1
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [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: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Lv R, Liu Z, Guo H, Zhang B, Wang X, Peng Y, Chang Y, Yang F, Xiong Y, Hao J, Gao X, Wang X, Xu Q, Shimosawa T, Qiang P. Fibroblast to macrophage-like cell transition in renal inflammatory injury through the MR/CSF1 pathway induced by aldosterone. Life Sci 2025; 372:123627. [PMID: 40216224 DOI: 10.1016/j.lfs.2025.123627] [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: 01/27/2025] [Revised: 03/25/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
AIMS Inflammatory injury promotes the chronic kidney disease (CKD) progression,with renal macrophage accumulation and proliferation of as typical manifestations of inflammatory injury. We aimed to verify fibroblast to macrophage-like cell transition as a new source of macrophages that participate in renal inflammatory injury. MATERIALS AND METHODS Wistar rats were divided into Sham, ALD (aldosterone infusion for 12 weeks), and ESA (aldosterone infusion and esaxerenone by diet for 12 weeks) groups. Rat kidney interstitial fibroblast (RKF) were cultured, induced with aldosterone or CSF1, and treated with antagonists in vitro. The proportions of FSP-1+ F4/80+ cells in the rat kidney and RKF, including M1 marker iNOS/CD86 and M2 marker CD206/CD163 were assessed by flow cytometry and immunofluorescence staining. Single-cell RNA sequencing was used to assess the origin of macrophages in the rat kidneys and related gene expression. Additionally, immunofluorescence was used to detect FSP-1+ F4/80+ cells in kidney biopsy samples from CKD patients. KEY FINDINGS Fibroblast to macrophage-like cell transition was observed in both the kidneys of aldosterone-infused rats and in vitro aldosterone-treated RKF, with a predominant differentiation into the M1 phenotype. This transformation was mediated through the MR/CSF1 signalling pathway, revealing a novel source of macrophages and providing significant insights into the mechanisms underlying organ fibrosis. SIGNIFICANCE Aldosterone induces fibroblast to macrophage-like cell transition through the MR/ CSF1 pathway.
Collapse
Affiliation(s)
- Ruyan Lv
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ziqian Liu
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haixia Guo
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Boya Zhang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xuan Wang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yunsong Peng
- Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang 050200, China
| | - Yi Chang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fan Yang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yunzhao Xiong
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Juan Hao
- Shijiazhuang Hospital of Traditional Chinese Medicine, Shijiazhuang 050200, China
| | - Xiaomeng Gao
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xiangting Wang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qingyou Xu
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita 286-8686, Japan.
| | - Panpan Qiang
- Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
3
|
Razazian M, Bahiraii S, Jannat I, Tiffner A, Beilhack G, Levkau B, Voelkl J, Alesutan I. Sphingosine kinase 1 inhibition aggravates vascular smooth muscle cell calcification. Pflugers Arch 2025; 477:815-826. [PMID: 39899071 DOI: 10.1007/s00424-025-03068-6] [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: 06/04/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Medial vascular calcification is common in chronic kidney disease patients and linked to hyperphosphatemia. Upon phosphate exposure, intricate signaling events orchestrate pro-calcific effects in the vasculature mediated by vascular smooth muscle cells (VSMCs). Sphingosine kinase 1 (SPHK1) produces sphingosine-1-phosphate (S1P) and is associated with complex effects in the vascular system. The present study investigated a possible involvement of SPHK1 in VSMC calcification. Experiments were performed in primary human aortic VSMCs under pro-calcific conditions, with pharmacological inhibition or knockdown of SPHK1 or SPNS2 (a lysolipid transporter involved in cellular S1P export), as well as in Sphk1-deficient and wild-type mice treated with cholecalciferol. In VSMCs, SPHK1 expression was up-regulated by pro-calcific conditions. Calcification medium up-regulated osteogenic marker mRNA expression and activity as well as calcification of VSMCs, effects significantly augmented by co-treatment with the SPHK1 inhibitor SK1-IN-1. SK1-IN-1 alone was sufficient to up-regulate osteogenic signaling in VSMCs during control conditions. Similarly, the SPHK1 inhibitor PF-543 and SPHK1 knockdown up-regulated osteogenic signaling in VSMCs and aggravated VSMC calcification. In contrast, co-treatment with the SPNS2 inhibitor SLF1081851 suppressed osteogenic signaling and calcification of VSMCs, effects abolished by silencing of SPHK1. In addition, Sphk1 deficiency aggravated vascular calcification and aortic osteogenic marker expression in mice after cholecalciferol overload. In conclusion, SPHK1 inhibition, knockdown, or deficiency aggravates vascular pro-calcific signaling and calcification. The reduced calcification after inhibition of S1P export suggests a possible involvement of intracellular S1P, but further studies are required to elucidate the complex roles of SPHKs and S1P signaling in calcifying VSMCs.
Collapse
MESH Headings
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors
- Phosphotransferases (Alcohol Group Acceptor)/metabolism
- Phosphotransferases (Alcohol Group Acceptor)/genetics
- Humans
- Vascular Calcification/metabolism
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Cells, Cultured
- Male
- Mice, Inbred C57BL
- Signal Transduction
- Anion Transport Proteins/metabolism
- Anion Transport Proteins/genetics
- Lysophospholipids/metabolism
- Sphingosine/metabolism
- Sphingosine/analogs & derivatives
- Osteogenesis/drug effects
- Methanol
- Pyrrolidines
- Sulfones
Collapse
Affiliation(s)
- Mehdi Razazian
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| | - Sheyda Bahiraii
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| | - Isratul Jannat
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| | - Adéla Tiffner
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Georg Beilhack
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, 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.
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| |
Collapse
|
4
|
Giaquinto A, Abate V, Vergatti A, Muscariello R, Iervolino A, Pucci M, Cavati G, Pirrotta F, De Filippo G, Esposito R, D'Elia L, Merlotti D, Gennari L, Rendina D. Standard and advanced echocardiographic study of patients with Paget's disease of bone: Evidence of a pagetic heart disease? J Intern Med 2025; 297:630-641. [PMID: 40342188 PMCID: PMC12087814 DOI: 10.1111/joim.20069] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
BACKGROUND Paget's disease of the bone (PDB) is a metabolic bone disorder involving one or more skeletal sites. Cardiovascular diseases (CVDs) have been described in patients with PDB but have not been systematically analysed. OBJECTIVES This study aimed to compare standard and advanced (speckle-tracking) echocardiographic parameters measured in patients with PDB and controls matched for age, weight, height and history of hypertension but without metabolic bone disorders. METHODS This multicentre case-control study included all patients with PDB referred to the Federico II and Siena Universities, Italy, from March 2019 to October 2022. During the same time, we enrolled at least one control for each patient, matched for age, sex, body mass index (BMI) and history of hypertension. RESULTS Sixty-nine patients with PDB and 115 healthy controls were enrolled in this study. All patients with PDB were treated with zoledronic acid at the time of diagnosis. Compared with controls, on standard echocardiography, patients with PDB showed a high prevalence of aortic and mitral valve calcifications and/or sclerosis, reduced left ventricular (LV) ejection fraction, stroke volume, cardiac output, increased interventricular septum thickness, posterior wall thickness, LV mass index, relative wall thickness, relative diastolic wall thickness, E/e' ratio and systemic vascular resistance. Using speckle-tracking echocardiography, patients with PDB showed a lower global longitudinal strain and global myocardial work efficiency than controls. There was no relationship between the PDB activity and extent and severity of cardiac abnormalities. CONCLUSION Overall, the myocardial function and structure were impaired in patients with PDB. Additionally, PDB was associated with early subclinical myocardial damage.
Collapse
Affiliation(s)
- Alfonso Giaquinto
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | - Veronica Abate
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | - Anita Vergatti
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | | | - Adelaide Iervolino
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | - Martina Pucci
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | - Guido Cavati
- Department of Medicine, Surgery and NeurosciencesUniversity of SienaSienaItaly
| | - Filippo Pirrotta
- Department of Medicine, Surgery and NeurosciencesUniversity of SienaSienaItaly
| | - Gianpaolo De Filippo
- Assistance Publique‐Hôpitaux de Paris, Hôpital Robert‐DebréService d'Endocrinologie‐DiabétologieParisFrance
| | - Roberta Esposito
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | - Lanfranco D'Elia
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| | - Daniela Merlotti
- Department of Medicine, Surgery and NeurosciencesUniversity of SienaSienaItaly
| | - Luigi Gennari
- Department of Medicine, Surgery and NeurosciencesUniversity of SienaSienaItaly
| | - Domenico Rendina
- Department of Clinical Medicine and SurgeryFederico II UniversityNaplesItaly
| |
Collapse
|
5
|
Lao XY, Sun YL, Zhao ZJ, Liu J, Ruan XF. Pharmacological effects of betulinic acid and its protective mechanisms on the cardiovascular system. Fitoterapia 2025; 183:106561. [PMID: 40288588 DOI: 10.1016/j.fitote.2025.106561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Betulinic acid (BA), a pentacyclic triterpenoid saponin widely found in plants, has attracted attention for its diverse pharmacological activities. Recent studies highlight its cardioprotective potential, promoting its relevance in cardiovascular research. AIM OF THE REVIEW This review summarizes BA's physicochemical properties, structure-activity relationships, natural sources, and synthesis strategies. It further discusses its pharmacokinetics and toxicity to evaluate its drug development potential, with emphasis on cardioprotective effects and related signaling pathways. METHODS Literature was collected from databases such as PubMed and Web of Science, focusing on studies addressing BA's chemical characteristics, biological activities, pharmacokinetics, and cardiovascular relevance. RESULTS BA exerts cardioprotective effects via multiple signaling pathways, including NRF2, NF-κB, MAPK, and NFAT. These contribute to its antioxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative actions, as well as its enhancement of endothelial function through nitric oxide signaling. BA also reduces lipid accumulation. Combined with favorable physicochemical properties and synthetic accessibility, these findings support BA as a promising multifunctional lead compound in cardiovascular pharmacology. CONCLUSION BA shows strong potential as a cardioprotective natural compound. Although further research is needed to validate its clinical efficacy and safety, its multi-target actions and structural versatility provide a solid basis for development in cardiovascular drug discovery.
Collapse
Affiliation(s)
- Xu Yuan Lao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuan Long Sun
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhe Jun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Liu
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Fen Ruan
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
6
|
Yu C, Huang J, Wang Y, Song J, Shen W. Dynamic deployment of H2A.Z positive nucleosome mediated transcriptomic plasticity within vascular smooth muscle cell. BMC Genomics 2025; 26:502. [PMID: 40389821 DOI: 10.1186/s12864-025-11679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 05/06/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND To maintain homeostasis in the mature human body, certain differentiated cells adopted high plasticity to refine their cellular functions. However, mechanisms that supported cellular plasticity still remained elusive. Here, through comprehensive transcriptomic and epigenetic studies of highly plastic vascular smooth muscle cells (SMCs), we aimed to decipher the chromatin basis that could mediate cellular plasticity. RESULTS In vascular smooth muscle cells, actively transcribed and highly adjustable genes tended to be associated with a continuously accessible region downstream of transcription start site (CAR-downTSS). This CAR-downTSS was located beyond the classic RNA polymerase II paused region, accessible at mono-nucleosome level and incorporated with histone variant H2A.Z. Depletion of H2A.Z reduced active histone modifications within CAR-downTSS, impaired RNA polymerase II transpassing when cells were stimulated, and consequently inhibited the ability of CAR-downTSS-associated genes to adjust their expression. Further in vitro and in vivo studies verified that this CAR-downTSS could be dynamically re-deployed onto different genes in vascular SMCs, whereas it was deployed in smaller quantities and remained quantitatively stable on genome within the quiescent cardiomyocytes. CONCLUSIONS Vascular SMCs dynamically deployed H2A.Z-positive nucleosomes extending continuously downstream transcription start sites on different genes to support their transcriptional adjustability, which served as an important mechanism mediating cellular plasticity.
Collapse
Affiliation(s)
- Chao Yu
- Institute of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jiaxin Huang
- Center for Diagnostics and Therapeutics, Amoytop Biotech Inc, Xiamen, China
| | - Yan Wang
- Institute of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Branch of National Clinical Research Center for Cardiovascular Diseases, Xiamen, China
| | - Jia Song
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China.
| | - Wei Shen
- Institute of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Fujian Branch of National Clinical Research Center for Cardiovascular Diseases, Xiamen, China.
| |
Collapse
|
7
|
Fu M, Lan Z, Ye Y, Gong Y, Liang Q, Li M, Feng L, Chen A, Dong Q, Li Y, Wang S, Liu X, Zhang X, Ou JS, Lu L, Yan J. The metabolite alpha-ketoglutarate inhibits vascular calcification partially through modulation of TET2/NLRP3 inflammasome signaling pathway. Kidney Int 2025:S0085-2538(25)00394-1. [PMID: 40383231 DOI: 10.1016/j.kint.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 03/30/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION Vascular calcification is prevalent in chronic kidney disease (CKD), but existing medical treatments fail to achieve satisfactory therapeutic effects. Vascular calcification is now recognized as an active multifactorial process involving diverse mechanisms. Alpha-ketoglutarate (AKG), an intermediate in tricarboxylic acid cycle, has been demonstrated to extend lifespan and ameliorate age-related osteoporosis. However, whether AKG inhibits vascular calcification remains unknown. METHODS Here, mineral deposition was studied with AKG treatment in rodent and human vascular smooth muscle cells (VSMCs) under osteogenic conditions in vivo and in vitro. RESULTS AKG treatment remarkably ameliorated calcification of rat and human arterial rings ex vivo and aortic calcification in CKD rats and mice. Mechanistically, AKG treatment upregulated DNA demethylase ten-eleven translocation 2 (TET2) expression during vascular calcification. Knockdown of TET2 by siRNA and pharmacological inhibition of TET2 by Bobcat339 promoted vascular calcification in rat VSMCs. Bobcat339 also enhanced rat aortic ring calcification. Conversely, TET2 overexpression ameliorated vascular calcification in rat VSMCs, rat aortic rings and CKD rats. Furthermore, VSMC-specific TET2 deficiency promoted aortic calcification in CKD mice. Both TET2 siRNA and Bobcat339 independently counteracted the inhibitory effect of AKG on vascular calcification of rat VSMCs. Inhibitory effect of AKG administration on vascular calcification was reduced in TET2 knockout mice. TET2 overexpression reduced the levels of the NLRP3 inflammasome pathway, cleaved Caspase-1 and IL-1β protein expression in VSMCs and NLRP3 agonist Nigericin-induced cell calcification. CONCLUSIONS Our study demonstrate that AKG attenuates vascular calcification partially via upregulation of TET2 and inhibition of NLRP3 inflammasome, indicating the critical role of epigenetic modifier in vascular calcification. Modulation of TET2 may become a promising strategy for the treatment of vascular calcification.
Collapse
Affiliation(s)
- Mingwei Fu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Yuan Gong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510665, China
| | - Mingxi Li
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Qianqian Dong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Yining Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Siyi Wang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Xiaoyu Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Xiuli Zhang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lihe Lu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou 510280, China.
| |
Collapse
|
8
|
Wang Y, Dou W, Qian X, Chen H, Zhang Y, Yang L, Wu Y, Xu X. Advancements in the study of short-chain fatty acids and their therapeutic effects on atherosclerosis. Life Sci 2025; 369:123528. [PMID: 40049368 DOI: 10.1016/j.lfs.2025.123528] [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: 12/01/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis (AS) remains a leading cause of cardiovascular disease and mortality globally. This chronic condition is characterized by inflammation, lipid accumulation, and the deposition of cellular components within arterial walls. Emerging evidence has highlighted the multifaceted therapeutic potential of short-chain fatty acids (SCFAs) in mitigating AS progression. SCFAs have demonstrated anti-inflammatory properties and the ability to regulate immune responses, metabolic pathways, vascular integrity, and intestinal barrier function in animal models of AS. Consequently, SCFAs have garnered significant attention as a promising approach for the prevention and treatment of AS. However, further clinical trials and studies are necessary to fully elucidate the underlying mechanisms and effects of SCFAs. Additionally, different types of SCFAs may exert distinct impacts, necessitating more in-depth investigation into their specific roles and mechanisms. This review provides an overview of the diverse cellular mechanisms contributing to AS formation, as well as a discussion of the significance of SCFAs in AS pathogenesis and their multifaceted therapeutic potential. Nonetheless, additional research is warranted to comprehensively understand and harness the potential of various SCFAs in the context of AS.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China; Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China; Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Wei Dou
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xin Qian
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Hao Chen
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Yi Zhang
- Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Liu Yang
- Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xiongfei Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
9
|
Huang L, Ding R, Yan K, Duan J, Sun Z. The Role of Endoplasmic Reticulum Stress in Fine Particulate Matter-Induced Phenotype Switching of Vascular Smooth Muscle Cells. Chem Res Toxicol 2025. [PMID: 40369400 DOI: 10.1021/acs.chemrestox.5c00056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
As a major component of air pollution, fine particulate matter (PM2.5) was the second global leading cause of death in 2021. Evidence from humans suggested that PM2.5 was associated with an enhanced coronary calcium score (CAC), and animal studies indicated that PM2.5 induced vascular calcification, while mechanisms remained largely unknown. In this study, PM2.5 enhanced the proliferative potential and migration capacity of human aortic vascular smooth muscle cells (VSMCs), as well as disturbing intracellular Ca2+ homeostasis. Subsequent transcriptomic analysis implicated that PM2.5 could influence genes involved in the IRE1α-mediated unfolded protein responses and reduce the expression of DNAJB9, a co-chaperone that formed a complex with BiP/IRE1α to inhibit the activation of endoplasmic reticulum (ER) stress. Further mechanistic investigations indicated that PM2.5 activated the IRE1α/XBP1 signaling pathway and enhanced the expression of osteogenic phenotype-related hallmarks. In contrast, pretreatment with an ER stress antagonist (4-PBA) could suppress PM2.5-associated calcium dysregulation and osteogenic transformation via alleviation of ER stress. Taken together, this study revealed the role of ER stress in the phenotype switching of VSMCs induced by PM2.5, highlighted the regulation of DNAJB9, provided insights into the mechanisms of air pollution-related vascular calcification, and pointed out molecules for future investigations.
Collapse
Affiliation(s)
- Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Kanglin Yan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
10
|
Qiu F, Miao HR, Hui HL, Qiu LJ, Chen Y, Luo M, Zhang JC, Lin YG, Li D, Ong SB, Hu XF, Jiang B, Zhang YQ. MHCII hiLYVE1 loCCR2 hi Interstitial Macrophages Promote Medial Fibrosis in Pulmonary Arterioles and Contribute to Pulmonary Hypertension. Circ Res 2025. [PMID: 40357547 DOI: 10.1161/circresaha.125.326173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/13/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a lethal disease characterized in part by progressive pulmonary arteriole (PA) remodeling. Excessive PA fibrosis and macrophage infiltration are often present in PH, but the potential associations are obscure. We investigated the link between interstitial macrophage (iMΦ) infiltration and PA fibrosis in PH and idiopathic pulmonary arterial hypertension. METHODS Lung tissue samples from patients with idiopathic pulmonary arterial hypertension and experimental PH animals were obtained to analyze the extent of fibrosis and iMΦ infiltration in the different layers of PAs and their correlation with disease severity. Single-cell RNA sequencing, lineage tracing, histological analyses, iMΦ and PA smooth muscle cell coculture, and transgenic animal experiments were used to investigate the cell heterogeneity and origins and molecular mechanisms by which iMΦs promote PA fibrosis. RESULTS We found that increased collagen deposition and fibrosis in the PA media were most strongly related to the severity of PH, and medial iMΦ infiltration may be involved in these pathological processes. Single-cell transcriptomics revealed that MHCIIhiLYVE1loCCR2hi iMΦs were the major type of iMΦ that expanded upon Sugen-5416 and hypoxia plus normoxia stimulation and were responsible for PA medial fibrosis. Lineage tracing experiments suggested that these medial iMΦs were largely from recruited monocytes. Mechanistically, MHCIIhiLYVE1loCCR2hi iMΦs promoted the transition of PA smooth muscle cells to a fibroblast-like phenotype through the WNT11 (wingless member 11)/planar cell polarity (PCP) pathway. Wnt11 deletion in iMΦs from PH rats normalized the fibrotic PA smooth muscle cell phenotype and decreased PA medial fibrosis, thereby improving vascular compliance and protecting against PH. Moreover, myeloid-specific Ccr2 deficiency in PH-PAs inhibited the medial infiltration of MHCIIhiLYVE1loCCR2hi iMΦs, which also relieved PH. CONCLUSIONS This study demonstrates that the recruitment of MHCIIhiLYVE1loCCR2hi iMΦs leads to medial fibrosis in PH-PAs associated with PH severity and that inhibition of their pathogenicity or recruitment reverses PA medial fibrosis and PH.
Collapse
Affiliation(s)
- Fan Qiu
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Hao-Ran Miao
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Hong-Liang Hui
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Lin-Jie Qiu
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Yi Chen
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Min Luo
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
| | - Jian-Chao Zhang
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Yan-Gui Lin
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
| | - Dan Li
- Community Health Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (D.L.)
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Chinese University of Hong Kong (CUHK) (S.-B.O.)
- Centre for Cardiovascular Genomics and Medicine, Lui Che Woo Institute of Innovative Medicine, Chinese University of Hong Kong (CUHK) (S.-B.O.)
- Neural, Vascular, and Metabolic Biology Thematic Research Program, School of Biomedical Sciences, Chinese University of Hong Kong (CUHK) (S.-B.O.)
- Hong Kong Hub of Paediatric Excellence, Hong Kong Children's Hospital, Kowloon Bay, China (S.-B.O.)
- Kunming Institute of Zoology-The Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences (S.-B.O.)
- CUHK Shenzhen Research Institute, China (S.-B.O.)
| | | | - Bo Jiang
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| | - Yi-Qian Zhang
- Department of Thoracic Cardiovascular Surgery, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., M.L., J.-C.Z., Y.-G.L., B.J., Y.-Q.Z.)
- Biological Laboratory of Hetao Shenzhen-Hong Kong Science and Technology Innovation Cooperation Zone, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China. (F.Q., H.-R.M., H.-L.H., L.-J.Q., Y.C., J.-C.Z., B.J., Y.-Q.Z.)
| |
Collapse
|
11
|
Zuo Y, NaveenKumar SK, Navaz S, Liang W, Sugur K, Kmetova K, Ayers CR, Kluge L, Chong E, Shah AM, Rohatgi A, Berry JD, Knight JS, de Lemos JA. Epidemiological and Translational Study of Calprotectin and Atherosclerotic Cardiovascular Disease. JAMA Cardiol 2025:2833589. [PMID: 40332890 PMCID: PMC12060021 DOI: 10.1001/jamacardio.2025.0945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 03/04/2025] [Indexed: 05/08/2025]
Abstract
Importance Innate immunity, particularly neutrophil activation, plays a crucial role in the pathogenesis of atherosclerotic cardiovascular disease (ASCVD). The potential of calprotectin, a biomarker of neutrophil activation, as a mechanistically informed biomarker for ASCVD in an ethnically diverse population requires further investigation. Objective To examine the prospective association between circulating calprotectin and ASCVD in a diverse, population-based cohort while also exploring calprotectin's mechanistic contributions to ASCVD in vitro. Design, Setting, and Participants Circulating calprotectin was measured in plasma collected from 2412 participants during phase 2 of the Dallas Heart Study, a multiethnic, population-based cohort study. The median follow-up after plasma collection was 8 years. Main Outcomes and Measures Associations with future ASCVD events (defined as first nonfatal myocardial infarction, nonfatal stroke, coronary revascularization, or death from a cardiovascular cause) were assessed using Cox proportional hazards models, adjusted for known cardiovascular disease risk factors as well as high-sensitivity C-reactive protein (hs-CRP), N-terminal pro-brain natriuretic peptide (NT-proBNP), and high-sensitivity cardiac troponin T (hs-cTnT). Results Higher calprotectin levels were associated with older age, male sex, Black race, hypertension, diabetes, and smoking history. Individuals with higher calprotectin had higher hemoglobin A1c, very low-density lipoprotein cholesterol, and triglycerides, and lower high-density lipoprotein cholesterol and cholesterol efflux capacity. Log-transformed calprotectin levels were associated with an increased risk of ASCVD events over 8 years (hazard ratio [HR], 1.98 per log increase [95% CI, 1.54-2.53]). This association remained statistically significant after adjusting for prior ASCVD and traditional risk factors (HR, 1.61 [95% CI, 1.22-2.13]) and hs-CRP, NT-proBNP, and hs-cTnT (HR, 1.43 [95% CI, 1.04-1.96]). Higher calprotectin also correlated with higher coronary artery calcium scores (P < .001). In vitro studies revealed that calprotectin impaired coronary endothelial integrity, diminished nitric oxide production, and fostered endothelial to mesenchymal transition, providing potential mechanisms for ASCVD progression. Conclusions and Relevance The findings suggest that calprotectin may serve as a mechanistically informed biomarker for ASCVD, independent of traditional and contemporary cardiovascular risk factors and biomarkers. However, its clinical utility warrants further evaluation.
Collapse
Affiliation(s)
- Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | | | - Sherwin Navaz
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Wenying Liang
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Kavya Sugur
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Katarina Kmetova
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Colby R. Ayers
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Lyndsay Kluge
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Emily Chong
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Amil M. Shah
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Anand Rohatgi
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Jarett D. Berry
- Department of Internal Medicine, University of Texas at Tyler
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - James A. de Lemos
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
12
|
Yu Q, Tian W. The role of SERCA in vascular diseases, a potential therapeutic target. Cell Calcium 2025; 129:103039. [PMID: 40367766 DOI: 10.1016/j.ceca.2025.103039] [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: 02/12/2025] [Revised: 04/19/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025]
Abstract
SERCA, the sarco/endoplasmic reticulum Ca2+-ATPase, is a pivotal protein that transports calcium ions (Ca2+) from the cytoplasm into the sarcoplasmic/endoplasmic reticulum (SR/ER), thus sustaining cellular Ca2+ homeostasis. A growing body of evidence indicates that SERCA dysfunction correlates with disrupted cellular Ca2+ homeostasis and ER stress, precipitating a spectrum of chronic diseases. As a regulator of Ca2+ homeostasis, SERCA emerges as a potential therapeutic target for conditions associated with Ca2+ imbalance. This review delineates the association between SERCA and a variety of vascular diseases.
Collapse
Affiliation(s)
- Qinghua Yu
- Department of Geriatric Cardiology, The First Hospital of China Medical University, China
| | - Wen Tian
- Department of Geriatric Cardiology, The First Hospital of China Medical University, China.
| |
Collapse
|
13
|
Brar S, Goli R, Barrios JP, Blaha MJ, Kianoush S, Pletcher MJ, Nomura SO, Tsai MY, Duan R, Budoff MJ, Szklo M, Tison GH. Association of Extracoronary Calcification and Incident Heart Failure in the Multiethnic Study of Atherosclerosis (MESA). JACC. HEART FAILURE 2025; 13:740-751. [PMID: 40146086 DOI: 10.1016/j.jchf.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/17/2024] [Accepted: 12/03/2024] [Indexed: 03/28/2025]
Abstract
BACKGROUND Extracoronary calcification (ECC) is a prevalent cardiovascular risk factor. OBJECTIVES The aim of this study was to examine the association between ECC and heart failure (HF), including heart failure with preserved ejection fraction (HFpEF). METHODS MESA (Multi-Ethnic Study of Atherosclerosis) participants with computed tomographic imaging at baseline for calcification of the aortic valve, aortic root, mitral valve, and thoracic aorta were included. ECC score was calculated by rescaling Agatston scores from 0 to 1 for each ECC site and summing the rescaled scores. Multivariable Cox proportional hazards regression was performed to examine the association between ECC quartiles and incident HF. RESULTS Of all MESA participants, 3,617 (53.1%) and 3,192 (46.9%) had ECC scores of 0 and >0, respectively. During a mean follow-up period of 12.9 ± 4.2 years, 358 HF events were observed, 179 HF with reduced ejection fraction and 135 HFpEF. After controlling for demographics and risk factors, the highest ECC quartile (compared with the lowest quartile) had 1.7-fold greater hazard of incident HF (adjusted HR: 1.72 [95% CI: 1.16-2.55]; P = 0.007), though this was attenuated to borderline significance after additional adjustment for coronary artery calcification. In contrast for HFpEF, the highest ECC quartile (compared with the lowest quartile) remained independently and statistically significantly associated with 3-fold greater hazard of incident HFpEF (adjusted HR: 3.09 [95% CI: 1.45-6.60]; P = 0.003) after full adjustment, including for coronary artery calcification. CONCLUSIONS ECC is associated with increased risk for HF, in particular HFpEF. If this finding is confirmed in other studies, ECC could help improve traditional risk factor estimation and clinical risk assessments for HF and HFpEF.
Collapse
Affiliation(s)
- Sandeep Brar
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | - Rahul Goli
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Joshua P Barrios
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Michael J Blaha
- Division of Cardiology, Department of Medicine, Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sina Kianoush
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Mark J Pletcher
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, San Francisco, California, USA
| | - Sarah O Nomura
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rong Duan
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Matthew J Budoff
- Division of Cardiology, Harbor-UCLA Medical Center, The Lundquist Institute, Los Angeles, California, USA
| | - Moyses Szklo
- Division of Cardiovascular and Clinical Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Geoffrey H Tison
- Division of Cardiology, Department of Medicine, University of California, San Francisco, San Francisco, California, USA; Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, California, USA; Center for Biosignal Research, University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
14
|
Miki S, Takashima M, Suzuki JI. Anti?atherosclerotic effect of aged garlic extract: Mode of action and therapeutic benefits (Review). Exp Ther Med 2025; 29:104. [PMID: 40171135 PMCID: PMC11959349 DOI: 10.3892/etm.2025.12854] [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: 09/25/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory disease characterized by plaque buildup within the arteries that obstructs blood flow and significantly increases the morbidity and mortality rates associated with cardiovascular diseases caused by impaired blood flow due to vascular stenosis or occlusion, such as angina and myocardial infarction. The development of atherosclerosis involves a complex interplay of endothelial dysfunction, accumulation of oxidized low-density lipoprotein and macrophage-driven inflammation. The risk factors for atherosclerosis include chronic inflammation, hyperlipidemia and hypertension. Effective management of these risk factors can prevent and delay the onset and progression of atherosclerosis. Garlic and its processed preparations have previously been utilized to mitigate cardiovascular risk factors and continue to be used in traditional medicine in several countries. Among these preparations, aged garlic extract (AGE) has been shown to improve atherosclerosis in clinical trials and animal studies. AGE contains various compounds with potential anti-atherosclerotic properties, such as S-1-propenylcysteine, S-allylcysteine and other sulfur-containing constituents, which may help prevent the development and progression of atherosclerosis. The present manuscript reviewed and discussed the anti-atherogenic effect of AGE and its constituents by highlighting their mode of action and potential benefits for prevention and therapy in the management of atherosclerosis.
Collapse
Affiliation(s)
- Satomi Miki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Miyuki Takashima
- Drug Discovery Laboratory, Wananga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| | - Jun-Ichiro Suzuki
- Central Research Institute, Wakunaga Pharmaceutical Co., Ltd., Hiroshima 739-1195, Japan
| |
Collapse
|
15
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2025; 71:317-335. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [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: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
16
|
Kozakova M, Morizzo C, Jamagidze G, Chiappino S, Chiappino D, Emdin M, Palombo C. Central pulse pressure, carotid artery remodeling and coronary artery calcifications. J Hypertens 2025; 43:774-780. [PMID: 39937055 DOI: 10.1097/hjh.0000000000003968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/27/2024] [Indexed: 02/13/2025]
Abstract
OBJECTIVES The aim of the study was to evaluate the role of central pulse pressure (PP) in carotid wall thickening and coronary artery calcification (CAC). METHODS In an asymptomatic general population ( N = 396, 163 men, 47-89 years), central PP was measured by applanation tonometry, CAC by computed tomography, and common carotid artery intima-media thickness (cIMT), pulse wave velocity (cPWV) and the power of the signal reflected from carotid media (cMP) by radiofrequency-based carotid ultrasound. High cIMT was defined as cIMT equal to or greater than the 75th percentile for given sex and age, and CAC presence as a CAC score greater than 0. RESULTS In the entire population, luminal diameter and cMP increased with increasing central PP ( r = 0.32 and 0.25; P < 0.0001). One hundred and ninety-seven individuals had high cIMT; individuals with high cIMT had higher central PP, luminal diameter, cMP and cPWV ( P = or <0.0001), but comparable wall tensile stress ( P = 0.23). In a logistic regression model, high cIMT was independently associated with luminal diameter and central PP. One hundred and fifty-two individuals had CAC score greater than 0; in a logistic regression model, CAC score greater than 0 was independently associated with sex, age, central PP, LDL-cholesterol, triglycerides and T2DM. CONCLUSION Our findings indicate that high central PP contributes both to an increase in cIMT and the development of CAC. However, while central PP was the only risk factor linked to high cIMT, multiple atherosclerotic risk factors were associated with CAC. Therefore, both high cIMT and CAC reflect the adverse impact of high pulsatile load on the vascular system, yet only CAC can be considered a marker of atherosclerosis.
Collapse
Affiliation(s)
- Michaela Kozakova
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa
- Esaote SpA, Genova
| | - Carmela Morizzo
- School of Medicine, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa
| | | | | | | | - Michele Emdin
- Fondazione Toscana G. Monasterio, Massa-Pisa
- Health Science Interdisciplinary Center, Sant'Anna School of Advanced Studies, Pisa, Italy
| | - Carlo Palombo
- School of Medicine, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa
| |
Collapse
|
17
|
Ding R, Ren X, Sun Q, liu S, Huang L, Sun Z, Duan J. Single-Cell Transcriptomics Revealed E-Cigarettes-Induced Vascular Remodeling by Enhancing Tcf21 Expression. MedComm (Beijing) 2025; 6:e70183. [PMID: 40248042 PMCID: PMC12004124 DOI: 10.1002/mco2.70183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| | - Qinglin Sun
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| | - Shiqian liu
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| | - Linyuan Huang
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| | - Junchao Duan
- Department of Toxicology and Sanitary ChemistrySchool of Public HealthCapital Medical UniversityBeijingPeople's Republic of China
- Laboratory for Clinical MedicineCapital Medical UniversityBeijingPeople's Republic of China
| |
Collapse
|
18
|
Liu F, Liang Q, Li L, Gong Y, Li M, Feng L, Chen A, Ye Y, Lan Z, Li Y, Ou JS, Lu L, Yan J. Thrombospondin-1 binds to integrin β3 to inhibit vascular calcification through suppression of NF-κB pathway. J Pathol 2025; 266:109-123. [PMID: 40084742 DOI: 10.1002/path.6417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 01/01/2025] [Accepted: 02/12/2025] [Indexed: 03/16/2025]
Abstract
Vascular calcification is an important risk factor related to all-cause mortality of cardiovascular events in patients with chronic kidney disease (CKD). Vascular extracellular matrix (ECM) proteins have been demonstrated to regulate vascular calcification. ECM protein thrombospondin 1 (THBS1/TSP-1) plays a critical role in the regulation of vascular diseases. However, whether THBS1 is involved in vascular calcification in CKD patients remains unclear. In this study, RNA sequencing datasets from the Gene Expression Omnibus (GEO) database GSE146638 showed that THBS1 was upregulated in the aortas of CKD rats. Enzyme-linked immunosorbent assay (elisa) revealed that serum THBS1 levels were increased in CKD patients with thoracic calcification. Western blotting and immunofluorescence analysis showed that THBS1 expression was increased in calcified vascular smooth muscle cells (VSMCs) and arteries. THBS1 knockdown exacerbated rat VSMC calcification induced by high phosphorus and calcium, as shown by Alizarin red staining and calcium content assays. Conversely, THBS1 overexpression attenuated VSMC calcification and abdominal aortic calcification in rats with CKD. Moreover, addition of recombinant THBS1 protein inhibited calcification of VSMCS and human arterial rings. Smooth muscle cell-specific knockout of THBS1 mice treated with vitamin D3 displayed aggravated aortic calcification. Mechanistically, the protein-protein interaction database STRING (http://string-db.org/) analysis and coimmunoprecipitation assays revealed THBS1 bound to integrin β3. Reduction of integrin β3 levels abrogated the protective effect of THBS1 on vascular calcification. RNA-seq analysis revealed that THBS1 overexpression modulated the nuclear factor-kappa B (NF-κB) signaling pathway. Of note, the inhibitory effect of THBS1 overexpression on the NF-κB signal was abolished by knockdown of integrin β3. In conclusion, THBS1 interacts with integrin β3 to inhibit vascular calcification through suppression of NF-κB signal, suggesting a promising therapeutic target for vascular calcification in CKD. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Thrombospondin 1/metabolism
- Thrombospondin 1/genetics
- Animals
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Vascular Calcification/prevention & control
- Humans
- NF-kappa B/metabolism
- Signal Transduction
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Integrin beta3/metabolism
- Integrin beta3/genetics
- Male
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Rats
- Mice
- Rats, Sprague-Dawley
- Disease Models, Animal
- Cells, Cultured
- Mice, Inbred C57BL
- Female
- Middle Aged
Collapse
Affiliation(s)
- Fang Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, PR China
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, PR China
| | - Yuan Gong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Mingxi Li
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, PR China
| | - Liyun Feng
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Yining Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| | - Jing-Song Ou
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC Key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Lihe Lu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, PR China
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease, Guangzhou, PR China
| |
Collapse
|
19
|
Zhou Z, Dong B, He D, Ma J, Kong Y, Zhu H, Xie C, Yang T, Zhen X, Zhang Z, He Z, Cheng J, Huang A, Chen J, Wu R, Yin H, Chen Y, Tao J, Huang H. GLS1-Mediated Redundancy in Glutamate Accelerates Arterial Calcification via Activating NMDAR/Ca 2+/β-Catenin Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414252. [PMID: 40289670 DOI: 10.1002/advs.202414252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/28/2025] [Indexed: 04/30/2025]
Abstract
Arterial calcification is a powerful predictor of both the events and mortality associated with cardiovascular diseases in chronic kidney disease (CKD) patients. GLS1 (glutaminase 1), a rate-limiting enzyme catalyzing the conversion of glutamine to glutamate, is disordered in various cardiovascular diseases. However, the potential interplay between GLS1-mediated glutamate production and arterial calcification remains poorly understood. Here, LC-MS/MS analysis of CKD patients' samples shows an abnormally elevated activity of GLS1, reflected by the increased glutamate/glutamine ratio. Moreover, GLS1 activity is positively correlated with arterial calcification progression, and its expression is upregulated in calcified arteries. Treatment with GLS1 inhibitors or knockdown of GLS1 alleviates osteogenic reprogramming. In contrast, glutamate administration boosts the development of arterial calcification. Mechanistically, GLS1 redundancy-regulated glutamate superfluity stimulates the activation of N-methyl-d-aspartate receptors (NMDAR), leading to Ca2+ influx and extracellular regulated protein kinases (ERK) phosphorylation, followed by the nuclear translocation of β-Catenin and acceleration of osteogenic reprogramming of vascular smooth muscle cells (VSMCs) in further. This research defines GLS1 as a key contributor to arterial calcification. Glutamate, a major product of GLS1-mediated glutamine metabolism, exerts a deleterious effect on arterial calcification by activating NMDAR and subsequently triggering Ca2+ influx, which in turn exacerbates β-Catenin-regulated osteogenic reprogramming in VSMCs.
Collapse
Affiliation(s)
- Ziting Zhou
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Bing Dong
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Dayu He
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Jianshuai Ma
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Yun Kong
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Huijin Zhu
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Chen Xie
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Tiecheng Yang
- Department of Nephrology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Xin Zhen
- Department of Nephrology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Zhengzhipeng Zhang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Zhaohui He
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Jinkun Cheng
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Aoran Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Jie Chen
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Ruo Wu
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Huiyong Yin
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Yanlian Chen
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| | - Jun Tao
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Hui Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518033, China
| |
Collapse
|
20
|
Kamboj K, Kumar V, Yadav AK. In Vitro Study of Vitamin D Effects on Immune, Endothelial, and Vascular Smooth Muscle Cells in Chronic Kidney Disease. Int J Mol Sci 2025; 26:3967. [PMID: 40362207 PMCID: PMC12071356 DOI: 10.3390/ijms26093967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Vitamin D has been shown to improve immunity as well as vascular function. We investigated the effect of cholecalciferol on T-cell phenotype in cultured peripheral blood mononuclear cells (PBMCs) from twenty vitamin D-deficient, non-diabetic chronic kidney disease (CKD) subjects. We also studied vitamin D effects on endothelial and vascular function markers in human aortic endothelial cells (HAECs) and in human aortic smooth muscle cells (HASMCs), respectively. We studied endothelial nitric oxide synthase (eNOS), mitogen-activated protein kinase 38 (p38 Map kinase), protein kinase B (Akt), and nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase) in HAECs and α-smooth muscle actin (α-SMA), smooth muscle calponin (SM-Calponin), smooth muscle myosin heavy chain (SM-MHC), and calcium-sensing receptor (CaSR) in HASMCs. Vitamin D receptors (VDRs) and CYP27B1 were studied in both cell types. In cultured PBMCs isolated from CKD subjects, the percentage of T helper 1(TH1) cells significantly decreased while that of T helper 2 (TH2) cells increased after cholecalciferol treatment. No significant change in intracellular and surface markers of T helper 17 (TH17) and T regulatory (Treg) cells was observed. In vitro treatment of HASMCs and HAECs with cholecalciferol led to significant and favorable alterations in mRNA expression of markers of vascular smooth muscle cells, i.e., α-SMA, SM-Calponin, and SM-MHC. Regarding endothelial cell markers, mRNA encoding eNOS, p38 Map kinase, protein kinase B (Akt), NADPH oxidase, VDR, and CYP27B1 were also significantly changed. Finally, the expression levels of the following proteins were notably altered: NADPH oxidase and protein kinase B (Akt) (in HAECs); SM-MHC and SM-Calponin (in HASMCs). In vitro treatment of PBMCs with cholecalciferol led to a favorable change in T-cell population, decreasing TH1 and increasing TH2 cell percentage, along with beneficial alterations in mRNA expression of HASMCs and HAECs' cell markers. This study provides evidence that cholecalciferol can influence immune and vascular function in CKD.
Collapse
MESH Headings
- Humans
- Renal Insufficiency, Chronic/immunology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/drug therapy
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/pathology
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Vitamin D/pharmacology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nitric Oxide Synthase Type III/metabolism
- Male
- Middle Aged
- Receptors, Calcitriol/metabolism
- Receptors, Calcitriol/genetics
- Cells, Cultured
- Cholecalciferol/pharmacology
- Female
- 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/metabolism
- 25-Hydroxyvitamin D3 1-alpha-Hydroxylase/genetics
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- p38 Mitogen-Activated Protein Kinases/metabolism
- Adult
- NADPH Oxidases/metabolism
Collapse
Affiliation(s)
- Kajal Kamboj
- Departments of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India; (K.K.); (V.K.)
| | - Vivek Kumar
- Departments of Nephrology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India; (K.K.); (V.K.)
| | - Ashok Kumar Yadav
- Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
21
|
Jeon Y, Kwon Y, Kim YJ, Jeon S, Ryu H, An K, Kim BC, Kim W, Lee SY, Bae JW, Hwang JY, Kang MG, Kang Y, Bhak J, Shin ES. Epigenetic Modulation of Vascular Smooth Muscle Cell Phenotype Switching in Early-Onset Acute Myocardial Infarction. Arterioscler Thromb Vasc Biol 2025. [PMID: 40242870 DOI: 10.1161/atvbaha.125.322503] [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: 01/19/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The epigenetic mechanisms underlying early-onset acute myocardial infarction (AMI) remain insufficiently characterized. The present study aims to elucidate the pathophysiology of early-onset AMI by investigating its epigenetic features as molecular indicators. METHODS A comparative differential methylation analysis was performed on whole blood samples from 298 patients with early-onset AMI with clinical follow-up and 247 controls using targeted bisulfite sequencing. Clusters of differentially methylated sites (CDMSs) were defined to highlight regions of concentrated methylation changes in patients with early-onset AMI. Cox proportional hazards regression was conducted to evaluate the prognostic significance of the methylation biomarkers. RESULTS A total of 692 differentially methylated sites (DMSs) were identified as biomarkers associated with early-onset AMI. Among these, 396 DMSs were grouped into 147 CDMSs. Notably, the UHRF1 and STIMATE genes, which regulate synthetic and osteoblast-like vascular smooth muscle cell phenotypes, respectively, contained CDMSs with the highest number of significant DMSs. UHRF1 demonstrated a CDMS with 10 significant DMSs within a 117-bp region, while STIMATE included a 264-bp CDMS with 10 significant DMSs. Both regions also exhibited consistent methylation patterns in coronary tissues, comparing human coronary plaque to normal coronary artery samples. Additionally, the HIPK3 gene, which modulates STAT3 expression, thereby promoting osteoblast-like transformation in vascular smooth muscle cells, showed a CDMS with 5 significant DMSs within a 123-bp region, with further validation in the corresponding tissues. Furthermore, over 66% biomarkers demonstrated significant associations with mortality in patients with early-onset AMI, providing evidence of the impact of these biomarkers on the pathophysiology of the disease. CONCLUSIONS This innovative epigenomic study into early-onset AMI not only identifies biomarkers associated with the disease and its mortality but also highlights the critical role of vascular smooth muscle cell phenotype regulation in early-onset AMI pathogenesis. Our findings suggest that changes in vascular smooth muscle cell phenotypes toward synthetic and osteoblast-like states play a crucial role in early-onset AMI.
Collapse
Affiliation(s)
- Yeonsu Jeon
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Yoonsung Kwon
- Korean Genomics Center, Ulsan National Institute of Science and Technology, Republic of Korea. (Y. Kwon, K.A., J.B.)
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, Republic of Korea. (Y. Kwon, K.A., J.B.)
| | - Yeo Jin Kim
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Sungwon Jeon
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Hyojung Ryu
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Kyungwhan An
- Korean Genomics Center, Ulsan National Institute of Science and Technology, Republic of Korea. (Y. Kwon, K.A., J.B.)
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, Republic of Korea. (Y. Kwon, K.A., J.B.)
| | - Byoung-Chul Kim
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Weon Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea (W.K.)
| | - Sang Yeub Lee
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University College of Medicine, Chung-Ang University Gwangmyeong Hospital, Republic of Korea (S.Y.L.)
| | - Jang-Whan Bae
- Department of Internal Medicine, Chungbuk National University Hospital, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea (J.-W.B.)
| | - Jin-Yong Hwang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea (J.-Y.H., M.G.K.)
| | - Min Gyu Kang
- Department of Internal Medicine, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Republic of Korea (J.-Y.H., M.G.K.)
| | - Younghui Kang
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
| | - Jong Bhak
- Clinomics, Inc, Osong, Republic of Korea (Y.J., Y.J.K., S.J., H.R., B.-C.K., Y. Kang, J.B.)
- Korean Genomics Center, Ulsan National Institute of Science and Technology, Republic of Korea. (Y. Kwon, K.A., J.B.)
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, Republic of Korea. (Y. Kwon, K.A., J.B.)
- Personal Genomics Institute, Genome Research Foundation, Osong, Republic of Korea (J.B.)
| | - Eun-Seok Shin
- Department of Cardiology, Ulsan University Hospital, University of Ulsan College of Medicine, Republic of Korea (E.-S.S.)
| |
Collapse
|
22
|
Schneider EH, Dopico AM, Bukiya AN. The Slo1 Y450F Substitution Modifies Basal Function and Cholesterol Response of Middle Cerebral Artery Smooth Muscle BK Channels in a Sexually Dimorphic Manner. Int J Mol Sci 2025; 26:3814. [PMID: 40332464 PMCID: PMC12028221 DOI: 10.3390/ijms26083814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Calcium- and voltage-gated potassium channels of large conductance (BK channels) in smooth muscle (SM) act as part of a negative feedback mechanism on SM contraction and associated decrease in cerebral artery diameter. Functional BK channels result from tetrameric association of α subunits encoded by KCNMA1 (Slo1). Ionic current from slo1 channels is inhibited by cholesterol in artificial lipid bilayers, an effect significantly reduced by the slo1 Y450F substitution. Whether such substitution affects cholesterol action on cerebral artery SM BK channel function and diameter remains unknown. Using the KCNMA1Y450F knock-in (K/I) mouse, we determined the effect of cholesterol enrichment on BK currents in native SM cells from middle cerebral artery using patch-clamp electrophysiology and the artery diameter ex vivo response to cholesterol. Results show that the KCNMA1Y450F K/I mutation modifies both basal function and the channel's response to cholesterol enrichment. Such modifications are detectable solely in SM cells from males, demonstrating sexual dimorphism. Unexpectedly, the modifications introduced by the Y450F substitution do not translate into observable changes in middle cerebral artery diameter ex vivo, suggesting that mechanisms at the SM level compensate for changes driven by the KCNMA1 point mutation under study.
Collapse
Affiliation(s)
| | | | - Anna N. Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
23
|
Ngo DHA, Hwang SB, Kwak HS. Impact of Carotid Artery Geometry and Clinical Risk Factors on Carotid Atherosclerotic Plaque Prevalence. J Pers Med 2025; 15:152. [PMID: 40278331 PMCID: PMC12029118 DOI: 10.3390/jpm15040152] [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: 02/27/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
Objectives: Carotid geometry and cardiovascular risk factors play a significant role in the development of carotid atherosclerotic plaques. This study aimed to investigate the correlation between carotid plaque formation and carotid artery geometry characteristics. Methods: A retrospective cross-sectional analysis was performed on 1227 patients, categorized into a normal group (n = 685) and carotid plaque groups causing either mild stenosis (<50% stenosis based on NASCET criteria, n = 385) or moderate-to-severe stenosis (>50%, n = 232). The left and right carotid were evaluated individually for each group. Patient data, including cardiovascular risk factors and laboratory test results, were collected. Carotid geometric measurements were obtained from 3D models reconstructed from cranio-cervical computed tomography angiography (CTA) using semi-automated software (MIMICS). The geometric variables analyzed included the vascular diameter and sectional area of the common carotid artery (CCA), internal carotid artery (ICA), external carotid artery (ECA), and carotid artery bifurcation (CAB), as well as the carotid bifurcation angles and carotid tortuosity. Results: Compared to the normal group, in both the right and left carotid arteries, patients with carotid plaques exhibited a significantly higher age (p < 0.001) and a greater prevalence of hypertension (p < 0.001) and diabetes mellitus (p < 0.001). Additionally, they demonstrated a larger CCA and a smaller carotid bifurcation dimension (p < 0.05). In the analysis of the left carotid artery, patients with carotid plaques also had a significantly smaller ICA dimension (p < 0.05) than the normal group. Conclusions: This study found that patients with carotid plaques were older and had a higher prevalence of hypertension and diabetes, larger CCAs, and smaller carotid bifurcations. The plaque-positive left ICA was significantly smaller than that of the plaque-negative group, suggesting a side-specific vulnerability. These findings highlight the role of carotid geometry in plaque formation and its potential clinical implications for personalized risk assessment and targeted interventions.
Collapse
Affiliation(s)
- Dac Hong An Ngo
- Department of Radiology, Research Institute of Clinical Medicine, Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, 20, Geonji-ro, Deokjin-gu, Jeonju-si 54907, Jeollabuk-do, Republic of Korea; (D.H.A.N.); (S.B.H.)
- Department of Radiology, University of Medicine and Pharmacy, Hue University, Hue 530000, Vietnam
| | - Seung Bae Hwang
- Department of Radiology, Research Institute of Clinical Medicine, Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, 20, Geonji-ro, Deokjin-gu, Jeonju-si 54907, Jeollabuk-do, Republic of Korea; (D.H.A.N.); (S.B.H.)
| | - Hyo Sung Kwak
- Department of Radiology, Research Institute of Clinical Medicine, Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, 20, Geonji-ro, Deokjin-gu, Jeonju-si 54907, Jeollabuk-do, Republic of Korea; (D.H.A.N.); (S.B.H.)
| |
Collapse
|
24
|
Savulescu-Fiedler I, Baz RO, Baz RA, Scheau C, Gegiu A. Coronary Artery Spasm: From Physiopathology to Diagnosis. Life (Basel) 2025; 15:597. [PMID: 40283152 PMCID: PMC12029111 DOI: 10.3390/life15040597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Coronary artery spasm (CAS) is a reversible vasoconstriction of normal or atherosclerotic epicardial coronary arteries with a subsequent reduction in myocardial blood flow, leading to myocardial ischemia, myocardial infarction, severe arrhythmias, or even sudden death. It is an entity that should be recognized based on a particular clinical presentation. Numerous differences exist between CAS and obstructive coronary disease in terms of mechanisms, risk factors, and therapeutic solutions. The gold standard for CAS diagnosis is represented by transitory and reversible occlusion of the coronary arteries at spasm provocation test, which consists of an intracoronary administration of Ach, ergonovine, or methylergonovine during angiography. The pathophysiology of CAS is not fully understood. However, the core of CAS is represented by vascular smooth muscle cell contraction, with a circadian pattern. The initiating event of this contraction may be represented by endothelial dysfunction, inflammation, or autonomic nervous system unbalance. Our study explores the intricate balance of these factors and their clinical relevance in the management of CAS.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Radu Octavian Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
- Department of Radiology and Medical Imaging, Faculty of Medicine, “Ovidius” University, 900527 Constanta, Romania
| | - Radu Andrei Baz
- Clinical Laboratory of Radiology and Medical Imaging, “Sf. Apostol Andrei” County Emergency Hospital, 900591 Constanta, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 030167 Bucharest, Romania
| | - Andrei Gegiu
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| |
Collapse
|
25
|
Zhang B, Liu Z, Chang Y, Lv R, Guo H, Qiang P, Shimosawa T, Xu Q, Yang F. Aldosterone-Induced Transformation of Vascular Smooth Muscle Cells into Macrophage-like Cells Participates in Inflammatory Vascular Lesions. Int J Mol Sci 2025; 26:3345. [PMID: 40244230 PMCID: PMC11989480 DOI: 10.3390/ijms26073345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the most abundant cell type in blood vessels, participating in cardiovascular diseases in various ways, among which their transformation into macrophage-like cells has become a research hotspot. In this study, rats were infused with aldosterone for 12 weeks, and VSMCs stimulated with aldosterone in vitro were used to observe aortic injury and the role of VSMC transformation. Vascular changes were detected via small animal ultrasound and H&E staining. Moreover, immunohistochemistry, immunofluorescence, Western blot, and flow cytometry were used to verify that the transformation of VSMCs into macrophage-like cells is regulated by mineralocorticoid receptor (MR) activation and macrophage colony-stimulating factor (M-CSF) and its receptor. Rat vasculature and in vitro cellular experiments revealed that VSMCs transformed into macrophage-like cells and secreted inflammatory factors such as interleukin-1β (IL-1β) and monocyte chemoattractant protein-1 (MCP-1), thereby exacerbating inflammatory vascular lesions, which was inhibited by the MR antagonist esaxerenone. These results reveal that increased levels of aldosterone activate MR, leading to the secretion of M-CSF by VSMCs. This further promotes the transformation of VSMCs into macrophage-like cells, which participate in inflammatory vascular lesions. Therefore, inhibiting the formation of macrophage-like cells can effectively reduce inflammatory vascular lesions.
Collapse
MESH Headings
- Animals
- Aldosterone/pharmacology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Rats
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Receptors, Mineralocorticoid/metabolism
- Male
- Inflammation/metabolism
- Inflammation/pathology
- Rats, Sprague-Dawley
- Cells, Cultured
- Macrophage Colony-Stimulating Factor/metabolism
- Interleukin-1beta/metabolism
- Chemokine CCL2/metabolism
Collapse
Affiliation(s)
- Boya Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ziqian Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yi Chang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ruyan Lv
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haixia Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Panpan Qiang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita 286-8686, Japan
| | - Qingyou Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fan Yang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| |
Collapse
|
26
|
Bolanle IO, Palmer TM. O-GlcNAcylation and Phosphorylation Crosstalk in Vascular Smooth Muscle Cells: Cellular and Therapeutic Significance in Cardiac and Vascular Pathologies. Int J Mol Sci 2025; 26:3303. [PMID: 40244145 PMCID: PMC11989994 DOI: 10.3390/ijms26073303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
More than 400 different types of post-translational modifications (PTMs), including O-GlcNAcylation and phosphorylation, combine to co-ordinate almost all aspects of protein function. Often, these PTMs overlap and the specific relationship between O-GlcNAcylation and phosphorylation has drawn much attention. In the last decade, the significance of this dynamic crosstalk has been linked to several chronic pathologies of cardiovascular origin. However, very little is known about the pathophysiological significance of this crosstalk for vascular smooth muscle cell dysfunction in cardiovascular disease. O-GlcNAcylation occurs on serine and threonine residues which are also targets for phosphorylation. A growing body of research has now emerged linking altered vascular integrity and homeostasis with highly regulated crosstalk between these PTMs. Additionally, a significant body of evidence indicates that O-GlcNAcylation is an important contributor to the pathogenesis of neointimal hyperplasia and vascular restenosis responsible for long-term vein graft failure. In this review, we evaluate the significance of this dynamic crosstalk and its role in cardiovascular pathologies, and the prospects of identifying possible targets for more effective therapeutic interventions.
Collapse
Affiliation(s)
| | - Timothy M. Palmer
- Biomedical Institute for Multimorbidity, Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| |
Collapse
|
27
|
Orlov I, Lenglet G, Avondo C, Beattie JH, Kamel S, Korichneva I. Copper Impedes Calcification of Human Aortic Vascular Smooth Muscle Cells Through Inhibition of Osteogenic Transdifferentiation and Promotion of Extracellular Matrix Stability. J Cell Physiol 2025; 240:e70035. [PMID: 40249000 PMCID: PMC12007077 DOI: 10.1002/jcp.70035] [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: 09/16/2024] [Revised: 01/20/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025]
Abstract
Vascular calcification (VC), a common pathological condition, is a strong predictor of cardiovascular events and associated mortality. Development and progression of VC heavily rely on vascular smooth muscle cells (VSMCs) and are closely related to oxidative stress, inflammation, and remodelling of extracellular matrix (ECM). Copper (Cu), an essential microelement, participates in these processes, but its involvement in pathophysiology of VC and VSMCs physiology remains poorly investigated. In the present study, we analysed the impact of Cu on the calcification of human aortic primary VSMCs induced in vitro by treatment with high calcium and phosphate levels. Supplementation with physiological micromolar doses of Cu significantly reduced the amount of calcium deposited on VSMCs as compared to moderate deficiency, Cu restriction with chelators or Cu excess. Moreover, optimal concentrations of Cu ions increased protein production by VSMCs, stimulated their metabolic activity, inhibited alkaline phosphatase activity associated with cell-conditioned medium and cellular lysates, and prevented osteogenic differentiation of VSMCs. RNA-seq results indicated that high calcium and phosphate treatments activated many pathways related to oxidative stress and inflammation in VSMCs at the initial stage of calcification. At the same time, expression of VSMCs-specific markers and certain components of ECM were downregulated. Supplementation of calcifying cells with 10 μM Cu prevented most of the transcriptomic alterations induced by high calcium and phosphate while chelation-mediated restriction of Cu greatly aggravated them. In summary, physiological concentration of Cu impedes in vitro calcification of VSMCs, prevents their osteogenic transition and minimises early phenotypic alterations induced by high calcium and phosphate, thereby underlining the importance of Cu homeostasis for the physiology of VSMCs, one of the cornerstones of cardiovascular health. Our data suggest that features of Cu metabolism and its status should be considered when developing preventive and therapeutic approaches for cardiovascular diseases.
Collapse
MESH Headings
- Humans
- Copper/pharmacology
- Extracellular Matrix/drug effects
- Extracellular Matrix/metabolism
- Cell Transdifferentiation/drug effects
- Osteogenesis/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/drug therapy
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Aorta/drug effects
- Aorta/pathology
- Aorta/metabolism
- Cells, Cultured
- Oxidative Stress/drug effects
- Calcium/metabolism
- Phosphates
Collapse
Affiliation(s)
- Iurii Orlov
- University of Picardie Jules VerneAmiensFrance
| | | | | | | | - Said Kamel
- University of Picardie Jules VerneAmiensFrance
| | | |
Collapse
|
28
|
You LL, Luo XB, Zhou WQ, Zhang RC, Li ZH, Xu JX, Ran J, Xu J. Aerobic exercise modulates aortic chondrogenesis and calcification via 5-methoxytryptophan and P38MAPK in atherosclerotic rats. Exp Gerontol 2025; 202:112722. [PMID: 40024450 DOI: 10.1016/j.exger.2025.112722] [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: 01/03/2025] [Revised: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND 5-Methoxytryptophan (5-MTP), a new endothelial factor with vasoprotective and anti-inflammatory effects, reduces aortic chondrogenesis and calcification during atherosclerosis. The aim of this study was to investigate the effects of aerobic exercise on aortic chondrogenesis and calcification during atherosclerosis in rats. To investigate the effect of aerobic exercise on the expression of 5-MTP/P38 MAPK signaling pathway. To lay a theoretical foundation for the therapeutic effect of exercise in rat atherosclerosis model. METHODS Establishment of a rat model of atherosclerosis using a high-fat diet combined with intraperitoneal injection of vitamin D3 (VD3). The aerobic exercise group underwent moderate-intensity aerobic exercise on an exercise treadmill for 8 weeks, while the atherosclerosis model group and the control group did not exercise. After exercise, blood and aortic samples were collected from all rats to evaluate the levels of serum triglyceride (TG), cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDLC), aortic chondrogenesis, calcification, 5-MTP level, collagen II, P38MAPK, pp38 MAPK, and IL-6 protein content. RESULTS (1)8 weeks of aerobic exercise significantly reduced aortic chondrogenesis, area of calcification, serum LDL-C, TC levels, atherosclerotic index and serum IL-6 levels in rats (p < 0.01), and lowered TG levels (p < 0.05);(2)8 weeks of aerobic exercise significantly increased aortic 5-MTP levels (p < 0.01) and decreased the content of aortic pp38MAPK, collagen II and IL-6 proteins significantly (p < 0.01). The pp38MAPK/P38MAPK ratio was also decreased (p < 0.01). CONCLUSION 8 weeks of aerobic exercise training improved dyslipidemia and reduced aortic chondrogenesis and calcification formation in AS rats. The mechanism may be related to increasing aortic 5-MTP levels and inhibiting the P38MAPK/ IL-6 signaling pathway.
Collapse
Affiliation(s)
| | - Xiao-Bing Luo
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China.
| | - Wen-Qi Zhou
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China.
| | - Rui-Chi Zhang
- Chengdu Sport University, School of Sports Medicine and Health, Chengdu, Sichuan 610041, China
| | - Zhong-Hao Li
- Chengdu Sport University, School of Sports Medicine and Health, Chengdu, Sichuan 610041, China
| | - Jia-Xin Xu
- Chengdu Sport University, School of Sports Medicine and Health, Chengdu, Sichuan 610041, China
| | - Jing Ran
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China
| | - Jie Xu
- Sichuan Orthopedic Hospital, Chengdu, Sichuan 610041, China
| |
Collapse
|
29
|
Calandri E, Verdoia M, Sirovich R, Giraudo MT, Pultrone M, Frantellizzi V, Viola O, Crivelli F, Gallina S, Serralunga M, Rognoni A, De Vincentis G. Combined single-photon emission computed tomography-myocardial perfusion imaging with coronary calcium score for assessing coronary disease. Nucl Med Commun 2025; 46:317-325. [PMID: 39773914 DOI: 10.1097/mnm.0000000000001947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
PURPOSE Coronary artery disease (CAD) underestimation represents a major pitfall of single-photon emission computed tomography-myocardial perfusion imaging (SPECT-MPI). Coronary artery calcium score (CACS) has emerged as a sensitive tool for the assessment of suspect CAD; however, the integration of SPECT-MPI with CACS has been seldom evaluated, so far, and was therefore the aim of the present study. METHODS Patients undergoing SPECT-MPI with CACS and subsequent coronary angiography were included. ROC curves were used to identify the CACS values best predictive for CAD. In SPECT-MPI negative patients, the formula: defined the optimal CACS cut-points. The Systematic Coronary Risk Evaluation 2 was applied for 10-year cardiovascular risk estimation. Significant CAD was defined for an epicardial coronary stenosis >70 or 50% for the left main. RESULTS Among 124 patients, 61 (49.19%) displayed positive SPECT-MPI, whereas 69 (56%) had significant CAD at angiography. Sensitivity, specificity, and positive predictive value (PPV) for SPECT-MPI were, respectively, 74, 82, and 84%. Considering 63 SPECT-MPI negative cases, the index values for CACS at the optimal cutoff value of 1949 were: sensitivity 28%, specificity 89%, and PPV 50%, allowing to further detect five (8%) of the patients with significant CAD. The increased discriminative power of the combined SPECT-MPI with CACS was not conditioned by the pretest cardiovascular risk. CONCLUSION Among patients with suspect CAD undergoing SPECT-MPI, the addition of CACS in negative cases allows to detect a consistent further 8% of patients with significant CAD, thus limiting the risk of disease underestimation and offering potential prognostic benefits.
Collapse
Affiliation(s)
| | - Monica Verdoia
- Department of Medicine and Urgency, Cardiology Unit, Ospedale degli Infermi, Ponderano,
| | | | | | - Mirco Pultrone
- Department of Medicine and Urgency, Nuclear Medicine Unit,
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Nuclear Medicine Unit, 'Sapienza' University, Rome and
| | - Orazio Viola
- Department of Medicine and Urgency, Cardiology Unit, Ospedale degli Infermi, Ponderano,
| | | | - Sonya Gallina
- Department of Medicine and Urgency, Nuclear Medicine Unit,
| | | | - Andrea Rognoni
- Department of Medicine and Urgency, Cardiology Unit, Ospedale degli Infermi, Ponderano,
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Nuclear Medicine Unit, 'Sapienza' University, Rome and
| |
Collapse
|
30
|
Roy M, Guo X, Wang Q, Stäb D, Jin N, Lim RP, Ooi A, Chakraborty S. Patient-specific prediction of arterial wall elasticity using medical image-informed in-silico simulations. Comput Biol Med 2025; 188:109849. [PMID: 39978097 DOI: 10.1016/j.compbiomed.2025.109849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Limitations in clinical cardiovascular research have driven the development of advanced simulations for patient-specific insights into arterial elasticity. However, uncertainties in model inputs, data resolution, and parameter estimation can compromise accuracy. Our research aimed to provide reliable estimates of the arterial wall elasticity non-invasively, where direct clinical measurement is difficult. By integrating patient-specific imaging with a simplified flow simulation model and uncertainty quantification, we sought to improve the reliability of these predictions as compared to the state-of-the-art. In a proof-of-concept study, we developed a simple area-averaged model of arterial hemodynamics, using Magnetic Resonance Angiogram (MRA)-derived geometries and input parameters based on the age, cuff blood pressure, and phase-contrast MRI data in five human subjects. This resulted in an in-silico model estimating the pressure and flow variations across the arterial-branches. Statistical uncertainties in the hemodynamic parameter predictions were quantified using non-intrusive Polynomial Chaos. Additionally, we developed a model to estimate the arterial elasticity by interlacing the results from fluid-structure interaction simulation for arterial hemodynamics with patient-specific clinical data. We found that the arterial elasticity values derived from our model, when used to predict the flowrates, closely matched the flow characteristics obtained from the patient-specific 4D flow MRI. The findings also showed zero or minimal positive/negative bias in our simulations, with no noticeable systematic error in predicting arterial elasticity values. Our results evidenced that accurate prediction of arterial wall elasticity is possible through use of an efficient simulation technique supplemented with clinically attainable imaging data. This has potential to predict cardiovascular-risk and guide individual patient management.
Collapse
Affiliation(s)
- Manideep Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Xiaojing Guo
- Department of Mechanical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Qingdi Wang
- Department of Mechanical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, VIC, 3010, Australia; Department of Biomedical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Daniel Stäb
- MR Research Collaborations, Siemens Healthcare Pty Limited, Melbourne, VIC, 3153, Australia
| | - Ning Jin
- Siemens Medical Solutions Inc. Malvern, PA, 19355, USA
| | - Ruth P Lim
- Departments of Radiology and Surgery, Melbourne Medical School, The University of Melbourne, Melbourne, VIC, 3010, Australia; Department of Radiology, Austin Health, Heidelberg VIC, 3084, Australia
| | - Andrew Ooi
- Department of Mechanical Engineering, Melbourne School of Engineering, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Suman Chakraborty
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India; Department of Mechanical Engineering, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| |
Collapse
|
31
|
Kawaguchi YO, Fujimoto S, Nozaki YO, Tomizawa N, Daida H, Minamino T. Current status and future perspective of coronary artery calcium score in asymptomatic individuals. J Cardiol 2025; 85:275-282. [PMID: 39631694 DOI: 10.1016/j.jjcc.2024.11.008] [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: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 12/07/2024]
Abstract
Atherosclerotic cardiovascular disease remains a major cause of death, and it is important to accurately estimate the cardiovascular events risk stratification even in asymptomatic patients. The coronary artery calcium score (CACS), which is quantitatively evaluated by electrocardiogram (ECG)-gated non-contrast chest computed tomography (CT) imaging, has been reported to be useful for cardiovascular event risk stratification in large studies. In the USA and Europe, guidelines recommend the use of the CACS in borderline or intermediate-risk asymptomatic individuals based on a high level of evidence. In Japan, however, the use of CACS in clinical practice is currently limited. Although it has been reported that the prevalence and distribution of coronary artery calcification (CAC) may differ by race and ethnicity, there are few data on its usefulness in stratifying the risk of cardiovascular events in asymptomatic Japanese individuals. While it is important to establish evidence for the usefulness of CACS in the Japanese population, for widespread clinical dissemination it would be beneficial to evaluate CAC and to perform accurate cardiovascular event risk stratification from non-ECG-gated non-contrast chest CT imaging performed during medical check-up and routine clinical practice. There have been reports on the usefulness of CAC assessed by non-ECG-gated chest CT imaging and on the relationship of CAC between ECG-gated and non-ECG-gated chest CT imaging. In recent years, a more accurate method of evaluating CACS from non-ECG-gated chest CT imaging has been developed using artificial intelligence, and further development is expected in the future.
Collapse
Affiliation(s)
- Yuko O Kawaguchi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinichiro Fujimoto
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Yui O Nozaki
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobuo Tomizawa
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Daida
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Radiological Technology, Juntendo University, Graduate School of Health Science, Tokyo, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
32
|
Li Y, Ma C, Sheng Y, Huang S, Sun H, Ti Y, Wang Z, Wang F, Chen F, Li C, Guo H, Tang M, Song F, Wang H, Zhong M. TRIB3 mediates vascular calcification by facilitating self-ubiquitination and dissociation of Smurf1 in chronic kidney disease. J Clin Invest 2025; 135:e175972. [PMID: 39932798 PMCID: PMC11957692 DOI: 10.1172/jci175972] [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: 10/03/2023] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
The osteogenic environment promotes vascular calcium phosphate deposition and aggregation of unfolded and misfolded proteins, resulting in ER stress in chronic kidney disease (CKD). Controlling ER stress through genetic intervention is a promising approach for treating vascular calcification. In this study, we demonstrated a positive correlation between ER stress-induced tribble homolog 3 (TRIB3) expression and progression of vascular calcification in human and rodent CKD. Increased TRIB3 expression promoted vascular smooth muscle cell (VSMC) calcification by interacting with the C2 domain of the E3 ubiquitin-protein ligase Smurf1, facilitating its K48-related self-ubiquitination at Lys381 and Lys383 and subsequent dissociation from the plasma membrane and nuclei. This degeneration of Smurf1 accelerated the stabilization of the osteogenic transcription factors RUNX family transcription factor 2 (Runx2) and SMAD family member 1 (Smad1). C/EBP homologous protein and activating transcription factor 4 are upstream transcription factors of TRIB3 in an osteogenic environment. Genetic KO of TRIB3 or rescue of Smurf1 ameliorated VSMC and vascular calcification by stabilizing Smurf1 and enhancing the degradation of Runx2 and Smad1. Our findings shed light on the vital role of TRIB3 as a scaffold in ER stress and vascular calcification and offer a potential therapeutic option for CKD.
Collapse
Affiliation(s)
- Yihui Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Chang Ma
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yanan Sheng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shanying Huang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Huaibing Sun
- Department of Organ Transplantation, Qilu Hospital, and
| | - Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhihao Wang
- Department of Geriatric Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Feng Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital, Jinan, Shandong, China
| | - Fangfang Chen
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Chen Li
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Haipeng Guo
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Mengxiong Tang
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, China
| | - Fangqiang Song
- Department of Critical Care Medicine, Affiliated Tengzhou Hospital of Xuzhou Medical University/Tengzhou Central People’s Hospital, Shandong, China
| | - Hao Wang
- Department of Critical Care Medicine, Qilu Hospital, Innovation Research Center for Sepsis and Multiple Organ Injury, Shandong University, Jinan, China
| | - Ming Zhong
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
33
|
Kim KA, Jung H, Kim M, Lee S, Lee D, Han D, Chang H, Choi S, Sung J, Chun EJ. The Role of Serum Calcium Levels in the Progression of Arterial Stiffness: Cross-Sectional and Longitudinal Analyses in a Multicenter Cohort. J Clin Hypertens (Greenwich) 2025; 27:e70055. [PMID: 40270347 PMCID: PMC12018888 DOI: 10.1111/jch.70055] [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: 01/22/2025] [Revised: 03/14/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
Increased arterial stiffness measured by brachial-ankle pulse wave velocity (baPWV) is a well-known risk factor for hypertension and future cardiovascular events. The relationship between serum calcium levels and increased arterial stiffness is not well understood. Individuals undergoing baPWV measurements as part of a generalized health examination, with normal serum calcium (8.5-10.5 mg/dL) and phosphate levels and no significant renal dysfunction, were selected from the Korea Initiatives on Coronary Artery (KOICA) registry. The cross-sectional relationship between serum calcium levels and baPWV, and the longitudinal effect of baseline serum calcium levels on baPWV progression in those with repeated baPWV measurements, were analyzed using multivariable regression models. A total of 9150 individuals with baseline baPWV and 2329 individuals (5451 PWV measurements) with follow-up baPWV were selected for cross-sectional and longitudinal analyses, respectively. After adjustment for confounders, higher serum calcium levels were associated with increased baseline baPWV (β-coefficient per 1 mg/dL increase, 19.61; 95% CI 7.77-31.45; p = 0.001). Higher serum calcium was also independently associated with a greater annualized baPWV progression rate longitudinally (β-coefficient per 1 mg/dL increase, 5.17; 95% CI, 1.82-8.67; p = 0.004). Subgroup analysis showed that the effect of serum calcium on baPWV progression had a significant interaction with baseline baPWV, systolic blood pressure, and the presence of diabetes (interaction p < 0.001). In conclusion, higher serum calcium levels within the normal range were associated with faster arterial stiffness progression measured by baPWV. Further studies are required to explore the potential for modulating calcium metabolism to slow arterial stiffness progression.
Collapse
Affiliation(s)
- Kyung An Kim
- Department of Internal MedicineDivision of CardiologyIncheon St. Mary's HospitalThe Catholic University of KoreaIncheonRepublic of Korea
- Catholic Research Institute for Intractable Cardiovascular DiseaseCollege of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - Hae‐Ok Jung
- Department of Internal MedicineDivision of CardiologySeoul St. Mary's HospitalThe Catholic University of KoreaSeoulRepublic of Korea
| | - Mi‐Jeong Kim
- Department of Internal MedicineDivision of CardiologyIncheon St. Mary's HospitalThe Catholic University of KoreaIncheonRepublic of Korea
- Catholic Research Institute for Intractable Cardiovascular DiseaseCollege of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
| | - So‐Young Lee
- Catholic Research Institute for Intractable Cardiovascular DiseaseCollege of MedicineThe Catholic University of KoreaSeoulRepublic of Korea
- Department of Internal MedicineDivision of CardiologySeoul St. Mary's HospitalThe Catholic University of KoreaSeoulRepublic of Korea
| | - Dong‐Hyeon Lee
- Health Promotion CenterSeoul St. Mary's HospitalThe Catholic University of KoreaSeoulRepublic of Korea
| | - Donghee Han
- Department of ImagingCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Hyuk‐Jae Chang
- Division of CardiologyYonsei Cardiovascular CenterYonsei University Health SystemSeoulRepublic of Korea
| | - Su‐Yeon Choi
- Division of CardiologySeoul National University Hospital Healthcare System Gangnam CenterSeoulRepublic of Korea
| | - Jidong Sung
- Division of CardiologyHeart Stroke & Vascular InstituteSamsung Medical CenterSeoulRepublic of Korea
| | - Eun Ju Chun
- Division of RadiologySeoul National University Bundang HospitalSeongnamRepublic of Korea
| |
Collapse
|
34
|
Naldi L, Peri A, Fibbi B. Apelin/APJ: Another Player in the Cancer Biology Network. Int J Mol Sci 2025; 26:2986. [PMID: 40243599 PMCID: PMC11988549 DOI: 10.3390/ijms26072986] [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: 12/30/2024] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The apelinergic system exerts multiple biological activities in human pathologies, including cancer. Overactivation of apelin/APJ, which has been detected in many malignant tumors, and the strong correlation with progression-free and overall survival, suggested the role of an oncogene for the apelin gene. Emerging evidence sheds new light on the effects of apelin on cellular functions and homeostasis in cancer cells and supports a direct role for this pathway on different hallmarks of cancer: "sustaining proliferative signaling", "resisting cell death", "activating invasion and metastasis", "inducing/accessing vasculature", "reprogramming cellular metabolism", "avoiding immune destruction" and "tumor-promoting inflammation", and "enabling replicative immortality". This article reviews the currently available literature on the intracellular processes regulated by apelin/APJ, focusing on those pathways correlated with tumor development and progression. Furthermore, the association between the activity of the apelinergic axis and the resistance of cancer cells to oncologic treatments (chemotherapy, immunotherapy, radiation) suggests apelin/APJ as a possible target to potentiate traditional therapies, as well as to develop diagnostic and prognostic applications. This issue will be also covered in the review.
Collapse
Affiliation(s)
- Laura Naldi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Alessandro Peri
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| | - Benedetta Fibbi
- “Pituitary Diseases and Sodium Alterations” Unit, AOU Careggi, 50139 Florence, Italy; (L.N.); (B.F.)
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50139 Florence, Italy
| |
Collapse
|
35
|
Wei G, Chen R, Liu S, Cai S, Feng Z. Telomere Length as Both Cause and Consequence in Type 1 Diabetes: Evidence from Bidirectional Mendelian Randomization. Biomedicines 2025; 13:774. [PMID: 40299325 PMCID: PMC12024553 DOI: 10.3390/biomedicines13040774] [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: 02/26/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Diabetes is the most prevalent metabolic disease globally, characterized by dysregulated glucose control and accompanied by multiple refractory complications. As a critical marker of cellular homeostasis, telomere length (TL) may be associated with the progression of diabetes. However, the causal relationship between diabetes and TL remains unclear, particularly whether cellular homeostasis imbalance acts as a consequence of diabetic complications or a precipitating factor in disease development. Methods: We performed a bidirectional Mendelian randomization (MR) analysis using genome-wide association study (GWAS) data. Following the three core assumptions of MR analysis, we conducted quality control on all instrumental variables to ensure methodological rigor. The inverse variance weighted (IVW) method served as the primary analytical method, supplemented by additional MR methods to evaluate the significance of the results. Furthermore, we performed sensitivity analyses to ensure the reliability and robustness of the findings. Results: Forward analysis revealed that shortened TL significantly increases the risk of broadly defined Type 1 diabetes (T1D) and unspecified types of diabetes (p < 0.05). Additionally, we identified a positive causal relationship between TL and several diabetes-related complications, including co-morbidities, diabetic nephropathy, and diabetic ketoacidosis (p < 0.05). Interestingly, the reverse analysis demonstrated a positive causal effect of T1D and its complications on TL (p < 0.05); however, this effect disappeared after adjusting for insulin use (p > 0.05). Conclusions: Bidirectional MR analyses revealed a complex relationship between TL and T1D, where shortened telomeres increase T1D risk while T1D itself may trigger compensatory mechanisms affecting telomere maintenance, with insulin playing a crucial regulatory role in this relationship. These findings suggest telomere biology may be fundamentally involved in T1D pathogenesis and could inform future therapeutic approaches.
Collapse
Affiliation(s)
- Guanping Wei
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Ruiping Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Shupeng Liu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China;
| | - Shenhua Cai
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhijun Feng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China;
| |
Collapse
|
36
|
Cheng Y, Zheng G, Huang H, Ni J, Zhao Y, Sun Y, Chang Y, Liu S, He F, Li D, Guo Y, Miao Y, Xu M, Wang D, Zhang Y, Hua Y, Yang S, Fan G, Ma C. GLSP mitigates vascular aging by promoting Sirt7-mediated Keap1 deacetylation and Keap1-Nrf2 dissociation. Theranostics 2025; 15:4345-4367. [PMID: 40225574 PMCID: PMC11984382 DOI: 10.7150/thno.110324] [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: 01/12/2025] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Background and Purpose: Vascular aging is a prior marker of human aging and a significant contributor to atherosclerosis and vascular calcification. However, there are limited pharmacological options available to mitigate vascular aging. Thus, understanding the mechanisms underlying vascular aging and age-related atherosclerosis and vascular calcification is crucial. This study investigates the targets of vascular aging and elucidates the role and mechanisms of Ganoderma lucidum spore powder (GLSP) in mitigating vascular aging and aging-associated atherosclerosis as well as vascular calcification. Methods: The anti-vascular aging effects of GLSP was determined in aged C57BL/6J mice and the targets of GLSP was identified through transcriptome sequencing. Additionally, the protective effects of GLSP on the aged vasculature were assessed by examining atherosclerosis in apoE-/- mice and vascular calcification in VD3 and nicotine-induced mice. In vitro, the protective effects of GLSP triterpenes against vascular aging and calcification was determined in vascular smooth muscle cells (VSMCs). Results: GLSP exerted anti-vascular aging effects by regulating the cell cycle and senescence-associated secretory phenotype (SASP), mitigating DNA damage, reducing oxidative stress, improving mitochondrial function and modulating metabolic levels. Furthermore, GLSP improved vascular aging-associated atherosclerosis and vascular calcification in vivo. Mechanistically, RNA sequencing revealed an upregulation of Sirt7 expression after GLSP treatment. Sirt7 inhibitor exacerbated VSMCs senescence and calcification in senescent VSMCs and abolished the anti-senescence and the inhibitory effect of GLSP triterpenes on VSMCs senescence and calcification. Innovatively, we found that Sirt7 interacted with Keap1 and facilitated Keap1 deacetylation, which promoted Keap1-Nrf2 dissociation and consequently enhanced Nrf2 nuclear translocation and activation. Conclusion: GLSP alleviates vascular aging by exerting antioxidant effects through the activation of the Sirt7-Nrf2 axis, providing a promising new strategy for delaying vascular aging, atherosclerosis and vascular calcification.
Collapse
Affiliation(s)
- Yanfei Cheng
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Guobin Zheng
- NHC Key Lab of Hormones and Development and Tianjin Key Lab of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin 300134, China
| | - Heming Huang
- Department of Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong 518020, China
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yun Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuting Sun
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yingxin Chang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shangjing Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Feng He
- Culture and Industry Research Center of Li Shizhen Traditional Chinese Medicine, Li Shizhen College of Traditional Chinese Medicine, Huanggang Normal University, Huanggang, 438000, China
| | - Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengxin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dongyue Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunsha Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunqing Hua
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Shu Yang
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
37
|
He J, Wan Y, Fan X, Yu H, Qin Y, Su J, Lu Y, Pan E, Hang D, Shen C, Zhou J, Wu M. Associations between kidney function with all-cause and cause-specific mortality in type 2 diabetes mellitus patients: a prospective cohort study in China. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:77. [PMID: 40083037 PMCID: PMC11907967 DOI: 10.1186/s41043-025-00809-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Abnormal kidney function is associated with adverse outcomes in patients with type 2 diabetes mellitus (T2DM). However, the evidence between kidney function and mortality among Chinese patients with T2DM were still limited. METHODS This cohort study included 19,919 participants with baseline T2DM from 2013 to 2014 in Jiangsu, China. Serum estimated glomerular filtration rate (eGFR), urea, and uric acid were measured at baseline, and Cox regression models were used to evaluate hazard ratios (HRs) and 95% confidential intervals (95%CIs) of all-cause and cause-specific mortality. Restricted cubic splines were used to analyze dose-response relationships, and we explored the best cut-off values by receiver operating characteristic (ROC) curves. RESULTS During a median follow-up of 9.77 years, 4,428 deaths occurred, including 1,542 (34.8%) due to cardiovascular disease (CVD), and 1,074 (24.3%) due to cancer. Compared to lowest quintile level (Q1), the highest quintile (Q5) of eGFR was negatively associated with all-cause (HR = 0.67, 95%CI: 0.58-0.77) and CVD mortality (HR = 0.57, 95%CI = 0.44-0.75). The higher levels of urea and uric acid were positively associated with all-cause mortality (Q5 vs. Q1: HR = 1.27, 95%CI: 1.16-1.39; HR = 1.21, 95%CI: 1.10-1.34), with an overall "U-shaped" dose-response relationships. Moreover, higher urea was negatively associated with cancer mortality (Q5 vs. Q1: HR = 0.79, 95%CI: 0.66-0.95). The best cut-off values with all-cause mortality were 88.50 ml/min/1.73m2, 6.95 mmol/L and 342.50 µmol/L for eGFR, urea, and uric acid, respectively. CONCLUSION We found abnormal kidney function was associated with mortality among people with T2DM. More clinical researches are needed to validate the effects and cut-off values of kidney function on mortality risk for T2DM prevention and management.
Collapse
Affiliation(s)
- Jialiu He
- Department of Epidemiology and Health Statistics, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Ya'nan Wan
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Xikang Fan
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Hao Yu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Yu Qin
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Jian Su
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
- School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yan Lu
- Department of Chronic Disease Prevention and Control, Suzhou City Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Enchun Pan
- Department of Chronic Disease Prevention and Control, Huai'an City Center for Disease Control and Prevention, Huai'an, 223021, China
| | - Dong Hang
- School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Chong Shen
- School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Jinyi Zhou
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China.
- School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Wu
- Department of Epidemiology and Health Statistics, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China.
| |
Collapse
|
38
|
Liu Z, Lv R, Guo H, Zhang B, Wang X, Qiang P, Xiong Y, Chang Y, Peng Y, Hao J, Wang X, Shimosawa T, Xu Q, Yang F. Proliferation of renal macrophage via MR/CSF1 pathway induced with aldosterone and inhibited by esaxerenone. Int Immunopharmacol 2025; 149:114208. [PMID: 39923576 DOI: 10.1016/j.intimp.2025.114208] [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: 01/07/2025] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/11/2025]
Abstract
Macrophage proliferation plays a critical role in kidney injury and repair, but due to their high plasticity and heterogeneity, the origins and subtypes of these proliferating cells remain unclear. This study investigates aldosterone-induced proliferation of renal macrophages, focusing on their origins, subtypes, and regulatory mechanisms using immunofluorescence, flow cytometry, and single-cell sequencing. The findings suggest that both resident and infiltrating macrophages proliferate in response to aldosterone, a significant proportion of which are renal resident macrophages, predominantly of the M1 subtype. The study also identifies the mineralocorticoid receptor/colony stimulation factor-1 (MR/CSF1) pathway as a key regulator of this process. Inhibition of this pathway through antagonists and inhibitors reduces macrophage proliferation and kidney damage, suggesting that targeting MR/CSF1 could be therapeutic against aldosterone-induced renal damage and inflammation.
Collapse
Affiliation(s)
- Ziqian Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ruyan Lv
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haixia Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Boya Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Xuan Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Panpan Qiang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yunzhao Xiong
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yi Chang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yunsong Peng
- Nephrology Department, Shijiazhuang Hospital of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050000, China
| | - Juan Hao
- Nephrology Department, Shijiazhuang Hospital of Traditional Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050000, China
| | - Xiangting Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita 286-8686, Japan
| | - Qingyou Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| | - Fan Yang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China; Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China.
| |
Collapse
|
39
|
Chen Y, Xu F, Chen F, Li S, Wu M, Chen S, Chen J, Yang Z, Sun Z, Chen Z. Hydrogen sulfide-mediated inhibition of ROCK 2 exerts a vasoprotective effect on ischemic brain injury. Am J Physiol Cell Physiol 2025; 328:C986-C1000. [PMID: 39716722 DOI: 10.1152/ajpcell.00708.2024] [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: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
As a gas molecule, hydrogen sulfide (H2S) exerts neuroprotective effects. Despite its recognized importance, there remains a need for a deeper understanding of H2S's impact on vascular smooth muscle cells and its role in ischemic brain injury. This study employs encompassing cultured primary cerebral vascular smooth muscle cells, oxygen-glucose deprivation/reoxygenation model, in vitro vascular tone assessments, in vivo middle cerebral artery occlusion and reperfusion experimentation in male rats, and the utilization of Rho-associated coiled-coil containing protein kinase 2 (ROCK2) knockout, to unravel the intricate relationship between H2S and cerebrovascular diastolic function. Our findings show that RhoA activation induces heightened vascular smooth muscle cell (VSMC) contraction, whereas the introduction of exogenous H2S mitigates the relaxant effect of the middle cerebral artery in rats through the downregulation of both ROCK1 and ROCK2, with ROCK2 exhibiting a more pronounced effect. Correspondingly, the attenuation of ROCK2 expression yields a more substantial reduction in the protective impact of H2S on cerebral blood flow, as well as learning and memory ability in ischemic injury, compared with the decrease in ROCK1 expression. Moreover, we demonstrate that H2S effectively mitigates the damage induced by oxygen-glucose deprivation/reoxygenation in male mouse primary vascular smooth muscle cells. This effect is characterized by enhanced cell proliferation, reduced lactate dehydrogenase leakage, elevated superoxide dismutase activity, and inhibited apoptosis. Notably, this protective effect is markedly diminished in cells derived from ROCK2 knockout mice. Our study reveals that H2S can relax cerebral vascular smooth muscle and ameliorate ischemic stroke injury by inhibiting the ROCK, with a particular emphasis on the role of ROCK2.NEW & NOTEWORTHY This study employs a diverse array of methods; our collective findings indicate that H2S safeguards against ischemic brain injury by inhibiting ROCK activity, thereby promoting relaxation of cerebral smooth muscle and mitigating the impairment of cerebral smooth muscle cell function caused by oxygen-glucose deprivation/reoxygenation. In addition, our data underscore the critical role of ROCK2 in mediating the cerebral protective effects of H2S, surpassing that of ROCK1.
Collapse
MESH Headings
- Animals
- rho-Associated Kinases/metabolism
- rho-Associated Kinases/genetics
- rho-Associated Kinases/antagonists & inhibitors
- Hydrogen Sulfide/pharmacology
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Rats
- Rats, Sprague-Dawley
- Mice
- Brain Ischemia/pathology
- Brain Ischemia/drug therapy
- Brain Ischemia/enzymology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Mice, Knockout
- Cerebrovascular Circulation/drug effects
- Infarction, Middle Cerebral Artery/enzymology
- Infarction, Middle Cerebral Artery/pathology
- Neuroprotective Agents/pharmacology
- Mice, Inbred C57BL
- Glucose/deficiency
- Cells, Cultured
- Reperfusion Injury
Collapse
Affiliation(s)
- Ye Chen
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Fangfang Xu
- Department of Phase I Clinical Trials Laboratory, The First Affiliated of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Fang Chen
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Shuaishuai Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, People's Republic of China
| | - Miao Wu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| | - Shuo Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| | - Jinhua Chen
- Department of Clinical Central Laboratory, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| | - Zhaoyi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, People's Republic of China
- Department of Phase I Clinical Trials Laboratory, The First Affiliated of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, People's Republic of China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
40
|
Wang Z, Liu J, Chen Y, Tang Y, Chen T, Zhou C, Wang S, Chang R, Chen Z, Yang W, Guo Z, Chen T. From physiology to pathology: Emerging roles of GPER in cardiovascular disease. Pharmacol Ther 2025; 267:108801. [PMID: 39889969 DOI: 10.1016/j.pharmthera.2025.108801] [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: 08/30/2024] [Revised: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Cardiovascular diseases (CVDs) are among the leading causes of death globally and pose a significant threat to public health. Factors such as prolonged high cholesterol levels, diabetes, smoking, unhealthy diet, and genetic predisposition could contribute to the occurrence and development of CVDs. Common CVDs include hypertension (HTN), atherosclerosis (AS), myocardial infarction (MI), myocardial ischemia-reperfusion injury (MIRI), heart failure (HF) and arrhythmia. Estrogen is recognized for its cardiovascular protective effects, resulting in lower incidence and mortality rates of CVDs in premenopausal women compared to men. The G protein-coupled estrogen receptor (GPER), a G protein-coupled receptor with a seven-transmembrane structure, exhibits unique structural characteristics and widespread tissue distribution. GPER activates intracellular signaling pathways through its interaction with G proteins, mediating estrogen's biological effects and participating in the regulation of cardiovascular function, metabolic balance, and nervous system. Although recent research has highlighted the significant role of GPER in the cardiovascular system, its specific mechanisms remain unclear. Therefore, this review summarizes the latest research on GPER in CVDs, including its fundamental characteristics, physiological functions in the cardiovascular system, and its roles and potential therapeutic applications in common CVDs such as HTN, AS, MI, MIRI, HF and arrhythmia. Exploring GPER's positive effects on cardiovascular health will provide new strategies and research directions for the treatment of CVDs.
Collapse
Affiliation(s)
- Zixuan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junren Liu
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ying Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yi Tang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ting Chen
- Hunan University of Chinese Medicine, The College of Acupuncture & Moxibustion and Tuina, Changsha 410208, China
| | - Chang Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shuo Wang
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617.China
| | - Ranbo Chang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhongshuai Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Wenqing Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhen Guo
- Hunan Provincial Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha 410219, China; Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China; Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha 410219, China
| | - Ting Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China; Hunan Provincial Key Laboratory of Traditional Chinese Medicine Powder and Innovative drug Research, Changsha 410208, China.
| |
Collapse
|
41
|
Whitehead M, Faleeva M, Oexner R, Cox S, Schmidt L, Mayr M, Shanahan CM. ECM Modifications Driven by Age and Metabolic Stress Directly Promote Vascular Smooth Muscle Cell Osteogenic Processes. Arterioscler Thromb Vasc Biol 2025; 45:424-442. [PMID: 39817328 PMCID: PMC11856005 DOI: 10.1161/atvbaha.124.321467] [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: 07/02/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND The ECM (extracellular matrix) provides the microenvironmental niche sensed by resident vascular smooth muscle cells (VSMCs). Aging and disease are associated with dramatic changes in ECM composition and properties; however, their impact on VSMC phenotype remains poorly studied. METHODS Here, we describe a novel in vitro model system that utilizes endogenous ECM to study how modifications associated with age and metabolic disease impact VSMC phenotype. ECM was synthesized using primary human VSMCs and modified during culture or after decellularization. Integrity, stiffness, and composition of the ECM was measured using superresolution microscopy, atomic force microscopy, and proteomics, respectively. VSMCs reseeded onto the modified ECM were analyzed for viability and osteogenic differentiation. RESULTS ECMs produced in response to mineral stress showed extracellular vesicle-mediated hydroxyapatite deposition and sequential changes in collagen composition and ECM properties. VSMCs seeded onto the calcified ECM exhibited increased extracellular vesicle release and Runx2 (Runt-related transcription factor 2)-mediated osteogenic gene expression due to the uptake of hydroxyapatite, which led to increased reactive oxygen species and the induction of DNA damage signaling. VSMCs seeded onto the nonmineralized, senescent ECM also exhibited increased Runx2-mediated osteogenic gene expression and accelerated calcification. In contrast, glycated ECM specifically induced increased ALP (alkaline phosphatase) activity, and this was dependent on RAGE (receptor for advanced glycation end products) signaling with both ALP and RAGE receptor inhibition attenuating calcification. CONCLUSIONS ECM modifications associated with aging and metabolic disease can directly induce osteogenic differentiation of VSMCs via distinct mechanisms and without the need for additional stimuli. This highlights the importance of the ECM microenvironment as a key driver of phenotypic modulation acting to accelerate age-associated vascular pathologies and provides a novel model system to study the mechanisms of calcification.
Collapse
Affiliation(s)
- Meredith Whitehead
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.W., M.F., R.O., L.S., M.M., C.M.S.), King’s College London, United Kingdom
| | - Maria Faleeva
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.W., M.F., R.O., L.S., M.M., C.M.S.), King’s College London, United Kingdom
| | - Rafael Oexner
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.W., M.F., R.O., L.S., M.M., C.M.S.), King’s College London, United Kingdom
| | - Susan Cox
- Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine (S.C.), King’s College London, United Kingdom
| | - Lukas Schmidt
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.W., M.F., R.O., L.S., M.M., C.M.S.), King’s College London, United Kingdom
| | - Manuel Mayr
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.W., M.F., R.O., L.S., M.M., C.M.S.), King’s College London, United Kingdom
| | - Catherine M. Shanahan
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.W., M.F., R.O., L.S., M.M., C.M.S.), King’s College London, United Kingdom
| |
Collapse
|
42
|
Sheng Y, Wei H, Lu S, Hong W. Alternative splicing of vascular calcification: Insights, opportunities, and challenges. Cell Signal 2025; 127:111626. [PMID: 39875046 DOI: 10.1016/j.cellsig.2025.111626] [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: 11/30/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Vascular calcification(VC) significantly increases the risk of cardiovascular events, leading to thickening of the myocardium and arteries, coronary heart disease, heart failure, and potentially triggering myocardial infarction and sudden cardiac death. Although VC is a reversible process, there are currently no methods or medications in clinical practice that can completely reverse or cure it. The current treatment strategies primarily focus on slowing the progression of VC and exploring new diagnostic and therapeutic approaches, making the identification of early diagnostic markers for VC particularly important. Alternative splicing(AS)has extensive potential in clinical applications as a biomarker, including in disease diagnosis and therapeutic targeting. This article provides an overview of the roles played by different isoforms of biomarkers in VC, with the aim of offering insights for early diagnosis and disease monitoring of VC.
Collapse
Affiliation(s)
- Yingkun Sheng
- Xingzhi College, Zhejiang Normal University, Jinhua 321100, China
| | - Hewen Wei
- Jinhua Key laboratory of Quality Evaluation and Standard Research of Traditional Chinese Medicine, Jinhua Food and Drug Inspection Research Institute, Jinhua 321000, China
| | - Shengmin Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Zhejiang Key Laboratory of Intelligent Food Logistic and Processing, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Weiling Hong
- Jinhua Advanced Research Institute, Jinhua 321019, China.
| |
Collapse
|
43
|
Mueller M, Charwat-Resl S, Schulze-Bauer HS, Nguyen C, Niessner A, Bartko PE, Giurgea GA, Zehetmayer S, Willfort-Ehringer A, Kautzky-Willer A, Koppensteiner R, Schlager O. Vascular strain of the aorta and peripheral arteries in patients with type 1 diabetes mellitus in comparison with healthy controls. Atherosclerosis 2025; 402:119106. [PMID: 39914324 DOI: 10.1016/j.atherosclerosis.2025.119106] [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: 01/25/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND AND AIMS Local variations of vascular strain may be related to the development of atherosclerotic lesions. Whether vascular strain of peripheral arteries without manifest atherosclerosis is affected by diabetes mellitus is not known. This study aimed to assess vascular strain of peripheral arteries and the abdominal aorta of young patients with type 1 diabetes mellitus (T1DM) in comparison with healthy controls. METHODS Vascular strain was determined by sonographic speckle tracking of the common carotid arteries (CCA), the abdominal aorta (AA), the common femoral arteries (CFA), and the popliteal arteries (PA) of patients with type 1 diabetes mellitus but without atherosclerosis and in controls. RESULTS Thirty-three patients with T1DM (mean age 33 years, SD 11.6) and 34 controls (mean age 24 years, SD 5.4) underwent sonographic determination of vascular strain in the CCA, AA, CFA, and PA. In total 4221 clips were processed for the analysis of vascular strain. To account for a potential impact of age on vascular strain, an age-matched model containing 18 patients with T1DM and 33 controls was used for the final analysis. In this age-matched model T1DM was independently related to vascular strain in the CFA (r = -0.48; p = 0.04), while no association was observed at other sites of the vascular tree. Intima media thickness was negatively correlated with vascular strain in the AA (r = -15.11) and the PA (r = -12.76, both p < 0.05). CONCLUSION T1DM appears to have an early impact on vascular strain of the CFA. Longitudinal observational studies are needed to further asses the course of these changes over time and to determine the impact of these early findings on patients' cardiovascular risk.
Collapse
Affiliation(s)
- Markus Mueller
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Austria.
| | - Silvia Charwat-Resl
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Austria
| | | | - Christina Nguyen
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Austria
| | - Alexander Niessner
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Austria
| | - Philipp E Bartko
- Division of Cardiology, Department of Medicine II, Medical University of Vienna, Austria
| | | | - Sonja Zehetmayer
- Center for Medical Data Science, Medical University of Vienna, Austria
| | | | | | - Renate Koppensteiner
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Austria
| | - Oliver Schlager
- Division of Angiology, Department of Medicine II, Medical University of Vienna, Austria
| |
Collapse
|
44
|
Zhang R, Zhou D. Effect of changes trajectory of serum phosphate levels on the 28-day mortality risk in patients with sepsis: a retrospective cohort study from the MIMIC-IV database. BMC Infect Dis 2025; 25:245. [PMID: 39984839 PMCID: PMC11844063 DOI: 10.1186/s12879-025-10547-9] [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: 07/16/2024] [Accepted: 01/23/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Serum phosphate levels have been reported to be linked to the prognosis in critically ill patients. The purpose of this study was to analyze the impact of the trajectory of changes in serum phosphate levels on the short-term mortality risk in patients with sepsis. METHODS This retrospective cohort study used data on patients with sepsis from the 2008-2019 Medical Information Mart for Intensive Care IV (MIMIC-IV) database. Serum phosphate level trajectories were constructed using a latent growth mixture modeling (LGMM) based on four measurements of serum phosphate at six-hour intervals within 24 h of admission to the intensive care unit (ICU). The relationship between serum phosphate levels at ICU admission and serum phosphate level trajectories and the risk of 28-day mortality in patients with sepsis was analyzed using Cox regression models, and hazard ratio (HR) and 95% confidence interval (CI) were calculated. RESULTS Of these 1,703 patients with sepsis included, 566 (33.24%) died within 28 days. The median serum phosphate levels of the patients were 4.10 (3.00, 5.50) mg/dL. Four serum phosphate level trajectories were classified: normal-level-steady trend (trajectory 1), high-level-steady trend (trajectory 2), high-level-decreasing trend (trajectory 3), and high-level-increasing trend (trajectory 4). High serum phosphate levels at admission were associated with a higher risk of 28-day mortality (HR = 1.05, 95%CI: 1.01-1.09) in patients with sepsis. For trajectories, trajectory 2 (HR = 1.27, 95%CI: 1.05-1.54) related to an increased risk of 28-day mortality compared with trajectory 1, whereas trajectory 4 (HR = 1.69, 95%CI: 0.99-2.91, P = 0.056) may be related. There was no significant difference in 28-day mortality between patients on trajectory 3 and trajectory 1 (P = 0.280). Subgroup analyses demonstrated that patients with trajectory 2 were linked to a higher risk of 28-day mortality in different population subgroups (P < 0.05). CONCLUSION Stable trajectories of high serum phosphate levels are an important risk factor for short-term mortality in patients with sepsis.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Emergency, Wuxi No.2 Chinese Medicine Hospital, No. 390 Xincheng Road, Binhu District, Wuxi, 214121, Jiangsu, P.R. China
| | - Dingxing Zhou
- Department of Emergency, Wuxi No.2 Chinese Medicine Hospital, No. 390 Xincheng Road, Binhu District, Wuxi, 214121, Jiangsu, P.R. China.
| |
Collapse
|
45
|
Ho CY, Wu MY, Thammaphet J, Ahmad S, Ho C.S. J, Draganova L, Anderson G, Jonnalagadda US, Hayward R, Shroff R, Wen WTL, Verhulst A, Foo RSY, Shanahan CM. Mineral Stress Drives Loss of Heterochromatin: An Early Harbinger of Vascular Inflammaging and Calcification. Circ Res 2025; 136:379-399. [PMID: 39840455 PMCID: PMC11825498 DOI: 10.1161/circresaha.124.325374] [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: 08/08/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Vascular calcification is a detrimental aging pathology markedly accelerated in patients with chronic kidney disease. PLA (prelamin A) is a biomarker of vascular smooth muscle cell aging that accelerates calcification however the mechanisms remain undefined. METHODS Vascular smooth muscle cells were transduced with PLA using an adenoviral vector and epigenetic modifications were monitored using immunofluorescence and targeted polymerase chain reaction array. Epigenetic findings were verified in vivo using immunohistochemistry in human vessels, in a mouse model of inducible prelamin A expression, and in a rat model of chronic kidney disease-induced calcification. Transcriptomic and chromatin immunoprecipitation followed by sequencing analyses were used to identify gene targets impacted by changes in the epigenetic landscape. Molecular tools and antibody arrays were used to monitor the effects of mineral dysregulation on heterochromatin, inflammation, aging, and calcification. RESULTS Here, we report that depletion of the repressive heterochromatin marks, H3K9me3 (histone H3, lysine 9, trimethylation) and H3K27me3 (histone H3, lysine 27,trimethylation), is an early hallmark of vascular aging induced by both nuclear lamina dysfunction and dysregulated mineral metabolism, which act to modulate the expression of key epigenetic writers and erasers. Global analysis of H3K9me3 and H3K27me3 marks and pathway analysis revealed deregulation of insulin signaling and autophagy pathways as well as cross-talking DNA damage and NF-κB (nuclear factor κB) inflammatory pathways consistent with early activation of the senescence-associated secretory phenotype. Expression of PLA in vivo induced loss of heterochromatin and promoted inflammation and osteogenic differentiation which preceded aging indices, such as DNA damage and senescence. Vessels from children on dialysis and rats with chronic kidney disease showed prelamin A accumulation and accelerated loss of heterochromatin before the onset of calcification. CONCLUSIONS Dysregulated mineral metabolism drives changes in the epigenetic landscape and nuclear lamina dysfunction that together promote early induction of inflammaging pathways priming the vasculature for downstream pathological change.
Collapse
MESH Headings
- Animals
- Heterochromatin/metabolism
- Heterochromatin/pathology
- Heterochromatin/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Humans
- Epigenesis, Genetic
- Rats
- Mice
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cellular Senescence
- Inflammation/metabolism
- Inflammation/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/genetics
- Histones/metabolism
- Mice, Inbred C57BL
- Cells, Cultured
- Disease Models, Animal
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Lamin Type A
Collapse
Affiliation(s)
- Chin Yee Ho
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Meng-Ying Wu
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Jirapath Thammaphet
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Sadia Ahmad
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - James Ho C.S.
- Nanyang Technological University, Singapore (J.H.C.S., U.S.J.)
| | - Lilia Draganova
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Grace Anderson
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | | | - Robert Hayward
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| | - Rukshana Shroff
- Nephrology Unit, Great Ormond Street Hospital and University College London Institute of Child Health, United Kingdom (R.S.)
| | - Wilson Tan Lek Wen
- Cardiovascular Disease Translational Research Programme, National University of Singapore Yong Loo Lin School of Medicine (W.T.L.W., R.F.)
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium (A.V.)
| | - Roger SY. Foo
- Cardiovascular Disease Translational Research Programme, National University of Singapore Yong Loo Lin School of Medicine (W.T.L.W., R.F.)
| | - Catherine M. Shanahan
- British Heart Foundation Centre for Research Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King’s College London, United Kingdom (C.Y.H., M.-Y.W., J.T., S.A., L.D., G.A., R.H., C.M.S.)
| |
Collapse
|
46
|
Tasouli-Drakou V, Ogurek I, Shaikh T, Ringor M, DiCaro MV, Lei K. Atherosclerosis: A Comprehensive Review of Molecular Factors and Mechanisms. Int J Mol Sci 2025; 26:1364. [PMID: 39941130 PMCID: PMC11818631 DOI: 10.3390/ijms26031364] [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: 01/13/2025] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Atherosclerosis, a condition characterized by the accumulation of lipids and a culprit behind cardiovascular events, has long been studied. However, in recent years, there has been an increase in interest in its initiation, with researchers shifting focus from traditional pathways involving the vascular infiltration of oxidized lipids and towards the novel presence of chronic inflammatory pathways. The accumulation of pro-inflammatory cytokines, in combination with the activation of transcription factors, creates a positive feedback loop that drives the creation and progression of atherosclerosis. From the upregulation of the nod-like receptor protein 3 (NLRP3) inflammasome and the Notch and Wnt pathways to the increased expression of VEGF-A and the downregulation of connexins Cx32, Cx37, and Cx40, these processes contribute further to endothelial dysfunction and plaque formation. Herein, we aim to provide insight into the molecular pathways and mechanisms implicated in the initiation and progression of atherosclerotic plaques, and to review the risk factors associated with their development.
Collapse
Affiliation(s)
- Vasiliki Tasouli-Drakou
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, NV 89106, USA; (I.O.); (T.S.); (M.R.); (M.V.D.)
| | - Ian Ogurek
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, NV 89106, USA; (I.O.); (T.S.); (M.R.); (M.V.D.)
| | - Taha Shaikh
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, NV 89106, USA; (I.O.); (T.S.); (M.R.); (M.V.D.)
| | - Marc Ringor
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, NV 89106, USA; (I.O.); (T.S.); (M.R.); (M.V.D.)
| | - Michael V. DiCaro
- Department of Internal Medicine, Kirk Kerkorian School of Medicine at the University of Nevada, Las Vegas, NV 89106, USA; (I.O.); (T.S.); (M.R.); (M.V.D.)
| | - KaChon Lei
- Department of Cardiovascular Medicine, University of Nevada, Las Vegas, NV 89106, USA;
| |
Collapse
|
47
|
Chen J, Yuan M, Wang J. Research progress of cysteine transporter SLC7A11 in endocrine and metabolic diseases. Mol Biol Rep 2025; 52:185. [PMID: 39899147 DOI: 10.1007/s11033-024-10193-5] [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: 09/29/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025]
Abstract
SLC7A11, often called xCT, belongs to the SLC family of transporters, which mediates the cellular influx of cystine and the efflux of glutamate. These transport processes are crucial for synthesizing GSH, enhancing the cell's ability to mitigate oxidative stress (OS). Emerging studies highlight the pivotal role of OS in triggering and exacerbating various metabolic and endocrine disorders, underlining the critical importance of regulating SLC7A11 expression levels. This study reviews the diverse roles of SLC7A11 in endocrine and metabolic diseases, examining its relationship with the metabolism of three key nutrients: proteins and amino acids, carbohydrates, and lipids. Additionally, the involvement of SLC7A11 in the onset and development of various common endocrine and metabolic disorders is analyzed. Additionally, it provides an overview of the current clinical and experimental use of SLC7A11 inhibitors and agonists. This review aims to offer insightful perspectives into the involvement of SLC7A11 in endocrine and metabolic pathologies and to foster the development of innovative therapeutic strategies that target SLC7A11.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Endocrinology and Metabolic Disease, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Mengzhu Yuan
- Department of Endocrinology and Metabolic Disease, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jianping Wang
- Department of Endocrinology and Metabolic Disease, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
- Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
48
|
Li Z, Wu H, Yao F, Li Y, Li Y, Xie SA, Yu F, Fu Y, Wang L, Zhou J, Kong W. Runx2-NLRP3 axis orchestrates matrix stiffness-evoked vascular smooth muscle cell inflammation. Am J Physiol Cell Physiol 2025; 328:C467-C482. [PMID: 39761974 DOI: 10.1152/ajpcell.00448.2024] [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: 07/02/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
Arterial stiffening is a hallmark of chronic kidney disease (CKD)-related cardiovascular events and is primarily attributed to the elevated matrix stiffness. Stiffened arteries are accompanied by low-grade inflammation, but the causal effects of matrix stiffness on inflammation remain unknown. For analysis of the relationship between arterial stiffness and vascular inflammation, pulse-wave velocity (PWV) and aortic inflammatory markers were analyzed in an adenine-induced mouse model of CKD in chronological order. Compared with their control littermates, mice with CKD showed elevated arterial stiffness at the early stage of disease progression, which preceded the onset of vascular inflammation. Correspondingly, the increase of matrix stiffness induced vascular smooth muscle cells (VSMCs) to transdifferentiate into an inflammatory phenotype, as indicated by the increased expression and secretion of MCP-1, IL-6, IL-1β, and IL-18. RNA-sequencing analysis of stiff matrix-cultured VSMCs and bioinformatics analysis with the ChIP-Atlas database revealed the potential involvement of the transcription factor Runx2. The expression and the nuclear localization of Runx2 were significantly increased in stiff matrix-cultured VSMCs. High-throughput ChIP-sequencing and promoter luciferase assays further revealed that NLRP3 was directly transcriptionally regulated by Runx2. The inhibition of Runx2 or NLRP3 inflammasome abrogated the proinflammatory effect of matrix stiffening on VSMCs. Together, these data revealed that arterial stiffness precedes vascular inflammatory responses in CKD mice and that the Runx2-NLRP3 axis orchestrates matrix stiffness and the VSMC inflammatory phenotype, which may contribute to the pathogenic role in arterial stiffness-related vascular inflammation and CKD-related cardiovascular complications.NEW & NOTEWORTHY As a hallmark of chronic kidney disease (CKD), arterial stiffening is related to increased vascular inflammation and cardiovascular morbidity, whereas the underlying mechanism is unclear. The study demonstrates that increased arterial stiffness precedes the onset of vascular inflammation, and matrix stiffness stimulates the transdifferentiation of vascular smooth muscle cells (VSMCs) to an inflammatory phenotype via activating Runx2-NLRP3 signaling, which provides novel insights into CKD-related cardiovascular disorder treatment.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Vascular Stiffness/physiology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Male
- Mice, Inbred C57BL
- Inflammation/metabolism
- Inflammation/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/chemically induced
- Signal Transduction
- Cells, Cultured
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Disease Models, Animal
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Fang Yao
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Si-An Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
49
|
Tariq H, Bukhari SZ, An R, Dong J, Ihsan A, Younis MR. Stem cell-derived exosome delivery systems for treating atherosclerosis: The new frontier of stem cell therapy. Mater Today Bio 2025; 30:101440. [PMID: 39866781 PMCID: PMC11758955 DOI: 10.1016/j.mtbio.2024.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide. As a chronic inflammatory disease with a complicated pathophysiology marked by abnormal lipid metabolism and arterial plaque formation, atherosclerosis is a major contributor to CVDs and can induce abrupt cardiac events. The discovery of exosomes' role in intercellular communication has sparked a great deal of interest in them recently. Exosomes are involved in strategic phases of the onset and development of atherosclerosis because they have been identified to control pathophysiologic pathways including inflammation, angiogenesis, or senescence. This review investigates the potential role of stem cell-derived exosomes in atherosclerosis management. We briefly introduced atherosclerosis and stem cell therapy including stem cell-derived exosomes. The biogenesis of exosomes along with their secretion and isolation have been elaborated. The design engineering of exosomes has been summarized to present how drug loading and surface modification with targeting ligands can improve the therapeutic and targeting capacity of exosomes, demonstrating atheroprotective action. Moreover, the mechanism of action (endothelial dysfunction, reduction of dyslipidemia, macrophage polarization, vascular calcification, and angiogenesis) of drug-loaded exosomes to treat atherosclerosis has been discussed in detail. In the end, a comparative and balanced viewpoint has been given regarding the current challenges and potential solutions to advance exosome engineering for cardiovascular therapeutic applications.
Collapse
Affiliation(s)
- Hassan Tariq
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| | - Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ruibing An
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Jian Dong
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Rizwan Younis
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
50
|
Ma H, Yu Y, Zhu Y, Wu H, Qiu H, Gu Y, Chen Q, Zuo C. Monitoring of microvascular calcification by time-resolved photoacoustic microscopy. PHOTOACOUSTICS 2025; 41:100664. [PMID: 39654983 PMCID: PMC11626619 DOI: 10.1016/j.pacs.2024.100664] [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: 08/24/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024]
Abstract
Monitoring of microvascular calcification (MC) is essential for the understanding of pathophysiological processes and the characterization of certain physiological states such as drug abuse, metabolic abnormality, and chronic nephrosis. In this work, we develop a novel and effective time-resolved photoacoustic microscopy (TR-PAM) technology, which can observe the obvious microvascular bio-elastic change in the development process of the MC owing to the calcium deposition along vascular walls.The feasibility of the TR-PAM imaging was validated using a group of agar phantoms and ex vivo tissues. Furthermore, MC pathological animal models were constructed and imaged in situ and in vivo by the TR-PAM to demonstrate its capability for the bio-mechanical monitoring and characterization of MC, and experimental results were consistent with the pathological knowledge. The feasibility study of monitoring MC by the TR-PAM proves that this technique has potential to be developed as a superficial microvascular bio-mechanical assessment method to supplement current clinical strategy for prediction and monitoring of some diseases.
Collapse
Affiliation(s)
- Haigang Ma
- Smart Computational Imaging Laboratory (SCILab), School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
- Smart Computational Imaging Research Institute (SCIRI) of Nanjing University of Science and Technology, Nanjing, Jiangsu 210019, China
- Jiangsu Key Laboratory of Spectral Imaging & Intelligent Sense, Nanjing, Jiangsu 210094, China
| | - Yinshi Yu
- Smart Computational Imaging Laboratory (SCILab), School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
- Smart Computational Imaging Research Institute (SCIRI) of Nanjing University of Science and Technology, Nanjing, Jiangsu 210019, China
- Jiangsu Key Laboratory of Spectral Imaging & Intelligent Sense, Nanjing, Jiangsu 210094, China
| | - Yahui Zhu
- Smart Computational Imaging Laboratory (SCILab), School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
- Smart Computational Imaging Research Institute (SCIRI) of Nanjing University of Science and Technology, Nanjing, Jiangsu 210019, China
- Jiangsu Key Laboratory of Spectral Imaging & Intelligent Sense, Nanjing, Jiangsu 210094, China
| | - Hongjun Wu
- Smart Computational Imaging Laboratory (SCILab), School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
- Smart Computational Imaging Research Institute (SCIRI) of Nanjing University of Science and Technology, Nanjing, Jiangsu 210019, China
- Jiangsu Key Laboratory of Spectral Imaging & Intelligent Sense, Nanjing, Jiangsu 210094, China
| | - Haixia Qiu
- Department of Laser medicine, the First Medical Center of PLA General Hospital, Beijing 100853, China
| | - Ying Gu
- Department of Laser medicine, the First Medical Center of PLA General Hospital, Beijing 100853, China
| | - Qian Chen
- Smart Computational Imaging Laboratory (SCILab), School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
- Jiangsu Key Laboratory of Spectral Imaging & Intelligent Sense, Nanjing, Jiangsu 210094, China
| | - Chao Zuo
- Smart Computational Imaging Laboratory (SCILab), School of Electronic and Optical Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu 210094, China
- Smart Computational Imaging Research Institute (SCIRI) of Nanjing University of Science and Technology, Nanjing, Jiangsu 210019, China
- Jiangsu Key Laboratory of Spectral Imaging & Intelligent Sense, Nanjing, Jiangsu 210094, China
| |
Collapse
|