1
|
Mei J, Ke H, Zhu J. Icariside II Alleviates Chondrocyte Inflammatory Injury by Inhibiting the TNIP2/NF-κB Pathway. Cell Biochem Biophys 2025:10.1007/s12013-024-01635-9. [PMID: 39775470 DOI: 10.1007/s12013-024-01635-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Icariside II exerts protective effects against various diseases; however, its specific effects on osteoarthritis (OA) remain unclear. Therefore, in this study, we aimed to investigate the effects of icariside II in an in vitro model of OA and analyze its action mechanisms. We established an in vitro OA model by treating a human chondrocyte cell line (CHON-001) with interleukin (IL)-1β, followed by treatment with different concentrations of icariside II. Cell viability was measured using the methyl thiazolyl tetrazolium assay, and the level of lactate dehydrogenase (LDH) released from cells was determined using the appropriate kit. Tumor necrosis factor (TNF)-α, IL-6, and IL-8 levels were determined via enzyme-linked immunosorbent assay. Flow cytometry was used to assess apoptosis. Apoptosisrelated protein expression levels and TNFAIP3-interacting protein 2 (TNIP2)/nuclear factor (NF)-κB signaling pathway were analyzed via reverse transcription-quantitative polymerase chain reaction and western blotting. Furthermore, TNIP2-small interfering RNA (siRNA) was used to determine whether the TNIP2/NF-κB pathway influences the effects of icariside II on OA. Results indicated that Icariside II did not exert any significant toxic effects on CHON-001 cells. It inhibited IL-1β-induced apoptosis and increase in LDH levels and enhanced the inflammatory response. Additionally, icariside II reversed the IL-1β-induced decrease in TNIP2 levels and increase in NF-κB phosphorylation. TNIP2-siRNA revealed that the TNIP2/NF-κB signaling pathway influenced the alleviating effects of icariside II on OA. In conclusion, our results revealed that icariside II attenuated IL-1β-induced inflammatory injury in chondrocytes by increasing TNIP2 expression and inhibiting NF-κB pathway activation, highlighting its therapeutic potential for OA.
Collapse
Affiliation(s)
- Jiajun Mei
- Department of Pain Medicine, Wuhan University of Science and Technology Affiliated Puren Hospital, Wuhan, China
| | - Hongkui Ke
- Department of Pain Medicine, Wuhan University of Science and Technology Affiliated Puren Hospital, Wuhan, China
| | - Junsong Zhu
- Department of Pain Medicine, Wuhan University of Science and Technology Affiliated Puren Hospital, Wuhan, China.
| |
Collapse
|
2
|
Wang Z, Gui Z, Zhang L, Wang Z. Advances in the mechanisms of vascular calcification in chronic kidney disease. J Cell Physiol 2025; 240:e31464. [PMID: 39392232 DOI: 10.1002/jcp.31464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Vascular calcification (VC) is common in patients with advanced chronic kidney disease (CKD).A series of factors, such as calcium and phosphorus metabolism disorders, uremic toxin accumulation, inflammation and oxidative stress and cellular senescence, cause osteoblast-like differentiation of vascular smooth muscle cells, secretion of extracellular vesicles, and imbalance of calcium regulatory factors, which together promote the development of VC in CKD. Recent advances in epigenetics have provided better tools for the investigation of VC etiology and new approaches for finding more accurate biomarkers. These advances have not only deepened our understanding of the pathophysiological mechanisms of VC in CKD, but also provided valuable clues for the optimization of clinical predictors and the exploration of potential therapeutic targets. The aim of this article is to provide a comprehensive overview of the pathogenesis of CKD VC, especially the new advances made in recent years, including the various key factors mentioned above. Through the comprehensive analysis, we expect to provide a solid theoretical foundation and research direction for future studies targeting the specific mechanisms of CKD VC, the establishment of clinical predictive indicators and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Zebin Gui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Lirong Zhang
- Department of Radiology, Affliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| |
Collapse
|
3
|
Cheng Y, Zhao C, Bin Y, Liu Y, Cheng L, Xia F, Tian X, Liu X, Liu S, Ying B, Shao Z, Yan W. The pathophysiological functions and therapeutic potential of GPR39: Focus on agonists and antagonists. Int Immunopharmacol 2024; 143:113491. [PMID: 39549543 DOI: 10.1016/j.intimp.2024.113491] [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: 09/03/2024] [Revised: 10/09/2024] [Accepted: 10/21/2024] [Indexed: 11/18/2024]
Abstract
G protein-coupled receptor 39 (GPR39), a member of the growth hormone-releasing peptide family, exhibits widespread expression across various tissues and demonstrates high constitutive activity, primarily activated by zinc ions. It plays critical roles in cell proliferation, differentiation, survival, apoptosis, and ion transport through the recruitment of Gq/11, Gs, G12/13, and β-arrestin proteins. GPR39 is involved in anti-inflammatory and antioxidant responses, highlighting its diverse pathophysiological functions. Recent discoveries of endogenous ligands have enhanced our understanding of GPR39's physiological roles. Aberrant expression and reactivation of GPR39 have been implicated in a range of diseases, particularly central nervous system disorders, endocrine disruptions, cardiovascular diseases, cancers, and liver conditions. These findings position GPR39 as a promising therapeutic target, with the efficacy of synthetic ligands validated in various in vivo models. Nonetheless, their clinical applicability remains uncertain, necessitating further exploration of novel agonists-especially biased agonists-and antagonists. This review examines the unique residues contributing to the high constitutive activity of GPR39, its endogenous and synthetic ligands, and its pathophysiological implications, aiming to elucidate its pharmacological potential for clinical application in disease treatment.
Collapse
Affiliation(s)
- Yuhui Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chang Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yan Bin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610000 China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaowen Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xinlei Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Sicen Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Binwu Ying
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhenhua Shao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Tianfu Jincheng Laboratory, Frontiers Medical Center, Chengdu 610212, Sichuan, China.
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Division of Nephrology and Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
4
|
Zhang Y, Huang L, Ou S. Research progress on the association between TMAO and vascular calcification in patients with chronic kidney disease. Ren Fail 2024; 46:2435485. [PMID: 39627031 PMCID: PMC11616764 DOI: 10.1080/0886022x.2024.2435485] [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: 08/08/2024] [Revised: 11/14/2024] [Accepted: 11/23/2024] [Indexed: 12/06/2024] Open
Abstract
Vascular calcification (VC) is a common complication in patients with chronic kidney disease (CKD) and a major risk factor for increased cardiovascular mortality in patients with CKD. Its pathology and pathogenesis are complex and have not been fully elucidated. Trimethylamine N-oxide (TMAO) is an enteric-borne uremic toxin that has been found to play a role in the progression of VC. This article mainly reviews the metabolism of TMAO, the relationship between TMAO and VC in CKD patients, and possible treatments for TMAO, aiming to further explore the mechanism of VC occurrence in CKD patients and provide potential diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
| | - Liangying Huang
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
| | - Santao Ou
- Department of Nephrology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
| |
Collapse
|
5
|
Takahashi A. Managing Zinc Supplementation in Hemodialysis Patients: Balancing and Preventing Deficiencies in Serum Copper and Zinc Levels with and Without HIF-PH Inhibitors. Nutrients 2024; 16:4135. [PMID: 39683529 DOI: 10.3390/nu16234135] [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: 11/04/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Zinc supplementation induces metallothionein, leading to reduced serum copper levels. Conversely, serum copper concentrations tend to rise with the use of HIF-PH inhibitors. METHODS To establish a safe level of zinc supplementation that avoids copper deficiency, serum copper and zinc concentrations measured every three months were retrospectively analyzed over five years in 50 patients undergoing hemodialysis. RESULTS At the initiation of the study, the median (IQR) concentrations were 100 (84.25-109) µg/dL for serum copper and 60.5 (50.5-70) µg/dL for serum zinc. All participants without zinc supplementation exhibited zinc deficiency (<80 µg/dL). After three months, copper deficiency (<71 µg/dL) was observed when serum copper concentrations were <98.6 µg/dL for patients with HIF-PH inhibitors and <90.3 µg/dL for patients without them. Reduced zinc supplementation may be necessary when serum copper falls below 90 µg/dL. Zinc levels remained deficient because supplementation was limited due to concerns about copper deficiency. Lowering the target zinc level to around 80 µg/dL instead of the conventional 80-120 µg/dL may be safer. CONCLUSIONS Regular monitoring of both copper and zinc levels, taking place at least every three months, is recommended to adjust zinc supplementation, especially in patients on HIF-PH inhibitors. Copper supplementation should also be considered alongside zinc supplementation to effectively treat hypozincemia.
Collapse
Affiliation(s)
- Akira Takahashi
- Dialysis Center, Tesseikai Neurosurgical Hospital, 28-1 Nakanohonmachi, Shijonawate 575-8511, Japan
| |
Collapse
|
6
|
Liao YR, Tsai YC, Hsieh TH, Tsai MT, Lin FY, Lin SJ, Lin CC, Chiang HY, Chu PH, Li SY. FHL2 in arterial medial calcification in chronic kidney disease. Nephrol Dial Transplant 2024; 39:2025-2039. [PMID: 38664060 PMCID: PMC11596093 DOI: 10.1093/ndt/gfae091] [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: 09/17/2023] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Arterial medial calcification (AMC) is a common complication in individuals with chronic kidney disease (CKD), which can lead to cardiovascular morbidity and mortality. The progression of AMC is controlled by a key transcription factor called runt-related transcription factor 2 (RUNX2), which induces vascular smooth muscle cells (VSMCs) transdifferentiation into an osteogenic phenotype. However, RUNX2 has not been targeted for therapy due to its essential role in bone development. The objective of our study was to discover a RUNX2 coactivator that is highly expressed in arterial VSMCs as a potential therapy for AMC. METHODS We employed transcriptomic analysis of human data and an animal reporter system to pinpoint four and a half LIM domains 2 (FHL2) as a potential target. Subsequently, we investigated the mRNA and protein expression patterns of FHL2 in the aortas of both human and animal subjects with CKD. To examine the role of FHL2 in the RUNX2 transcription machinery, we conducted coimmunoprecipitation and chromatin immunoprecipitation experiments. Next, we manipulated FHL2 expression in cultured VSMCs to examine its impact on high phosphate-induced transdifferentiation. Finally, we employed FHL2-null mice to confirm the role of FHL2 in the development of AMC in vivo. RESULTS Among all the potential RUNX2 cofactors, FHL2 displays selective expression within the cardiovascular system. In the context of CKD subjects, FHL2 undergoes upregulation and translocation from the cytosol to the nucleus of arterial VSMCs. Once in the nucleus, FHL2 interacts structurally and functionally with RUNX2, acting as a coactivator of RUNX2. Notably, the inhibition of FHL2 expression averts transdifferentiation of VSMCs into an osteogenic phenotype and mitigates aortic calcification in uremic animals, without causing any detrimental effects on the skeletal system. CONCLUSION These observations provide evidence that FHL2 is a promising target for treating arterial calcification in patients with CKD.
Collapse
MESH Headings
- Animals
- LIM-Homeodomain Proteins/metabolism
- LIM-Homeodomain Proteins/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/complications
- Humans
- Mice
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/etiology
- Vascular Calcification/genetics
- Male
- Cell Transdifferentiation
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Knockout
Collapse
Affiliation(s)
- Yuan-Ru Liao
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Cheng Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yen Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hou-Yu Chiang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Guang University, Taoyuan, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taiwan
| | - Szu-Yuan Li
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
7
|
Ma DJ, Oh BL, Bak E, Kim JS, Lee J, Choi HJ. A Comprehensive Health Screening Program Reveals the Prevalence of and Risk Factors for Age-Related Macular Degeneration: A Cross-Sectional Analysis. Biomedicines 2024; 12:2681. [PMID: 39767587 PMCID: PMC11727633 DOI: 10.3390/biomedicines12122681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: We investigated the prevalence of age-related macular degeneration (AMD) and associated risk factors in Korean subjects who underwent comprehensive health screening examinations. Methods: This single health screening center-based cross-sectional study included a total of 73,574 consecutive participants older than 30 years who underwent a health screening examination, including fundus photography, between October 2003 and December 2010. Weighted prevalence and risk factors for AMD were evaluated. Logistic regression was used to identify AMD risk factors. Results: The weighted prevalence of AMD was 15.42%, with a prevalence of 3.34% among people in their 30s. Advanced age significantly increased the risk for both early/intermediate AMD (p < 0.001 across the age groups of 40, 50, 60, and 70+ years) and advanced AMD (p <0.001 for the age groups of 60 and 70+ years). The male sex was strongly associated with an increased risk of both early/intermediate and advanced AMD (p < 0.001 for both). Retinal arteriosclerosis, whether low- or high-grade, was linked to early/intermediate AMD (p < 0.001 for both grades), whereas only high-grade arteriosclerosis was linked to advanced AMD (p < 0.001). Additionally, hypertension (p < 0.001), the hepatitis B carrier status (p < 0.001), elevated mean corpuscular volume (p < 0.001), and lower serum uric acid levels (p = 0.014) were associated with early/intermediate AMD. Higher education levels protected against early/intermediate AMD (p = 0.004 for high school graduates, p < 0.001 for ≥college graduates). Higher serum inorganic phosphate levels (p = 0.002) and lower total serum ALB levels (p = 0.005) were significant risk factors for advanced AMD. Conclusions: Korean individuals as young as 30 years old are at risk of AMD. This study newly identified associations between retinal arteriosclerosis and both early/intermediate and advanced AMD, as well as associations between serum inorganic phosphate levels and total ALB levels with advanced AMD.
Collapse
Affiliation(s)
- Dae Joong Ma
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.J.M.)
- Department of Ophthalmology, Hallym University Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Baek-Lok Oh
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.J.M.)
- Genome Insight, Inc., Daejeon 34051, Republic of Korea
| | - Eunoo Bak
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.J.M.)
- Department of Ophthalmology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu 11759, Republic of Korea
| | - Jin-Soo Kim
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.J.M.)
- Department of Ophthalmology, Chungnam National University School of Medicine, Daejeon 34134, Republic of Korea
| | - Jinho Lee
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.J.M.)
- Hana Seoul Eye Clinic, Bucheon 14537, Republic of Korea
| | - Hyuk Jin Choi
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; (D.J.M.)
- Department of Ophthalmology, Seoul National University Hospital Healthcare System Gangnam Center, Seoul 06236, Republic of Korea
| |
Collapse
|
8
|
Balogh E, Tóth A, Csiki DM, Jeney V. Zinc Ameliorates High Pi and Ca-Mediated Osteogenic Differentiation of Mesenchymal Stem Cells. Nutrients 2024; 16:4012. [PMID: 39683406 DOI: 10.3390/nu16234012] [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: 10/23/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Zinc is the second most abundant trace element in the human body, stored mainly in the bones. Zinc is required for bone growth and homeostasis and is also a crucial cofactor for numerous proteins that play key roles in maintaining microstructural integrity and bone remodeling. Bone marrow-derived mesenchymal stem cells (BMSCs) are multipotent progenitors found in the bone marrow stroma and can differentiate along multiple lineage pathways. In this study, we investigated the effect of zinc on the osteogenic differentiation of BMSCs. We stimulated the osteogenic differentiation of BMSCs with high phosphate and Ca-containing osteogenic medium (PiCa) in the presence or absence of zinc. We followed calcification by measuring ECM mineralization, the Ca content of the ECM, mRNA, and the protein expression of the osteo-chondrogenic transcription factor RUNX2 and SOX9 and its targets OCN and ALP. Zinc dose-dependently abolished PiCa-induced ECM mineralization and decreased the expression of RUNX2, SOX9, OCN, and ALP. Serum albumin did not alter the inhibitory effect of zinc on BMSC mineralization. Our further analysis with the zinc-chelator TPEN and ZnCl2 confirmed the specific inhibitory effect of free zinc ions on BMSC mineralization. Zinc inhibited phosphate uptake and PiCa-induced upregulation of the sodium-dependent phosphate cotransporters (PiT-1 and PiT-2). Zinc attenuated the PiCa-induced increase in ROS production. Taken together, these data suggest that zinc inhibits PiCa-induced BMSC calcification by regulating phosphate uptake and ROS production.
Collapse
Affiliation(s)
- Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Dávid Máté Csiki
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
9
|
Mitsis A, Khattab E, Christodoulou E, Myrianthopoulos K, Myrianthefs M, Tzikas S, Ziakas A, Fragakis N, Kassimis G. From Cells to Plaques: The Molecular Pathways of Coronary Artery Calcification and Disease. J Clin Med 2024; 13:6352. [PMID: 39518492 PMCID: PMC11545949 DOI: 10.3390/jcm13216352] [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: 09/22/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024] Open
Abstract
Coronary artery calcification (CAC) is a hallmark of atherosclerosis and a critical factor in the development and progression of coronary artery disease (CAD). This review aims to address the complex pathophysiological mechanisms underlying CAC and its relationship with CAD. We examine the cellular and molecular processes that drive the formation of calcified plaques, highlighting the roles of inflammation, lipid accumulation, and smooth muscle cell proliferation. Additionally, we explore the genetic and environmental factors that contribute to the heterogeneity in CAC and CAD presentation among individuals. Understanding these intricate mechanisms is essential for developing targeted therapeutic strategies and improving diagnostic accuracy. By integrating current research findings, this review provides a comprehensive overview of the pathways linking CAC to CAD, offering insights into potential interventions to mitigate the burden of these interrelated conditions.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Elina Khattab
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Evi Christodoulou
- Cardiology Department, Limassol General Hospital, State Health Services Organization, Limassol 3304, Cyprus;
| | - Kimon Myrianthopoulos
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Michael Myrianthefs
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (A.M.); (E.K.); (K.M.); (M.M.)
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| |
Collapse
|
10
|
Wang Z, Tang F, Zhao B, Yan H, Shao X, Yang Q. Composite dietary antioxidant index and abdominal aortic calcification: a national cross-sectional study. Nutr J 2024; 23:130. [PMID: 39438863 PMCID: PMC11494755 DOI: 10.1186/s12937-024-01029-w] [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: 04/26/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
PURPOSE The Composite Dietary Antioxidant Index (CDAI) is a novel, inclusive measure for evaluating the antioxidant potential of diets. We aim to explore the link between the CDAI and abdominal aortic calcification (AAC) in U.S. adults aged ≥ 40 years. METHODS This cross-sectional study collected dietary and AAC data for individuals aged ≥ 40 years from the 2013-2014 National Health and Nutrition Examination Survey (NHANES) database. The CDAI was calculated using six dietary antioxidants. AAC was evaluated using a semi-quantitative scoring system known as AAC-24, with an AAC score greater than 6 as severe AAC (SAAC). To examine the association between CDAI and AAC, including SAAC, liner/logistic regression analyses and smooth curve fitting were applied. RESULTS A total of 2,640 participants were included in this study, and significant decreases in AAC score and SAAC prevalence were observed with ascending CDAI levels (P < 0.01). After adjusting for confounding factors, a clear link was established between the CDAI and both AAC score (β = -0.083, 95% CI -0.144-0.022, P = 0.008) and SAAC (OR = 0.883, 95% CI 0.806-0.968, P = 0.008), respectively. Further smooth curve fitting indicated a negative correlation between CDAI and both AAC score and SAAC. CONCLUSIONS Dietary antioxidant consumption, as quantified by the CDAI, shows an inverse relationship with AAC risk. Additional longitudinal and intervention studies are essential.
Collapse
Affiliation(s)
- Zhaoxiang Wang
- Department of Endocrinology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Fengyan Tang
- Department of Endocrinology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Bo Zhao
- Department of Cardiology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, 215300, China
| | - Han Yan
- Department of Endocrinology, Affiliated Wujin Hospital of Jiangsu University, Changzhou, Jiangsu, 213017, China
- Department of Endocrinology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213017, China
| | - Xuejing Shao
- Department of Endocrinology, Affiliated Wujin Hospital of Jiangsu University, Changzhou, Jiangsu, 213017, China
- Department of Endocrinology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213017, China
| | - Qichao Yang
- Department of Endocrinology, Affiliated Wujin Hospital of Jiangsu University, Changzhou, Jiangsu, 213017, China.
- Department of Endocrinology, Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213017, China.
| |
Collapse
|
11
|
Li K, Yan J, Wang S, Zhu C, Zhu Q, Lu S, Hu P, Dessie T, Kim IH, Ahmed AA, Liu HY, Ennab W, Cai D. Transcriptome analysis provides new insights into the response of canine intestinal epithelial cells treated by sulforaphane: a natural product of cruciferous origin. Front Vet Sci 2024; 11:1460500. [PMID: 39415954 PMCID: PMC11479859 DOI: 10.3389/fvets.2024.1460500] [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: 07/06/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
This study presents a comprehensive transcriptome analysis of canine intestinal epithelial cells following treatment with sulforaphane (SFN), a naturally occurring compound found in cruciferous vegetables with established anti-inflammatory and antioxidant properties. Through high-throughput sequencing, we identified 29,993 genes, among which 1,612 were differentially expressed, with 792 up-regulated and 820 down-regulated in response to SFN treatment. Our analysis revealed significant enrichment of genes in pathways associated with the inflammatory response, lipid metabolism, oxidative stress response, and T-cell mediated immunity, suggesting SFN's potential in modulating these biological processes. Notably, the PPARγ gene, which plays a crucial role in the body's oxidative stress and inflammatory response, was highly up-regulated, indicating its possible centrality in SFN's effects. Gene-gene interaction analysis further supported SFN's role in alleviating inflammation through PPARγ, with key genes in oxidative stress and inflammatory response pathways showing significant correlations with PPARγ. Overall, our findings provide molecular evidence for SFN's protective effects on canine intestinal health, potentially through the modulation of inflammatory and oxidative stress pathways, with PPARγ emerging as a critical mediator.
Collapse
Affiliation(s)
- Kaiqi Li
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jin Yan
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Shiqi Wang
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chuyang Zhu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Qi Zhu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Sichen Lu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ping Hu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tadelle Dessie
- International Livestock Research Institute, Addis Ababa, Ethiopia
| | - In Ho Kim
- Department of Animal Resource and Science, Dankook University, Cheonan, Republic of Korea
| | - Abdelkareem A. Ahmed
- Department of Veterinary Biomedical Sciences, Botswana University of Agriculture and Agriculture and Natural Resources, Gaborone, Botswana
| | - Hao-Yu Liu
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Wael Ennab
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Demin Cai
- Laboratory of Animal Physiology and Molecular Nutrition, Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- International Joint Research Laboratory in Universities of Jiangsu Province of China for Domestic Animal Germplasm Resources and Genetic Improvement, Yangzhou, Jiangsu, China
| |
Collapse
|
12
|
Mao J, Sun Z, Wang S, Bi J, Xue L, Wang L, Wang H, Jiao G, Chen Y. Multifunctional Bionic Periosteum with Ion Sustained-Release for Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403976. [PMID: 39225563 PMCID: PMC11497021 DOI: 10.1002/advs.202403976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/28/2024] [Indexed: 09/04/2024]
Abstract
In this study, a novel bionic periosteum (BP)-bioactive glass fiber membrane (BGFM) is designed. The introduction of magnesium ion (Mg2+) and zinc ion (Zn2+) change the phase separation during the electrospinning (ES) jet stretching process. The fiber's pore structure transitions from connected to closed pores, resulting in a decrease in the rapid release of metal ions while also improving degradation via reducing filling quality. Additionally, the introduction of magnesium (Mg) and zinc (Zn) lead to the formation of negative charged tetrahedral units (MgO4 2- and ZnO4 2-) in the glass network. These units effectively trap positive charged metal ions, further inhibiting ion release. In vitro experiments reveal that the deigned bionic periosteum regulates the polarization of macrophages toward M2 type, thereby establishing a conducive immune environment for osteogenic differentiation. Bioinformatics analysis indicate that BP enhanced bone repair via the JAK-STAT signaling pathway. The slow release of metal ions from the bionic periosteum can directly enhance osteogenic differentiation and vascularization, thereby accelerating bone regeneration. Finally, the bionic periosteum exhibits remarkable capabilities in angiogenesis and osteogenesis, demonstrating its potential for bone repair in a rat calvarial defect model.
Collapse
Affiliation(s)
- Junjie Mao
- Liquid‐Solid Structural Evolution & Processing of Materials (Ministry of Education)School of Materials Science and EngineeringShandong UniversityJinanShandong250061P. R. China
| | - Zhenqian Sun
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandong250012P. R. China
- The First Clinical Medical SchoolShandong UniversityJinanShandong250012P. R. China
| | - Shidong Wang
- Musculoskeletal Tumor CenterPeking University People's HospitalBeijing100044P. R. China
| | - Jianqiang Bi
- Liquid‐Solid Structural Evolution & Processing of Materials (Ministry of Education)School of Materials Science and EngineeringShandong UniversityJinanShandong250061P. R. China
| | - Lu Xue
- Shandong Second Medical UniversityWeifangShandong261000P. R. China
- Shanxian Central HospitalHezeShandong274300P. R. China
| | - Lu Wang
- Liquid‐Solid Structural Evolution & Processing of Materials (Ministry of Education)School of Materials Science and EngineeringShandong UniversityJinanShandong250061P. R. China
| | - Hongliang Wang
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandong250012P. R. China
| | - Guangjun Jiao
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandong250012P. R. China
| | - Yunzhen Chen
- Department of OrthopaedicsQilu Hospital of Shandong UniversityJinanShandong250012P. R. China
| |
Collapse
|
13
|
Chen W, Chen A, Lian G, Yan Y, Liu J, Wu J, Gao G, Xie L. Zinc attenuates monocrotaline-induced pulmonary hypertension in rats through upregulation of A20. J Mol Cell Cardiol 2024; 195:24-35. [PMID: 39002608 DOI: 10.1016/j.yjmcc.2024.07.003] [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: 01/18/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Pulmonary hypertension (PH) is characterized by excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), in which inflammatory signaling caused by activation of the NF-κB pathway plays an important role. A20 is an important negative regulator of the NF-κB pathway, and zinc promotes the expression of A20 and exerts a protective effect against various diseases (e.g. COVID19) by inhibiting the inflammatory signaling. The role of A20 and intracellular zinc signaling in PH has been explored, but the extracellular zinc signaling is not well understood, and whether zinc has protective effects on PH is still elusive. Using inductively coupled plasma mass spectrometry (ICP-MS), we studied the alteration of trace elements during the progression of monocrotaline (MCT)-induced PH and found that serum zinc concentration was decreased with the onset of PH accompanied by abnormalities of other three elements, including copper, chromium, and magnesium. Zinc chloride injection with the dosage of 5 mg/kg intraperitoneally partially corrected this abnormality and inhibited the progression of PH. Zinc supplementation induced the expression of A20 in lung tissue and reduce the inflammatory responses. In vitro, zinc supplementation time-dependently upregulated the expression of A20 in PASMCs, therefore correcting the excessive proliferation and migration of cells caused by hypoxia. Using genetically encoded-FRET based zinc probe, we found that these effects of zinc ions are not achieved by entering cells, but most likely by activating cell surface zinc receptor (ZnR/GPR39). These results provide the first evidence of the effectiveness of zinc supplementation in the treatment of PH.
Collapse
Affiliation(s)
- Weixiao Chen
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Ai Chen
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Guili Lian
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Yan Yan
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Junping Liu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Jingying Wu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Gufeng Gao
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Clinical Research Center for Geriatric Hypertension Disease of Fujian province, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China; Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fujian Province, Fuzhou, People's Republic of China; Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China.
| |
Collapse
|
14
|
Nakatani S, Shoji T, Morioka F, Nakaya R, Ueda M, Uedono H, Tsuda A, Morioka T, Fujii H, Yoshida H, Mori K, Emoto M. Association between Serum Zinc and All-Cause Mortality in Patients Undergoing Maintenance Hemodialysis: The Osaka Dialysis Complication Study (ODCS). Nutrients 2024; 16:3270. [PMID: 39408237 PMCID: PMC11478467 DOI: 10.3390/nu16193270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Zinc is an essential microelement, and its deficiency is common in patients undergoing hemodialysis. However, the association between serum zinc and mortality in these patients remains unclear. The aim of this study was to explore the possible association between serum zinc levels and all-cause mortality in prevalent patients with kidney failure on maintenance hemodialysis. METHODS This was a prospective cohort study of maintenance hemodialysis patients followed up for 5 years. The key exposure was serum zinc level measured at baseline, and the outcome was all-cause mortality. Their association was analyzed using Cox proportional hazard models. RESULTS Among 1662 eligible patients selected for this analysis, 468 (28%) died. Lower serum zinc levels were associated with a higher risk for mortality, independent of the major demographic factors and factors including mineral and bone disorder and renal anemia. However, this association was no longer significant when adjusted for serum albumin. Because there was a close correlation between serum zinc and albumin levels, we performed further analyses in which participants were categorized into four groups by median serum zinc (68 µg/dL) and albumin (3.7 g/dL) levels. In the lower serum albumin groups, risk of death was significantly higher in those with lower zinc than those with higher zinc levels, whereas such a difference was not significant in the high serum albumin groups. CONCLUSIONS In patients undergoing maintenance hemodialysis with lower serum albumin levels, a lower serum zinc level was associated with a higher risk of mortality.
Collapse
Affiliation(s)
- Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
| | - Tetsuo Shoji
- Department of Vascular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan;
- Vascular Science Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Fumiyuki Morioka
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
| | - Rino Nakaya
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
| | - Mayuko Ueda
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (M.U.); (H.Y.)
| | - Hideki Uedono
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
| | - Akihiro Tsuda
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
| | - Tomoaki Morioka
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
| | - Hisako Fujii
- Department of Health and Medical Innovation, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan;
| | - Hisako Yoshida
- Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (M.U.); (H.Y.)
| | - Katsuhito Mori
- Department of Nephrology, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan;
| | - Masanori Emoto
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan; (F.M.); (R.N.); (H.U.); (A.T.); (T.M.); (M.E.)
- Vascular Science Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan
- Department of Nephrology, Osaka Metropolitan University Graduate School of Medicine, Osaka 545-8585, Japan;
| |
Collapse
|
15
|
Geroldinger-Simic M, Sohail A, Razazian M, Krennmayr B, Pernsteiner V, Putz T, Lackner HK, Pasch A, Sepp N, Alesutan I, Voelkl J. Accelerated calciprotein crystallization time (T50) is correlated with impaired lung diffusion capacity in systemic sclerosis. Front Immunol 2024; 15:1425885. [PMID: 39399492 PMCID: PMC11466802 DOI: 10.3389/fimmu.2024.1425885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024] Open
Abstract
Background Systemic sclerosis (SSc) is a complex auto-immune disease characterized by vascular damage, inflammation, fibrosis and calcinosis, where pulmonary involvement is the leading cause of mortality. Calciprotein particles (CPPs) are increasingly formed upon disbalance of the physiological mineral buffering system and induce pro-inflammatory effects. This exploratory study investigated whether functional indicators of the endogenous mineral buffering system are dysregulated in SSc and linked to disease activity. Methods T50 (calciprotein crystallization test or serum calcification propensity) and hydrodynamic radius of secondary CPPs (CPP2) were determined in serum samples from 78 SSc patients and 44 controls without SSc, and were associated with disease activity markers of SSc. Results T50 was reduced and CPP2 radius was increased in SSc patients as compared to controls, indicating a deranged mineral buffering system. This was accompanied by slightly higher serum phosphate and PTH levels in SSc patients, while iFGF23 was not significantly modified. Longitudinally, all parameters remained unchanged over time in SSc patients, only iFGF23 increased. While the modified Rodnan skin score showed some inconsistent correlations with mineral buffering indicators, their association was not independent of other factors. However, lower T50 was significantly correlated to reduced lung diffusion capacity and this association remained significant in a multivariate linear regression model. Conclusion This study provides indications for a disturbed mineral buffering system in SSc. Increased serum calcification propensity (lower T50) is correlated with impaired lung diffusion capacity, suggesting a possible role of deranged mineral buffering in disease progression. Further studies are required to confirm these observations in larger cohorts and to investigate a putative functional relevance.
Collapse
Affiliation(s)
- Marija Geroldinger-Simic
- Department of Dermatology and Venereology, Ordensklinikum Linz Elisabethinen, Linz, Austria
- Faculty of Medicine, Johannes Kepler University, Linz, Austria
| | - Azmat Sohail
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Mehdi Razazian
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Beatrice Krennmayr
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Victoria Pernsteiner
- Department of Dermatology and Venereology, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Thomas Putz
- Department of Dermatology and Venereology, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Helmut K. Lackner
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Calciscon AG, Biel, Switzerland
| | - Norbert Sepp
- Department of Dermatology and Venereology, Ordensklinikum Linz Elisabethinen, Linz, Austria
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
16
|
Liang X, Li Y, Wang P, Liu H. Key regulators of vascular calcification in chronic kidney disease: Hyperphosphatemia, BMP2, and RUNX2. PeerJ 2024; 12:e18063. [PMID: 39308809 PMCID: PMC11416758 DOI: 10.7717/peerj.18063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Vascular calcification is quite common in patients with end-stage chronic kidney disease and is a major trigger for cardiovascular complications in these patients. These complications significantly impact the survival rate and long-term prognosis of individuals with chronic kidney disease. Numerous studies have demonstrated that the development of vascular calcification involves various pathophysiological mechanisms, with the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) being of utmost importance. High phosphate levels, bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (RUNX2) play crucial roles in the osteogenic transdifferentiation process of VSMCs. This article primarily reviews the molecular mechanisms by which high phosphate, BMP2, and RUNX2 regulate vascular calcification secondary to chronic kidney disease, and discusses the complex interactions among these factors and their impact on the progression of vascular calcification. The insights provided here aim to offer new perspectives for future research on the phenotypic switching and osteogenic transdifferentiation of VSMCs, as well as to aid in optimizing clinical treatment strategies for this condition, bearing significant clinical and scientific implications.
Collapse
Affiliation(s)
- Xinhua Liang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Yankun Li
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Peng Wang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| | - Huafeng Liu
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| |
Collapse
|
17
|
Sohail A, Obereigner J, Mitter G, Schmid T, Hofer AS, Schuster G, Hügl A, Dorninger AH, Mandl M, Pasch A, Lackner HK, Papousek I, Dieplinger B, Suessner S, Antlanger M, Cejka D, Alesutan I, Voelkl J. Association of serum zinc with mineral stress in chronic kidney disease. Clin Kidney J 2024; 17:sfae258. [PMID: 39286240 PMCID: PMC11403325 DOI: 10.1093/ckj/sfae258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Indexed: 09/19/2024] Open
Abstract
Background The excessive cardiovascular mortality of patients with chronic kidney disease (CKD) could be linked to mineral stress, the biological consequence of calcium-phosphate nanoparticle exposure. This study investigated whether zinc is associated with mineral stress markers in CKD. Methods Zinc and T50 (serum calcification propensity) as well as hydrodynamic radius of secondary calciprotein particles (CPP2) were measured in blood donors and CKD patients with/out dialysis. Results Serum zinc concentrations and T50 were reduced, while CPP2 radius was increased in CKD patients. Serum zinc levels positively correlated with T50 and inversely correlated with CPP2 radius. In a hierarchical linear regression model, T50 was associated with age, calcium, phosphate, magnesium and albumin. Addition of zinc significantly improved prediction of the model, confirming an additional contribution of zinc to T50. Similar observations were made for the association of zinc and CPP2 radius, but spiking experiments indicated that zinc may stronger modify T50 than CPP2 radius. Also, urinary zinc excretion was increased in patients with kidney disease and correlated to T50 and CPP2 radius. Serum zinc further correlated with markers of arterial stiffness in blood donors and CKD patients, but these associations did not remain significant in a multivariate linear regression model. Conclusions Reduced serum zinc levels in CKD appear directly linked to lower T50 and associated with larger CPP2 radius. Further studies on the associations of zinc and mineral stress as well as putative therapeutic benefits of zinc supplementation are required.
Collapse
Affiliation(s)
- Azmat Sohail
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Obereigner
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Gregor Mitter
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | | | - Anna-Sofie Hofer
- Department of Medicine III - Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz, Linz, Austria
| | - Gerhard Schuster
- Red Cross Transfusion Service of Upper Austria, Austrian Red Cross, Linz, Austria
| | - Astrid Hügl
- Red Cross Transfusion Service of Upper Austria, Austrian Red Cross, Linz, Austria
| | - Angelika H Dorninger
- Red Cross Transfusion Service of Upper Austria, Austrian Red Cross, Linz, Austria
| | - Markus Mandl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Andreas Pasch
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Calciscon AG, Biel, Switzerland
| | - Helmut K Lackner
- Division of Physiology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Ilona Papousek
- Institute of Psychology, Biological Psychology Unit, University of Graz, Graz, Austria
| | - Benjamin Dieplinger
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder Linz and Ordensklinikum Linz, Linz, Austria
| | - Susanne Suessner
- Red Cross Transfusion Service of Upper Austria, Austrian Red Cross, Linz, Austria
| | - Marlies Antlanger
- Department of Internal Medicine 2, Kepler University Hospital and Johannes Kepler University, Linz, Austria
| | - Daniel Cejka
- Department of Medicine III - Nephrology, Hypertension, Transplantation Medicine, Rheumatology, Geriatrics, Ordensklinikum Linz, Linz, Austria
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| |
Collapse
|
18
|
Liu C, Zhang H, Yang Y, Cao Y, Liang D. Association Between Dietary Zinc Intake and Increased Renal Function in US Adults. Biol Trace Elem Res 2024; 202:3871-3885. [PMID: 38015328 DOI: 10.1007/s12011-023-03969-6] [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: 08/15/2023] [Accepted: 11/19/2023] [Indexed: 11/29/2023]
Abstract
We aimed to investigate the association between the dietary zinc intake and the risk of albuminuria, low estimated glomerular filtration rate (eGFR), and chronic kidney disease (CKD) in the US general population. This study was a cross-sectional study utilizing the data from the 2003-2018 National Health and Nutrition Examination Survey. Albuminuria was defined as urinary albumin:creatinine ratio (ACR) > 30 mg/g. Low eGFR was defined as an eGFR of less than 60 mL/min/1.73 m2. CKD is characterized by albuminuria or low eGFR. Multivariate logistic regression analysis, subgroup analyses, interaction tests, and restricted cubic spline (RCS) analysis were performed in this study. For 37,195 enrolled participants in this study, the mean dietary zinc intake was 11.85 ± 0.07 mg/day, and the rate of albuminuria, low eGFR, and CKD was 9.37%, 6.68%, and 14.10%, respectively. Participants with a higher dietary zinc intake showed a lower risk of albuminuria, low eGFR, and CKD. In the fully adjusted model, we found that participants in the highest dietary zinc intake quartile had 26% lower odds of the rate of CKD than those in quartile 1. Subgroup analyses showed that dietary zinc intake was positively associated with the risk of low eGFR in participants who were now smokers. The potential nonlinear relationship between dietary zinc intake and the risk of CKD and albuminuria was also revealed. Higher dietary zinc intake was significantly associated with a lower likelihood of CKD, which might be helpful in kidney function protection among the general population.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Hao Zhang
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuwei Yang
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Cao
- The Eighth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Dan Liang
- Department of Endocrine, People's Hospital of Chongqing Liangjiang New Area, Chongqing, China.
- West China Clinical Medical College of Sichuan University, Sichuan, China.
| |
Collapse
|
19
|
Liu G, Liao W, Lv X, Zhu M, Long X, Xie J. Comprehensive analysis of hypoxia-related genes in diagnosis and immune infiltration in acute myocardial infarction: based on bulk and single-cell RNA sequencing data. Front Mol Biosci 2024; 11:1448705. [PMID: 39234566 PMCID: PMC11371776 DOI: 10.3389/fmolb.2024.1448705] [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: 06/14/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Background Hypoxia has been found to cause cellular dysfunction and cell death, which are essential mechanisms in the development of acute myocardial infarction (AMI). However, the impact of hypoxia-related genes (HRGs) on AMI remains uncertain. Methods The training dataset GSE66360, validation dataset GSE48060, and scRNA dataset GSE163956 were downloaded from the GEO database. We identified hub HRGs in AMI using machine learning methods. A prediction model for AMI occurrence was constructed and validated based on the identified hub HRGs. Correlations between hub HRGs and immune cells were explored using ssGSEA analysis. Unsupervised consensus clustering analysis was used to identify robust molecular clusters associated with hypoxia. Single-cell analysis was used to determine the distribution of hub HRGs in cell populations. RT-qPCR verified the expression levels of hub HRGs in the human cardiomyocyte model of AMI by oxygen-glucose deprivation (OGD) treatment in AC16 cells. Results Fourteen candidate HRGs were identified by differential analysis, and the RF model and the nomogram based on 8 hub HRGs (IRS2, ZFP36, NFIL3, TNFAIP3, SLC2A3, IER3, MAFF, and PLAUR) were constructed, and the ROC curves verified its good prediction effect in training and validation datasets (AUC = 0.9339 and 0.8141, respectively). In addition, the interaction between hub HRGs and smooth muscle cells, immune cells was elucidated by scRNA analysis. Subsequently, the HRG pattern was constructed by consensus clustering, and the HRG gene pattern verified the accuracy of its grouping. Patients with AMI could be categorized into three HRG subclusters, and cluster A was significantly associated with immune infiltration. The RT-qPCR results showed that the hub HRGs in the OGD group were significantly overexpressed. Conclusion A predictive model of AMI based on HRGs was developed and strongly associated with immune cell infiltration. Characterizing patients for hypoxia could help identify populations with specific molecular profiles and provide precise treatment.
Collapse
Affiliation(s)
- Guoqing Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wang Liao
- Department of Cardiology, The First People's Hospital of Yulin, Yulin, Guangxi, China
| | - Xiangwen Lv
- Department of Cardiology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Miaomiao Zhu
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xingqing Long
- The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jian Xie
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
20
|
Li Q, Zhou Q, Li S, Li S, Liao W, Yu L, Liu C, Li M, Xia H. Target analysis and identification of curcumin against vascular calcification. Sci Rep 2024; 14:17344. [PMID: 39069521 PMCID: PMC11284211 DOI: 10.1038/s41598-024-67776-2] [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: 01/29/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024] Open
Abstract
To investigate the mechanism of curcumin (CUR) on vascular calcification (VC), we screen for common targets of CUR and atherosclerosis and verify the targets genes in vivo and in vitro experiments. The common targets of CUR and AS were screened and obtained using different databases. These target genes were analyzed by GO and KEGG pathway enrichment analysis. PPI network analysis was performed and to analyze the key targets. A rat VC model was constructed and CUR was fed for three weeks. The changes of vascular structure and calcium salt deposition were observed in H&E and Von Kossa staining. Further, the expression of these target proteins was detected in the primary VSMCs of VC. The 31 common targets were obtained. GO functional enrichment analysis obtained 1284 terms and KEGG pathway enriched 66 pathways. The key genes were identified in the cytoHubba plugin. The molecular docking analysis showed that CUR bound strongly to EGFR, STAT3 and BCL2. The animal experiments showed the deposition calcium salt reduced by the CUR administration. These proteins BMP2, RUNX2, EGFR, STAT3 and BAX expression were upregulated in VC group and CUR attenuated the upregulated expression. The signal protein Akt and p65 expression increased in VC group and decreased in CUR group. We identified some common target genes of CUR and AS and identified these key genes. The anti-VC effect of CUR was associated with the inhibition of upregulation of EGFR, STAT3 and RUNX2 expression in VSMCs.
Collapse
Affiliation(s)
- Qingjie Li
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
- The Central Hospital of Zhoukou, Zhoukou, 466001, People's Republic of China
| | - Qiaofeng Zhou
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Shihuan Li
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Suqin Li
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Wenli Liao
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Liangzhu Yu
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Mincai Li
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
| | - Hongli Xia
- School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
- The Central Hospital of Xianning, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China.
| |
Collapse
|
21
|
Padoan F, Piccoli E, Pietrobelli A, Moreno LA, Piacentini G, Pecoraro L. The Role of Zinc in Developed Countries in Pediatric Patients: A 360-Degree View. Biomolecules 2024; 14:718. [PMID: 38927121 PMCID: PMC11201578 DOI: 10.3390/biom14060718] [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: 05/14/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Zinc is an important trace element for growth and health at pediatric ages. Zinc is fundamental in inflammatory pathways, oxidative balance, and immune function. Zinc exhibits anti-inflammatory properties by modulating Nuclear Factor-kappa (NF-κB) activity and reducing histamine release from basophils, leukocytes, and mast cells. Furthermore, its antioxidant activity protects against oxidative damage and chronic diseases. Finally, zinc improves the ability to trigger effective immune responses against pathogens by contributing to the maturation of lymphocytes, the production of cytokines, and the regulation of apoptosis. Given these properties, zinc can be considered an adjunctive therapy in treating and preventing respiratory, nephrological, and gastrointestinal diseases, both acute and chronic. This review aims to deepen the role and metabolism of zinc, focusing on the role of supplementation in developed countries in pediatric diseases.
Collapse
Affiliation(s)
- Flavia Padoan
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics University of Verona, 37126 Verona, Italy
| | - Elena Piccoli
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics University of Verona, 37126 Verona, Italy
| | - Angelo Pietrobelli
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics University of Verona, 37126 Verona, Italy
| | - Luis A. Moreno
- Growth, Exercise, Nutrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Universidad de Zaragoza, 50001 Zaragoza, Spain
| | - Giorgio Piacentini
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics University of Verona, 37126 Verona, Italy
| | - Luca Pecoraro
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics University of Verona, 37126 Verona, Italy
| |
Collapse
|
22
|
Padoan F, Guarnaroli M, Brugnara M, Piacentini G, Pietrobelli A, Pecoraro L. Role of Nutrients in Pediatric Non-Dialysis Chronic Kidney Disease: From Pathogenesis to Correct Supplementation. Biomedicines 2024; 12:911. [PMID: 38672265 PMCID: PMC11048674 DOI: 10.3390/biomedicines12040911] [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: 03/16/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Nutrition management is fundamental for children with chronic kidney disease (CKD). Fluid balance and low-protein and low-sodium diets are the more stressed fields from a nutritional point of view. At the same time, the role of micronutrients is often underestimated. Starting from the causes that could lead to potential micronutrient deficiencies in these patients, this review considers all micronutrients that could be administered in CKD to improve the prognosis of this disease.
Collapse
Affiliation(s)
| | | | - Milena Brugnara
- Pediatric Unit, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy (A.P.)
| | | | | | | |
Collapse
|
23
|
Atefipour N, Dianat M, Badavi M, Radan M, Mard SA. The Role of Rosmarinic Acid in the Protection Against Inflammatory Factors in Rats Model With Monocrotaline-Induced Pulmonary Hypertension: Investigating the Signaling Pathway of NFκB, OPG, Runx2, and P-Selectin in Heart. J Cardiovasc Pharmacol 2024; 83:258-264. [PMID: 38151743 DOI: 10.1097/fjc.0000000000001534] [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: 08/09/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023]
Abstract
ABSTRACT Shortness of breath and syncope are common symptoms of right ventricular failure caused by pulmonary arterial hypertension (PAH), which is the result of blockage and increased pressure in the pulmonary arteries. There is a significant amount of evidence supporting the idea that inflammation and vascular calcification (VC) are important factors in PAH pathogenesis. Therefore, we aimed to investigate the features of the inflammatory process and gene expression involved in VC in monocrotaline (MCT)-induced PAH rats. MCT (60 mg/kg, i.p.) was used to induce PAH. Animals were given normal saline or rosmarinic acid (RA) (10, 15, and 30 mg/kg, gavage) for 21 days. An increase in right ventricular systolic pressure was evaluated as confirming PAH. To determine the level of inflammation in lung tissue, pulmonary edema and the total and differential white blood cell counts in the bronchoalveolar lavage fluid were measured. Also, the expression of NFκB, OPG, Runx2, and P-selectin genes was investigated to evaluate the level of VC in the heart. Our experiment showed that RA significantly decreased right ventricular hypertrophy, inflammatory factors, NFκB, Runx2, and P-selectin gene expression, pulmonary edema, total and differential white blood cell count, and increased OPG gene expression. Therefore, our research showed that RA protects against MCT-induced PAH by reducing inflammation and VC in rats.
Collapse
Affiliation(s)
- Narges Atefipour
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; and
| | - Mahin Dianat
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; and
- Persian Gulf Physiology Research Center, Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; and
- Persian Gulf Physiology Research Center, Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Radan
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; and
- Persian Gulf Physiology Research Center, Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; and
- Persian Gulf Physiology Research Center, Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
24
|
Wu W, Fan H, Cen J, Huang P, Li G, Tan Y, Liu G, Hong B. Novel diagnostic biomarkers related to necroptosis and immune infiltration landscape in acute myocardial infarction. PeerJ 2024; 12:e17044. [PMID: 38426147 PMCID: PMC10903340 DOI: 10.7717/peerj.17044] [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: 06/29/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
Background Acute myocardial infarction (AMI) can occur suddenly, which may induce deadly outcomes, and the population suffering from AMI presents a younger trend. Necroptosis, the new cell necrosis type, is associated with the pathogenic mechanisms of diverse cardiovascular diseases (CVDs). Its diagnostic value and molecular mechanisms in AMI are still unclear. Objective: This study focused on determining key necroptosis-related genes as well as immune infiltration in AMI. Methods We first examined the GSE66360 dataset for identifying necroptosis-related differentially expressed genes (NRDEGs). Thereafter, GO and functional annotation were performed, then a PPI network was built. In addition, "CIBERSORT" in R was applied in comparing different immune infiltration degrees in AMI compared with control groups. The receiver operating characteristic (ROC) curve was plotted to evaluate whether hub NRDEGs could be used in AMI diagnosis. Associations of immune cells with candidate NRDEGs biomarkers were examined by Spearman analysis. Finally, hub NRDEGs were validated by cell qPCR assays and another two datasets. Results A total of 15 NRDEGs were identified and multiple enrichment terms associated with necroptosis were discovered through GO and KEGG analysis. Upon module analysis, 10 hub NRDEGs were filtered out, and the top six hub NRDEGs were identified after ROC analysis. These top six NRDEGs might have a certain effect on modulating immune infiltrating cells, especially for mast cells activated, NK cells activated and neutrophils. Finally, two AMI datasets and qPCR assay came to identical findings. Conclusion Our results offer the reliable molecular biomarkers and new perspectives for necroptosis in AMI, which lay a certain foundation for developing novel anti-AMI therapeutic targets.
Collapse
Affiliation(s)
- Wenfa Wu
- General Practice, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Hongxing Fan
- Neurology, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Junlin Cen
- General Practice, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Pei Huang
- General Practice, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Guidong Li
- General Practice, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Yanping Tan
- Neurology, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Gen Liu
- General Practice, Guangzhou Red Cross Hospital, Guangzhou, China
| | - Baoshan Hong
- General Practice, Guangzhou Red Cross Hospital, Guangzhou, China
| |
Collapse
|
25
|
He M, Wei W, Zhang Y, Xiang Z, Peng D, Kasimumali A, Rong S. Gut microbial metabolites SCFAs and chronic kidney disease. J Transl Med 2024; 22:172. [PMID: 38369469 PMCID: PMC10874542 DOI: 10.1186/s12967-024-04974-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/11/2024] [Indexed: 02/20/2024] Open
Abstract
The global incidence of Chronic Kidney Disease (CKD) is steadily escalating, with discernible linkage to the intricate terrain of intestinal microecology. The intestinal microbiota orchestrates a dynamic equilibrium in the organism, metabolizing dietary-derived compounds, a process which profoundly impacts human health. Among these compounds, short-chain fatty acids (SCFAs), which result from microbial metabolic processes, play a versatile role in influencing host energy homeostasis, immune function, and intermicrobial signaling, etc. SCFAs emerge as pivotal risk factors influencing CKD's development and prognosis. This paper review elucidates the impact of gut microbial metabolites, specifically SCFAs, on CKD, highlighting their role in modulating host inflammatory responses, oxidative stress, cellular autophagy, the immune milieu, and signaling cascades. An in-depth comprehension of the interplay between SCFAs and kidney disease pathogenesis may pave the way for their utilization as biomarkers for CKD progression and prognosis or as novel adjunctive therapeutic strategies.
Collapse
Affiliation(s)
- Meng He
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenqian Wei
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yichen Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhouxia Xiang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Dan Peng
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ayijiaken Kasimumali
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
26
|
Gao J, Liu L, Wu Z, Gan H. Zic family member 3 attenuates oxidative stress-induced vascular smooth muscle cell apoptosis in patients with chronic kidney disease. Tissue Cell 2024; 86:102286. [PMID: 38091851 DOI: 10.1016/j.tice.2023.102286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/03/2023] [Accepted: 12/06/2023] [Indexed: 01/21/2024]
Abstract
Neointimal hyperplasia is reportedly essential for arteriovenous fistulas (AVF) in patients undergoing hemodialysis. Oxidative stress is vital in the progression of uremic venous intimal hyperplasia. Studies have suggested that zinc ions obstruct vascular calcification in patients with chronic kidney disease (CKD). Recent studies have shown that the zinc finger protein, Zic family member 3 (ZIC3), is crucial for the earliest cardiovascular progenitors. ZIC3 mutations are associated with congenital heart disease. However, the mechanism of action of ZIC3 in vascular intimal hyperplasia in CKD remains unelucidated. Venous specimens were collected during primary AVF surgery and traumatic amputation, and serum samples were collected from patients with CKD and healthy controls. Mouse vascular smooth muscle cells (VSMCs) were treated with hydrogen peroxide (H2O2) to clarify the role of ZIC3 in CKD. ZIC3 expression was reduced in the veins of patients with uremia and the serum of those with CKD. Zic3 and Bcl2 levels were significantly decreased in mouse VSMCs treated with H2O2·H2O2 inhibited mouse VSMC activity, upregulated Bax, and cleaved caspase 3 expression. Following Zic3 overexpression, Bcl2 expression level and cell viability were elevated, whereas Bax and cleaved caspase 3 expression levels were downregulated. In contrast, Zic3 knockdown yielded the opposite results. Therefore, ZIC3 could be a new therapeutic target in venous neointimal hyperplasia of CKD.
Collapse
Affiliation(s)
- Jianya Gao
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nephrology, Chongqing University Three Gorges Hospital, Chongqing 404100, China
| | - Lei Liu
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nephrology, Chongqing University Three Gorges Hospital, Chongqing 404100, China
| | - Zecheng Wu
- Department of Nephrology, Chongqing University Three Gorges Hospital, Chongqing 404100, China
| | - Hua Gan
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
27
|
Wang X, Wang Z, He J. Similarities and Differences of Vascular Calcification in Diabetes and Chronic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:165-192. [PMID: 38222032 PMCID: PMC10788067 DOI: 10.2147/dmso.s438618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
Presently, the mechanism of occurrence and development of vascular calcification (VC) is not fully understood; a range of evidence suggests a positive association between diabetes mellitus (DM) and VC. Furthermore, the increasing burden of central vascular disease in patients with chronic kidney disease (CKD) may be due, at least in part, to VC. In this review, we will review recent advances in the mechanisms of VC in the context of CKD and diabetes. The study further unveiled that VC is induced through the stimulation of pro-inflammatory factors, which in turn impairs endothelial function and triggers similar mechanisms in both disease contexts. Notably, hyperglycemia was identified as the distinctive mechanism driving calcification in DM. Conversely, in CKD, calcification is facilitated by mechanisms including mineral metabolism imbalance and the presence of uremic toxins. Additionally, we underscore the significance of investigating vascular alterations and newly identified molecular pathways as potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiabo Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
28
|
Blaj LA, Cucu AI, Tamba BI, Turliuc MD. The Role of the NF-kB Pathway in Intracranial Aneurysms. Brain Sci 2023; 13:1660. [PMID: 38137108 PMCID: PMC10871091 DOI: 10.3390/brainsci13121660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
The pathophysiology of intracranial aneurysms (IA) has been proven to be closely linked to hemodynamic stress and inflammatory pathways, most notably the NF-kB pathway. Therefore, it is a potential target for therapeutic intervention. In the present review, we investigated alterations in the vascular smooth muscle cells (VSMCs), extracellular matrix, and endothelial cells by the mediators implicated in the NF-kB pathway that lead to the formation, growth, and rupture of IAs. We also present an overview of the NF-kB pathway, focusing on stimuli and transcriptional targets specific to IAs, as well as a summary of the current strategies for inhibiting NF-kB activation in IAs. Our report adds to previously reported data and future research directions for treating IAs using compounds that can suppress inflammation in the vascular wall.
Collapse
Affiliation(s)
- Laurentiu Andrei Blaj
- Department of Neurosurgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.A.B.); (M.D.T.)
- “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Andrei Ionut Cucu
- “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
- Faculty of Medicine and Biological Sciences, University Stefan cel Mare of Suceava, 720229 Suceava, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Department of Pharmacology, Clinical Pharmacology and Algesiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Dana Turliuc
- Department of Neurosurgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.A.B.); (M.D.T.)
- “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| |
Collapse
|
29
|
Takahashi A. Co-Administration of Roxadustat and Zinc Stabilizes Both Serum Copper and Zinc Concentrations in Patients Undergoing Hemodialysis. Nutrients 2023; 15:4887. [PMID: 38068745 PMCID: PMC10708076 DOI: 10.3390/nu15234887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Patients undergoing hemodialysis often require zinc supplementation owing to hypozincemia, which may reduce serum copper concentrations. However, hypoxia-inducible factor-prolyl hydroxylase inhibitors (HIF-PHIs), which are used to treat renal anemia, have been reported to increase serum copper. Therefore, this study investigates the effectiveness of a combination of HIF-PHIs and zinc for the stabilization of serum copper and zinc concentrations during zinc supplementation for patients undergoing hemodialysis with renal anemia and hypozincemia. The serum zinc and copper concentrations were retrospectively compared over an 8-month period in 20 patients being administered roxadustat (an HIF-PHI) and 20 controls. The changes in concentrations were tracked in participants taking roxadustat who initiated or increased zinc supplementation. The serum zinc concentrations of the participants were significantly higher (p < 0.001) during zinc supplementation, regardless of roxadustat administration. Post-roxadustat, the serum copper concentrations were significantly higher than those pre-roxadustat or in non-roxadustat-treated participants, irrespective of zinc supplementation (p < 0.005). Even post-roxadustat, the serum copper concentrations were significantly lower, with no increase during zinc supplementation (p < 0.040). When zinc supplementation was initiated or increased in participants taking roxadustat, copper and zinc concentrations were normalized. Thus, combining zinc supplementation with roxadustat prevents both an excessive increase in serum copper and a decrease in serum zinc.
Collapse
Affiliation(s)
- Akira Takahashi
- Dialysis Center, Tesseikai Neurosurgical Hospital, 28-1 Nakanohonmachi, Shijonawate 575-8511, Japan
| |
Collapse
|
30
|
Kamei Y, Okumura Y, Adachi Y, Mori Y, Sakai M, Ohnishi K, Ohminami H, Masuda M, Yamanaka-Okumura H, Taketani Y. Humoral and cellular factors inhibit phosphate-induced vascular calcification during the growth period. J Clin Biochem Nutr 2023; 73:198-204. [PMID: 37970550 PMCID: PMC10636584 DOI: 10.3164/jcbn.23-11] [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: 02/07/2023] [Accepted: 02/25/2023] [Indexed: 11/17/2023] Open
Abstract
Hyperphosphatemia is an independent and non-classical risk factor of cardiovascular disease and mortality in patients with chronic kidney disease (CKD). Increased levels of extracellular inorganic phosphate (Pi) are known to directly induce vascular calcification, but the detailed underlying mechanism has not been clarified. Although serum Pi levels during the growth period are as high as those observed in hyperphosphatemia in adult CKD, vascular calcification does not usually occur during growth. Here, we have examined whether the defence system against Pi-induced vascular calcification can exist during the growth period using mice model. We found that calcification propensity of young serum (aged 3 weeks) was significantly lower than that of adult serum (10 months), possibly due to high fetuin-A levels. In addition, when the aorta was cultured in high Pi medium in vitro, obvious calcification was observed in the adult aorta but not in the young aorta. Furthermore, culture in high Pi medium increased the mRNA level of tissue-nonspecific alkaline phosphatase (TNAP), which degrades pyrophosphate, only in the adult aorta. Collectively, our findings indicate that the aorta in growing mouse may be resistant to Pi-induced vascular calcification via a mechanism in which high serum fetuin-A levels and suppressed TNAP expression.
Collapse
Affiliation(s)
- Yuki Kamei
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Department of Food and Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yosuke Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuichiro Adachi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuki Mori
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maiko Sakai
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kohta Ohnishi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hirokazu Ohminami
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Department of Food Science and Nutrition, Doshisha Women’s College of Liberal Arts, Teramachi Nishi-iru, Imadegawa-dori, Kamigyo-ku, Kyoto 602-0893, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
31
|
Wang C, Liang Q, He S, Zhu J, Lin X, Lin G, Wu D, Zhang W, Wang Z. Role of inflammation and immunity in vascular calcification: a bibliometric and visual analysis, 2000-2022. Front Cardiovasc Med 2023; 10:1258230. [PMID: 37965089 PMCID: PMC10642504 DOI: 10.3389/fcvm.2023.1258230] [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: 07/13/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Background In recent years, a great deal of research has been done on vascular calcification (VC), and inflammation and immunity have been displayed to play important roles in the mechanism of VC. However, to date, no comprehensive or systematic bibliometric analyses have been conducted on this topic. Methods Articles and reviews on the roles of inflammation and immunity in VC were obtained from the Web of Science Core Collection on August 5, 2022. Four scientometric software packages-HistCite, CiteSpace, VOSviewer, and R-bibliometrix-were used for the bibliometric and knowledge mapping analyses. Results The obtained 1,868 papers were published in 627 academic journals by 9,595 authors of 2,217 institutions from 69 countries. The annual number of publications showed a clear growth trend. The USA and China were the most productive countries. Karolinska Institutet, Harvard University, and the University of Washington were the most active institutions. Stenvinkel P published the most articles, whereas Demer LL received the most citations. Atherosclerosis published the most papers, while Circulation was the most highly cited journal. The largest cluster among the 22 clusters, based on the analysis of co-citations, was osteo-/chondrogenic transdifferentiation. "Vascular calcification," "inflammation," "chronic kidney disease," and "expression" were the main keywords in the field. The keyword "extracellular vesicle" attracted great attention in recent years with the strongest citation burst. Conclusions Osteo-/chondrogenic transdifferentiation is the primary research topic in this field. Extracellular vesicles are expected to become a new research focus for exploring the inflammatory and immune mechanisms of VC.
Collapse
Affiliation(s)
- Chen Wang
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Siyi He
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jie Zhu
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiafei Lin
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Guanwen Lin
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Duozhi Wu
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wenqi Zhang
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhihua Wang
- Department of Anesthesiology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
32
|
Qian X, Wang Y, Li X, Li Y, Li L. TNFAIP3 interacting protein 2 relieves lipopolysaccharide (LPS)-induced inflammatory injury in endometritis by inhibiting NF-kappaB activation. Immun Inflamm Dis 2023; 11:e970. [PMID: 37904691 PMCID: PMC10571501 DOI: 10.1002/iid3.970] [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: 03/10/2023] [Revised: 07/18/2023] [Accepted: 07/21/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Endometritis seriously affects the health of women, and it is important to identify new targets for its treatment. OBJECTIVE This study aimed to explore the role of TNFAIP3 interacting protein 2 (TNIP2) in endometritis through human endometrial epithelial cells (hEECs) stimulated by lipopolysaccharide (LPS). METHODS hEECs were induced with LPS to build a cellular model of endometritis. Cell growth and apoptosis were detected by cell counting kit-8 and flow cytometry. The TNIP2 mRNA and protein levels were measured using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis, respectively. The caspase3 activity was calculated using a Caspase3 activity kit. Interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) levels were determined by enzyme-linked-immunosorbent-assay. The reactive oxygen species (ROS), lactate dehydrogenase (LDH), catalase (CAT), and superoxide dismutase (SOD) levels were determined using the corresponding kits. Nuclear factor-kappaB (NF-κB) pathway was determined by western blot assay. RESULTS TNIP2 was downregulated in the LPS-induced endometritis cell model. Cell viability was reduced, apoptosis was enhanced, and IL-6, IL-1β, and TNF-α levels increased in LPS-induced hEECs. Additionally, LDH activity and ROS concentration were upregulated, whereas CAT and SOD activities were downregulated in LPS-induced hEECs. These results were reversed by TNIP2 overexpression. Moreover, the results hinted that NF-κB was involved in the effects of TNIP2 on the LPS-induced endometritis cell model. CONCLUSION TNIP2 alleviated endometritis by inhibiting the NF-κB pathway, suggesting a potential therapeutic target for endometritis.
Collapse
Affiliation(s)
- Xinxin Qian
- Department of GynecologyThe Third Affiliated Hospital of Qiqihar Medical CollegeQiqiharChina
| | - Yan Wang
- Department of GynecologyThe Third Affiliated Hospital of Qiqihar Medical CollegeQiqiharChina
| | - Xingmei Li
- Department of GynecologyThe Third Affiliated Hospital of Qiqihar Medical CollegeQiqiharChina
| | - Yuewen Li
- Department of GynecologyThe Third Affiliated Hospital of Qiqihar Medical CollegeQiqiharChina
| | - Liping Li
- Department of GynecologyThe Third Affiliated Hospital of Qiqihar Medical CollegeQiqiharChina
| |
Collapse
|
33
|
Hu Y, Zhao Y, Li P, Lu H, Li H, Ge J. Hypoxia and panvascular diseases: exploring the role of hypoxia-inducible factors in vascular smooth muscle cells under panvascular pathologies. Sci Bull (Beijing) 2023; 68:1954-1974. [PMID: 37541793 DOI: 10.1016/j.scib.2023.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 08/06/2023]
Abstract
As an emerging discipline, panvascular diseases are a set of vascular diseases with atherosclerosis as the common pathogenic hallmark, which mostly affect vital organs like the heart, brain, kidney, and limbs. As the major responser to the most common stressor in the vasculature (hypoxia)-hypoxia-inducible factors (HIFs), and the primary regulator of pressure and oxygen delivery in the vasculature-vascular smooth muscle cells (VSMCs), their own multifaceted nature and their interactions with each other are fascinating. Abnormally active VSMCs (e.g., atherosclerosis, pulmonary hypertension) or abnormally dysfunctional VSMCs (e.g., aneurysms, vascular calcification) are associated with HIFs. These widespread systemic diseases also reflect the interdisciplinary nature of panvascular medicine. Moreover, given the comparable proliferative characteristics exhibited by VSMCs and cancer cells, and the delicate equilibrium between angiogenesis and cancer progression, there is a pressing need for more accurate modulation targets or combination approaches to bolster the effectiveness of HIF targeting therapies. Based on the aforementioned content, this review primarily focused on the significance of integrating the overall and local perspectives, as well as temporal and spatial balance, in the context of the HIF signaling pathway in VSMC-related panvascular diseases. Furthermore, the review discussed the implications of HIF-targeting drugs on panvascular disorders, while considering the trade-offs involved.
Collapse
Affiliation(s)
- Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yongchao Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Peng Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Hua Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Shanghai Clinical Research Center for Interventional Medicine, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China; Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
34
|
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: 6] [Impact Index Per Article: 3.0] [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
|
35
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
36
|
Movérare-Skrtic S, Voelkl J, Nilsson KH, Nethander M, Luong TTD, Alesutan I, Li L, Wu J, Horkeby K, Lagerquist MK, Koskela A, Tuukkanen J, Tobias JH, Lerner UH, Henning P, Ohlsson C. B4GALNT3 regulates glycosylation of sclerostin and bone mass. EBioMedicine 2023; 91:104546. [PMID: 37023531 PMCID: PMC10102813 DOI: 10.1016/j.ebiom.2023.104546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Global sclerostin inhibition reduces fracture risk efficiently but has been associated with cardiovascular side effects. The strongest genetic signal for circulating sclerostin is in the B4GALNT3 gene region, but the causal gene is unknown. B4GALNT3 expresses the enzyme beta-1,4-N-acetylgalactosaminyltransferase 3 that transfers N-acetylgalactosamine onto N-acetylglucosaminebeta-benzyl on protein epitopes (LDN-glycosylation). METHODS To determine if B4GALNT3 is the causal gene, B4galnt3-/- mice were developed and serum levels of total sclerostin and LDN-glycosylated sclerostin were analysed and mechanistic studies were performed in osteoblast-like cells. Mendelian randomization was used to determine causal associations. FINDINGS B4galnt3-/- mice had higher circulating sclerostin levels, establishing B4GALNT3 as a causal gene for circulating sclerostin levels, and lower bone mass. However, serum levels of LDN-glycosylated sclerostin were lower in B4galnt3-/- mice. B4galnt3 and Sost were co-expressed in osteoblast-lineage cells. Overexpression of B4GALNT3 increased while silencing of B4GALNT3 decreased the levels of LDN-glycosylated sclerostin in osteoblast-like cells. Mendelian randomization demonstrated that higher circulating sclerostin levels, genetically predicted by variants in the B4GALNT3 gene, were causally associated with lower BMD and higher risk of fractures but not with higher risk of myocardial infarction or stroke. Glucocorticoid treatment reduced B4galnt3 expression in bone and increased circulating sclerostin levels and this may contribute to the observed glucocorticoid-induced bone loss. INTERPRETATION B4GALNT3 is a key factor for bone physiology via regulation of LDN-glycosylation of sclerostin. We propose that B4GALNT3-mediated LDN-glycosylation of sclerostin may be a bone-specific osteoporosis target, separating the anti-fracture effect of global sclerostin inhibition, from indicated cardiovascular side effects. FUNDING Found in acknowledgements.
Collapse
Affiliation(s)
- Sofia Movérare-Skrtic
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria; Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Karin H Nilsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Maria Nethander
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Trang Thi Doan Luong
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Lei Li
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Jianyao Wu
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Karin Horkeby
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marie K Lagerquist
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Antti Koskela
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Juha Tuukkanen
- Department of Anatomy and Cell Biology, Faculty of Medicine, Institute of Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Jon H Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, and Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ulf H Lerner
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Petra Henning
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
37
|
Zhang YY, Ren KD, Luo XJ, Peng J. COVID-19-induced neurological symptoms: focus on the role of metal ions. Inflammopharmacology 2023; 31:611-631. [PMID: 36892679 PMCID: PMC9996599 DOI: 10.1007/s10787-023-01176-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/10/2023]
Abstract
Neurological symptoms are prevalent in both the acute and post-acute phases of coronavirus disease 2019 (COVID-19), and they are becoming a major concern for the prognosis of COVID-19 patients. Accumulation evidence has suggested that metal ion disorders occur in the central nervous system (CNS) of COVID-19 patients. Metal ions participate in the development, metabolism, redox and neurotransmitter transmission in the CNS and are tightly regulated by metal ion channels. COVID-19 infection causes neurological metal disorders and metal ion channels abnormal switching, subsequently resulting in neuroinflammation, oxidative stress, excitotoxicity, neuronal cell death, and eventually eliciting a series of COVID-19-induced neurological symptoms. Therefore, metal homeostasis-related signaling pathways are emerging as promising therapeutic targets for mitigating COVID-19-induced neurological symptoms. This review provides a summary for the latest advances in research related to the physiological and pathophysiological functions of metal ions and metal ion channels, as well as their role in COVID-19-induced neurological symptoms. In addition, currently available modulators of metal ions and their channels are also discussed. Collectively, the current work offers a few recommendations according to published reports and in-depth reflections to ameliorate COVID-19-induced neurological symptoms. Further studies need to focus on the crosstalk and interactions between different metal ions and their channels. Simultaneous pharmacological intervention of two or more metal signaling pathway disorders may provide clinical advantages in treating COVID-19-induced neurological symptoms.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
38
|
Cao B, Wang J, Feng J. Signaling pathway mechanisms of neurological diseases induced by G protein-coupled receptor 39. CNS Neurosci Ther 2023; 29:1470-1483. [PMID: 36942516 PMCID: PMC10173710 DOI: 10.1111/cns.14174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND G protein-coupled receptor 39 (GPR39) is a transmembrane zinc receptor with two splice variants, which belongs to the G-protein-coupled receptor growth hormone-releasing peptide family. Its expression is induced by zinc, which activates GPR39, and its activation mediates cell proliferation, ion homeostasis, and anti-inflammatory, antioxidant, and other pathophysiological effects via different signaling pathways. AIMS The article reviews the latest literature in this field. In particular, the role of GPR39 in nervous system is discussed. MATERIALS AND METHODS GPR39 can be a promising target in neurological diseases for targeted therapy, which will help doctors overcome the associated problems. DISCUSSION GPR39 is expressed in vivo at several sites. Increasing evidence suggests that GPR39 plays an important role as a neuroprotective agent in vivo and regulates various neurological functions, including neurodegeneration, neuroelectrophysiology, and neurovascular homeostasis. CONCLUSION This review aims to provide an overview of the functions, signal transduction pathways, and pathophysiological role of GPR39 in neurological diseases and summarize the GPR39 agonists that have been identified in the recent years.
Collapse
Affiliation(s)
- Bin Cao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
39
|
Zhang H, Wang S, Gu X, Qiu H, Zhang Y. L-shaped association between dietary zinc intake and the risk of developing cardiovascular disease in Chinese adults: A cohort study. Front Nutr 2023; 10:1032048. [PMID: 37006929 PMCID: PMC10064069 DOI: 10.3389/fnut.2023.1032048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
Background Although the association of zinc (Zn) with cardiovascular disease (CVD) has been studied, no consensus has been reached on this relationship, particularly dietary Zn intake. The purpose of this study was to assess the effect of dietary Zn intake on the risk of CVD and to analyze whether this effect varied according to zinc consumption using representative data from China. Methods 11,470 adults from the China Health and Nutrition Survey (CHNS) were eventually enrolled. The dietary information was collected by the 3 day 24-h dietary recalls combined with dietary weighting method. CVD was defined as participants with self-reported physician-diagnosed apoplexy and/or myocardial infarction during the follow-up. Cox regression was used to calculate the hazard ratios (HRs) of CVD with 95% confidence intervals. Restricted cubic spline function plus Cox regression was used to visualize the influence trend of dietary Zn intake on new-onset CVD and to test whether this trend is linear. 2-segment Cox regression was established to address the nonlinear trend. Results 431 participants developed CVD, including 262 strokes and 197 myocardial infarctions. Compared with the lowest quintile (Q1), the adjusted hazard ratios and 95% confidence interval (CI) of CVD in Q2 to Q5 of dietary Zn intake were 0.72 (0.54, 0.97), 0.59 (0.42, 0.81), 0.50 (0.34, 0.72) and 0.44 (0.27, 0.71), respectively. The influence trend of dietary Zn intake on new-onset CVD was nonlinear and L-shaped. When dietary Zn intake <13.66 mg/day, increased dietary Zn intake was significantly associated with decreased risk of developing CVD (HR = 0.87, 95% CI: 0.82-0.92, p-value <0.0001). Conclusion An L-shaped trend was observed between dietary Zn intake and the risk of developing CVD, indicating that dietary Zn intake should be improved moderately, but not excessively, for the benefit of cardiovascular disease.
Collapse
Affiliation(s)
- Huanxiang Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jiamusi University, Jiamusi, China
- The first Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Shanjie Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xia Gu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongbin Qiu
- Department of Epidemiology and Biostatistics, School of Public Health, Jiamusi University, Jiamusi, China
- The first Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yiying Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jiamusi University, Jiamusi, China
- The first Affiliated Hospital of Jiamusi University, Jiamusi, China
| |
Collapse
|
40
|
Emri E, Cappa O, Kelly C, Kortvely E, SanGiovanni JP, McKay BS, Bergen AA, Simpson DA, Lengyel I. Zinc Supplementation Induced Transcriptional Changes in Primary Human Retinal Pigment Epithelium: A Single-Cell RNA Sequencing Study to Understand Age-Related Macular Degeneration. Cells 2023; 12:773. [PMID: 36899910 PMCID: PMC10000409 DOI: 10.3390/cells12050773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Zinc supplementation has been shown to be beneficial to slow the progression of age-related macular degeneration (AMD). However, the molecular mechanism underpinning this benefit is not well understood. This study used single-cell RNA sequencing to identify transcriptomic changes induced by zinc supplementation. Human primary retinal pigment epithelial (RPE) cells could mature for up to 19 weeks. After 1 or 18 weeks in culture, we supplemented the culture medium with 125 µM added zinc for one week. RPE cells developed high transepithelial electrical resistance, extensive, but variable pigmentation, and deposited sub-RPE material similar to the hallmark lesions of AMD. Unsupervised cluster analysis of the combined transcriptome of the cells isolated after 2, 9, and 19 weeks in culture showed considerable heterogeneity. Clustering based on 234 pre-selected RPE-specific genes divided the cells into two distinct clusters, we defined as more and less differentiated cells. The proportion of more differentiated cells increased with time in culture, but appreciable numbers of cells remained less differentiated even at 19 weeks. Pseudotemporal ordering identified 537 genes that could be implicated in the dynamics of RPE cell differentiation (FDR < 0.05). Zinc treatment resulted in the differential expression of 281 of these genes (FDR < 0.05). These genes were associated with several biological pathways with modulation of ID1/ID3 transcriptional regulation. Overall, zinc had a multitude of effects on the RPE transcriptome, including several genes involved in pigmentation, complement regulation, mineralization, and cholesterol metabolism processes associated with AMD.
Collapse
Affiliation(s)
- Eszter Emri
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
- Section Ophthalmogenetics, Department of Human Genetics, Queen Emma Centre for Precision Medicine, Amsterdam UMC, Location AMC, 1105AZ Amsterdam, The Netherlands
| | - Oisin Cappa
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Caoimhe Kelly
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Elod Kortvely
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - John Paul SanGiovanni
- Biosciences Research Laboratories, BIO5 Institute, University of Arizona, 1230 North Cherry Avenue, Tucson, AZ 85724, USA
| | - Brian S. McKay
- Department of Ophthalmology and Vision Science, University of Arizona, 1656 E. Mabel Street, Tucson, AZ 85724, USA
| | - Arthur A. Bergen
- Section Ophthalmogenetics, Department of Human Genetics, Queen Emma Centre for Precision Medicine, Amsterdam UMC, Location AMC, 1105AZ Amsterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN-KNAW), 1105AZ Amsterdam, The Netherlands
| | - David A. Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| |
Collapse
|
41
|
Clinical Significance of Trace Element Zinc in Patients with Chronic Kidney Disease. J Clin Med 2023; 12:jcm12041667. [PMID: 36836202 PMCID: PMC9964431 DOI: 10.3390/jcm12041667] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/02/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
The trace element zinc is essential for diverse physiological processes in humans. Zinc deficiency can impair growth, skin reproduction, immune function, maintenance of taste, glucose metabolism, and neurological function. Patients with chronic kidney disease (CKD) are susceptible to zinc deficiency, which is associated with erythropoiesis-stimulating agent (ESA) hypo-responsive anemia, nutritional problems, and cardiovascular diseases as well as non-specific symptoms such as dermatitis, prolonged wound healing, taste disturbance, appetite loss, or cognitive decline. Thus, zinc supplementation may be useful for the treatment of its deficiency, although it often causes copper deficiency, which is characterized by several severe disorders including cytopenia and myelopathy. In this review article, we mainly discuss the significant roles of zinc and the association between zinc deficiency and the pathogenesis of complications in patients with CKD.
Collapse
|
42
|
Pan W, Jie W, Huang H. Vascular calcification: Molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e200. [PMID: 36620697 PMCID: PMC9811665 DOI: 10.1002/mco2.200] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Abstract
Vascular calcification (VC) is recognized as a pathological vascular disorder associated with various diseases, such as atherosclerosis, hypertension, aortic valve stenosis, coronary artery disease, diabetes mellitus, as well as chronic kidney disease. Therefore, it is a life-threatening state for human health. There were several studies targeting mechanisms of VC that revealed the importance of vascular smooth muscle cells transdifferentiating, phosphorous and calcium milieu, as well as matrix vesicles on the progress of VC. However, the underlying molecular mechanisms of VC need to be elucidated. Though there is no acknowledged effective therapeutic strategy to reverse or cure VC clinically, recent evidence has proved that VC is not a passive irreversible comorbidity but an active process regulated by many factors. Some available approaches targeting the underlying molecular mechanism provide promising prospects for the therapy of VC. This review aims to summarize the novel findings on molecular mechanisms and therapeutic interventions of VC, including the role of inflammatory responses, endoplasmic reticulum stress, mitochondrial dysfunction, iron homeostasis, metabolic imbalance, and some related signaling pathways on VC progression. We also conclude some recent studies on controversial interventions in the clinical practice of VC, such as calcium channel blockers, renin-angiotensin system inhibitions, statins, bisphosphonates, denosumab, vitamins, and ion conditioning agents.
Collapse
Affiliation(s)
- Wei Pan
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Wei Jie
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Joint Laboratory of Guangdong‐Hong Kong‐Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic DiseaseSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
43
|
Li A, Li Y, Wang Y, Wang Y, Li X, Qubi W, Xiong Y, Zhu J, Liu W, Lin Y. ACADL Promotes the Differentiation of Goat Intramuscular Adipocytes. Animals (Basel) 2023; 13:281. [PMID: 36670821 PMCID: PMC9854987 DOI: 10.3390/ani13020281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Intramuscular fat (IMF) deposits help improve meat quality such as marbling, juicy, flavor and tenderness. Long-chain acyl-CoA dehydrogenase (ACADL) is a key enzyme for catalyzing fatty acid oxidation, and studies have shown ACADL is involved in the deposition and differentiation of intramuscular adipocytes. However, the effect of ACADL on intramuscular adipocytes differentiation in goats needs further study. In this study, to explore the mechanism of ACADL on the development of goat intramuscular adipocytes, we constructed an over-expression plasmids and a SI-RNA of ACADL to explore the function of ACADL on the development of goat IMF. It was found that overexpression of ACADL promoted the differentiation of goat intramuscular adipocytes, and promoted the expression of fat cell differentiation marker genes lipoprotein lipase (LPL), peroxisome proliferator activated receptor gamma (PPARγ), APETALA-2-like transcription factor gene (AP2), CCAT enhancer binding protein (CEBPα), preadipocyte Factor 1 (Pref-1) and CCAT enhancer binding protein (CEBPβ), and the opposite trend occurred after interference. In addition, we screened of this related tumor necrosis factor (TNF) signaling pathway by RNA-Seq. So, we validate the signaling pathway with inhibitor of TNF signaling pathway. In summary, these results indicate that ACADL promotes intramuscular adipocytes differentiation through activation TNF signaling pathway. This study provides an important basis for the mechanism of IMF development.
Collapse
Affiliation(s)
- An Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Yanyan Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Youli Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Yong Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Xin Li
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Wuqie Qubi
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
| | - Wei Liu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| | - Yaqiu Lin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu 610041, China; (A.L.); (Y.L.); (Y.W.); (Y.W.); (X.L.); (W.Q.); (Y.X.); (J.Z.); (W.L.)
- Key Laboratory of Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation, Southwest Minzu University, Chengdu 610041, China
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China
| |
Collapse
|
44
|
Ishioka K, Hidaka S, Fujiwara N, Yamano M, Mochida Y, Oka M, Maesato K, Moriya H, Ohtake T, Kobayashi S. Association between zinc deficiency and aorta stiffness in non-diabetic hemodialysis patients. PLoS One 2023; 18:e0268875. [PMID: 36607966 PMCID: PMC9821515 DOI: 10.1371/journal.pone.0268875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES Zinc deficiency (Zn < 60 μg/dL) is known to play an important role for vascular calcification. However, little data is available regarding the association between zinc deficiency and aorta stiffness in dialysis patients. Thus, we studied the relationship between zinc deficiency and aorta stiffness in non-diabetic hemodialysis (HD) patients. METHODS Of 150 patients receiving maintenance HD at our hospital, we included 79 non-diabetic HD patients (age: 70±11 years, 49 men) after excluding 71 diabetic HD patients. Zinc deficiency was defined as Zn <60 μg/dL during pre-HD blood sampling. The association between zinc deficiency and aorta stiffness was analyzed. Aorta stiffness was evaluated as brachial-ankle pulse wave velocity (baPWV). Other surrogate markers for cardiovascular complications were also measured. RESULTS The zinc deficiency group (ZD group) included 45 patients (57.0%). Compared to the zinc non-deficiency group (ZND group), patients with ZD group were significantly older, higher levels of CRP and hypoalbuminemia. Moreover, they had significantly higher levels of baPWV, and lower levels of ankle-brachial pressure index (ABI) (p<0.05). After adjusting for hypoalbuminemia, and CRP, multivariate analysis showed that age and zinc level were independent predictors of baPWV. CONCLUSION The study suggested that zinc deficiency may be an independent risk factor for aorta stiffness, even after adjusting for malnutrition and inflammation.
Collapse
Affiliation(s)
- Kunihiro Ishioka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
- * E-mail:
| | - Sumi Hidaka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Naoki Fujiwara
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Mizuki Yamano
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Yasuhiro Mochida
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Machiko Oka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Kyoko Maesato
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Hidekazu Moriya
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Takayasu Ohtake
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Shuzo Kobayashi
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kamakura, Japan
| |
Collapse
|
45
|
Xie Y, Liu F, Zhang X, Jin Y, Li Q, Shen H, Fu H, Mao J. Benefits and risks of essential trace elements in chronic kidney disease: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1400. [PMID: 36660676 PMCID: PMC9843383 DOI: 10.21037/atm-22-5969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/19/2022] [Indexed: 01/01/2023]
Abstract
Background and Objective Chronic kidney disease (CKD) is an important public health concern. With the decline of renal function, CKD patients gradually progress to end-stage kidney disease and need to undergo dialysis or kidney transplantation to maintain life, bringing a heavy economic burden to the family and society. Therefore, it is necessary to effectively prevent and delay the progression of CKD. Essential trace elements play an indispensable role in CKD, and the objective of this study is to systematically review their benefits in the disease and summarize the risks of their excess. Methods The keywords "trace elements", "chronic kidney disease", "dialysis", "inflammation", and "fibrosis" and their combinations were used to search for relevant literature published in the PubMed database and Web of Science. We then summarized the role of trace element abnormalities in CKD patients in anemia, oxidative stress, inflammation, and chronic fibrosis, and the risk of their excess. Key Content and Findings Imbalance of essential trace elements is a common complication of CKD and a risk factor for CKD progression, cardiovascular events, and death. This article reviews the effects of essential trace elements (iron, zinc, selenium, copper, iodine, and manganese) on CKD. We analyze literature and discuss the advantages and disadvantages of various essential trace elements. Conclusions Research shows CKD patients have an imbalance of essential trace elements, and treatment based on these is an important direction for future exploration. A knowledge of the homeostasis of trace elements is important to improving the prognosis of CKD patients and delaying the progression of the disease.
Collapse
Affiliation(s)
- Yi Xie
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Fei Liu
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaojing Zhang
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yanyan Jin
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiuyu Li
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Huijun Shen
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Haidong Fu
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
46
|
Luo F, Guo W, Liu W. Exosomes derived from bone marrow mesenchymal stem cells inhibit human aortic vascular smooth muscle cells calcification via the miR-15a/15b/16/NFATc3/OCN axis. Biochem Biophys Res Commun 2022; 635:65-76. [DOI: 10.1016/j.bbrc.2022.09.076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
|
47
|
Yao M, Liu Y, Sun M, Qin S, Xin W, Guan X, Zhang B, He T, Huang Y. The molecular mechanisms and intervention strategies of mitophagy in cardiorenal syndrome. Front Physiol 2022; 13:1008517. [PMID: 36353377 PMCID: PMC9638141 DOI: 10.3389/fphys.2022.1008517] [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: 07/31/2022] [Accepted: 10/13/2022] [Indexed: 11/15/2022] Open
Abstract
Cardiorenal syndrome (CRS) is defined as a disorder of the heart and kidney, in which acute or chronic injury of one organ may lead to acute or chronic dysfunction of the other. It is characterized by high morbidity and mortality, resulting in high economic costs and social burdens. However, there is currently no effective drug-based treatment. Emerging evidence implicates the involvement of mitophagy in the progression of CRS, including cardiovascular disease (CVD) and chronic kidney disease (CKD). In this review, we summarized the crucial roles and molecular mechanisms of mitophagy in the pathophysiology of CRS. It has been reported that mitophagy impairment contributes to a vicious loop between CKD and CVD, which ultimately accelerates the progression of CRS. Further, recent studies revealed that targeting mitophagy may serve as a promising therapeutic approach for CRS, including clinical drugs, stem cells and small molecule agents. Therefore, studies focusing on mitophagy may benefit for expanding innovative basic research, clinical trials, and therapeutic strategies for CRS.
Collapse
Affiliation(s)
- Mengying Yao
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yong Liu
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengjia Sun
- Department of Cardiology, Institute of Cardiovascular Diseases of PLA, The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shaozong Qin
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wang Xin
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xu Guan
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bo Zhang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ting He
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yinghui Huang, ; Ting He,
| | - Yinghui Huang
- Department of Nephrology, The key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yinghui Huang, ; Ting He,
| |
Collapse
|
48
|
Pluquet M, Kamel S, Choukroun G, Liabeuf S, Laville SM. Serum Calcification Propensity Represents a Good Biomarker of Vascular Calcification: A Systematic Review. Toxins (Basel) 2022; 14:toxins14090637. [PMID: 36136575 PMCID: PMC9501050 DOI: 10.3390/toxins14090637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Vascular calcification contributes to cardiovascular morbidity and mortality. A recently developed serum calcification propensity assay is based on the half-transformation time (T50) from primary calciprotein particles (CPPs) to secondary CPPs, reflecting the serum’s endogenous capacity to prevent calcium phosphate precipitation. We sought to identify and review the results of all published studies since the development of the T50-test by Pasch et al. in 2012 (whether performed in vitro, in animals or in the clinic) of serum calcification propensity. To this end, we searched PubMed, Elsevier EMBASE, the Cochrane Library and Google Scholar databases from 2012 onwards. At the end of the selection process, 57 studies were analyzed with regard to the study design, sample size, characteristics of the study population, the intervention and the main results concerning T50. In patients with primary aldosteronism, T50 is associated with the extent of vascular calcification in the abdominal aorta. In chronic kidney disease (CKD), T50 is associated with the severity and progression of coronary artery calcification. T50 is also associated with cardiovascular events and all-cause mortality in CKD patients, patients on dialysis and kidney transplant recipients and with cardiovascular mortality in patients on dialysis, kidney transplant recipients, patients with ischemic heart failure and reduced ejection fraction, and in the general population. Switching from acetate-acidified dialysate to citrate-acidified dialysate led to a longer T50, as did a higher dialysate magnesium concentration. Oral administration of magnesium (in CKD patients), phosphate binders, etelcalcetide and spironolactone (in hemodialysis patients) was associated with a lower serum calcification propensity. Serum calcification propensity is an overall marker of calcification associated with hard outcomes but is currently used in research projects only. This assay might be a valuable tool for screening serum calcification propensity in at-risk populations (such as CKD patients and hemodialyzed patients) and, in particular, for monitoring changes over time in T50.
Collapse
Affiliation(s)
- Maxime Pluquet
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
| | - Said Kamel
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Department of Biochemistry, Amiens University Medical Center, F-80000 Amiens, France
| | - Gabriel Choukroun
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Department of Nephrology, Amiens University Medical Center, F-80000 Amiens, France
| | - Sophie Liabeuf
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens University Medical Center, F-80000 Amiens, France
- Correspondence:
| | - Solène M. Laville
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, F-80000 Amiens, France
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens University Medical Center, F-80000 Amiens, France
| |
Collapse
|
49
|
Study on the Effects of Different Doses of Dahuang Zhechong Pills on the Ubiquitin Proteasome Pathway/Nuclear Factor-κB in Rats with Atherosclerosis and Its Mechanism. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6056829. [PMID: 36134116 PMCID: PMC9482499 DOI: 10.1155/2022/6056829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/07/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022]
Abstract
In order to investigate the effects of different doses of Dahuang Zhechong pills on the ubiquitin proteasome pathway/nuclear factor-κB (UPP-NF-κB) in rats with atherosclerosis (AS), 58-week-old male Wistar rats were selected and randomly divided into the normal group, model group, control group, low-dose group, and high-dose group. The model group and the drug group are given intraperitoneal injections of vitamins, and the model group and the drug group are given a high-fat diet. Rats in the low-dose group and high-dose group are given low-dose and high-dose Dahuang Zhechong pill lavage solution, respectively. Besides, the control group is given simvastatin solution by gavage, and intervention is performed once a day for 12 weeks. Ubiquitin (Ub) protein expression, ubiquitin activase (UBE1), nuclear factor-κB, nuclear inhibitory factor-κB (IκB) gene expression, total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), and serum tumor necrosis factor-α (TNF-α) are compared. The experimental result shows that Dahuang Zhechong pills can reduce inflammation and prevent and treat AS by blocking the activation of the UPP/NF-κB signaling pathway and can be used as a proteasome inhibitor in the clinical treatment of AS.
Collapse
|
50
|
Periostin Augments Vascular Smooth Muscle Cell Calcification via β-Catenin Signaling. Biomolecules 2022; 12:biom12081157. [PMID: 36009051 PMCID: PMC9405747 DOI: 10.3390/biom12081157] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/26/2022] Open
Abstract
Medial vascular calcification is common in chronic kidney disease (CKD) and is closely linked to hyperphosphatemia. Vascular smooth muscle cells (VSMCs) can take up pro-calcific properties and actively augment vascular calcification. Various pro-inflammatory mediators are able to promote VSMC calcification. In this study, we investigated the effects and mechanisms of periostin, a matricellular signaling protein, in calcifying human VSMCs and human serum samples. As a result, periostin induced the mRNA expression of pro-calcific markers in VSMCs. Furthermore, periostin augmented the effects of β-glycerophosphate on the expression of pro-calcific markers and aggravated the calcification of VSMCs. A periostin treatment was associated with an increased β-catenin abundance as well as the expression of target genes. The pro-calcific effects of periostin were ameliorated by WNT/β-catenin pathway inhibitors. Moreover, a co-treatment with an integrin αvβ3-blocking antibody blunted the pro-calcific effects of periostin. The silencing of periostin reduced the effects of β-glycerophosphate on the expression of pro-calcific markers and the calcification of VSMCs. Elevated serum periostin levels were observed in hemodialysis patients compared with healthy controls. These observations identified periostin as an augmentative factor in VSMC calcification. The pro-calcific effects of periostin involve integrin αvβ3 and the activation of the WNT/β-catenin pathway. Thus, the inhibition of periostin may be beneficial to reduce the burden of vascular calcification in CKD patients.
Collapse
|