling. Previous studies indicate osteopontin (OPN) and more recently survivin (SURV) overexpression as the hallmarks of heart failure although SURV function in the heart is not completely clarified. In this study, we investigated the involvement of SURV in intracellular signaling of hypertrophic cardiomyocytes and the impact of its transcriptional silencing, laying the foundation for novel target gene therapy in cardiac hypertrophy. Oligonucleotide-based molecules, like theranostic optical nanosensors (molecular beacons) and siRNAs, targeting SURV and OPN mRNAs, were developed. Their diagnostic and therapeutic potential was evaluated in vitro in hypertrophic FGF23-induced human cardiomyocytes and in vivo in transverse aortic constriction hypertrophic mouse model. Engineered erythrocyte was used as shuttle to selectively target and transfer siRNA molecules into unhealthy cardiac cells in vivo. The results highlight how the SURV knockdown could negatively influence the expression of genes involved in myocardial fibrosis in vitro and restores structural, functional, and morphometric features in vivo. Together, these data suggested that SURV is a key factor in inducing cardiomyocytes hypertrophy, and its shutdown is crucial in slowing disease progression as well as reversing cardiac hypertrophy. In the perspective, targeted delivery of siRNAs through engineered erythrocytes can represent a promising therapeutic strategy to treat cardiac hypertrophy. Theranostic SURV molecular beacon (MB-SURV), transfected into FGF23-induced hypertrophic human cardiomyocytes, significantly dampened SURV overexpression. SURV down-regulation determines the tuning down of MMP9, TIMP1 and TIMP4 extracellular matrix remodeling factors while induces the overexpression of the cardioprotective MCAD factor, which counterbalance the absence of pro-survival and anti-apoptotic SURV activity to protect cardiomyocytes from death. In transverse aortic constriction (TAC) mouse model, the SURV silencing restores the LV mass levels to values not different from the sham group and counteracts the progressive decline of EF, maintaining its values always higher with respect to TAC group. These data demonstrate the central role of SURV in the cardiac reverse remodeling and its therapeutic potential to reverse cardiac hypertrophy.
Collapse
Affiliation(s)
- Claudia Kusmic
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy
| | - Alessio Vizzoca
- Institute of Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via Gobetti 101, 40129, Bologna, Italy
| | - Monia Taranta
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy
| | - Lorena Tedeschi
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy
| | - Lisa Gherardini
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy
| | - Gualtiero Pelosi
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy
| | - Ambra Giannetti
- Institute of Applied Physics, Nello Carrara"(IFAC), National Research Council of Italy (CNR), Florence, Italy
| | - Sara Tombelli
- Institute of Applied Physics, Nello Carrara"(IFAC), National Research Council of Italy (CNR), Florence, Italy
| | - Settimio Grimaldi
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Rome, Italy
| | - Francesco Baldini
- Institute of Applied Physics, Nello Carrara"(IFAC), National Research Council of Italy (CNR), Florence, Italy
| | - Claudio Domenici
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy
| | - Maria Giovanna Trivella
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy.
| | - Caterina Cinti
- Institute of Clinical Physiology (IFC), National Research Council of Italy (CNR), via Moruzzi 1, 56124, Pisa, Italy.
- Institute of Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via Gobetti 101, 40129, Bologna, Italy.
| |
Collapse
|
44
|
van der Vaart A, Yeung S, van Dijk P, Bakker S, de Borst M. Phosphate and fibroblast growth factor 23 in diabetes. Clin Sci (Lond) 2021; 135:1669-1687. [PMID: 34283205 PMCID: PMC8302806 DOI: 10.1042/cs20201290] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022]
Abstract
Diabetes is associated with a strongly elevated risk of cardiovascular disease, which is even more pronounced in patients with diabetic nephropathy. Currently available guideline-based efforts to correct traditional risk factors are only partly able to attenuate this risk, underlining the urge to identify novel treatment targets. Emerging data point towards a role for disturbances in phosphate metabolism in diabetes. In this review, we discuss the role of phosphate and the phosphate-regulating hormone fibroblast growth factor 23 (FGF23) in diabetes. We address deregulations of phosphate metabolism in patients with diabetes, including diabetic ketoacidosis. Moreover, we discuss potential adverse consequences of these deregulations, including the role of deregulated phosphate and glucose as drivers of vascular calcification propensity. Finally, we highlight potential treatment options to correct abnormalities in phosphate and FGF23. While further studies are needed to more precisely assess their clinical impact, deregulations in phosphate and FGF23 are promising potential target in diabetes and diabetic nephropathy.
Collapse
Affiliation(s)
- Amarens van der Vaart
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
- Department of Medicine, Division of Endocrinology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Stanley M.H. Yeung
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
| | - Peter R. van Dijk
- Department of Medicine, Division of Endocrinology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Stephan J.L. Bakker
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
| | - Martin H. de Borst
- Department of Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen,The Netherlands
| |
Collapse
|
45
|
McCarty MF, Lerner A, DiNicolantonio JJ, Iloki-Assanga SB. High Intakes of Bioavailable Phosphate May Promote Systemic Oxidative Stress and Vascular Calcification by Boosting Mitochondrial Membrane Potential-Is Good Magnesium Status an Antidote? Cells 2021; 10:1744. [PMID: 34359914 PMCID: PMC8303439 DOI: 10.3390/cells10071744] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic kidney disease is characterized by markedly increased risk for cardiovascular mortality, vascular calcification, and ventricular hypertrophy, and is associated with increased systemic oxidative stress. Hyperphosphatemia, reflecting diminished glomerular phosphate (Pi) clearance, coupled with a compensatory increase in fibroblast growth factor 23 (FGF23) secretion are thought to be key mediators of this risk. Elevated serum and dietary Pi and elevated plasma FGF23 are associated with increased cardiovascular and total mortality in people with normal baseline renal function. FGF23 may mediate some of this risk by promoting cardiac hypertrophy via activation of fibroblast growth factor receptor 4 on cardiomyocytes. Elevated serum Pi can also cause a profound increase in systemic oxidative stress, and this may reflect the ability of Pi to act directly on mitochondria to boost membrane potential and thereby increase respiratory chain superoxide production. Moreover, elevated FGF23 likewise induces oxidative stress in vascular endothelium via activation of NADPH oxidase complexes. In vitro exposure of vascular smooth muscle cells to elevated Pi provokes an osteoblastic phenotypic transition that is mediated by increased mitochondrial oxidant production; this is offset dose-dependently by increased exposure to magnesium (Mg). In vivo, dietary Mg is protective in rodent models of vascular calcification. It is proposed that increased intracellular Mg opposes Pi's ability to increase mitochondrial membrane potential; this model could explain its utility for prevention of vascular calcification and predicts that Mg may have a more global protective impact with regard to the direct pathogenic effects of hyperphosphatemia.
Collapse
Affiliation(s)
- Mark F. McCarty
- Catalytic Longevity Foundation, 811 B Nahant Ct., San Diego, CA 92109, USA;
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer 5262000, Israel
| | | | - Simon B. Iloki-Assanga
- Department of Research and Postgraduate in Food Science, Universidad de Sonora, Hermosillo 83000, Mexico;
| |
Collapse
|
46
|
Memmos E, Papagianni A. New Insights into the Role of FGF-23 and Klotho in Cardiovascular Disease in Chronic Kidney Disease Patients. Curr Vasc Pharmacol 2021; 19:55-62. [PMID: 32310050 DOI: 10.2174/1570161118666200420102100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/29/2022]
Abstract
Alterations of fibroblast growth factor 23 (FGF-23) and Klotho levels are considered to be the earliest biochemical abnormality of chronic kidney disease - mineral and bone disease (CKDMBD) syndrome. Moreover, emerging data suggests that the dysregulated FGF-23 and Klotho axis has many effects on the cardiovascular (CV) system and contributes significantly to the increased CV morbidity and mortality rates of CKD patients. This review examines recent evidence on the role of FGF-23 and Klotho in the development and progression of CV complications of uremia namely cardiac hypertrophy, uremic cardiomyopathy, and atherosclerotic and arteriosclerotic vascular lesions. Moreover, the available evidence on their associations with adverse clinical outcomes are summarized. Undoubtedly, more studies are needed to further elucidate the effects of FGF-23 and Klotho on the heart and vessels and to gain insights into their prognostic value as CV risk factors. Finally, large prospective studies are required to test the hypothesis that modification of their levels would have a favourable impact on the unacceptably high mortality rates of these patient populations.
Collapse
Affiliation(s)
- Evangelos Memmos
- Department of Nephrology, Aristotle University of Thessaloniki, General Hospital "Hippokratio", Thessaloniki, Greece
| | - Aikaterini Papagianni
- Department of Nephrology, Aristotle University of Thessaloniki, General Hospital "Hippokratio", Thessaloniki, Greece
| |
Collapse
|
47
|
Discharge FGF23 level predicts one year outcome in patients admitted with acute heart failure. Int J Cardiol 2021; 336:98-104. [PMID: 34019969 DOI: 10.1016/j.ijcard.2021.05.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients with acute heart failure (AHF) show high levels of fibroblast growth factor-23 (FGF23) on admission. We examined if plasma FGF23 changes during an episode of AHF, and if FGF23 holds prognostic significance in this setting. METHODS Consecutive AHF patients were enrolled. Blood samples were collected on admission and at discharge. Patients were then followed for all-cause death or HF hospitalization. RESULTS Patients (n = 125; median age 76 years [interquartile interval 71-83], 63% men, left ventricular ejection fraction 35% [25%-56%]) had median admission FGF23 70 ng/L (47-100), N-terminal pro-B-type natriuretic peptide (NT-proBNP) 5844 ng/L (2,503-10,468), high-sensitivity troponin T (hs-TnT) 40 ng/L (25-72), and soluble suppression of tumorigenesis-2 (sST2) 26 ng/mL (17-37). While other biomarkers decreased, FGF23 increased by 15% from admission to discharge (p = 0.033), with a significant correlation with percent changes in estimated glomerular filtration rate (rho = 0.306, p = 0.001). Over a 12-month follow-up, 64 patients (51%) experienced the endpoint. They were more often men, older, with higher systolic pulmonary artery pressure (sPAP), higher NT-proBNP, hs-TnT and discharge FGF23. The best FGF23 cut-off at discharge from receiver operating characteristics analysis was 78 ng/L. Both discharge FGF23 and the 78 ng/L cut-off independently predicted outcome in models including gender, sPAP, age, and 1) admission NT-proBNP, 2) discharge NT-proBNP, 3) admission NT-proBNP and hs-TnT, 4) discharge NT-proBNP and hs-TnT. The 78 ng/L cut-off also refined risk reclassification. CONCLUSIONS During an AHF episode, FGF23 increases from admission to discharge, and patients with higher discharge FGF23 have a higher risk of worse outcome.
Collapse
|
48
|
FGF23: A Review of Its Role in Mineral Metabolism and Renal and Cardiovascular Disease. DISEASE MARKERS 2021; 2021:8821292. [PMID: 34055103 PMCID: PMC8149241 DOI: 10.1155/2021/8821292] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/01/2020] [Accepted: 05/04/2021] [Indexed: 01/03/2023]
Abstract
FGF23 is a hormone secreted mainly by osteocytes and osteoblasts in bone. Its pivotal role concerns the maintenance of mineral ion homeostasis. It has been confirmed that phosphate and vitamin D metabolisms are related to the effect of FGF23 and its excess or deficiency leads to various hereditary diseases. Multiple studies have shown that FGF23 level increases in the very early stages of chronic kidney disease (CKD), and its concentration may also be highly associated with cardiac complications. The present review is limited to some of the most important aspects of calcium and phosphate metabolism. It discusses the role of FGF23, which is considered an early and sensitive marker for CKD-related bone disease but also as a novel and potent cardiovascular risk factor. Furthermore, this review gives particular attention to the reliability of FGF23 measurement and various confounding factors that may impact on the clinical utility of FGF23. Finally, this review elaborates on the clinical usefulness of FGF23 and evaluates whether FGF23 may be considered a therapeutic target.
Collapse
|
49
|
Ishigami J, Grams ME, Michos ED, Lutsey PL, Matsushita K. 25-hydroxyvitamin D, Fibroblast Growth Factor 23, and Risk of Acute Kidney Injury Over 20 Years of Follow-Up. Kidney Int Rep 2021; 6:1299-1308. [PMID: 34013108 PMCID: PMC8116771 DOI: 10.1016/j.ekir.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/11/2021] [Accepted: 02/01/2021] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Low serum 25-hydroxyvitamin D levels have been identified as a risk factor for acute kidney injury (AKI) among critically ill patients. Whether low 25-hydroxyvitamin D levels are associated with long-term incidence of hospitalization with AKI in the general population is unknown. METHODS Among 12,380 participants (mean age, 57 years; 24% black) in the Atherosclerosis Risk in Communities (ARIC) Study who attended visit 2 (1990-1992), we explored the association of serum 25-hydroxyvitamin D with incident hospitalization with AKI. Multivariable Cox models were used to estimate hazard ratios (HRs). We also examined the association of serum fibroblast growth factor 23 (FGF23) with AKI. RESULTS During a median follow-up of 24.3 years, 2145 participants had incident hospitalization with AKI (crude incidence rate: 8.3; 95% confidence interval [CI]: 8.0-8.7, per 1,000 person-years). In multivariable Cox models (including adjustment for kidney function), lower 25-hydroxyvitamin D and higher FGF23 levels were each significantly associated with an increased risk of AKI (HR: 1.35; 95% CI: 1.17-1.54, for lowest vs. highest quartile for 25-hydroxyvitamin D, and HR: 1.19; 95% CI: 1.05-1.36, for highest vs. lowest quartile for FGF23). The association was consistent across demographic and clinical subgroups, regardless of whether AKI was the primary diagnosis for hospitalization, and when adjusting for incident chronic kidney disease (CKD) or cardiovascular disease (CVD) as a time-varying covariate. CONCLUSION Among middle- to older-age adults in the community, low 25-hydroxyvitamin D and high FGF23 levels were independently associated with an increased risk of AKI. Future studies should explore underlying mechanisms linking these bone mineral metabolism markers with kidney injury.
Collapse
Affiliation(s)
- Junichi Ishigami
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erin D. Michos
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Pamela L. Lutsey
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
50
|
Graves JM, Vallejo JA, Hamill CS, Wang D, Ahuja R, Patel S, Faul C, Wacker MJ. Fibroblast growth factor 23 (FGF23) induces ventricular arrhythmias and prolongs QTc interval in mice in an FGF receptor 4-dependent manner. Am J Physiol Heart Circ Physiol 2021; 320:H2283-H2294. [PMID: 33929896 DOI: 10.1152/ajpheart.00798.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factor 23 (FGF23) is a phosphate regulating protein hormone released by osteocytes. FGF23 becomes markedly elevated in chronic kidney disease (CKD), for which the leading cause of death is cardiovascular disease, particularly sudden cardiac death. Previously, we found that FGF23 increases intracellular Ca2+ in cardiomyocytes and alters contractility in mouse ventricles ex vivo via FGF receptor 4 (FGFR4). In the present study, we demonstrate that FGF23 induces cardiac arrhythmias and prolongs QTc interval in mice, and we tested whether these effects are mediated through FGFR4. In isolated Langendorff perfused hearts, FGF23 perfusion increased mechanical arrhythmias in the form of premature ventricular beats (PVBs), and induced runs of ventricular tachycardia in 6 of 11 animals, which were attenuated with pretreatment of an anti-FGFR4 blocking antibody. Ex vivo ECG analysis of isolated intact hearts showed increased ventricular arrhythmias and QTc prolongation after FGF23 infusion compared with vehicle. In vivo, injection of FGF23 into the jugular vein led to the emergence of premature ventricular contractions (PVCs) in 5 out of 11 experiments. FGF23 also produced a significant lengthening effect upon QTc interval in vivo. In vivo FGFR4 blockade ameliorated the arrhythmogenic and QTc prolonging effects of FGF23. Finally, FGF23 increased cardiomyocyte Ca2+ levels in intact left ventricular muscle which was inhibited by FGR4 blockade. We conclude that FGF23/FGFR4 signaling in the heart may contribute to ventricular arrhythmogenesis and repolarization disturbances commonly observed in patients with CKD via Ca2+ overload and may be an important therapeutic target to reduce cardiac mortality in CKD.NEW & NOTEWORTHY Here we provide direct evidence that fibroblast growth factor 23 (FGF23), a phosphaturic hormone elevated in chronic kidney disease, is proarrhythmic. FGF23 acutely triggered ventricular arrhythmias and prolonged corrected QT interval (QTc) in isolated mouse hearts and in vivo. FGF23 also increased Ca2+ levels in ventricular muscle tissue. Blockade of the FGF receptor 4 signaling pathway using a monoclonal antibody ameliorated ventricular arrhythmias, QTc prolongation, and elevated ventricular Ca2+ induced by FGF23, and may represent a potential therapeutic target in chronic kidney disease.
Collapse
Affiliation(s)
- Jonah M Graves
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Julian A Vallejo
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Chelsea S Hamill
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Derek Wang
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Rohan Ahuja
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Shaan Patel
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Christian Faul
- Division of Nephrology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Michael J Wacker
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| |
Collapse
|