1
|
Zeng H, Zhao W, Wang R, Wang X, Luo P, Zhang X, Zeng X. Reliability and validity of the Geriatric Self-Care Scale among Chinese older adults. Ann Med 2025; 57:2478480. [PMID: 40083155 PMCID: PMC11912289 DOI: 10.1080/07853890.2025.2478480] [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: 08/05/2024] [Revised: 09/27/2024] [Accepted: 02/19/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Although the Geriatric Self-Care Scale (GSS) has been widely used in community investigations in China, its reliability and validity have yet to be analyzed. OBJECTIVES This study aimed to examine the reliability and validity of the GSS in community-dwelling Chinese older adults. METHODS This prospective observational study included 29428 older adults. Content validity was analyzed using the Content Validity Index (CVI). Reliability analysis included internal consistency and test-retest reliability. Differences in participants who could live independently versus those who could not were analyzed for discriminant validity. The Barthel Index was used as the gold standard. Spearman's correlation and Receiver Operating Characteristic (ROC) analysis were used to assess convergent validity. RESULTS The CVI for the GSS was 0.920, with CVI values of 1.000, 0.900, 1.000, 0.800 and 0.900. The Cronbach's alpha for the GSS was 0.869 (for all participants) and 0.867 (for participants aged ≥ 65 years) and the item Cronbach's alpha coefficients were all >0.8. The Pearson and intraclass correlation coefficients for both the scale and each item were greater than 0.8. There were significant differences (p < 0.05) between participants who could live independently and those who could not. There were significant correlations (p < 0.05) between the GSS and Barthel Index for each item and the total score. ROC analysis revealed that all areas under the curve were greater than 0.8, with a sensitivity and specificity exceeding 0.8. CONCLUSIONS The GSS showed good reliability and validity among community-dwelling older adults in China.
Collapse
Affiliation(s)
- Hongji Zeng
- School of Public Health, Zhengzhou University, Zhengzhou, China
- Dysphagia Research Institution, Zhengzhou University, Zhengzhou, China
| | - Weijia Zhao
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Rui Wang
- School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xin'ao Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengchao Luo
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuyang Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xi Zeng
- Dysphagia Research Institution, Zhengzhou University, Zhengzhou, China
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Wang L, Song Y, Shu Y, Xue B, Yu F, Yin Y, Feng Z, Ma X, Yao Y, Pan Y, Jin S. CAVIN-2 positively correlates with diabetic PAD and promotes LDL transcytosis by inhibiting eNOS activation. Ann Med 2025; 57:2457526. [PMID: 39887709 PMCID: PMC11789226 DOI: 10.1080/07853890.2025.2457526] [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: 08/25/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVE Caveolae are closely linked to the onset and progression of atherosclerosis. The pivotal involvement of caveolin-1 (CAV1) within the caveolae in atherosclerosis development has been consistently supported. However, the potential contributions of additional caveolae proteins to atherosclerosis necessitate further exploration. Therefore, this research aimed to afford clinical evidence linking CAVIN-2 to diabetic peripheral artery disease (PAD) and its role in low-density lipoprotein (LDL) transcytosis. METHODS Blood samples were collected from a total of 115 participants, including 36 patients without diabetes (ND), 26 patients with type 2 diabetes mellitus (T2DM), and 53 patients with T2DM and PAD (DM-PAD). The plasma levels of CAV1, CAVIN-1, and CAVIN-2 were measured by ELISA. The correlation between CAV1, CAVIN-1, CAVIN-2, and diabetic PAD was examined using Spearman correlation analysis. The predictive effect of CAV1 and CAVIN-2 were analyzed by receiver operating characteristic (ROC) curves. Cellular experiments were used to investigate the effect and mechanism of CAVIN-2 on LDL transcytosis. RESULTS Elevated CAV1 and CAVIN-2 levels were observed in T2DM and DM-PAD groups, with a positive correlation to DM-PAD and PAD severity. Both CAV1 and CAVIN-2 emerged as predictors of DM-PAD. In vitro, CAVIN-2 knockdown decreased LDL transcytosis, while CAVIN-2 overexpression increased it. Additionally, CAVIN-2 was found to inhibit eNOS activation and nitric oxide (NO) production, thereby promoting LDL transcytosis and atherosclerosis progression. CONCLUSION CAVIN-2 was positively correlated with DM-PAD and promoted LDL transcytosis through the inhibition of eNOS activation, contributing to atherosclerosis development. This study provided clinical evidence linking CAVIN-2 to diabetic PAD and suggested its potential as a biomarker for disease progression.
Collapse
Affiliation(s)
- Li Wang
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Song
- Department of Endocrinology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Shu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Baorui Xue
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fangyang Yu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yao Yin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ziyun Feng
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiang Ma
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yulin Yao
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yangze Pan
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
3
|
Chriqui LE, Cavin S, Perentes JY. Dual implication of endothelial adhesion molecules in tumor progression and cancer immunity. Cell Adh Migr 2025; 19:2472308. [PMID: 40071851 PMCID: PMC11913389 DOI: 10.1080/19336918.2025.2472308] [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: 02/15/2024] [Revised: 10/16/2024] [Accepted: 01/19/2025] [Indexed: 03/19/2025] Open
Abstract
Adhesion molecules are proteins expressed at the surface of various cell types. Their main contribution to immunity is to allow the infiltration of immune cells in an inflamed site. In cancer, adhesion molecules have been shown to promote tumor dissemination favoring the development of metastasis. While adhesion molecule inhibition approaches were unsuccessful for cancer control, their importance for the generation of an immune response alone or in combination with immunotherapies has gained interest over the past years. Currently, the balance of adhesion molecules for tumor promotion/inhibition is unclear. Here we review the role of selectins, intercellular adhesion molecules (ICAM) and vascular cell adhesion molecules (VCAM) from the perspective of the dual contribution of adhesion molecules in tumor progression and immunity.
Collapse
Affiliation(s)
- Louis-Emmanuel Chriqui
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| | - Sabrina Cavin
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| | - Jean Yannis Perentes
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Shalaby YM, Nakhal MM, Afandi B, Al-Zohily B, Majed L, Kumar KK, Emerald BS, Sadek B, Akour A, Akawi N. Impact of sodium-glucose cotransporter-2 inhibitors on aging biomarkers and plasma ceramide levels in type 2 diabetes: beyond glycemic control. Ann Med 2025; 57:2496795. [PMID: 40289660 PMCID: PMC12039402 DOI: 10.1080/07853890.2025.2496795] [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: 01/19/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Aging is a complex biological process marked by the decline of physiological functions and heightened susceptibility to chronic illnesses, notably cardiometabolic disorders. Ceramides (Cer) are lipid derivatives linked to aging and metabolic diseases. Sodium-Glucose Cotransporter-2 inhibitors (SGLT2i), widely used in managing type 2 diabetes, have an unclear impact on aging biomarkers and Cer profiles. OBJECTIVE This study explored the association between SGLT2i use, plasma Cer levels (CerC16:0, CerC18:0, CerC22:0, CerC24:0, and CerC24:1), and aging biomarkers-Human Insulin-Like Growth Factor 1 (IGF-1), mammalian target of rapamycin (mTOR), 5-Methylcytosine (5MC), and Human H2AFX (Histone H2AX) in patients with type 2 diabetes mellitus (T2DM). METHODS In this retrospective study, 95 participants were divided into three groups: patients on SGLT2i (n = 34), patients on non-SGLT2i anti-diabetic treatments (n = 36), and healthy controls (n = 25). Plasma Cer and aging biomarkers were quantified using Liquid Chromatography with tandem mass spectrometry (LC-MS-MS) and ELISA, respectively. Principal component analysis (PCA) assessed group-based clustering, while ANCOVA evaluated group differences with confounder adjustment. RESULTS SGLT2i-treated patients showed significantly lower CerC16:0, CerC22:0, and CerC24:1 levels (p < 0.01) and decreased 5MC and H2AX (p < 0.05) compared to non-SGLT2i patients. IGF-1 was significantly elevated in the SGLT2i group (p < 0.01), suggesting a possible protective effect on metabolic health. PCA distinguished control from diabetic groups but revealed overlap between SGLT2i and non-SGLT2i groups. CONCLUSION Beyond glucose control, SGLT2i may improve plasma Cer and aging markers in diabetic patients, supporting their broader therapeutic potential in aging and age-related diseases. Further large-scale studies are warranted to confirm these effects and underlying mechanisms.
Collapse
Affiliation(s)
- Youssef M. Shalaby
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Mohammed Moutaz Nakhal
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Bachar Afandi
- Department of Endocrinology, Tawam Hospital, Al-Ain, UAE
- Department of Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Bashar Al-Zohily
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Lina Majed
- Department of Endocrinology, Tawam Hospital, Al-Ain, UAE
| | - Kukkala Kiran Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Bassem Sadek
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Amal Akour
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Nadia Akawi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| |
Collapse
|
5
|
Jin Z, Li X, Liu H, He T, Jiang W, Peng L, Wu X, Chen M, Fan Y, Lu Z, Fan D, Wang H. MEGF9 prevents lipopolysaccharide-induced cardiac dysfunction through activating AMPK pathway. Redox Rep 2025; 30:2435252. [PMID: 39737911 DOI: 10.1080/13510002.2024.2435252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVE Inflammation and oxidative damage play critical roles in the pathogenesis of sepsis-induced cardiac dysfunction. Multiple EGF-like domains 9 (MEGF9) is essential for cell homeostasis; however, its role and mechanism in sepsis-induced cardiac injury and impairment remain unclear. METHODS Adenoviral and adeno-associated viral vectors were applied to overexpress or knock down the expression of MEGF9 in vivo and in vitro. To stimulate septic injury, cardiomyocytes and mice were treated lipopolysaccharide (LPS). To clarify the necessity of AMP-activated protein kinase (AMPK), global AMPK knockout mice were used. RESULTS We found that MEGF9 expressions were reduced in cardiomyocytes and mice by LPS stimulation. Compared with negative controls, plasma MEGF9 levels were also decreased in septic patients, and negatively correlated with LPS-induced cardiac dysfunction. In addition, MEGF9 overexpression attenuated, while MEGF9 knockdown aggravated LPS-induced inflammation and oxidative damage in vivo and in vitro, thereby regulating LPS-induced cardiac injury and impairment. Mechanistic studies revealed that MEGF9 overexpression alleviated LPS-induced cardiac dysfunction through activating AMPK pathway. CONCLUSION We for the first time demonstrate that MEGF9 prevents LPS-related inflammation, oxidative damage and cardiac injury through activating AMPK pathway, and provide a proof-of-concept for the treatment of LPS-induced cardiac dysfunction by targeting MEGF9.
Collapse
Affiliation(s)
- Zhili Jin
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Xianqing Li
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Huixia Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Tao He
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Wanli Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Li Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Xiaoyan Wu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Ming Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Yongzhen Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Di Fan
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| | - Hairong Wang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
6
|
Ye Z, Liu R, Wang H, Zuo A, Jin C, Wang N, Sun H, Feng L, Yang H. Neuroprotective potential for mitigating ischemia-reperfusion-induced damage. Neural Regen Res 2025; 20:2199-2217. [PMID: 39104164 PMCID: PMC11759025 DOI: 10.4103/nrr.nrr-d-23-01985] [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: 02/24/2024] [Revised: 04/09/2024] [Accepted: 06/22/2024] [Indexed: 08/07/2024] Open
Abstract
Reperfusion following cerebral ischemia causes both structural and functional damage to brain tissue and could aggravate a patient's condition; this phenomenon is known as cerebral ischemia-reperfusion injury. Current studies have elucidated the neuroprotective role of the sirtuin protein family (Sirtuins) in modulating cerebral ischemia-reperfusion injury. However, the potential of utilizing it as a novel intervention target to influence the prognosis of cerebral ischemia-reperfusion injury requires additional exploration. In this review, the origin and research progress of Sirtuins are summarized, suggesting the involvement of Sirtuins in diverse mechanisms that affect cerebral ischemia-reperfusion injury, including inflammation, oxidative stress, blood-brain barrier damage, apoptosis, pyroptosis, and autophagy. The therapeutic avenues related to Sirtuins that may improve the prognosis of cerebral ischemia-reperfusion injury were also investigated by modulating Sirtuins expression and affecting representative pathways, such as nuclear factor-kappa B signaling, oxidative stress mediated by adenosine monophosphate-activated protein kinase, and the forkhead box O. This review also summarizes the potential of endogenous substances, such as RNA and hormones, drugs, dietary supplements, and emerging therapies that regulate Sirtuins expression. This review also reveals that regulating Sirtuins mitigates cerebral ischemia-reperfusion injury when combined with other risk factors. While Sirtuins show promise as a potential target for the treatment of cerebral ischemia-reperfusion injury, most recent studies are based on rodent models with circadian rhythms that are distinct from those of humans, potentially influencing the efficacy of Sirtuins-targeting drug therapies. Overall, this review provides new insights into the role of Sirtuins in the pathology and treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Zi Ye
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Runqing Liu
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hangxing Wang
- Division of Infectious Diseases, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Aizhen Zuo
- The Clinical Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Cen Jin
- School of Medical Imaging, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Nan Wang
- Division of Gastroenterology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huiqi Sun
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| | - Luqian Feng
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hua Yang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
7
|
Li J, Gao Q, Liu H, Liu S, Wang Y, Sun X, Zheng J, Yang H, Hu B. Integrating 16S rDNA sequencing analysis and targeted metabolomics to explore the mechanism of Xiexin Tang in treating atherosclerosis mice induced by high-fat diet. J Pharm Biomed Anal 2025; 259:116760. [PMID: 40014894 DOI: 10.1016/j.jpba.2025.116760] [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: 12/11/2024] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Xiexin Tang (XXT) is a classic Chinese medicine formula that can be used to treat Atherosclerosis (AS). This study aimed to investigate the mechanism by which XXT regulated AS lipid levels. Firstly, the mixture components of XXT were analyzed by High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Then, the AS model based on Apolipoprotein E knockout (ApoE-/-) mice was established. Cytokines related to lipid metabolism and bile acid metabolism were detected by Quantitative Real-time PCR (qRT-PCR). 16S rDNA gene sequencing was performed to analyze differential bacterial populations, and the mechanism of XXT regulation of bile acids affecting lipid metabolism was further explored by targeted metabolomics. Further, antibiotic-treated mice were used to investigate the role of gut microbiota in the anti-AS effect of XXT. The results showed that XXT attenuated the lipid levels and reversed the abnormal elevation of cytokines, such as hepatic lipid metabolism and inflammatory reaction in AS mice. XXT also repaired the gut barrier damage and reversed gut microbiota disorders in AS mice. Furthermore, the metabolic levels of bile acids were reshaped by XXT. Whereas, in the absence of gut microbiota, XXT failed to attenuate lipid levels and inhibit the expression of cytokines related to inflammation and bile acid metabolism in AS mice and failed to play a role in ultimately treating AS. In conclusion, XXT could effectively inhibit the inflammatory reaction and lipid accumulation in AS mice, and this effect was closely related to its remodeling of gut microbiota to regulate bile acid metabolism.
Collapse
MESH Headings
- Animals
- Drugs, Chinese Herbal/pharmacology
- Atherosclerosis/drug therapy
- Atherosclerosis/metabolism
- Mice
- Gastrointestinal Microbiome/drug effects
- Metabolomics/methods
- Lipid Metabolism/drug effects
- Diet, High-Fat/adverse effects
- Male
- Bile Acids and Salts/metabolism
- Mice, Inbred C57BL
- Chromatography, High Pressure Liquid/methods
- Tandem Mass Spectrometry/methods
- RNA, Ribosomal, 16S/genetics
- Mice, Knockout, ApoE
- Disease Models, Animal
- DNA, Ribosomal/genetics
- Cytokines/metabolism
- Mice, Knockout
- Liver/metabolism
- Liver/drug effects
Collapse
Affiliation(s)
- Junling Li
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Qianru Gao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Songlin Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Yanchun Wang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Xiongjie Sun
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China; Key Laboratory of Chinese Medicinal Resource and Chinese Herbal Compound of the Ministry of Education, Huangjiahu West Road 16, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Huangjiahu West Road 16, Wuhan 430065, PR China.
| |
Collapse
|
8
|
Rodrigues MM, Falcão LM. Pathophysiology of heart failure with preserved ejection fraction in overweight and obesity - Clinical and treatment implications. Int J Cardiol 2025; 430:133182. [PMID: 40120824 DOI: 10.1016/j.ijcard.2025.133182] [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: 02/04/2025] [Revised: 03/09/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with vast prevalence worldwide. Despite recent advances in understanding its pathophysiology, HFpEF remains under-diagnosed in clinical practice. Obesity-related HFpEF is a distinct and frequent phenotype with an additionally challenging diagnosis. We address the importance of overweight and obesity in HFpEF, focusing on the influence of adipose tissue in inflammation and neurohormonal activity. We also discuss atrial and ventricular remodelling in obesity-related HFpEF and potential clinical implications. Obesity is an independent risk factor for HFpEF. Adipose tissue synthesizes aldosterone, causing lower levels of natriuretic peptide. Adipocytes dysfunction promotes a pro-inflammatory state and leads to extracellular matrix remodelling and consequently stiffening of the heart and vessels. Thus, the quantity, distribution and quality of the excess fat influences cardiovascular risk. Visceral and epicardial adipose tissue are often associated with an increased likelihood of developing HFpEF. Obesity-related HFpEF presents higher risk of left ventricular concentric remodelling and inadequate accommodation of the expanded volume due to the obesity, resulting in higher left ventricular filling pressure. Nevertheless, microvascular endothelium inflammation modifies cardiomyocyte elasticity and increases collagen deposition, which enhances myocardial fibrosis and results in HFpEF. Furthermore, neurohormonal activation may also contribute to cardiac remodelling by inducing plasma volume expansion. In turn, leptin also stimulates aldosterone synthesis and enhances renin-angiotensin-aldosterone system. Obesity-related HFpEF presents worse overall prognosis, with increased risk of heart failure hospitalization and all-cause mortality. Intentional weight loss through caloric restriction, physical activity, pharmacological intervention and/or bariatric surgery are promising strategies.
Collapse
Affiliation(s)
- Mariana M Rodrigues
- Faculty of Medicine, University of Lisbon Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - L Menezes Falcão
- Faculty of Medicine, University of Lisbon, Cardiovascular Center University of Lisbon (CCUL@RISE), Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
9
|
Yuan J, Deng T, Yang Q, Lv D, Zhou Z, You L, Feng Q, Meng X, Pang Q, Li H, Zhu B. Loss of LSD1 ameliorates myocardial infarction by regulating angiogenesis via transcriptional activation of Vegfa. Life Sci 2025; 372:123613. [PMID: 40210117 DOI: 10.1016/j.lfs.2025.123613] [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: 01/15/2025] [Revised: 03/13/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
AIMS Our study aims to explore the regulatory role and underlying mechanisms of Lysine-specific demethylase 1 (LSD1) in angiogenesis following myocardial infarction (MI). MATERIALS AND METHODS We generated inducible cardiomyocyte-specific Lsd1 knockout (Lsd1-cKO) mice and established a MI model. The function of LSD1 in cardiac angiogenesis in MI mice was assessed through echocardiography, histopathological staining, and immunofluorescence analysis. In vitro, Lsd1 silencing in cardiomyocytes was achieved by transfecting small interfering RNA (siRNA), followed by hypoxic treatment to simulate the in vivo MI model. The above cardiomyocyte-conditioned medium was collected and used to treat endothelial cells to observe changes in endothelial function. Additionally, we employed Cleavage Under Targets and Tagmentation sequencing (CUT&Tag-seq) to investigate the potential mechanisms by which LSD1 exerts its effects. KEY FINDINGS We found that the absence of LSD1 protected against cardiac dysfunction and promoted angiogenesis in mice with MI. Lsd1-silenced cardiomyocytes enhance the migration and tube formation function of endothelial cells by releasing vascular endothelial growth factor A (VEGF-A) under hypoxic conditions. The combined analysis of CUT&Tag-seq data revealed that silencing of Lsd1 promoted the monomethylation of H3K4 at the Vegfa promoter and region, leading to the transcriptional activation of Vegfa mRNA in cardiomyocytes. SIGNIFICANCE Our research indicates that lowered level of LSD1 in cardiomyocytes enhances VEGF-A paracrine secretion and improves endothelial cell function through cross-talk, ultimately promoting angiogenesis. These findings suggest that targeting LSD1 might be an effective therapeutic approach to protect against MI.
Collapse
Affiliation(s)
- Jinghan Yuan
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingshan Yang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Danyi Lv
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenfang Zhou
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qipu Feng
- Animal Experiment Center, Sichuan University West China Hospital, Chengdu, Sichuan, China
| | - Xiangmin Meng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiuyu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bingmei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Michaud M, Sailler L, Lemeu M, Castel B, Vacheret F, Prudhomme L, Pugnet G. Efficacy and safety of Anakinra in idiopathic recurrent pericarditis: Results from the UPACITER study. Int J Cardiol 2025; 430:133204. [PMID: 40164317 DOI: 10.1016/j.ijcard.2025.133204] [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: 02/19/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Idiopathic recurrent pericarditis (IRP) is a chronic autoinflammatory condition characterized by relapsing episodes of pericardial inflammation. Standard treatment involves colchicine and non-steroidal anti-inflammatory drugs (NSAIDs), with corticosteroids as a second-line therapy. However, many patients develop corticosteroid dependency or resistance, necessitating alternative treatments. Anakinra, an interleukin-1 (IL-1) receptor antagonist, has shown promising results as a third-line therapy. METHODS The UPACITER multicenter study is a retrospective study conducted on patients with IRP and treated with anakinra between 2018 and 2024 in Midi-Pyrénées region of France. Patients met the 2015 ESC criteria for recurrent pericarditis. Clinical features, treatment responses, and side effects were collected. Primary endpoint was complete response, defined as the resolution of symptoms, normalization of C-reactive protein levels, and absence of pericardial effusion. RESULTS Twelve patients were included, with a median age of 46 years (interquartile range: 29.7-52.7). All patients achieved a complete response and corticosteroid discontinuation (from 17.9 ± 10.68 mg to 0 mg, p < 0.005). The recurrence-free survival rates were 100 % (95 % CI [100;100]) at month 6, 88.9 % (95 % CI [70.6;100]) at month 12, and 77.8 % (95 % CI [54.9;100]) at months 18 and 24. Three patients experienced a relapse of pericarditis: 2 during the spacing of injection and one after discontinuation of anakinra, with a median time before relapse of 12 months (IQR 9.5-13). One serious infection was reported. CONCLUSION Anakinra is a safe and effective third-line therapy for patients with IRP resistant to conventional treatments, leading to a reduction in recurrences and corticosteroid discontinuation.
Collapse
Affiliation(s)
- Martin Michaud
- Department of Internal Medicine, Clinique Saint Exupery, Toulouse, France; Department of internal medicine, Toulouse University Hospital, Toulouse, France.
| | - Laurent Sailler
- Department of internal medicine, Toulouse University Hospital, Toulouse, France
| | - Mélanie Lemeu
- Department of Internal Medicine, Albi Hospital, Albi, France
| | - Brice Castel
- Department of Internal Medicine, Tarbes Hospital, Tarbes, France
| | - Fabienne Vacheret
- Department of Internal Medicine, Perpignan Hospital, Perpignan, France
| | | | - Grégory Pugnet
- Department of internal medicine, Toulouse University Hospital, Toulouse, France
| |
Collapse
|
11
|
Guan T, Lu Z, Tai R, Guo S, Zhang Z, Deng S, Ye J, Chi K, Zhang B, Chen H, Deng Z, Ke Y, Huang A, Chen P, Wang C, Ou C. Silicified curcumin microspheres Combats cardiovascular diseases via Nrf2/HO-1 pathway. Bioact Mater 2025; 49:378-398. [PMID: 40144796 PMCID: PMC11937612 DOI: 10.1016/j.bioactmat.2025.03.007] [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: 01/17/2025] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Diabetes and chemotherapy frequently give rise to severe cardiovascular complications, including chemotherapy-induced cardiotoxicity and diabetes-associated vascular remodeling. Nevertheless, the precise epidemiological features of these cardiovascular ailments remain incompletely elucidated, resulting in a dearth of effective therapeutic strategies in clinical settings. To tackle this intricate challenge, we have delved extensively into database resources, conducted comprehensive analyses of pertinent epidemiological data, and designed silicified curcumin (Si/Cur) microspheres as a novel therapeutic approach for cardiovascular diseases. By harnessing the alkaline microenvironment generated by silicon (Si), Si/Cur markedly elevates the bioavailability of curcumin (Cur). Further investigations have elucidated that Si/Cur exerts its therapeutic actions primarily via the Nrf2/HO-1 signaling pathway, effectively suppressing vascular remodeling and mitigating myocardial injury, thus disrupting the vicious cycle of persistent cardiovascular damage. In conclusion, this study integrates clinical cohort research to dissect epidemiological characteristics, directs the design and application of biomaterials, and paves the way for a novel and efficacious therapeutic avenue for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Tianwang Guan
- Guangdong Engineering Research Center of Boron Neutron Therapy and Application in Malignant Tumors, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, Dongguan Engineering Research Center for Innovative Boron Drugs and Novel Radioimmune Drugs, Cancer Center, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Guangdong, 523059, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
| | - Zhenxing Lu
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Rundong Tai
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Shuai Guo
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Zhaowenbin Zhang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shaohui Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Jujian Ye
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Kaiyi Chi
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510182, China
| | - Binghua Zhang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, China
| | - Huiwan Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Zhilin Deng
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Yushen Ke
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Andong Huang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, 510180, China
| | - Peier Chen
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, 510280, China
- The Tenth Affiliated Hospital (Dongguan People's Hospital), Southern Medical University, Dongguan, 523059, China
| |
Collapse
|
12
|
Tao H, Wang C, Zou C, Zhu H, Zhang W. Unraveling the potential of neuroinflammation and autophagy in schizophrenia. Eur J Pharmacol 2025; 997:177469. [PMID: 40054715 DOI: 10.1016/j.ejphar.2025.177469] [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: 10/22/2024] [Revised: 02/03/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
Schizophrenia (SCZ) is a complex and chronic psychiatric disorder that affects a significant proportion of the global population. Although the precise etiology of SCZ remains uncertain, recent studies have underscored the involvement of neuroinflammation and autophagy in its pathogenesis. Neuroinflammation, characterized by hyperactivated microglia and markedly elevated pro-inflammatory cytokines, has been observed in postmortem brain tissues of SCZ patients and is closely associated with disease severity. Autophagy, a cellular process responsible for eliminating damaged components and maintaining cellular homeostasis, is believed to play a pivotal role in neuronal health and the onset of SCZ. This review explores the roles and underlying mechanisms of neuroinflammation and autophagy in SCZ, with a particular focus on their intricate interplay. Additionally, we provide an overview of potential therapeutic strategies aimed at modulating neuroinflammation and autophagy, including nutritional interventions, anti-inflammatory drugs, antipsychotics, and plant-derived natural compounds. The review also addresses the dual effects of antipsychotics on autophagy. Our objective is to translate these insights into clinical practice, expanding the therapeutic options available to improve the overall health and well-being of individuals with SCZ.
Collapse
Affiliation(s)
- Hongxia Tao
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Congyin Wang
- Department of Emergency Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Chuan Zou
- Department of General Practice, Chengdu Fifth People's Hospital, Chengdu, Sichuan, 611130, China
| | - Hongru Zhu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Wei Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Vlad ML, Mares RG, Jakobsson G, Manea SA, Lazar AG, Preda MB, Popa MA, Simionescu M, Schiopu A, Manea A. Therapeutic S100A8/A9 inhibition reduces NADPH oxidase expression, reactive oxygen species production and NLRP3 inflammasome priming in the ischemic myocardium. Eur J Pharmacol 2025; 996:177575. [PMID: 40180274 DOI: 10.1016/j.ejphar.2025.177575] [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: 10/09/2024] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Oxidative stress and alterations in redox signalling have been implicated in the pathophysiology of myocardial infarction (MI). NADPH oxidase (Nox) is an important source of reactive oxygen species (ROS) in the infarcted myocardium. Alarmin S100A8/A9 amplifies acute myocardial inflammation in MI and has been shown to be a promising therapeutic target to improve cardiac function post-MI. We aimed to elucidate the underlying mechanisms linking S100A8/A9, oxidative stress and the inflammatory response in MI. MI was induced by permanent left coronary artery ligation in C57BL/6J mice, followed by treatment with the S100A8/A9 inhibitor ABR-238901 (30 mg/kg) or PBS for 3 days. The in-vivo experiments were complemented with mechanistic studies on cultured macrophages (Mac), important cellular effectors in MI. Compared to sham-operated animals, we detected significant increases in the Nox1, Nox2, Nox4 catalytic subunits at mRNA and protein levels, and NADPH-dependent ROS production in the left ventricle of MI mice. S100A8/A9 blockade prevented the up-regulation of Nox1/2/4 expression, reduced ROS formation, suppressed NF-kB activation and prevented NLRP3 inflammasome priming and activation, leading to reduced levels of active IL-1β. In-vitro, S100A8/A9 induced gene expression of Nox catalytic subtypes and NLRP3 in Mac in a TLR4-dependent and dose-dependent manner. These effects were counteracted by pharmacological inhibition of S100A8/9, TLR4, Nox1/4 and Nox2. In conclusion, we show that Nox upregulation and ROS formation triggered by S100A8/A9 contributes to NLRP3 inflammasome priming and increased IL-1β production in the infarcted myocardium. These mechanisms can be therapeutically targeted to prevent inflammatory and oxidant myocardial damage in acute MI.
Collapse
Affiliation(s)
- Mihaela-Loredana Vlad
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania; Department of Cardiology II, Emergency Clinical County Hospital, Targu Mures, Romania.
| | - Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Malmö, Sweden.
| | - Simona-Adriana Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Alexandra-Gela Lazar
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Mihai Bogdan Preda
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Mirel Adrian Popa
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Alexandru Schiopu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania; Department of Translational Medicine, Lund University, Malmö, Sweden; Department of Internal Medicine, Skåne University Hospital, Lund, Sweden.
| | - Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| |
Collapse
|
14
|
Prathumwon C, Anuchapreeda S, Kiattisin K, Panyajai P, Wichayapreechar P, Surh YJ, Ampasavate C. Curcumin and EGCG combined formulation in nanostructured lipid carriers for anti-aging applications. Int J Pharm X 2025; 9:100323. [PMID: 40115962 PMCID: PMC11923819 DOI: 10.1016/j.ijpx.2025.100323] [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: 11/20/2024] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/23/2025] Open
Abstract
Curcumin (Cur) and epigallocatechin gallate (EGCG), the primary active compounds in turmeric and green tea, respectively, have been investigated for their anti-aging potential. The Cur and EGCG combination was encapsulated in sustained-release nanostructured lipid carriers (NLCs) to enhance their bioactivities and pharmaceutical properties. A significant enhancement in the antioxidant activities of the Cur and EGCG combination was observed at an optimal ratio, as demonstrated by the 2,2-diphenyl-1-picrylhydrazyl radical scavenging assay (118.83 ± 3.78 %), ferric ion reducing antioxidant power assay (217.25 ± 13.45 %), and lipid peroxidation inhibition assay (106.08 ± 12.93 %), compared to Cur alone without compromising the antioxidant activities and total phenolic content of EGCG. This is due to the enhancement of total phenolic content of the combination of 218.83 ± 10.57 %. For anti-aging activities, the combination exhibited stimulation of SIRT1 protein and inhibition of collagenase and elastase of 27.53 ± 0.73 %, 43.70 ± 1.05 % and 51.76 ± 6.52 % compared with that achieved with Cur alone, respectively. The incorporation of the Cur and EGCG combination into NLCs resulted in high entrapment efficiencies of 98.60 ± 0.05 % for Cur and 98.40 ± 0.08 % for EGCG, with corresponding loading capacities of 0.789 ± 0.001 % and 3.935 ± 0.003 %, respectively. When formulated NLCs into an emulgel base, the system demonstrated sustained release profiles over 48 h, with 12.82 ± 0.99 % release of Cur and 63.77 ± 5.76 % release of EGCG. Significant skin retention was also observed after 24 h, with 23.88 ± 1.71 % Cur and 22.79 ± 4.65 % EGCG retained in the skin. Therefore, Cur: EGCG-loaded NLCs in emulgel can deliver the active compounds into the dermis, enhancing skin penetration, sustained delivery, and anti-aging activity superior to each conventional single active compound in topical formulations.
Collapse
Affiliation(s)
- Chidchanok Prathumwon
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Songyot Anuchapreeda
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokwan Kiattisin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pawaret Panyajai
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Panikchar Wichayapreechar
- Department of Cosmetic Sciences, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 151-741, South Korea
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
15
|
Fateh ST, Shiraseb F, Hajinasab MM, Noori S, Clark CCT, Mirzaei K. Interaction between 3-SNP genetic risk score and dietary fats intake on inflammatory markers among overweight and obese women. J Diabetes Metab Disord 2025; 24:80. [PMID: 40093786 PMCID: PMC11909376 DOI: 10.1007/s40200-024-01542-z] [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: 06/29/2024] [Accepted: 09/14/2024] [Indexed: 03/19/2025]
Abstract
Objectives This study, for the first time, sought to investigate whether the interaction between the GRS consists of three SNPs (CAV-1, CRY-1, MC4R) and fat intake is associated with inflammatory markers among Iranian overweight and obese women. Methods This cross-sectional study was conducted with 246 overweight and obese women, aged 18-48 years. Three SNPs, including CAV-1 rs3807992, CRY-1 rs2287161, and MC4R rs17782313, were genotyped using PCR-RFLP to calculate the genetic risk score (GRS) for each participant. Dietary fat intake was measured using a validated semi-quantitative food frequency questionnaire (FFQ). C-reactive protein (CRP), interleukin-1β (IL-1β), transforming growth factor-β (TGF-β), monocyte chemoattractant protein-1 (MCP-1), plasminogen activator inhibitor-1 (PAI-1), and Galectin-3 (Gal-3) were assessed as the primary outcomes of the study. Results After controlling for confounding variables, a significant interaction between high total fat intake and high GRS, compared to the reference group, was found for TGF-β level (P-value: 0.028). A significant positive interaction between high GRS and high intakes of SFA intake (P-value: 0.013). A significant interaction between high GRS and high intakes of MUFA, compared to the reference group, was found for ghrelin level (P-value: 0.040) and MCP-1 level (P-value: 0.075). There was a significant interaction between high GRS and intakes of DHA, compared to the reference group, for Gal-3 level (P-value: 0.013) MCP-1 level (P-value: 0.020). Conclusions Consuming different types of fats can influence the interaction between GRS and inflammatory markers, suggesting further research is needed to fully understand this relationship. Supplementary information The online version contains supplementary material available at 10.1007/s40200-024-01542-z.
Collapse
Affiliation(s)
| | - Farideh Shiraseb
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science (TUMS), Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Mohammad Mahdi Hajinasab
- Department of Nutrition, Electronic Health and Statistics Surveillance Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Noori
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Cain C. T. Clark
- Centre for Intelligent Healthcare, Coventry University, CV1 5FB Coventry, U.K
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Science (TUMS), Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| |
Collapse
|
16
|
Manzoor N, Samad N, Khaliq S, Bin Khatab Abbasi B, Ahmad S, Irfan A, Raish M, Bin Jardan YA. Acipimox mitigates depression like behavior following high fat rich diet in rats. Brain Res Bull 2025; 225:111342. [PMID: 40216031 DOI: 10.1016/j.brainresbull.2025.111342] [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: 01/05/2025] [Revised: 02/19/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Acipimox (ACPX), a niacin derivative, has demonstrated antioxidant activity In vitro and In vivo; however, it has not been widely used in treating neurological problems. The present study examined the effects of Acipimox on body weight, dietary intake, depressive symptoms, oxide-neuroinflammation, 5-HT metabolism, and 5-HT1A receptor expression in hypothalamus of rats. Forty eight (n = 8) male albino rats were randomly divided into six groups (i) Vehicle (Veh)+ normal diet (ND) (ii) ND + ACPX (25 mg/mL/kg; low dose) (iii) ND+ ACPX (50 mg/mL/kg; high dose) (iv) Veh +High fat rich diet (HFRD) (v) HFRD+ACPX (25 mg/mL/kg; low dose (vi) HFRD+ACPX (50 mg/mL/kg; high dose). Animals were given their respective treatment for 8 weeks. After that, behavioral tests i.e. tail suspension test (TST) and forced swim test (FST) performed for depression-like behavior assessment. Animals were decapitated and the hypothalamus was isolated from the brain for biochemical and neurochemical analysis. Results showed that, HFRD induced depression like behavior and increased body weight and food intake was prevented by repeated administration of ACPX (both doses). HFRD induced increased oxido-neuroinflammation, altered serotonin metabolism and serotonin-1A receptor relative expression in the hypothalamus were regulated by ACPX (both doses). In conclusion, HFRD-induced behavioral deficits (depression like behavior) mitigated by ACPX through its antioxidant, anti-inflammatory, and neuromodulatory properties. It is recommended that use of ACPX could be helpful for HFRD-induced behavioral impairment i.e. depression.
Collapse
Affiliation(s)
- Natasha Manzoor
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan 60800, Pakistan.
| | - Noreen Samad
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan 60800, Pakistan.
| | - Saima Khaliq
- Department of Biochemistry, Federal Urdu University of Arts, Science and Technology, Karachi, Pakistan.
| | - Bakar Bin Khatab Abbasi
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA.
| | - Saara Ahmad
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan.
| | - Ali Irfan
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan.
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
17
|
Stevens DL, Bryant AE. Complexities of cardiomyopathy in septic shock. Curr Opin Infect Dis 2025; 38:214-221. [PMID: 40127058 DOI: 10.1097/qco.0000000000001102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
PURPOSE OF REVIEW This review highlights the complex pathophysiology of myocardial dysfunction in septic shock and emphasizes the need for early and repeated hemodynamic assessments to improve outcome. RECENT FINDINGS Septic cardiomyopathy is a complex, dynamic process driven by multiple mechanisms such as direct myocardial depression induced by host immune mediators (e.g., cytokines, nitric oxide) and/or bacterial toxins, and mitochondrial metabolic dysfunction. Recent echocardiography studies have described multiple unique hemodynamic clusters (phenotypes) that correlated with clinical outcomes. Similarly, serial echocardiography findings and mean arterial pressure abnormalities in patients with Streptococcal Toxic Shock Syndrome (StrepTSS) yielded three distinct hemodynamic groups that predicted mortality and morbidity. Because excessive use of fluids and/or vasopressors can be detrimental, especially in patients with microvascular injury or cardiomyopathy, application of the cardiovascular performance criteria of these different phenotypes could better inform management decisions in real time and improve outcome. SUMMARY Septic cardiomyopathy is a dynamic, multidimensional response of the myocardium to infection involving both normal and dysregulated immune responses in which the measurable changes in myocardial function predict outcomes. This current paradigm mandates that functional parameters of cardiac performance be measured early and repeatedly throughout the disease course using echocardiography to guide treatment and improve outcome.
Collapse
Affiliation(s)
- Dennis L Stevens
- University of Washington School of Medicine, Seattle, Washington
| | | |
Collapse
|
18
|
Leist L, Hausmann L, Pohlink C. Poor communication impairs optimal care of patients with heart failure and erectile dysfunction. ESC Heart Fail 2025; 12:1932-1941. [PMID: 39828426 PMCID: PMC12055399 DOI: 10.1002/ehf2.15201] [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: 09/13/2023] [Revised: 10/23/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
AIMS Heart failure (HF) and erectile dysfunction (ED) share numerous risk factors and pathogenetic mechanisms, resulting in a high prevalence of ED among male patients with HF. This questionnaire-based study aimed to better understand the patient's journey from a patient perspective and that of healthcare professionals (HCPs) regarding communication, education and treatment of ED as a recognized comorbid condition. METHODS Two independent online surveys were conducted between November 2021 and January 2022 in Germany. Analysis of the first patient-targeting survey comprised 927 male patients with HF. As part of this survey, 176 patients with HF and comorbid ED provided further information about their experiences and ED-related communication with HCPs. The second online survey collected the perspectives of 78 physicians including general practitioners (GPs), cardiologists and internists throughout Germany. RESULTS The average age of participating male patients with HF was 55 years. Both patients and HCPs considered sexual life as an important aspect of patients' quality of life (QoL). Fifty-six per cent of all patients with HF rated their sexual life as important, and 43% were unsatisfied with it. Patients' suffering due to ED was reported as moderate to severe by nearly all HCPs (92%) and patients themselves (82%). A communication gap and perceived imbalance in education were identified, with 27% of patients reporting consultations about ED, while 58% of HCPs claimed to proactively address the issue of ED during counselling. Thirty-nine per cent of HCPs felt that their patients were uncomfortable talking about ED, but only 7% of patients reported discomfort. As a possible result, 69% of male HF patients with ED felt left alone, and 74% stated that they would like to talk to an HCP. Due to inadequate education together with patients not considering their HF doctor as a contact for ED management, as few as 20% of patients with ED receive prescription drugs for ED management. HCPs reported that 32% of their patients are non-compliant with HF medication due to ED, highlighting the importance of sexual health for patients' QoL. CONCLUSIONS The results of our surveys suggest that most of the male HF patients may be receiving incomplete ED management also affecting HF treatment, most likely due to impaired communication with their HCPs with barriers from both sides. HCPs giving patients advice on this topic are urgently needed to improve patients' QoL, reduce patient suffering and avoid discontinuation of HF medications for fear of side effects.
Collapse
Affiliation(s)
- Lea Leist
- Novartis Pharma GmbHNurembergGermany
| | | | | |
Collapse
|
19
|
Jenabi Ghods M, Amirabadizadeh A, Delbari A, Naserpour M, Saatchi M. Prevalence of macro-vascular complications among type 2 diabetic adults aged 50 and over: results from Ardakan cohort study on aging (ACSA). J Diabetes Metab Disord 2025; 24:39. [PMID: 39801689 PMCID: PMC11711917 DOI: 10.1007/s40200-024-01556-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Objective Type 2 diabetes mellitus (T2DM) is a common condition that can lead to adverse macrovascular complications. This study aims to determine the prevalence of macrovascular complications in adults aged ≥ 50 with T2DM in Ardakan city, using data from the Ardakan Cohort Study on Aging (ACSA). Methods A cross-sectional investigation involved 5933 participants from the ACSA; of those assessed, 2340 had T2DM. Macrovascular complications, specifically coronary artery disease (CAD), cerebrovascular disease (CVD), and peripheral artery disease(PAD) were identified through medical records and physician assessment. Logistic regression was used to identify risk factors for these complications. Results The prevalence of CAD and CVD were 16.9% (95% CI:16.0-19.0) and 4% (95% CI:3.3-5.0), respectively. risk factors for CAD included age over 60 (OR = 1.47, 95% CI: 1.08-2.01, p = 0.01), male gender (OR = 1.87, 95% CI: 1.33-2.62, p < 0.001), former smoking (OR = 1.96, 95% CI: 1.30-2.95, p = 0.001), hypertension (OR = 3.16, 95% CI: 2.23-4.46, p < 0.001), and over ten years of diabetes duration(OR = 2.04, 95% CI: 1.39-2.99, p < 0.001) and For CVD, significant risk factors included male gender (OR = 2.61, 95% CI: 1.52-4.51, p = 0.001) and hypertension (OR = 2.36, 95% CI: 1.27-4.39, p = 0.006). Conclusion This study highlights the high prevalence of macrovascular complications in adults over 50 with T2DM in Ardakan. It emphasizes the importance of managing key risk factors such as hypertension and quitting smoking, especially in older adults and males.
Collapse
Affiliation(s)
- Mariye Jenabi Ghods
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
- Department of Nursing, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Alireza Amirabadizadeh
- Student Research Committee, Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Delbari
- Iranian Research Center on Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mahshad Naserpour
- Department of Nursing, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Mohammad Saatchi
- Department of Biostatistics and Epidemiology, University of Social Welfare and Rehabilitation Science, Tehran, Iran
- Health in Emergency and Disaster Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| |
Collapse
|
20
|
Li G, He W, Wang DW. Immune cell dynamics in heart failure: implicated mechanisms and therapeutic targets. ESC Heart Fail 2025; 12:1739-1758. [PMID: 39905753 PMCID: PMC12055366 DOI: 10.1002/ehf2.15238] [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: 10/24/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
The relationship between heart failure (HF) and immune activation has garnered significant interest. Studies highlight the critical role of inflammation in HF, affecting cardiac structure and function. Despite promising anti-inflammatory therapies, clinical trials have faced challenges, indicating an incomplete understanding of immune mechanisms in HF. Immune cells, which are key cytokine sources, are pivotal in HF progression. In this review, the authors provide a comprehensive overview of the complex role of different types of immune cells and their cell subtypes in HF. In addition, the authors summarize the available targets and animal experimental evidence for targeting immune cells for the treatment of HF. Future research directions will focus on the roles of immune cells and their interrelationships at different stages of HF, aiming to develop more targeted therapeutic strategies that can achieve more precise interventions in the pathological process of HF.
Collapse
Affiliation(s)
- Gen Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Wu He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological DisordersWuhan430000China
| |
Collapse
|
21
|
İnce H, Yılmaz Z, Karabulut A, Aydın E, Yıldırım Y, Kara AV, Kadiroğlu AK, Aydın FY. Evaluation of risk factors associated with epicardial fat tissue thickness in patients with stage 4 and stage 5 chronic kidney disease. Int Urol Nephrol 2025; 57:1977-1982. [PMID: 39893350 DOI: 10.1007/s11255-025-04386-5] [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: 10/07/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE Chronic kidney disease (CKD) is a global public health issue, often associated with high mortality and morbidity, especially due to cardiovascular diseases. Epicardial adipose tissue (EAT), the visceral fat surrounding the heart, has been recognized as a significant factor in cardiovascular risk. This study aims to assess the relationship between EAT and body composition in patients with stage 4 and 5 CKD using bioelectrical impedance analysis (BIA). METHODS The study included 80 patients with stage 4 and 5 CKD who were not on dialysis. Body composition was measured using BIA, and EAT was assessed using transthoracic echocardiography(ECHO). Exclusion criteria included heart failure, morbid obesity, pregnancy, and pacemaker use, among others. Various demographic, clinical, and biochemical parameters were also recorded. RESULTS Patients with stage 5 CKD (6.7 ± 0.12) had significantly higher EAT compared to stage 4 CKD (5.9 ± 0.09) patients. EAT showed a positive correlation with age, BMI, blood pressure, C-reactive protein (CRP), and triglyceride levels, and a negative correlation with albumin and HDL levels. Multivariate analysis revealed that increased systolic blood pressure(SBP)(p:0.019),fat tissue mass (FTM)(p < 0.001), low HDL(p: 0.027), and low albumin(p < 0.001) were independent predictors of EAT. CONCLUSION EAT is higher in advanced CKD patients and is associated with several cardiovascular risk factors. Measuring EAT in CKD patients using non-invasive methods like ECHO could be valuable in predicting cardiovascular risks. Addressing the factors that contribute to increased EAT may improve clinical outcomes for CKD patients.
Collapse
Affiliation(s)
- Hasan İnce
- Medical Faculty Department of Nephrology, Dicle University, Diyarbakır, Turkey.
| | - Zülfükar Yılmaz
- Medical Faculty Department of Nephrology, Dicle University, Diyarbakır, Turkey
| | - Aziz Karabulut
- Department of Cardiology, Memorial Hospital, Diyarbakır, Turkey
| | - Emre Aydın
- Medical Faculty Department of Nephrology, Dicle University, Diyarbakır, Turkey
| | - Yaşar Yıldırım
- Medical Faculty Department of Nephrology, Dicle University, Diyarbakır, Turkey
| | - Ali Veysel Kara
- Faculty of Medicine Department of Nephrology, Erzincan University, Erzincan, Turkey
| | | | - Fatma Yılmaz Aydın
- Faculty Department of Internal Medicine Diyarbakır, Dicle University Medical, Diyarbakır, Turkey
| |
Collapse
|
22
|
Gao Y, Gao K, Shi R, Huang X, Dang P, Liu H, Zheng X, Xue Y. Association between phenotypic age and in-hospital outcomes in patients with acute myocardial infarction: A retrospective observational study. IJC HEART & VASCULATURE 2025; 58:101670. [PMID: 40235940 PMCID: PMC11997336 DOI: 10.1016/j.ijcha.2025.101670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025]
Abstract
Background Phenotypic age (PhenoAge) has emerged as a superior predictor of age-related morbidity and mortality. This study aimed to assess the associations between PhenoAge and in-hospital outcomes in patients with acute myocardial infarction (AMI). Methods 2896 AMI patients admitted to the First Affiliated Hospital of Xi'an Jiaotong University from 2019 to 2022 were analyzed in this retrospective study. PhenoAge was calculated by using the phenotypic age calculator, an equation for chronologic age and 9 clinical biomarkers, and Phenotypic Age Accelerate (PhenoAgeAccel) was measured using the residuals of regression PhenoAge on chronological age. Clinical outcomes were defined as in-hospital major adverse cardiovascular events (MACEs), including cardiogenic shock, malignant arrhythmia, acute heart failure, and mechanical complications. Results Overall, patients with high PhenoAge had a higher Gensini score and a higher likelihood of receiving supportive care, as well as worse clinical outcomes. The same results were observed in patients with positive PhenoAgeAccel. Moreover, PhenoAge and PhenoAgeAccel were significantly associated with in-hospital MACEs even after adjusting for multiple traditional risk factors. The area under the curve for PhenoAge was 0.714 (P < 0.001), which significantly outperformed chronologic age (AUC: 0.601, P < 0.001) and other cardiovascular risk factors. Re-examination of the ROC curves using different combinations of variables, PhenoAge was also able to significantly improve the predictive value of several models. Conclusions PhenoAge is significantly associated with clinical outcomes and reliably predicts in-hospital MACEs. Compared with chronological age, PhenoAge is a better complementary biomarker for predicting the risk of in-hospital adverse cardiovascular events in patients with AMI.
Collapse
Affiliation(s)
- Yajie Gao
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| | - Ke Gao
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| | - Ruijuan Shi
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| | - Xiaorui Huang
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| | - Peizhu Dang
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| | - Hui Liu
- Biobank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
| | - Xiaopu Zheng
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| | - Yanbo Xue
- Department of Cardiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061 Shaanxi, China
- Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi’an 710061 Shaanxi, China
| |
Collapse
|
23
|
Chokkalla AK, Arruri V, Mehta SL, Vemuganti R. Loss of Epitranscriptomic Modification N 6-Methyladenosine (m 6A) Reader YTHDF1 Exacerbates Ischemic Brain Injury in a Sexually Dimorphic Manner. Transl Stroke Res 2025; 16:831-847. [PMID: 38869772 DOI: 10.1007/s12975-024-01267-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
N6-Methyladenosine (m6A) is a neuronal-enriched, reversible post-transcriptional modification that regulates RNA metabolism. The m6A-modified RNAs recruit various m6A-binding proteins that act as readers. Differential m6A methylation patterns are implicated in ischemic brain damage, yet the precise role of m6A readers in propagating post-stroke m6A signaling remains unclear. We presently evaluated the functional significance of the brain-enriched m6A reader YTHDF1, in post-stroke pathophysiology. Focal cerebral ischemia significantly increased YTHDF1 mRNA and protein expression in adult mice of both sexes. YTHDF1-/- male, but not female, mice subjected to transient middle cerebral artery occlusion (MCAO) showed worsened motor function recovery and increased infarction compared to sex-matched YTHDF1+/+ mice. YTHDF1-/- male, but not female, mice subjected to transient MCAO also showed significantly perturbed expression of genes related to inflammation, and increased infiltration of peripheral immune cells into the peri-infarct cortex, compared with sex-matched YTHDF1+/+ mice. Thus, this study demonstrates a sexual dimorphism of YTHDF1 in regulating post-ischemic inflammation and pathophysiology. Hence, post-stroke epitranscriptomic regulation might be sex-dependent.
Collapse
Affiliation(s)
- Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Suresh L Mehta
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
24
|
Hillard PJA. Artificial Intelligence (AI) and The Journal of Pediatric and Adolescent Gynecology (JPAG). J Pediatr Adolesc Gynecol 2025; 38:301-303. [PMID: 40345793 DOI: 10.1016/j.jpag.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
|
25
|
Zhang N, Tian X, Sun D, Tse G, Xie B, Zhao Z, Liu T. Clonal hematopoiesis, cardiovascular disease and cancer treatment-induced cardiotoxicity. Semin Cancer Biol 2025; 111:89-114. [PMID: 40023267 DOI: 10.1016/j.semcancer.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 01/05/2025] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
Clonal hematopoiesis (CH) arises when a substantial proportion of mature blood cells is derived from a single hematopoietic stem cell lineage. It is considered to be a premalignant state that predisposes individuals to an increased risk of cancers. Recently, emerging evidence has demonstrated a strong association between CH and both the incidence and mortality of cardiovascular diseases (CVD), with the relative risks being comparable to those attributed to traditional cardiovascular risk factors. In addition, CH has been suggested to play a role in CVD and anti-cancer treatment-related cardiotoxicity amongst cancer survivors. Moreover, certain forms of chemotherapy and radiation therapy have been shown to promote the clonal expansion of specific CH-related mutations. Consequently, CH may play a substantial role in the realm of cardio-oncology. In this review, we discuss the association between CH with cancer and CVD, with a special focus on anti-cancer treatment-related cardiotoxicity, discuss possible future research avenues and propose a systematic approach for clinical practice.
Collapse
Affiliation(s)
- Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xu Tian
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dongkun Sun
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, China
| | - Bingxin Xie
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
26
|
Wang H, Zhang M, Xin M, Yue X, Piao J, Zhao L, Bi H, Wang S, Jin C, Nan Y, Jin X, Cheng XW. Dexmedetomidine Reduces Chronic Stress-Related Thrombosis in a Mouse FeCl 3 Model. FASEB J 2025; 39:e70546. [PMID: 40304859 PMCID: PMC12042889 DOI: 10.1096/fj.202500724r] [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: 03/08/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
Chronic psychological stress (CPS) is a significant risk factor for thrombotic cardio-cerebrovascular diseases (TCVDs). Clinical data suggest that the α2-adrenergic receptor (AdR-α2) agonist dexmedetomidine (Dex) can influence coagulation in stress-exposed intensive care unit patients. Given the important role of protease-activated receptor-2 (PAR-2) in vascular pathobiology, we aimed to investigate the potential effects of Dex on stress-related thrombus formation, focusing on the PAR-2 signaling pathway. Eight-week-old male mice underwent non-stress and immobilization stress with Dex treatment for 2 weeks and were then subjected to carotid artery thrombosis induction using ferric chloride (FeCl3). On Day 14 post-stress, the mice exhibited increased thrombus weight and length, along with harmful alterations in the plasma levels of von Willebrand factor and metalloproteinase with thrombospondin Type 13 motifs. Additionally, arterial protein and/or mRNA levels of PAR-2, p-Akt, Bcl-2, cleaved caspase-3, cytochrome c, gp91phox, TNF-α, MCP-1, ICAM-1, VCAM-1, and TLR-4 were altered, accompanied by arterial endothelial loss. Dex treatment reversed these changes. Conversely, AdR-α2 blockage with yohimbine diminished the benefits of Dex. In vitro, Dex reduced stress serum-induced reactive oxygen species production and endothelial apoptosis, along with beneficial alterations in PAR-2, Bcl-2, and cytochrome c protein levels. Yohimbine diminished these effects. Thus, α2-adrenergic receptor activation appeared to mitigate stress-related thrombus formation in mice undergoing FeCl3-induced surgery, possibly by negatively regulating PAR-2 signaling. These findings suggest a potential therapeutic strategy for CPS-related thrombotic events in patients with TCVDs.
Collapse
Affiliation(s)
- Huazhen Wang
- Department of AnesthesiologyYanbian University HospitalYanjiJilinP.R. China
| | - Meiping Zhang
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
| | - Minglong Xin
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
| | - Xueling Yue
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
| | - Jinshun Piao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
| | - Longguo Zhao
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
| | - Hehui Bi
- Department of AnesthesiologyYanbian University HospitalYanjiJilinP.R. China
| | - Shiyan Wang
- Department of AnesthesiologyYanbian University HospitalYanjiJilinP.R. China
| | - Chunzi Jin
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
| | - Yongshan Nan
- Department of AnesthesiologyYanbian University HospitalYanjiJilinP.R. China
| | - Xianglan Jin
- Department of AnesthesiologyYanbian University HospitalYanjiJilinP.R. China
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Jilin Provincial Key Laboratory of Stress and Cardiovascular DiseaseYanbian University HospitalYanjiJilinP.R. China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of EducationYanbian UniversityYanjiJilinP.R. China
- Department of Community Healthcare and GeriatricsNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
27
|
Ishikawa J, Futami S, Toba A, Yamamoto A, Kobayashi K, Takani K, Ono H, Maeda T, Kawano M, Kiyomizu M, Tamura Y, Araki A, Mori H, Harada K. Splenic and portal venous flow associated with frailty and sarcopenia in older outpatients with cardiovascular disease. BMC Geriatr 2025; 25:319. [PMID: 40346462 DOI: 10.1186/s12877-025-05973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Older patients with cardiovascular disease often experience frailty and sarcopenia. We evaluated whether a reduced blood flow in the splenic and portal vein is associated with frailty and sarcopenia in older patients with cardiovascular disease. METHODS Blood flow in the splenic and portal vein was evaluated using EPIQ7 (Philips) in older patients (aged ≥ 65 years, 123 patients) with cardiovascular disease, who visited the frailty outpatient clinic. Frailty was assessed using the Japanese version of Cardiovascular Health Study (J-CHS) criteria and the Kihon Checklist (KCL), while sarcopenia was assessed using the Asian Working Group of Sarcopenia 2019 criteria. RESULTS The mean age of the patients was 81.6 ± 6.6 years (42.3% female). Frailty was observed in 34.2% of patients using the J-CHS criteria and 36.9% using the KCL criteria, while severe sarcopenia was identified in 20.2% of patients. In the KCL criteria, the splenic venous flow decreased with the severity of frailty (248.3 ± 148.4, 202.1 ± 177.9, 139.2 ± 81.1 mL/min, P = 0.007), Additionally, the splenic venous flow was significantly lower in frail patients than in robust patients (P = 0.006). This association remained significant even after adjusting for confounding factors such as age, sex, body mass index, habitual drinking, smoking history, diabetes, dyslipidemia, hypertension, systolic blood pressure, atrial fibrillation, heart failure, and history of stroke (P = 0.039). In a parallel analysis, the splenic venous flow was remarkably decreased in patients with sarcopenia (232.0 ± 172.8 vs. 145.0 ± 91.9 mL/min, P = 0.003); however, no significant relationship was found between the severity of frailty and splenic venous flow according to the J-CHS criteria (P = 0.159). Among the J-CHS criteria sub-items, the splenic venous flow was decreased in patients with a decreased appendicular skeletal muscle index (ASMI) (332.9 ± 41.6 vs. 98.5 ± 43.5 mL/min, P = 0.005); however, there was no significant difference in the splenic venous flow between patients with and without decreased walking speed (P = 0.064) or reduced grip strength (P = 0.369). The portal venous flow was not significantly associated with frailty or sarcopenia. CONCLUSION In older patients with cardiovascular disease, a decreased splenic venous flow was observed in those with frailty by the KCL criteria, those with sarcopenia, and those with a decreased ASMI.
Collapse
Affiliation(s)
- Joji Ishikawa
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan.
| | - Shutaro Futami
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Ayumi Toba
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Aya Yamamoto
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Keisho Kobayashi
- Department of Laboratory Testing, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Kana Takani
- Department of Laboratory Testing, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Hideko Ono
- Department of Laboratory Testing, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Teppei Maeda
- Department of Laboratory Testing, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masuyo Kawano
- Department of Laboratory Testing, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Masaru Kiyomizu
- Department of Laboratory Testing, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshiaki Tamura
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism, and Endocrinology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Hideaki Mori
- Department of Medical Education, Kyorin University School of Medicine, Tokyo, Japan
| | - Kazumasa Harada
- Department of Cardiology, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| |
Collapse
|
28
|
Costantino S, Mohammed SA, Ambrosini S, Telesca M, Mengozzi A, Walavalkar K, Gorica E, Herwig M, van Heerebeek L, Xia J, Karsai G, Hornemann T, Dzemali O, Santoro R, Lin Q, Ruschitzka F, Hamdani N, Paneni F. Chromatin Rewiring by SETD2 Drives Lipotoxic Injury in Cardiometabolic HFpEF. Circ Res 2025; 136:1079-1095. [PMID: 40211947 DOI: 10.1161/circresaha.124.325310] [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: 07/25/2024] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Cardiometabolic heart failure with preserved ejection fraction (cHFpEF) is a highly prevalent and deadly condition. Histone 3 trimethylation at lysine 36 (H3k36me3)-a chromatin signature induced by the histone methyltransferase SETD2 (SET domain containing 2)-correlates with changes in gene expression in human failing hearts; however, its role remains poorly understood. This study investigates the role of SETD2 in cHFpEF. METHODS Chromatin immunoprecipitation sequencing and RNA sequencing were used to investigate H3k36me3-related transcriptional regulation. Mice with cardiomyocyte-specific deletion of SETD2 (c-SETD2-/-) were generated and subjected to high-fat diet feeding and L-NAME treatment for 15 weeks to induce cHFpEF. Cardiac function and exercise tolerance were assessed by echocardiography and treadmill exhaustion test. A selective pharmacological inhibitor of SETD2, EZM0414, was also tested in cHFpEF mice. Mechanistic experiments were performed in cultured cardiomyocytes exposed to palmitic acid. SETD2 signaling and the effects of EZM0414 were also investigated in cardiomyocytes from patients with cHFpEF and control donors. RESULTS SETD2 was upregulated in cHFpEF mouse hearts, and its chromatin mark H3k36me3 was involved in lipid metabolism and highly enriched on the promoter of the Srebf1 gene, encoding for SREBP1 (sterol regulatory binding protein 1). SETD2 activation in cHFpEF led to SREBP1 upregulation, triglyceride accumulation, and lipotoxic damage. Of note, cardiomyocyte-specific deletion of SETD2 in mice prevented heart failure with preserved ejection fraction-related hypertrophy, diastolic dysfunction, and lung congestion while improving exercise tolerance. SETD2 deletion blunted H3K36me3 enrichment on Srebf1 promoter, thus leading to a marked rewiring of the cardiac lipidome and restoration of autophagic flux. In vivo treatment with the SETD2 inhibitor EZM0414 recapitulated the effects of SETD2 deletion. Silencing of SETD2 in palmitic acid-treated cardiomyocytes prevented SREBP1 upregulation, whereas SETD2 overexpression mirrored lipotoxic damage. Finally, SETD2 was upregulated in left ventricle specimens from patients with cHFpEF while EZM0414 attenuated cardiomyocyte stiffness. CONCLUSIONS Targeting SETD2 might prevent lipotoxic injury in cHFpEF.
Collapse
Affiliation(s)
- Sarah Costantino
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Shafeeq A Mohammed
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Samuele Ambrosini
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Marialucia Telesca
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy (M.T.)
| | - Alessandro Mengozzi
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (A.M.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Kaivalya Walavalkar
- Department of Molecular Mechanisms of Disease, DMMD, University of Zurich, Switzerland (K.W., R.S.), University of Zurich, Switzerland
| | - Era Gorica
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
| | - Melissa Herwig
- Institute of Physiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Molecular and Experimental Cardiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital (M.H., N.H.), Ruhr University, Bochum, Germany
| | | | - Junyan Xia
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, China (J.X., Q.L.)
| | - Gergely Karsai
- Institute of Clinical Chemistry, University Hospital Zurich (G.K., T.H.), University of Zurich, Switzerland
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University Hospital Zurich (G.K., T.H.), University of Zurich, Switzerland
| | - Omer Dzemali
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- Department of Cardiac Surgery (O.D.), University Hospital Zurich, Switzerland
- Departement of Cardiac Surgery, City Hospital Zurich Triemli, Switzerland (O.D.)
| | - Raffaella Santoro
- Department of Molecular Mechanisms of Disease, DMMD, University of Zurich, Switzerland (K.W., R.S.), University of Zurich, Switzerland
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, China (J.X., Q.L.)
| | - Frank Ruschitzka
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- University Heart Center, Cardiology (F.R., F.P.), University Hospital Zurich, Switzerland
| | - Nazha Hamdani
- Institute of Physiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Molecular and Experimental Cardiology (M.H., N.H.), Ruhr University, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital (M.H., N.H.), Ruhr University, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Hungary (N.H.)
- Department of Physiology, University Maastricht, the Netherlands (N.H.)
| | - Francesco Paneni
- Department of Cardiology, Center for Translational and Experimental Cardiology, University Hospital Zurich (S.C., S.A.M., S.A., M.T., A.M., E.G., J.X., O.D., F.R., F.P.), University of Zurich, Switzerland
- University Heart Center, Cardiology (F.R., F.P.), University Hospital Zurich, Switzerland
| |
Collapse
|
29
|
Ma YN, Ma SR, Yang L, Wu J, Wang YR, Bao LJ, Ma L, Wu QQ, Wang ZH. Diagnostic biomarkers and immune infiltration profiles common to COVID-19, acute myocardial infarction and acute ischaemic stroke using bioinformatics methods and machine learning. BMC Neurol 2025; 25:201. [PMID: 40340571 PMCID: PMC12060493 DOI: 10.1186/s12883-025-04212-6] [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: 11/13/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND COVID-19 is a disease that affects people globally. Beyond affecting the respiratory system, COVID-19 patients are at an elevated risk for both venous and arterial thrombosis. This heightened risk contributes to an increased probability of acute complications, including acute myocardial infarction (AMI) and acute ischemic stroke (AIS). Given the unclear relationship between COVID-19, AMI, and AIS, it is crucial to gain a deeper understanding of their associations and potential molecular mechanisms. This study aims to utilize bioinformatics to analyze gene expression data, identify potential therapeutic targets and biomarkers, and explore the role of immune cells in the disease. METHODS This study employed three Gene Expression Omnibus (GEO) datasets for analysis, which included data on COVID-19, AMI and AIS. We performed enrichment analysis on the co-DEGs for these three diseases to clarify gene pathways and functions, and also examined the relationship between co-DEGs and immune infiltration. Machine learning techniques and protein-protein interaction networks (PPI) were used to identify hub genes within the co-DEGs. Finally, we employed a dual validation strategy integrating independent GEO datasets and in vitro experiments with human blood samples to comprehensively assess the reliability of our experimental findings. RESULTS We identified 88 co-DEGs associated with COVID-19, AMI and AIS. Enrichment analysis results indicated that co-DEGs were significantly enriched in immune inflammatory responses related to leukocytes and neutrophils. Immune infiltration analysis revealed significant differences in immune cell populations between the disease group and the normal group. Finally, genes selected through machine learning methods included: CLEC4E, S100A12, and IL1R2. Based on the PPI network, the top ten most influential DEGs were identified as MMP9, TLR2, TLR4, ITGAM, S100A12, FCGR1A, CD163, FCER1G, FPR2, and CLEC4D. The integration of the protein-protein interaction (PPI) network with machine learning techniques facilitated the identification of S100A12 as a potential common biomarker for early diagnosis and a therapeutic target for all three diseases. Ultimately, validation of S100A12 showed that it was consistent with our experimental results, confirming its reliability as a biomarker. Moreover, it demonstrated good diagnostic performance for the three diseases. CONCLUSION We employed bioinformatics methods and machine learning to investigate common diagnostic biomarkers and immune infiltration characteristics of COVID-19, AMI and AIS. Functional and pathway analyses indicated that the co-DEGs were primarily enriched in immune inflammatory responses related to leukocytes and neutrophils. Through two machine learning approaches and the PPI network, and subsequent validation and evaluation, we identified S100A12 as a potential common therapeutic target and biomarker related to immune response that may influence these three diseases.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Si-Rong Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li Yang
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Juan Wu
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Ya-Rong Wang
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li-Jia Bao
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Li Ma
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Qing-Qiu Wu
- Department of Geriatrics and Specialty Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Zhen-Hai Wang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China.
- Diagnosis and Treatment Engineering Technology Research Center of Nervous System Diseases of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China.
- Neurology Center, Ningxia Medical University General Hospital, Yinchuan, Ningxia, China.
| |
Collapse
|
30
|
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
|
31
|
Wang Q, Zhang X, Han C, Lv Z, Zheng Y, Liu X, Du Z, Liu T, Xue D, Li T, Wang L. Immunodynamic axis of fibroblast-driven neutrophil infiltration in acute pancreatitis: NF-κB-HIF-1α-CXCL1. Cell Mol Biol Lett 2025; 30:57. [PMID: 40335899 PMCID: PMC12060353 DOI: 10.1186/s11658-025-00734-6] [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: 11/01/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is a sterile inflammation, and 10-20% of cases can progress to severe acute pancreatitis (SAP), which seriously threatens human life and health. Neutrophils and their extracellular traps (NETs) play an important role in the progression of AP. However, the immunodynamic factors between the excessive infiltration of neutrophils during the occurrence of AP have not been fully elucidated. METHODS Adult male C57BL/6 J mice were selected. An AP model was induced by cerulein, and a control group was set up. Single-cell sequencing technology was used to reveal the cell atlas of AP pancreatitis tissue. In vivo, the model mice were treated with anti-Ly6G antibody, DNase I, SC75741, PX-478, and SRT3109 respectively. In vitro, human pancreatic stellate cells were treated with hypoxia, H2O2, NAC, and JSH-2, and co-cultured with neutrophils in Transwell chambers. The severity of inflammation was evaluated, and the molecular mechanism by which fibroblasts exacerbate AP was revealed through techniques such as cell colony formation assay, cell migration assay, cell transfection, immunofluorescence, flow cytometry, Western blot, reverse-transcription quantitative polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (co-IP). RESULTS The study showed that the elimination of neutrophils and NETs could significantly improve AP. Single-cell RNA sequencing (scRNA-seq) indicated that both neutrophils and fibroblasts in pancreatic tissue exhibited heterogeneity during AP. Among them, neutrophils highly expressed CXCR2, and fibroblasts highly expressed CXCL1. Further experimental results demonstrated that the infiltration of neutrophils in the early stage of AP was related to the activation of fibroblasts. The activation of fibroblasts depended on the nuclear factor kappa B (NF-κB) signaling pathway induced by hypoxia. NF-κB enhanced the activation of pancreatic stellate cells (PSCs) and the secretion of CXCL1 by directly promoting the transcription of HIF-1α and indirectly inhibiting PHD2, resulting in the accumulation of HIF-1α protein. The NF-κB-HIF-1α signal promoted the secretion of CXCL1 by fibroblasts through glycolysis and induced the infiltration of neutrophils. Finally, blocking the NF-κB-HIF-1α-CXCL1 signaling axis in vivo reduced the infiltration of neutrophils and improved AP. CONCLUSIONS This study, for the first time, demonstrated that activation of fibroblasts is one of the immunological driving factors for neutrophil infiltration and elucidated that glycolysis driven by the NF-κB-HIF-1α pathway is the intrinsic molecular mechanism by which fibroblasts secrete CXCL1 to chemotactically attract neutrophils. This finding provides a highly promising target for the treatment of AP.
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Chenglong Han
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenyi Lv
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zheng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuxu Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiwei Du
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianming Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongbo Xue
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Tao Li
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China.
| | - Liyi Wang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
32
|
Agbadua OG, Kúsz N, Berkecz R, Vass E, Csámpai A, Tóth G, Balogh GT, Marcourt L, Wolfender JL, Queiroz EF, Hunyadi A. New Insights into the French Paradox: Free Radical Scavenging by Resveratrol Yields Cardiovascular Protective Metabolites. J Med Chem 2025. [PMID: 40331971 DOI: 10.1021/acs.jmedchem.4c03061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Resveratrol was subjected to a diversity-oriented synthesis using oxidative transformations by various biorelevant, biomimetic, or biomimetic-related chemical reagents. Using a combined strategy of ultrahigh-resolution profiling, bioactivity screening, and bioactivity-guided isolation, 19 metabolites were obtained. The compounds were tested for their in vitro enzyme inhibitory activity on angiotensin-1 converting enzyme (ACE), cyclooxygenase-1 and -2, and 15-lipoxygenase (LOX), and evaluated for their relevant drug-like properties in silico. The compounds demonstrated a generally increased cardiovascular protective and anti-inflammatory potential and better drug-likeness compared to resveratrol. Trans-δ-viniferin (6) was identified as a competitive, C-domain-selective ACE inhibitor that is over 20 times more potent than resveratrol. Further, trans-ε-viniferin (2) acted as an over 40 times stronger LOX inhibitor than resveratrol. While our results cannot be directly translated to the health benefits of dietary resveratrol consumption without further studies, it is demonstrated that biologically relevant oxidative environments transform resveratrol into potent cardiovascular protective and anti-inflammatory metabolites.
Collapse
Affiliation(s)
- Orinamhe G Agbadua
- Institute of Pharmacognosy, University of Szeged, Eötvös str. 6, H-6720 Szeged, Hungary
| | - Norbert Kúsz
- Institute of Pharmacognosy, University of Szeged, Eötvös str. 6, H-6720 Szeged, Hungary
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, University of Szeged, Somogyi str. 4, H-6720 Szeged, Hungary
| | - Elemér Vass
- Department of Organic Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/a, H-1117 Budapest, Hungary
| | - Antal Csámpai
- Department of Organic Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/a, H-1117 Budapest, Hungary
| | - Gábor Tóth
- NMR Group, Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - György T Balogh
- Department of Pharmaceutical Chemistry, Semmelweis University, H-1092 Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, H-1085 Budapest, Hungary
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, Eötvös str. 6, H-6720 Szeged, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Eötvös str. 6, H-6720 Szeged, Hungary
- Graduate Institute of Natural Products, Shih-Chuan first Rd. 100, Kaohsiung 807, Taiwan
| |
Collapse
|
33
|
Zhu T, Ding Y, Wu X, Li Y, Cheng G, Wang N, Yang Q, Zhang W, Chen X, Liu X. Pentraxin 3 promotes the expression of pro-inflammatory cytokines and the migration of macrophages in myocarditis. BMC Cardiovasc Disord 2025; 25:354. [PMID: 40335910 PMCID: PMC12060373 DOI: 10.1186/s12872-025-04790-w] [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: 11/24/2024] [Accepted: 04/21/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND This study aims to investigate the expression of Pentraxin 3 (PTX3) and Nod-like receptor family pyrin domain-containing 3 (NLRP3) in myocarditis and to elucidate their roles and potential interplay in the pathogenesis of myocarditis. METHODS Immunofluorescence staining was performed on myocardial autopsy specimens from deceased patients with severe myocarditis or severe trauma. H9C2 cardiomyocytes were divided into five groups: Control, Lipopolysaccharide (LPS), LPS + PTX3 overexpression, LPS + small interfering RNA targeting PTX3 (si-PTX3), and LPS + PTX3 overexpression + si-NLRP3. The expression levels of PTX3 and NLRP3 at the RNA level were quantified using quantitative real-time polymerase chain reaction (qPCR), while protein expression was assessed via western blot. The concentrations of interleukin-1β (IL-1β) and IL-18 were determined by enzyme-linked immunosorbent assay (ELISA). Macrophages migration was evaluated using Transwell assays. RESULTS Immunofluorescence staining revealed co-localization and increased expression of PTX3 and NLRP3 in the myocardium of patients with severe myocarditis. In vitro experiments demonstrated that PTX3 enhanced the expression of NLRP3, IL-1β, and IL-18 in LPS-stimulated cardiomyocytes. Furthermore, PTX3 was shown to promote macrophage migration by regulating NLRP3 expression, as assessed by Transwell assays. CONCLUSION Our findings suggest that PTX3-mediated NLRP3 activation contributes to inflammatory responses and promotes macrophage migration in myocarditis. This study provides a foundation for future investigations into PTX3-targeted therapies for myocarditis.
Collapse
Affiliation(s)
- Tianyu Zhu
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Ying Ding
- Department of Nephrology, The Second Medical Center of Chinese PLA General Hospital, National Clinical Research Centre for Geriatric Diseases, Beijing, 100853, P.R. China
| | - Xiaohui Wu
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Yan Li
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Guanliang Cheng
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Ning Wang
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Quan Yang
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Wenchao Zhang
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| | - Xuezhi Chen
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China.
| | - Xiaohui Liu
- Department of Cardiology, Peking University International Hospital, Beijing, 102206, P.R. China
| |
Collapse
|
34
|
Zhao Z, Wei TT, Zhang WX, Zhang SS, Wu R, Li F, Yang H, Zhang Q, Xi J, Zhou Y, Wang T, Du J, Lu QB, Ge Q. Association of homoarginine with arginine and disease severity in COVID-19 patients. Amino Acids 2025; 57:24. [PMID: 40332615 PMCID: PMC12058869 DOI: 10.1007/s00726-025-03453-6] [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: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
This study explored the relationship between the concentrations of homoarginine and arginine and between homoarginine concentration and laboratory parameters in coronavirus disease 2019 (COVID-19) patients with different severity to demonstrate the role of homoarginine in the progress of COVID-19. The laboratory-confirmed COVID-19 patients were included from Peking University Third Hospital during December 2022 to January 2023. Serum, urine, and stool samples were collected from the patients and detected by liquid chromatography-mass spectrometry. Totally 46 patients were recruited, including 18 in the mild group, 19 in the severe group, and 9 fatal. The concentration of homoarginine was positively correlated with the concentration of arginine in serum (r = 0.50), urine (r = 0.55), and stool samples (r = 0.39), respectively (all P < 0.001). The serum concentration and urine concentration of homoarginine were lower in severe patients than in mild patients (both P < 0.05). 13 indicators reflecting immunity and coagulation, including but not limited to T cell, white blood cell, natural killer cell, interleukin 6 (IL-6), and IL-8, had statistically significant correlations with both disease severity and the homoarginine concentration. Patients with hypertension were significantly associated with the decreased serum homoarginine (odds ratio 10.905, 95% confidence interval 1.454 - 137.144). Our results suggest that the homoarginine plays a role in the progress of COVID-19, which may be achieved by influencing arginine metabolism.
Collapse
Affiliation(s)
- Zhiling Zhao
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Ting-Ting Wei
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Wan-Xue Zhang
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Shan-Shan Zhang
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Rui Wu
- Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Fei Li
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Han Yang
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Qiang Zhang
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Jingjing Xi
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Yiguo Zhou
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Tiehua Wang
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Juan Du
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China
| | - Qing-Bin Lu
- Department of Laboratorial Science and Technology and Vaccine Research Center, School of Public Health, Peking University, Beijing, China.
- Center for Infectious Disease and Policy Research and Global Health and Infectious Diseases Group, Peking University, Beijing, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China.
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China.
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
- Center for Infectious Disease and Policy Research and Department of Laboratorial of Science and Technology, School of Public Health, Peking University, No 38 Xue-Yuan Rd, Haidian District, Beijing, 100191, China.
| | - Qinggang Ge
- Department of Intensive Care Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing, 100191, China.
| |
Collapse
|
35
|
Delk SC, Gurgis FW, Reddy ST. Mechanisms and Applications of Apolipoproteins and Apolipoprotein Mimetic Peptides: Common Pathways in Cardiovascular Disease and Cancer. Semin Cancer Biol 2025:S1044-579X(25)00057-4. [PMID: 40345461 DOI: 10.1016/j.semcancer.2025.05.006] [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: 02/16/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Apolipoproteins are the defining functional component of lipoproteins and play critical roles in lipid transport and metabolism. High-density lipoprotein (HDL) and its primary functional constituent, apolipoprotein A-I, are of particular importance because of anti-inflammatory and antioxidant properties. Apolipoprotein mimetic peptides are short-chain amino acids designed to mimic the functions and alpha-helical structure of endogenous apolipoproteins and have demonstrated efficacy in ameliorating animal models of cardiovascular disease (CVD) and cancer. The mechanisms underlying the mimetics are yet to be fully elucidated, but a comprehensive review of the literature suggests that the peptides attack pathways shared in the pathophysiology of both diseases. This review also discusses the many pre-clinical studies on the mimetic peptides, highlighting possible mechanisms at work in each. Proposed mechanisms of protection against CVD and cancer include binding and removal of pro-inflammatory oxidized lipids, reduction in reactive oxygen species, and modulation of immune cell populations. Additionally, nanoparticles (NP) formulations incorporating apolipoprotein mimetic peptides or recombinant apolipoproteins have exhibited anti-atherogenic and anti-cancer activity. To date, clinical trials to assess the effect of reconstituted HDL NPs on CVD outcomes have not shown significant improvement. The large body of successful animal studies on apolipoproteins and apolipoprotein mimetic peptides presents a disconnect between pre-clinical and clinical efficacy, highlighting the need for a more complete understanding of the underlying pathways and mechanisms.
Collapse
Affiliation(s)
- Samuel C Delk
- Environmental and Molecular Toxicology Interdepartmental Degree Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA
| | - Faheem W Gurgis
- Department of Emergency Medicine, University of Florida College of Medicine, Gainesville, Florida, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA
| | - Srinivasa T Reddy
- Environmental and Molecular Toxicology Interdepartmental Degree Program, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA; Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095 USA.
| |
Collapse
|
36
|
Xu J, Wang H, Han B, Zhang X. Mechanisms through which laparoscopic sleeve gastrectomy mitigates atherosclerosis risk: a focus on visceral adipose tissue. Eur J Med Res 2025; 30:370. [PMID: 40336107 PMCID: PMC12057030 DOI: 10.1186/s40001-025-02635-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Bariatric surgery is currently considered the key treatment method for patients with obesity and related complications. Among the various surgeries, laparoscopic sleeve gastrectomy (LSG) is the most widely used. Obesity is a multifactor chronic disease characterized by the accumulation of visceral adipose tissue (VAT), leading to susceptibility to cardiac metabolic diseases. Many mechanisms, including abnormal lipid metabolism, insulin resistance, inflammation, endothelial dysfunction, adipocytokine imbalance and inflammasome activation, have been identified as the basis for the relationship between obesity and atherosclerosis. Bariatric surgery, such as LSG, reduces the risk of atherosclerosis in people living with obesity by reducing energy intake, disrupting energy balance and reducing the secretion of intestinal hormones to intervene in these risk factors. This review explores the current understanding of how LSG affects VAT and its impact on the risk of atherosclerosis.
Collapse
Affiliation(s)
- Juan Xu
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| | - Heyue Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Bin Han
- Shanxi Provincial People's Hospital, Taiyuan, 030032, China
| | - Xiaomin Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| |
Collapse
|
37
|
Yu B, Sopic M, Sluimer JC. Single-cell RNA sequencing (scRNA-seq) and its insights into cellular heterogeneity in atherosclerosis. Vascul Pharmacol 2025:107499. [PMID: 40345606 DOI: 10.1016/j.vph.2025.107499] [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: 01/22/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of cellular diversity in human biology, providing novel insights into disease mechanisms. In cardiovascular disease (CVD), scRNA-seq enables precise mapping of complex cell populations, uncovering unique cell types and states that influence disease progression and suggest new therapeutic targets. In atherosclerosis (AS), scRNA-seq has redefined plaque pathology by identifying distinct cell types, including endothelial cells (ECs), smooth muscle cells (SMCs), fibroblasts, macrophages, T cells, and B cells, each with specific roles in plaque stability, inflammation, and disease progression. In our review, we summarized these major cellular populations and their cellular heterogeneity in non-diseased and atherosclerotic aorta, as identified by scRNA-seq in mice and human tissues. We discussed conserved and species-specific subpopulations, their defining markers, and their functional implications in plaque progression. In addition, we integrated findings from scRNA-seq with experimental studies to highlight key molecular targets with therapeutic potential. In the future, these insights offer a refined cellular and molecular framework of atherosclerosis and may help the development of targeted interventions to promote plaque stabilization and reduce cardiovascular risk.
Collapse
Affiliation(s)
- Baixue Yu
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands.
| | - Miron Sopic
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg; Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia.
| | - Judith C Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, 6229 HX Maastricht, the Netherlands; Aachen Maastricht Institute for CardioRenal research (AMICARE), 52074 Aachen, Germany; British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
38
|
Mroueh A, Algara-Suarez P, Fakih W, Gong DS, Matsushita K, Park SH, Amissi S, Auger C, Kauffenstein G, Meyer N, Ohlmann P, Jesel L, Pieper MP, Marchandot B, Morel O, Mazzucotelli JP, Schini-Kerth VB. SGLT2 expression in human vasculature and heart correlates with low-grade inflammation and causes eNOS-NO/ROS imbalance. Cardiovasc Res 2025; 121:643-657. [PMID: 39739876 DOI: 10.1093/cvr/cvae257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/31/2024] [Accepted: 11/12/2024] [Indexed: 01/02/2025] Open
Abstract
AIMS Sodium-glucose co-transporter 2 inhibitors (SGLT2i) show a cardioprotective effect in heart failure and myocardial infarction, pathologies often associated with low-grade inflammation. This cross-sectional study aims to investigate whether low-grade inflammation regulates SGLT2 expression and function in human vasculature, heart, and endothelial cells (ECs). METHODS AND RESULTS Human internal thoracic artery (ITA), left ventricle (LV) specimens, and cultured porcine coronary artery ECs were used. Expression of target molecules was assessed using RT-qPCR, western blot analysis, and immunofluorescence staining, and the generation of reactive oxygen species (ROS) and nitric oxide (NO) using fluorescent probes. The function of SGLT2 was investigated using empagliflozin and SGLT1 or 2 siRNA. SGLT2 mRNA and protein levels in ITA and LV specimens were correlated with the level of low-grade inflammation, markers of the angiotensin system, and EC activation. SGLT2 staining was observed in the ITA endothelium and smooth muscle, the coronary microcirculation, and cardiomyocytes. Elevated ROS formation in high SGLT2-expressing specimens was reduced by inhibition of the angiotensin system, SGLT2, and TNF-α. Exposure of ECs to IL-1ß, IL-6, and TNF-α led to an increase in SGLT1 and SGLT2 mRNA and protein expression, up-regulation of components of the angiotensin system, enhanced ROS and decreased NO formation, and activation of NF-κB. The stimulatory effect of TNF-α was prevented by N-acetylcysteine and inhibition of the angiotensin system, SGLT2 but not SGLT1, and NF-κB. CONCLUSION Low-grade inflammation is closely associated with SGLT2 expression in human vasculature and heart, and this response contributes to a feedforward mechanism with the AT1R/NADPH oxidase pathway to cause eNOS-NO/ROS imbalance.
Collapse
Affiliation(s)
- Ali Mroueh
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Paola Algara-Suarez
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
- Faculty of Pharmacy, Strasbourg University, Strasbourg 67000, France
| | - Walaa Fakih
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Dal-Seong Gong
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Kensuke Matsushita
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
- Division of Cardiovascular Medicine, Nouvel Hoôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Sin-Hee Park
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Said Amissi
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Cyril Auger
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Gilles Kauffenstein
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
| | - Nicolas Meyer
- Department of Biostatistics, Strasbourg University Hospital, Strasbourg, France
| | - Patrick Ohlmann
- Division of Cardiovascular Medicine, Nouvel Hoôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Laurence Jesel
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
- Division of Cardiovascular Medicine, Nouvel Hoôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | | | - Benjamin Marchandot
- Division of Cardiovascular Medicine, Nouvel Hoôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Olivier Morel
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
- Division of Cardiovascular Medicine, Nouvel Hoôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Jean-Philippe Mazzucotelli
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
- Division of Cardiac Surgery and Heart Transplant, Nouvel Hoôpital Civil, Strasbourg University Hospital, Strasbourg 67091, France
| | - Valérie B Schini-Kerth
- Translational Cardiovascular Medicine UR 3074, FMTS, 1 rue Eugène Boeckel, Strasbourg 67084, France
- Faculty of Pharmacy, Strasbourg University, Strasbourg 67000, France
| |
Collapse
|
39
|
Oryani MA, Mohammad Al-Mosawi AK, Javid H, Tajaldini M, Karimi-Shahri M. A Bioligical Perspective on the role of miR-206 in Colorectal cancer. Gene 2025:149552. [PMID: 40339768 DOI: 10.1016/j.gene.2025.149552] [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: 11/18/2024] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
MicroRNAs (miRs) have emerged as pivotal regulators in the development and progression of colorectal cancer (CRC), and MicroRNA-206 (miR-206) has garnered attention as a potentially influential factor. However, the specific biological functions and complete mechanistic understanding of miR-206 in CRC remain largely uncharacterized. This study aims to bridge this research gap by providing a comprehensive analysis of miR-206's role in CRC. An exploration of the molecular mechanisms regulated by miR-206, its intricate interplay with target genes, and its significant impact on cellular processes highlights its potential utility as both a diagnostic marker and a therapeutic target. The significance of this research lies in potentially enabling the development of innovative therapeutic approaches, ultimately aiming to improve prognosis and survival rates in CRC patients by elucidating the functions of miR-206. Critical pathways, such as c-Met and PTEN/AKT, play crucial roles within the regulatory network of miR-206 in CRC and impact various cellular processes involved in CRC pathogenesis, metastasis, and treatment response. Understanding the complex interactions between miR-206 and key signaling pathways like c-Met and PTEN/AKT is crucial for understanding the underlying mechanisms driving CRC initiation and progression. This knowledge can inform the development of targeted therapeutic interventions, potentially leading to improved patient outcomes and advances in CRC management.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboubeh Tajaldini
- Ischemic Disorder Research Center, Golestan University of Medical Sciences. Gorgan, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
40
|
Arindkar SK, Singh S, Kumar MJM, Nagarajan P. The impact of leptin receptor (LepR) mutation on the development of MASLD in a murine model. Gene 2025:149550. [PMID: 40339770 DOI: 10.1016/j.gene.2025.149550] [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: 01/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
Genes play an important role in regulating insulin signaling, adipokines, oxidative stress, lipid metabolism, and inflammation in susceptibility and progression of Metabolic dysfunction-associated steatotic liver disease (MASLD). Among various genes, the LepR gene influences insulin sensitivity and controls lipid metabolism, contributing to the development of MASLD. Our previous study reported that a novel congenic mouse (WSB.db) with a LepR mutation exhibited resistance to MASLD. To further evaluate this strain for resistance, we fed this new mouse strain with LepR mutation and B6.db mice, the mouse model of metabolic disease with a high-fat diet as a second hit for 12 weeks and evaluated the pathophysiology, serum biochemistry, Quantitative real-time polymerase chain reaction (qPCR) to determine the expression of specific genes involved in the development of fatty changes in the liver and hepatic transcriptome signatures in liver tissue. In contrast to db/db (B6.db) mice, which exhibited all the pathological hallmarks for MASLD, the LepR mutant congenic strain was still resistant to developing liver steatosis. Transcriptome analysis with KEGG PATHWAY: hsa04932 revealed significant upregulation of AMPKγ3 and MApk10 (JNK3) in WSB.db mice, suggesting that congenic mice with the LepR mutation are resistant to MASLD without the liver pathology to effect. These results propose that the LepR mutation has a different impact on liver pathology depending on genetic background, indicating upregulation of specific genes in the development of MASLD. This study will facilitate the identification of therapeutic targets against MASLD with LepR mutation.
Collapse
Affiliation(s)
| | - Surender Singh
- BRIC NII - Experimental Animal Facility, National Institute of Immunology, New Delhi 100 067, India
| | | | - Perumal Nagarajan
- BRIC NII - Experimental Animal Facility, National Institute of Immunology, New Delhi 100 067, India.
| |
Collapse
|
41
|
Suleman S, Ängquist L, Linneberg A, Hansen T, Grarup N. Exploring the genetic intersection between obesity-associated genetic variants and insulin sensitivity indices. Sci Rep 2025; 15:15761. [PMID: 40328835 PMCID: PMC12056085 DOI: 10.1038/s41598-025-98507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
Insulin sensitivity (IS) is a key determinant of metabolic health and may share genetic factors with obesity-related traits. Previous large-scale genetic studies have identified variants associated with IS as well as obesity related traits like body mass index (BMI) and waist-to-hip ratio (WHR). Notably, many of these associations are shared across traits, indicating a potential genetic overlap. However, the genetic intersection between IS and obesity-related traits remains underexplored. To explore this gap, we investigated associations between six IS indices, including fasting and post-glucose load measures, and genetic variants linked to BMI and WHR to determine their influence on IS and related cardiometabolic traits. To achieve this, we calculated six IS indices using fasting and oral glucose tolerance test (OGTT) data from 5,007 non-diabetic individuals, grouping them into fasting, OGTT0,120, and OGTT0,30,120 categories. A total of 678 BMI-associated and 265 WHR-associated genetic variants were analysed using linear regression, adjusting for age and sex, with sex-specific analyses for WHR. Analyses were conducted with and without BMI adjustments and corrected for multiple testing (padj). Additionally, we explored the relationship between IS-linked variants and their associations with type 2 diabetes (T2D), coronary artery disease (CAD) and stroke. Among the 678 BMI-associated variants, 100 showed nominal associations (p < 0.05) with at least one IS index; and 20 remained significant after multiple testing correction (padj < 0.05) when not adjusting for BMI. After adjusting for BMI, 70 variants retained nominal associations, and six remained significant (padj < 0.05). In sex-specific analyses of the 265 WHR-associated variants, 12 variants were associated in females when adjusted for BMI, whereas no significant associations were observed in males. Furthermore, BMI- and WHR-associated variants linked to decreased IS, such as those in FTO and VPS13C loci, were also associated with increased T2D and stroke risk, whereas IS-increasing variants, including those in VPS13C and PPARG, were linked to lower T2D and stroke risk, with some, like THADA, showing opposing effects on CAD. This study offers insights into genetic variants that influence both IS and obesity-related traits, revealing BMI- and WHR-associated variants with both positive and negative effects on IS and their potential impact on cardiometabolic health.
Collapse
Affiliation(s)
- Sufyan Suleman
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Human Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Lars Ängquist
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
42
|
Obmann D, Münch P, Graf B, von Jouanne-Diedrich H, Zausig YA. Comparison of different AI systems for diagnosing sepsis, septic shock, and cardiogenic shock: a retrospective study. Sci Rep 2025; 15:15850. [PMID: 40328810 PMCID: PMC12056228 DOI: 10.1038/s41598-025-00830-9] [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: 12/27/2024] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Sepsis, septic shock, and cardiogenic shock are life-threatening conditions associated with high mortality rates, but differentiating them is complex because they share certain symptoms. Using the Medical Information Mart for Intensive Care (MIMIC)-III database and artificial intelligence (AI), we aimed to increase diagnostic precision, focusing on Bayesian network classifiers (BNCs) and comparing them with other AI methods. Data from 5970 adults, including 950 patients with cardiogenic shock, 1946 patients with septic shock, and 3074 patients with sepsis, were extracted for this study. Of the original 51 variables included in the data records, 12 were selected for constructing the predictive model. The data were divided into training and validation sets at an 80:20 ratio, and the performance of the BNCs was evaluated and compared with that of other AI models, such as the one rule classifier (OneR), classification and regression tree (CART), and an artificial neural network (ANN), in terms of accuracy, sensitivity, specificity, precision, and F1-score. The BNCs exhibited an accuracy of 87.6% to 91.5%. The CART model demonstrated a notable 91.6% accuracy when only three decision levels were used, whereas the intricate ANN model reached 90.5% accuracy. Both the BNCs and the CART model allowed clear interpretation of the predictions. BNCs have the potential to be valuable tools in diagnostic tasks, with an accuracy, sensitivity, and precision comparable, in some cases, to those of ANNs while demonstrating superior interpretability.
Collapse
Affiliation(s)
- Dirk Obmann
- Department of Anaesthesiology and Critical Care, Klinikum Aschaffenburg-Alzenau, Aschaffenburg, Germany.
- Department of Anaesthesiology, University of Regensburg, Regensburg, Germany.
| | - Philipp Münch
- Faculty of Engineering, Competence Centre for Artificial Intelligence, TH Aschaffenburg (University of Applied Sciences), Aschaffenburg, Germany
| | - Bernhard Graf
- Department of Anaesthesiology, University of Regensburg, Regensburg, Germany
| | - Holger von Jouanne-Diedrich
- Faculty of Engineering, Competence Centre for Artificial Intelligence, TH Aschaffenburg (University of Applied Sciences), Aschaffenburg, Germany
| | - York A Zausig
- Department of Anaesthesiology and Critical Care, Klinikum Aschaffenburg-Alzenau, Aschaffenburg, Germany
- Department of Anaesthesiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
43
|
Mannheimer B, Lindh JD, Fahlén CB, Issa I, Falhammar H, Skov J. Drug-induced hyponatremia in clinical care. Eur J Intern Med 2025:S0953-6205(25)00175-X. [PMID: 40328519 DOI: 10.1016/j.ejim.2025.04.034] [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: 02/28/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
PURPOSE Over the last decades, advances in understanding of previously described associations have important implications for diagnosis and workup of hyponatremia. In addition, new drug groups potentially affecting sodium balance and water homeostasis have evolved. The aim of this review is to summarize current evidence on drug-induced hyponatremia in clinical care. METHODS We searched PubMed using the string "Inappropriate ADH Syndrome/chemically induced"[Mesh] OR "Inappropriate ADH Syndrome/diagnosis"[Mesh]) OR ("Hyponatremia/chemically induced"[Mesh] OR "Hyponatremia/diagnosis"[Mesh]), January 1st, 2008, to September 2nd 2024. In total 2003 articles were found and reviewed. Relevant articles referenced herein were subsequently traced backwards and also reviewed. RESULTS Drugs associated with hyponatremia, including selective serotonin reuptake inhibitors, antipsychotics, antiepileptic drugs and proton pump inhibitors, typically cause hyponatremia shortly after initiation of treatment. For thiazide diuretics, the number one culprit in drug-induced hyponatremia, the risk for hyponatremia is highest the first weeks after initiation and then gradually decreases to a stable but still increased level after around 3 months. Several drugs that promote a negative water balance such as loop diuretics, lithium and of sodium-glucose cotransporter-2 inhibitors appear to decrease the risk for hyponatremia. Treatment with immune checkpoint inhibitors is associated with an increased risk of hypophysitis and adrenalitis resulting in hyponatremia due to secondary and primary cortisol deficiency. CONCLUSION For most drugs associated with hyponatremia, including thiazides, the cause-effect relationship is tightly linked to newly initiated treatment. Further research is warranted to characterize the association between hyponatremia and newly developed drugs such as sodium-glucose cotransporter-2 inhibitors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Buster Mannheimer
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Department of Medicine, Södersjukhuset, Stockholm, Sweden.
| | - Jonatan D Lindh
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Bergh Fahlén
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Department of Medicine, Södersjukhuset, Stockholm, Sweden
| | - Issa Issa
- Department of Clinical Science and Education at Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Department of Medicine, Södersjukhuset, Stockholm, Sweden
| | - Henrik Falhammar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | - Jakob Skov
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Medicine, Karlstad Central Hospital, Region Värmland, Sweden
| |
Collapse
|
44
|
Lacoste B, Prat A, Freitas-Andrade M, Gu C. The Blood-Brain Barrier: Composition, Properties, and Roles in Brain Health. Cold Spring Harb Perspect Biol 2025; 17:a041422. [PMID: 38951020 PMCID: PMC12047665 DOI: 10.1101/cshperspect.a041422] [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: 07/03/2024]
Abstract
Blood vessels are critical to deliver oxygen and nutrients to tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier (BBB), which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and protects the neural tissue from toxins and pathogens, and alterations of this barrier are important components of the pathogenesis and progression of various neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the brain endothelial cells (ECs) that form the walls of the blood vessels. These properties are regulated by interactions between different vascular, perivascular, immune, and neural cells. Understanding how these cell populations interact to regulate barrier properties is essential for understanding how the brain functions in both health and disease contexts.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Moises Freitas-Andrade
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
| | - Chenghua Gu
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
45
|
Cheng CN, Lee CW, Lee CH, Tang SC, Kuo CH. Elucidating stroke etiology through lipidomics of thrombi and plasma in acute ischemic stroke patients undergoing endovascular thrombectomy. J Cereb Blood Flow Metab 2025:271678X251327944. [PMID: 40322967 PMCID: PMC12052910 DOI: 10.1177/0271678x251327944] [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: 10/15/2024] [Revised: 02/04/2025] [Accepted: 02/27/2025] [Indexed: 05/08/2025]
Abstract
Acute ischemic stroke (AIS) requires detailed etiology information to guide optimal management. Given the pivotal role of lipids in AIS, we conducted a comprehensive lipidomics analysis of paired thrombi and plasma from AIS patients, correlating the findings with stroke etiology. Patients were recruited across four etiologies: cardioembolism (CE), large artery atherosclerosis (LAA), active cancer (Cancer), and undetermined. Plasma and thrombi were collected before and during endovascular thrombectomy and analyzed using in-house targeted lipidomics. Among 51 patients (37 CE, 7 LAA, 4 Cancer, and 3 undetermined), we identified 37 and 70 lipid species significantly different between thrombi in CE and LAA, and CE and Cancer, respectively (FDR-corrected P < 0.05). No significant differences were observed in plasma. Notably, 21 diacylglycerols and 11 polyunsaturated triacylglycerols were depleted (2.5 to 12 folds) in LAA compared to CE, while 10 ceramides and 57 glycerophospholipids were elevated in Cancer. With 80% validation accuracy, 29 and 59 lipids distinguished LAA and Cancer from CE, respectively. A neural network model using these lipids effectively classified undetermined patients. This study emphasizes the significance of thrombus lipids in distinguishing between LAA, CE, and Cancer etiologies in AIS, enhancing our understanding of stroke pathophysiology and informing future clinical managements.
Collapse
Affiliation(s)
- Chih-Ning Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chung-Wei Lee
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Hua Lee
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sung-Chun Tang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
46
|
Zhao Y, Wen J, Yang Y, Jia L, Ma Q, Jia W, Qi W. In vitro fermentation characteristics of polysaccharide from Scrophularia ningpoensis and its effects on type 2 diabetes mellitus gut microbiota. PeerJ 2025; 13:e19374. [PMID: 40343088 PMCID: PMC12060902 DOI: 10.7717/peerj.19374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Background Increasing evidence has shown a close relation between the pathogenesis of type 2 diabetes mellitus (T2DM), which is a global health problem with multifactorial etiopathogenesis, and gut microbiota. Methods During in-vitro fermentation of Scrophularia ningpoensis (known as Xuanshen) polysaccharide (SNP) by T2DM gut microbiota, effects of SNP on the gas content, production of short-chain fatty acids (SCFAs), metabolite profile and microbiota composition were studied. Results Analysis of chemical compositions indicates that the total sugar content of SNP was found to be as high as 87.35 ± 0.13% (w/w). SNP treatment significantly improved the gas volume and composition in T2DM fecal matter. Moreover, intestinal flora degraded SNP to produce SCFAs, thus regulating SCFA production and composition. Metabolomic analysis implied that SNP shows potential to regulate the five gut metabolites (L-valine, L-leucine, L-isoleucine, L-alanine, and xylitol) in T2DM fecal matter. Furthermore, dysbiosis of gut microbiota induced by T2DM was reversed by SNP. The evidence includes decreasing Firmicutes/Bacteroidota ratio at phylum level promoting proliferation of the bacterial abundance of Dorea, Parabacteroides, Faecalibacterium, and Lachnospira and decreased bacterial abundance of Escherichia-Shigella. Based on these findings, the action mechanism of SNP against T2DM was clarified by reshaping microbiota and regulating intestinal metabolites, and a novel target was provided for interventions of T2DM.
Collapse
Affiliation(s)
- Yang Zhao
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
| | - Juwei Wen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
| | - Yu Yang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
| | - Lina Jia
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Qian Ma
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Weiguo Jia
- The Center of Gerontology and Geriatrics, National Clinical Research Center of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Qi
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, China
| |
Collapse
|
47
|
Cotter G, Petrie MC, Butler J, Davison B, Chioncel O, Biegus J, Pagnesi M, Voors AA, Metra M, Ponikowski P, Mann D, Bhatt DL. Obesity and inflammation in chronic and acute heart failure. Heart Fail Rev 2025:10.1007/s10741-025-10518-x. [PMID: 40319221 DOI: 10.1007/s10741-025-10518-x] [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] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Obesity and inflammation have been associated with an increased incidence of heart failure (HF) and death. However, until recent years, no therapy directed towards reducing inflammation and reducing obesity has been shown to reduce those adverse outcomes. Over the past few years, a few small studies have suggested that improving obesity-and in even smaller studies, reducing inflammation-may help improve HF severity, congestion, quality of life, and possibly outcomes. Larger studies that are being planned and executed, which will report their results within the next 2-3 years, should help further clarify the effects of weight and inflammation reduction in patients with HF.
Collapse
Affiliation(s)
- Gad Cotter
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France.
- Momentum Research Inc, Durham, NC, USA.
- , Heart Initiative, Durham, NC, USA.
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, Scotland, UK
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- Department of Medicine, University of Mississippi, Jackson, MS, USA
| | - Beth Davison
- Université Paris Cité, INSERM UMR-S 942 (MASCOT), Paris, France
- Momentum Research Inc, Durham, NC, USA
- , Heart Initiative, Durham, NC, USA
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases "Prof C C Iliescu", University of Medicine "Carol Davila", Bucharest, Romania
| | - Jan Biegus
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Matteo Pagnesi
- Cardiology, Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Marco Metra
- Cardiology, Cardiology, ASST Spedali Civili and Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Brescia, Italy
| | - Piotr Ponikowski
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Douglas Mann
- Cardiovascular Division, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Deepak L Bhatt
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Affiliation(s)
- Robert Sykes
- School of Cardiovascular and Metabolic Health, University of Glasgow, University Place, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Agamemnon Street, Clydebank G81 4DY, UK
| | - Daniel T Y Ang
- School of Cardiovascular and Metabolic Health, University of Glasgow, University Place, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Agamemnon Street, Clydebank G81 4DY, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, University Place, Glasgow G12 8TA, UK
- West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Agamemnon Street, Clydebank G81 4DY, UK
| |
Collapse
|
49
|
Skalante O, Hachimi Idrissi M, Cherkaoui S, Benaissa E, Ben Lahlou Y, Chadli M. Cardiac tamponade revealing tuberculous pericarditis: a case report. Access Microbiol 2025; 7:000983.v4. [PMID: 40321681 PMCID: PMC12048714 DOI: 10.1099/acmi.0.000983.v4] [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: 01/19/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Tuberculosis remains a major public health issue in Morocco. Pulmonary tuberculosis is the most common form, but various extrapulmonary forms exist. Tuberculous pericarditis is a rare form of extrapulmonary tuberculosis that can be complicated by cardiac tamponade, pericardial constriction or their combination, which can threaten the patient's life. Its clinical and radiological signs are nonspecific, and the clinical presentation can be misleading and incomplete, sometimes even with an initial tamponade. We report the case of a 68-year-old female patient admitted for intense retrosternal chest pain associated with acute dyspnoea, evolving in the context of unquantified weight loss and general deterioration. Additionally, she reported a history of fever and night sweats. Clinical examination revealed a conscious, febrile, hypotensive, tachycardic, polypneic patient with good oxygen saturation, signs of right heart failure and muffled heart sounds on auscultation. Chest X-ray revealed cardiomegaly, and the ECG showed diffuse low voltage. Given the presence of Beck's triad suggestive of cardiac tamponade, a transthoracic echocardiogram was performed, revealing a large pericardial effusion with a 'swinging heart'. A chest CT scan also confirmed the large pericardial effusion. The diagnosis of cardiac tamponade was made based on the clinical and radiological findings, and pericardial drainage was performed, after which the patient showed clinical improvement. PCR GenXpert MTB/RIF Ultra detected the presence of Mycobacterium tuberculosis in the pericardial fluid, with no resistance to rifampicin. Culture was positive for M. tuberculosis. The diagnosis of tuberculous pericarditis was, thus, confirmed, and the patient was started on quadruple antituberculosis therapy with good clinical progress.
Collapse
Affiliation(s)
- Oumaima Skalante
- Department of Bacteriology, Mohammed V Military Teaching Hospital, Rabat, Morocco
| | | | - Soukaina Cherkaoui
- Department of Cardiology, Mohammed V Military Teaching Hospital, Rabat, Morocco
| | - Elmostafa Benaissa
- Department of Bacteriology, Mohammed V Military Teaching Hospital, Rabat, Morocco
| | - Yassine Ben Lahlou
- Department of Bacteriology, Mohammed V Military Teaching Hospital, Rabat, Morocco
| | - Mariama Chadli
- Department of Bacteriology, Mohammed V Military Teaching Hospital, Rabat, Morocco
| |
Collapse
|
50
|
Gigase FAJ, Boekhorst MMGBM, Suleri A, Rommel AS, Breen M, Muetzel RL, Hillegers MHJ, Elovitz MA, Steegers EAP, De Witte LD, Bergink V. Maternal Immune Activation During Pregnancy and Obstetric Outcomes: A Population-Based Cohort Study. BJOG 2025. [PMID: 40313214 DOI: 10.1111/1471-0528.18191] [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: 09/23/2024] [Revised: 03/19/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVE Maternal immune activation has been proposed as a mechanism for adverse pregnancy outcomes, yet the mechanisms and effects of timing remain unclear. Immune disruption in early gestation may be particularly detrimental as this is an important period for placental development, which has been associated with the pathology of adverse obstetric outcomes. To increase our understanding of risk factors for adverse obstetric outcomes, we aim to investigate the association between multiple inflammatory and angiogenic markers during early pregnancy and adverse pregnancy outcomes in a large population-based cohort. DESIGN Prospective population-based pregnancy cohort study (n = 7513). SETTING Rotterdam, the Netherlands. POPULATION Pregnant women in Rotterdam between April 2002 and January 2006. METHODS Serum inflammatory markers (high-sensitivity (HS)-C-reactive protein (CRP), interleukin (IL)-1β, IL-6, IL-17a, IL-23, interferon (IFN)-γ) and angiogenic factors (sFlt-1 and PlGF) were analysed in repeated measures around 13-20 weeks gestation. A cytokine index was created using principal component analysis. MAIN OUTCOME MEASURES Hypertensive disorders of pregnancy, spontaneous preterm birth and small for gestational age at birth. RESULTS HS-CRP, but not the cytokine index, was associated with increased risk of spontaneous preterm birth after multiple testing correction. We found no association of HS-CRP or the cytokine index with hypertensive disorders of pregnancy and small for gestational age at birth after multiple testing correction. Inflammatory and angiogenic factors were associated with each other, yet effect sizes were small. CONCLUSIONS We found no strong evidence of a link between early gestation typical inflammatory marker levels and the risk of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Frederieke A J Gigase
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
- The Generation R Study Group, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | | | - Anna Suleri
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
- The Generation R Study Group, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Anna-Sophie Rommel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Michael Breen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Manon H J Hillegers
- Department of Child and Adolescent Psychiatry, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Michal A Elovitz
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Women's Biomedical Research Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Eric A P Steegers
- Department of Obstetrics and Gynecology, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Lot D De Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Human Genetics, Radboud UMC, Nijmegen, the Netherlands
- Department of Psychiatry, Radboud UMC, Nijmegen, the Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Psychiatry, Erasmus MC University Medical Centre Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|