201
|
Li B, Wang J, Xu J, Xie J, Liu Q, Yang C, Zhang Z. Association between dyslipidemia and vitamin D deficiency: a cross-sectional study in Chinese healthy population. Front Endocrinol (Lausanne) 2025; 16:1450924. [PMID: 40370783 PMCID: PMC12074960 DOI: 10.3389/fendo.2025.1450924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 03/31/2025] [Indexed: 05/16/2025] Open
Abstract
Background Given the global prevalence of vitamin D deficiency, this study examines the association between serum 25-hydroxyvitamin D (25(OH)D) levels and lipid profiles, including total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides (TG). Methods In a cross-sectional analysis at the Yuhuan People's Hospital Physical Examination Center, 1535 asymptomatic individuals underwent anthropometric assessment and blood sampling. Serum 25(OH)D levels were quantified by a chemiluminescent immunoassay, while lipid parameters were evaluated by an enzymatic method on a fully automated analyzer. Results The mean serum 25(OH)D level of the participants was 49.6 ± 15.3 nmol/L, with 54.7% demonstrating vitamin D deficiency. Female participants had significantly lower mean 25(OH)D levels (46.2 ± 13.7 nmol/L) than males (54.1 ± 16.0 nmol/L, P < 0.001). After adjustment for age and BMI, multivariate ordinal logistic analysis revealed a 1.71-fold higher dyslipidemia-associated risk of vitamin D deficiency in the population (OR=1.71, 95% CI=1.07-2.79), more pronounced in females(OR=2.84, 95% CI=1.17-8.09) compared to males (OR=1.44, 95% CI=0.82-2.58). Notably, vitamin D deficiency was significantly associated with elevated LDL-C levels in females (OR=5.17, 95% CI=1.47-32.96), in contrast to males. Conclusion This study highlights a significant association between 25(OH)D deficiency and an increased risk of dyslipidemia, especially in females, highlighting the importance of considering gender-specific risk factors in the management of 25(OH)D deficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhengmao Zhang
- Department of orthopaedics, Yuhuan people’s hospital, Taizhou, Zhejiang, China
| |
Collapse
|
202
|
Muttiah B, Hanafiah A. Gut Microbiota and Cardiovascular Diseases: Unraveling the Role of Dysbiosis and Microbial Metabolites. Int J Mol Sci 2025; 26:4264. [PMID: 40362500 PMCID: PMC12072866 DOI: 10.3390/ijms26094264] [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: 04/09/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Cardiovascular diseases (CVDs), including heart failure (HF), hypertension, myocardial infarction (MI), and atherosclerosis, are increasingly linked to gut microbiota dysbiosis and its metabolic byproducts. HF, affecting over 64 million individuals globally, is associated with systemic inflammation and gut barrier dysfunction, exacerbating disease progression. Similarly, hypertension and MI correlate with reduced microbial diversity and an abundance of pro-inflammatory bacteria, contributing to vascular inflammation and increased cardiovascular risk. Atherosclerosis is also influenced by gut dysbiosis, with key microbial metabolites such as trimethylamine-N-oxide (TMAO) and short-chain fatty acids (SCFAs) playing crucial roles in disease pathogenesis. Emerging evidence highlights the therapeutic potential of natural compounds, including flavonoids, omega-3 fatty acids, resveratrol, curcumin, and marine-derived bioactives, which modulate the gut microbiota and confer cardioprotective effects. These insights underscore the gut microbiota as a critical regulator of cardiovascular health, suggesting that targeting dysbiosis may offer novel preventive and therapeutic strategies. Further research is needed to elucidate underlying mechanisms and optimize microbiome-based interventions for improved cardiovascular outcomes.
Collapse
Affiliation(s)
- Barathan Muttiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
- GUT Research Group, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
203
|
Hsieh FR, Lee WK, Chen SSY, Sun CK, Yeh WB, Hung YM, Tsai FJ, Chang R, Pan LF. Varicose vein is associated with an increased risk of ischemic stroke: a population-based matched cohort study. Postgrad Med J 2025:qgaf063. [PMID: 40302491 DOI: 10.1093/postmj/qgaf063] [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: 12/31/2024] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 05/02/2025]
Abstract
IMPORTANCE The association between varicose veins (VVs) and stroke, and the influence of VV management on the risk of stroke remained unclear. OBJECTIVE To evaluate whether VVs are associated with an elevated risk of stroke and assess if interventions for VVs alter this risk. DESIGN From 1 January 2000 to 31 December 2015, adults with VVs were matched by propensity score with those without VVs. Patients with prior strokes were excluded. Follow-up continued until 31 December 2018. OUTCOMES Relative hazards through comparing incidence rates of ischemic stroke (IS) and hemorrhagic stroke (HS) between the two groups using Cox proportional hazards models. RESULTS Comparison of the VV group (n = 23 238, mean [SD] age = 54.3 [15.6] years; 70% female) with the non-VV group (n = 92 952, mean [SD] age = 54.1 [15.7] years; 71% female) revealed a higher incidence rate of IS in the former than the latter (13.15 vs. 11.16 per 1000 person-years; 2555 vs. 8799 cases, respectively). The adjusted hazard ratio (aHR) for overall stroke in the VV group was 1.16 (95% CI = 1.11-1.21). Both females (aHR = 1.18 [95% CI = 1.11-1.25]) and males (aHR = 1.15 [95% CI = 1.07-1.24]) with VVs showed an increased IS risk. In patients aged ≥50, VVs were associated with a higher IS risk (aHR = 1.17 [95% CI = 1.12-1.23]). No significant correlation was found between VVs and HS. The association of VV management with a reduced risk of stroke (aHR = 0.82 [95% CI = 0.70-0.96]) was supported by sensitivity analyses. CONCLUSIONS Adults with VVs showed an increased risk of IS but not HS. Key message What is already known on this topic. Varicose veins (VVs) are linked to cardiovascular conditions, but their association with stroke risk and the impact of VV interventions remain unclear. What this study adds. This study demonstrated that adults with VVs, especially those aged ≥50, have a higher ischemic stroke (IS) risk. VV interventions may reduce this risk. How this study might affect research, practice, or policy. Stroke risk assessment and VV management should be considered in older patients to improve preventive care.
Collapse
Affiliation(s)
- Feng-Renn Hsieh
- Department of Emergency Medicine, Kaohsiung Municipal United Hospital, No. 976, Jhonghua 1st Rd., Gushan Dist., Kaohsiung City 80457, Taiwan
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd., Zuoying Dist., Kaohsiung City 813414, Taiwan
| | - Wei-Kai Lee
- Department of Emergency Medicine, Sinying Hospital, Ministry of Health and Welfare, No. 73, Xinyi Street, Xinying District, Tainan City 73042, Taiwan
| | - Sunny Ssu-Yu Chen
- School of Medicine, Chung Shan Medical University, No. 110, Section 1, Jianguo North Road, Taichung City 40201, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou Dist., Taipei City 112304, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Dachang Hospital, I-Shou University, No. 132-1, Kaisyuan 2nd Rd., Lingya Dist., Kaohsiung City 807066, Taiwan
- School of Medicine for International Students, College of Medicine, I-Shou University, No. 8, Yida Rd., Jiaosu Village Yanchao District,Kaohsiung City 82445, Taiwan
| | - Wen-Bin Yeh
- Department of Emergency Medicine, Kaohsiung Municipal Min-Sheng Hospital, No. 134, Kaixuan 2nd Road, Lingya District, Kaohsiung City 802511, Taiwan
| | - Yao-Min Hung
- Department of Internal Medicine, Taitung Hospital, Ministry of Health and Welfare, No. 1, Wuquan Street, Taitung City, Taitung County, 95043, Taiwan
- Master Program in Biomedicine, College of Science and Engineering, National Taitung University, No. 369, Sec. 2, University Rd., Taitung City, Taitung County, 950309, Taiwan
- College of Health and Nursing, Meiho University, No. 23, Pingguang Road, Pingtung County, 912009, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, No. 100, Section 1, Jingmao Rd, Beitun District, Taichung City 406040, Taiwan
- Department of Medical Research, China Medical University Hospital, No. 2, Yude Road, North District, Taichung City 40447, Taiwan
- Division of Medical Genetics, China Medical University Children's Hospital, No. 2, Yude Road, North District, Taichung City 40447, Taiwan
- Department of Biotechnology and Bioinformatics, Asia University, No. 500, Lioufeng Rd., Wufeng, Taichung 413305, Taiwan
| | - Renin Chang
- School of Medicine, Chung Shan Medical University, No. 110, Section 1, Jianguo North Road, Taichung City 40201, Taiwan
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd., Zuoying Dist., Kaohsiung City 813414, Taiwan
- Department of Recreation Sports Management, Tajen University, No. 20, Weixin Rd., Yanpu Township, Pingtung County 907101, Taiwan
| | - Li-Fei Pan
- Department of General Affair Office, Kaohsiung Veterans General Hospital, No. 386, Dazhong 1st Rd., Zuoying Dist., Kaohsiung City 813414, Taiwan
| |
Collapse
|
204
|
Zhu M, Yang Y, Tang X, Hou H, Zhang Y, Chen R. Exploring the role of the CD74 + cardiac macrophage subset in trastuzumab cardiotoxicity and its mechanisms. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167875. [PMID: 40316056 DOI: 10.1016/j.bbadis.2025.167875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/24/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
Trastuzumab (TRZ) is the standard treatment for human epidermal growth factor receptor 2 (HER-2) positive breast cancer, but its cardiotoxicity significantly impacts the prognosis and quality of survival of patients, and the underlying mechanism of TRZ-related cardiotoxicity remains incompletely understood. Macrophage subsets better reflect macrophage heterogeneity than the traditional macrophage M1/M2 type polarization classification. CD74, a receptor with strong binding affinity for macrophage migration inhibitory factor, plays an important role in macrophage activation. After successfully constructing a mouse TRZ cardiotoxicity model, flow cytometry indicated that CD74+ cardiac macrophages (CMφs) were significantly elevated in the TRZ group. Single-cell data were utilized to identify CD74+ CMφs, GO and KEGG analyses of the DEGs were conducted to further validate the CD74/STAT1 signaling pathway. Analyses using RT-PCR, immunofluorescence, and western blot revealed a marked increase in the expression of genes and proteins linked to this pathway in TRZ-treated group. Additionally, levels of inflammation-related factors and the expression of apoptotic proteins was elevated following TRZ treatment. CD74-knockdown RAW 264.7 macrophages cell line were constructed via Lentiviruses carrying CD74 (hU6-MCS-CBh-gcGFP-IRES-puromycin) transfection and co-cultured with HL-1 cardiomyocytes to establish an in vitro TRZ cardiotoxicity model. Western blot analysis of CD74/STAT1 signaling pathway protein levels demonstrated that CD74 knockdown rescued TRZ-induced cellular damage. These findings suggest that TRZ may promote inflammation and apoptosis in cardiomyocytes, leading to cardiotoxicity through the CD74+ CMφ subset, which regulates the CD74/STAT1 signaling pathway. CD74+ CMφs are anticipated to be a novel intervention target and therapeutic strategy for addressing TRZ-induced cardiotoxicity.
Collapse
Affiliation(s)
- Minyan Zhu
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pharmacology, Soochow University, Suzhou, China
| | - Yaping Yang
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pharmacology, Soochow University, Suzhou, China
| | - Xinchen Tang
- Department of Health Sciences in Physical Education, Macao Polytechnic University, Macao
| | - Huan Hou
- Department of Pharmacy, Dazhou Central Hospital, Dazhou, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Women's Hospital of Jiangnan University, Wuxi, China.
| | - Rong Chen
- Department of Pharmacy, the First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pharmacology, Soochow University, Suzhou, China.
| |
Collapse
|
205
|
La Torre F, Meliota G, Civino A, Campanozzi A, Cecinati V, Rosati E, Sacco E, Santoro N, Vairo U, Cardinale F. Advancing multidisciplinary management of pediatric hyperinflammatory disorders. Front Pediatr 2025; 13:1553861. [PMID: 40370972 PMCID: PMC12075326 DOI: 10.3389/fped.2025.1553861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 04/03/2025] [Indexed: 05/16/2025] Open
Abstract
Pediatric hyperinflammatory diseases, including Still's disease, Kawasaki disease (KD), multisystem inflammatory syndrome in children (MIS-C), and recurrent pericarditis (RP), represent a spectrum of conditions characterized by immune dysregulation and systemic inflammation. Each disorder exhibits distinct pathophysiological mechanisms and clinical features, yet their overlapping presentations often pose diagnostic challenges. Early and accurate differentiation is critical to mitigate complications such as macrophage activation syndrome (MAS), coronary artery aneurysms, and myocardial dysfunction. This narrative review explores the pathophysiology, diagnostic criteria, and management of these conditions, emphasizing the utility of advanced biomarkers, imaging modalities, and genetic testing. For Still's disease, the review highlights the transformative role of biologic therapies targeting IL-1 and IL-6 in reducing systemic inflammation and improving outcomes. In KD, timely administration of intravenous immunoglobulin (IVIG) and combination with high-dose steroids in high-risk patients is pivotal for preventing coronary complications. MIS-C, associated with SARS-CoV-2 infection, requires tailored immunomodulatory approaches, including corticosteroids and biologics, to address severe hyperinflammation and multiorgan involvement. RP management prioritizes NSAIDs, colchicine, and IL-1 inhibitors to reduce recurrence and corticosteroid dependence. The review advocates for a multidisciplinary approach, integrating standardized diagnostic algorithms and disease-specific expertise to optimize patient care. Future research directions include the identification of predictive biomarkers, exploration of novel therapeutic targets, and development of evidence-based treatment protocols to enhance long-term outcomes in pediatric inflammatory diseases.
Collapse
Affiliation(s)
- Francesco La Torre
- Department of Pediatrics, Giovanni XXIII Pediatric Hospital, University of Bari, Bari, Italy
| | - Giovanni Meliota
- Pediatric Cardiology, Giovanni XXIII Pediatric Hospital, Bari, Italy
| | - Adele Civino
- Pediatric Rheumatology and Immunology Unit, “Vito Fazzi” Hospital, Lecce, Italy
| | - Angelo Campanozzi
- Pediatric Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Valerio Cecinati
- Pediatric and Pediatric Oncohematology Unit, Santissima Annunziata Hospital, Taranto, Italy
| | - Enrico Rosati
- Neonatology and Intensive Care Unit, “Vito Fazzi” Hospital, Lecce, Italy
| | - Emanuela Sacco
- Pediatric Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Nicola Santoro
- Pediatric and Oncohematology Unit, Azienda Ospedaliero Universitaria “Policlinico Consorziale”, Bari, Italy
| | - Ugo Vairo
- Pediatric Cardiology, Giovanni XXIII Pediatric Hospital, Bari, Italy
| | - Fabio Cardinale
- Department of Pediatrics, Giovanni XXIII Pediatric Hospital, University of Bari, Bari, Italy
| |
Collapse
|
206
|
Li Y, Luo M, Ye C, Bao H. Recent advancements in dual-locked optical probe for precise imaging of atherosclerosis. Org Biomol Chem 2025; 23:4024-4035. [PMID: 40196887 DOI: 10.1039/d5ob00312a] [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: 04/09/2025]
Abstract
Atherosclerosis (AS) is the primary pathological basis for cardiovascular and cerebrovascular events, making early diagnosis and dynamic risk assessment crucial for reducing the incidence of thrombotic occurrences. Traditional imaging techniques, such as CT, MRI, and PET, provide morphological information about plaques; however, they are limited in their ability to detect the molecular activity characteristics of these plaques, which hampers the assessment of thrombotic risk. In recent years, molecular optical probes have offered new insights into plaque activity detection by targeting specific biomarkers. However, single-target probes often produce false positive signals due to the cross-expression of biomarkers, which limits their clinical application. To overcome this challenge, dual-lock optical probes have been developed, achieving dual-target synergistic activation, which significantly enhances the specificity and signal-to-noise ratio of imaging. This article reviews the targeted imaging strategies and recent advancements in dual-lock optical probes in the context of AS, with a particular emphasis on their application in lipid droplet enrichment, oxidative stress, and specific enzymes. Although the technology still faces challenges regarding sensitivity, specificity, and multi-target design, its potential for future development is substantial. Through interdisciplinary collaboration and technological innovation, dual-lock optical probes are poised to evolve from 'imaging tools' to 'integrated diagnosis and treatment platforms', thereby advancing the precise diagnosis and treatment of complex diseases.
Collapse
Affiliation(s)
- Yulong Li
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350117, P. R. of China
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. of China
| | - Mengyao Luo
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350117, P. R. of China
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. of China
| | - Changqing Ye
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
| | - Hongli Bao
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, State Key Laboratory of Structural Chemistry, Center for Excellence in Molecular Synthesis, Chinese Academy of Sciences, 155 Yangqiao Road West, Fuzhou 350002, Fujian, P. R. of China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. of China
- Fujian College, University of Chinese Academy of Sciences, Fuzhou, Fujian 350002, P. R. of China
| |
Collapse
|
207
|
Chen MH, Chagari B, Abramson AM, Shi LJ, He J, Shi W. The Genetic Elements of the Obesity Paradox in Atherosclerosis Identified in an Intercross Between Hyperlipidemic Mouse Strains. Int J Mol Sci 2025; 26:4241. [PMID: 40362477 PMCID: PMC12072963 DOI: 10.3390/ijms26094241] [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: 03/24/2025] [Revised: 04/22/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Overweight and obese individuals show lower mortality rates or better prognoses than those of normal weight in a variety of diseases, a phenomenon called the "obesity paradox". An inverse association of adiposity with atherosclerosis has been observed in both humans and mice. To dissect phenotypic and genetic connections between the traits, 154 female and 145 male F2 mice were generated from an intercross between BALB/cJ and LP/J apolipoprotein E-deficient mice and fed a Western diet for 12 weeks. Atherosclerotic lesion size in the aortic root, body weight, plasma lipids, and glucose were measured, and genotyping was performed on miniMUGA SNP arrays. Quantitative trait locus (QTL) analyses on all F2 mice with sex as a covariate revealed four significant QTLs on chromosomes (Chr) 3, 6, 13, and 15 for atherosclerosis and three significant QTLs on Chr2, 7, and 15 for body weight. Chr15 QTL for atherosclerosis overlapped with one for body weight near 36 Mb. After adjusting for variation in body weight, Chr15 atherosclerosis QTL was downgraded from significant to suggestive linkage. Body weight was inversely correlated with atherosclerotic lesion sizes and accounted for more variance than a single other risk factor for atherosclerosis among F2 mice. Analysis of public data collected from two backcross cohorts revealed strong correlations between body weight and fat mass in adult mice (r ≥ 0.93; p ≤ 1.6 × 10-136). Thus, the obesity paradox in atherosclerosis is partially attributable to shared genetic components that have an opposite effect on adiposity and atherosclerosis.
Collapse
Affiliation(s)
- Mei-Hua Chen
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (M.-H.C.); (B.C.); (A.M.A.); (L.J.S.); (J.H.)
| | - Bilhan Chagari
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (M.-H.C.); (B.C.); (A.M.A.); (L.J.S.); (J.H.)
| | - Ashley M. Abramson
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (M.-H.C.); (B.C.); (A.M.A.); (L.J.S.); (J.H.)
| | - Lisa J. Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (M.-H.C.); (B.C.); (A.M.A.); (L.J.S.); (J.H.)
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (M.-H.C.); (B.C.); (A.M.A.); (L.J.S.); (J.H.)
| | - Weibin Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22903, USA; (M.-H.C.); (B.C.); (A.M.A.); (L.J.S.); (J.H.)
- Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
208
|
Ahmad H, Gopakumar J, Nachun DC, Ma L, D’Addabbo J, Huang X, Koyano T, Boyd J, Woo YPJ, Fong R, Aalami O, Nguyen PK, Jaiswal S. Single cell RNA sequencing of haematopoietic cells in fresh and frozen human atheroma tissue. Cardiovasc Res 2025; 121:396-404. [PMID: 39907372 PMCID: PMC12038232 DOI: 10.1093/cvr/cvaf014] [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: 07/07/2021] [Revised: 09/22/2024] [Accepted: 12/05/2024] [Indexed: 02/06/2025] Open
Abstract
AIMS Single-cell RNA sequencing (scRNA-seq) is a powerful method for exploring the cellular heterogeneity within human atheroma but typically requires fresh tissue to preserve cell membrane integrity, limiting the feasibility of large-scale biobanking for later analysis. The aim of this study was to determine whether cryopreservation of fragile and necrotic atheroma tissue affects the viability and transcriptomic profiles of haematopoietic cells in subsequent scRNA-seq analysis, enabling the use of cryopreserved atheroma samples for future research. METHODS AND RESULTS We performed scRNA-seq on five paired fresh and cryopreserved atheroma samples-three from coronary arteries and two from carotid arteries. Each sample was enzymatically digested, sorted for CD45+ haematopoietic cells, and processed using the 10× Genomics scRNA-seq workflow. Half of each sample was processed immediately, while the other half was cryopreserved in liquid nitrogen for an average of 5 weeks before thawing and processing. In carotid artery samples, we noted the absence of LYVE1+ macrophages, likely due to the loss of the adventitial layer during endarterectomy procedures. Our results indicated that cryopreservation modestly affected cellular integrity, leading to an increase in the relative abundance of mitochondrial RNA in frozen samples. Minimal differences were observed between fresh and cryopreserved samples in uniquely detected transcripts, cell clustering, or transcriptional profiles within haematopoietic populations. CONCLUSIONS Our study demonstrates that cryopreserved human atheroma samples can be successfully profiled using scRNA-seq, with comparable transcriptomic data to that obtained from fresh samples. These findings suggest that cryopreservation is a viable method for biobanking atheroma tissues, facilitating large-scale studies without the need for immediate sample processing.
Collapse
Affiliation(s)
- Herra Ahmad
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
- Department of Cardiology, Charité Universitätsmedizin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Jayakrishnan Gopakumar
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Daniel C Nachun
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Lisa Ma
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - Jessica D’Addabbo
- Department of Medicine (Cardiovascular Medicine), Stanford University, Stanford, CA, USA
| | - Xianxi Huang
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Tiffany Koyano
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Jack Boyd
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Yi-Ping Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Robyn Fong
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Oliver Aalami
- Department of Surgery, Division of Vascular Surgery, Stanford University, Stanford, CA, USA
| | - Patricia K Nguyen
- Department of Medicine (Cardiovascular Medicine), Stanford University, Stanford, CA, USA
- School of Medicine and the School of Engineering, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Medicine (Division of Cardiovascular Medicine), Palo Alto VA Medical Center, Palo Alto, CA, USA
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
- School of Medicine and the School of Engineering, Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Department of Developmental Biology, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford Cancer Institute (SCI), Stanford University, Stanford, CA, USA
| |
Collapse
|
209
|
Lin X, Xia L, Zhou Y, Xie J, Tuo Q, Lin L, Liao D. Crosstalk Between Bile Acids and Intestinal Epithelium: Multidimensional Roles of Farnesoid X Receptor and Takeda G Protein Receptor 5. Int J Mol Sci 2025; 26:4240. [PMID: 40362481 PMCID: PMC12072030 DOI: 10.3390/ijms26094240] [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: 03/16/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Bile acids and their corresponding intestinal epithelial receptors, the farnesoid X receptor (FXR), the G protein-coupled bile acid receptor (TGR5), play crucial roles in the physiological and pathological processes of intestinal epithelial cells. These acids and receptors are involved in the regulation of intestinal absorption, signal transduction, cellular proliferation and repair, cellular senescence, energy metabolism, and the modulation of gut microbiota. A comprehensive literature search was conducted using PubMed, employing keywords such as bile acid, bile acid receptor, FXR (nr1h4), TGR5 (gpbar1), intestinal epithelial cells, proliferation, differentiation, senescence, energy metabolism, gut microbiota, inflammatory bowel disease (IBD), colorectal cancer (CRC), and irritable bowel syndrome (IBS), with a focus on publications available in English. This review examines the diverse effects of bile acid signaling and bile receptor pathways on the proliferation, differentiation, senescence, and energy metabolism of intestinal epithelial cells. Additionally, it explores the interactions between bile acids, their receptors, and the microbiota, as well as the implications of these interactions for host health, particularly in relation to prevalent intestinal diseases. Finally, the review highlights the importance of developing highly specific ligands for FXR and TGR5 receptors in the context of metabolic and intestinal disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Duanfang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (X.L.); (L.X.); (Y.Z.); (J.X.); (Q.T.); (L.L.)
| |
Collapse
|
210
|
Huang L, Feng Z, Yang W, Zhu Y, Li J, Huang L, Wang R, Peng L, He M, Tang Y, Chen P, Lan C, Zhou X, Zhou L, Ye C, Zhang L, Jiang J, Ye Y, Wang R, He Y, Liu Y, Gong H, Xiong H, Xia L, Xu H, Zhang B, Tu R, Du C, Cui L, Gao J, Huang Z, Tang C. Parecoxib sequential with imrecoxib for occurrence and remission of severe acute pancreatitis: a multicentre, double-blind, randomised, placebo-controlled trial. Gut 2025:gutjnl-2024-334038. [PMID: 40301118 DOI: 10.1136/gutjnl-2024-334038] [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: 10/07/2024] [Accepted: 04/12/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND There is no effective drug treatment for the organ failure (OF) caused by severe acute pancreatitis (SAP). OBJECTIVE We aimed to evaluate the efficacy of cyclooxygenase-2 inhibitors (COX-2-Is) on the treatment of SAP and its safety. DESIGN In this multicentre, double-blind, randomised, placebo-controlled, investigator-initiated trial, 348 patients with acute pancreatitis aged 18-75 years, <1 week from onset of illness to admission, and Acute Physiology and Chronic Health Evaluation II Score ≥7 or modified Marshall Score ≥2, were randomly assigned (1:1) to the COX-2-Is group (parecoxib sequential with imrecoxib) or the placebo group. SAP occurrence, duration of OF, local complications, clinical outcomes and serum inflammatory mediators were measured. RESULTS Compared with the placebo group, SAP occurrence was reduced by 20.7% (77.6% vs 61.5%, p=0.001) and the persistent OF duration in SAP was shortened by 2 days (p<0.001) after COX-2-Is treatment. For patients enrolled within or after 48 hours from symptom onset, SAP occurrence was reduced by 23.8% (p=0.001) and 8.5% (p=0.202), and the persistent OF duration in SAP was shortened by 3 days (p=0.001) and 2 days (p=0.010) after COX-2-Is treatment, respectively. The occurrence of local complications in the COX-2-Is group was significantly lower than those in the placebo group, 33.7% vs 49.1%, p=0.004. The serum levels of inflammatory mediators and 30-day mortality (from 8.6% to 3.4%) were significantly reduced after COX-2-Is treatment, p<0.05. The incidence of adverse events was similar between the two treatment groups. CONCLUSION Parecoxib sequential with imrecoxib was effective and well tolerated in reducing the occurrence and duration of SAP and local complications through suppression of systemic inflammatory response, leading to decreased morbidity.
Collapse
Affiliation(s)
- Luming Huang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Lab of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhe Feng
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Wenjuan Yang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yin Zhu
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jing Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Libin Huang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lan Peng
- Department of Gastroenterology, Wenjiang District People's Hospital, Chengdu, Sichuan, China
| | - Mingshun He
- Department of Gastroenterology, Nanbu Country People's Hospital, Nanchong, Sichuan, China
| | - Yingmei Tang
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ping Chen
- Department of Gastroenterology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Cheng Lan
- Department of Gastroenterology, Hainan Medical University Affiliated Hainan Hospital, Haikou, Hainan, China
| | - Xiaoqing Zhou
- Department of Gastroenterology, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Lin Zhou
- Department of Gastroenterology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Nanjing, Jiangsu, China
| | - Cheng Ye
- Department of Gastroenterology, Zhejiang Provincial Hospital of Chinese Medicine, Hangzhou, Zhejiang, China
| | - Linhao Zhang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jingsun Jiang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yanting Ye
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Rui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yan He
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yan Liu
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Hui Gong
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Huifang Xiong
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Liang Xia
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Haiyan Xu
- Department of Gastroenterology, Wenjiang District People's Hospital, Chengdu, Sichuan, China
| | - Bin Zhang
- Department of Gastroenterology, Nanbu Country People's Hospital, Nanchong, Sichuan, China
| | - Rongfang Tu
- Department of Gastroenterology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chun Du
- Department of Gastroenterology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Lujia Cui
- Department of Gastroenterology, Hainan Medical University Affiliated Hainan Hospital, Haikou, Hainan, China
| | - Jinhang Gao
- Lab of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhiyin Huang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Lab of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chengwei Tang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- Lab of Gastroenterology and Hepatology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
211
|
Psaltis PJ, Nguyen MT, Singh K, Sinhal A, Wong DTL, Alcock R, Rajendran S, Dautov R, Barlis P, Patel S, Salagaras T, Marathe JA, Bursill CA, Montarello NJ, Nidorf SM, Thompson PL, Butters J, Cuthbert AR, Yelland LN, Ottaway JL, Kataoka Y, Di Giovanni G, Nicholls SJ. Optical coherence tomography assessment of the impact of colchicine on non-culprit coronary plaque composition after myocardial infarction. Cardiovasc Res 2025; 121:468-478. [PMID: 39189611 DOI: 10.1093/cvr/cvae191] [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: 04/09/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 08/28/2024] Open
Abstract
AIMS Low-dose colchicine reduces the risk of cardiovascular events after myocardial infarction (MI). The purpose of this study was to assess the effect of colchicine post-MI on coronary plaque morphology in non-culprit segments by optical coherence tomography (OCT). METHODS AND RESULTS COCOMO-ACS was a double-blind, placebo-controlled trial that randomized 64 patients (median age 61.5 years; 9.4% female) with acute non-ST-segment elevation MI to colchicine 0.5 mg daily or placebo for a median of 17.8 months in addition to guideline-recommended therapy. Participants underwent serial OCT imaging within a matched segment of non-culprit coronary artery that contained at least one lipid-rich plaque causing ≥20% stenosis. The primary outcome was the change in minimum fibrous cap thickness (FCT) in non-culprit segments from baseline to final visit. Of those randomized, 57 (29 placebo, 28 colchicine) had evaluable imaging at baseline and follow-up. Overall, colchicine had no effect on relative (placebo +48.0 ± 35.1% vs. colchicine +62.4 ± 38.1%, P = 0.18) or absolute changes in minimum FCT (+29.2 ± 20.9 µm vs. + 37.2 ± 21.3 µm, P = 0.18), or change in maximum lipid arc (-38.8 ± 32.2° vs. -54.8 ± 46.9°, P = 0.18) throughout the imaged non-culprit segment. However, in patients assigned colchicine, cap rupture was less frequent (placebo 27.6% vs. colchicine 3.6%, P = 0.03). In post hoc analysis of 43 participants who had been followed for at least 16 months, minimum FCT increased to a greater extent in the colchicine group (placebo +38.7 ± 25.4% vs. colchicine +64.7 ± 34.1%, P = 0.005). CONCLUSION In this study, OCT failed to detect an effect of colchicine on the minimum FCT or maximum lipid arc of plaques in non-culprit segments post-MI. The post hoc observation that minimum FCT increased to a greater extent with colchicine after more prolonged treatment suggests that longer-term studies may be required to detect the effect of anti-inflammatory therapies on plaque morphology by OCT. CLINICAL TRIAL NUMBER Australian New Zealand Clinical Trials Registry Identifier, ACTRN12618000809235, registered on the 11 May 2018.
Collapse
Affiliation(s)
- Peter J Psaltis
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Mau T Nguyen
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Kuljit Singh
- Department of Cardiology, Gold Coast University Hospital, Gold Coast, Australia
| | - Ajay Sinhal
- Department of Cardiology, Flinders Medical Center, Adelaide, Australia
| | - Dennis T L Wong
- Victorian Heart Institute, Monash University, Clayton, Australia
| | - Richard Alcock
- Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | | | - Rustem Dautov
- Department of Cardiology, The Prince Charles Hospital, Brisbane, Australia
| | - Peter Barlis
- Department of Cardiology, The Northern Hospital, Melbourne, Australia
| | - Sanjay Patel
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | - Thalia Salagaras
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
| | - Jessica A Marathe
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Christina A Bursill
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Nicholas J Montarello
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
| | - Stefan M Nidorf
- Advara HeartCare, Western Australia, Perth, Australia
- Cardiovascular Science and Diabetes Program, Harry Perkins Institute of Medical Research, Perth, Australia
| | - Peter L Thompson
- Advara HeartCare, Western Australia, Perth, Australia
- Cardiovascular Science and Diabetes Program, Harry Perkins Institute of Medical Research, Perth, Australia
- Heart Research Institute, Sir Charles Gairdner Hospital, Perth, Australia
| | - Julie Butters
- Victorian Heart Institute, Monash University, Clayton, Australia
| | - Alana R Cuthbert
- SAHMRI Women & Kids, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Lisa N Yelland
- SAHMRI Women & Kids, South Australian Health and Medical Research Institute, Adelaide, Australia
- School of Public Health, The University of Adelaide, Adelaide, Australia
| | - Juanita L Ottaway
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
| | - Yu Kataoka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Giuseppe Di Giovanni
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Victorian Heart Institute, Monash University, Clayton, Australia
| | | |
Collapse
|
212
|
Mille-Hamard L, Momken I, Koralsztein JP, Billat VL. Cardiovascular, Hemodynamic, and Anthropometric Adaptations Induced by Walking Training at FATmax in Obese Males and Females over 45 Years Old. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:701. [PMID: 40427817 PMCID: PMC12111642 DOI: 10.3390/ijerph22050701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025]
Abstract
AIMS AND OBJECTIVES The present study aimed to examine the effects of 6 months of unsupervised training, walking at maximal fat oxidation (FATmax), on body composition and cardiovascular function at rest and exercise, in middle aged obese subjects. METHODS AND RESULTS A single group with pre-test/post-test study design was conducted. Eighteen obese subjects (11 males and 7 females) over 45 were engaged in a non-supervised walking training for 6 months, 40 min, 3 times per week, at the targeted HR corresponding to FATmax (5.5 ± 0.6 km·h-1). This training modality led to a reduction in obesity-related indicators among participants, including weight (-3.7 ± 3.4 kg), BMI (-1.4 ± 1.3 kg/m2), waist circumference (-5.6 ± 4.7 cm), and body fat percentage (-2.1 ± 2.7%). However, we observed a great variability in this response to training according to individuals. Furthermore, heart rate and rate of pressure product (RPP) at rest significantly decreased (6% and 11% respectively) as well as the cardiac load during exercise (RPP -11% and cardiac cost -8%) after training. In conclusion, walking at FATmax is an efficient non-supervised training modality, allowing improvement in both body parameters and cardiovascular markers at rest and during exercise in middle age obese subjects. Even if body parameter changes were modest, the cardiac load decrease is an important factor for reducing the risk of cardiovascular diseases in this population.
Collapse
Affiliation(s)
- Laurence Mille-Hamard
- Unité de Biologie Intégrative des Adapations à l’Exercice (UBIAE), EA 1374, Université d’Evry-Paris Saclay, 91000 Evry-Courcouronnes, France; (I.M.); (V.L.B.)
- Informatique, Bio-Informatique et Systèmes Complexes (IBISC), EA 4526, Université Paris-Saclay, 91020 Evry, France
| | - Iman Momken
- Unité de Biologie Intégrative des Adapations à l’Exercice (UBIAE), EA 1374, Université d’Evry-Paris Saclay, 91000 Evry-Courcouronnes, France; (I.M.); (V.L.B.)
- INSERM UMR-S 1180, Faculté de Pharmacie, Université Paris-Saclay, 91400 Orsay, France
| | | | - Véronique Louise Billat
- Unité de Biologie Intégrative des Adapations à l’Exercice (UBIAE), EA 1374, Université d’Evry-Paris Saclay, 91000 Evry-Courcouronnes, France; (I.M.); (V.L.B.)
- Informatique, Bio-Informatique et Systèmes Complexes (IBISC), EA 4526, Université Paris-Saclay, 91020 Evry, France
- EA 4445-Movement, Balance, Performance, and Health Laboratory, Université de Pau et des Pays de l’Adour, 65000 Tarbes, France
| |
Collapse
|
213
|
Jolfayi AG, Beheshti AT, Hosseini SM, Fakhrabadi AA, Mohebbi B, Malakootian M, Maleki M, Pouraliakbar H, Hosseini S, Arabian M. Epicardial adipose tissue features as a biomarker and therapeutic target in coronary artery disease. Sci Rep 2025; 15:14786. [PMID: 40295726 PMCID: PMC12037875 DOI: 10.1038/s41598-025-99600-w] [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] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
This study aimed to examine the interplay between epicardial adipose tissue (EAT) features, macrophage polarization, and the cytokines Resistin and Apelin in the context of coronary artery disease (CAD). Using a case-control design with 21 CAD and 20 non-CAD individuals, the study collected demographic data, cardiovascular risk factors, and medical histories. Metabolic risk factors were assessed through laboratory tests, and CAD presence was confirmed by imaging studies. Detailed measurements of epicardial adipose characteristics were obtained through CT scans. Blood samples were analyzed for Resistin and Apelin levels, and tissue samples from EAT for macrophage polarization. The results revealed no significant differences in EAT volume and density between CAD and non-CAD groups, but the CAD group exhibited a significantly higher Calcium score. Apelin and Resistin mRNA expression levels in the right ventricular epicardial and atrioventricular fat tissue showed significantly lower Apelin and higher Resistin levels in CAD patients. CD206 expression levels in EAT were substantially lower in the CAD group, while CD11c expression levels were significantly higher. The CAD group exhibited a significantly higher CD11c/CD206 ratio in adipose tissue macrophages. This investigation highlights the significance of molecular characteristics of EAT in CAD development. While no significant differences were found in EAT volume and density, lower Apelin and higher Resistin mRNA expression in CAD patients' right ventricular fat tissue were observed. Changes in macrophage polarization markers, lower CD206 and higher CD11c, along with a higher CD11c/CD206 ratio in the macrophages of CAD patients have been shown in two investigated regions of EAT.
Collapse
Affiliation(s)
| | | | | | | | - Bahram Mohebbi
- Cardiovascular Research Center, Rajaie Cardiovascular Institute, Tehran, Iran
| | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Institute, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Institute, Tehran, Iran
| | | | - Saeid Hosseini
- Heart Valve Disease Research Center, Rajaie Cardiovascular Institute, Tehran, Iran
| | - Maedeh Arabian
- Cardiogenetic Research Center, Rajaie Cardiovascular Institute, Tehran, Iran.
| |
Collapse
|
214
|
Guo Y, Li J, Liu X, Ding H, Zhang W. Potential therapeutic targets for ischemic stroke in pre-clinical studies: Epigenetic-modifying enzymes DNMT/TET and HAT/HDAC. Front Pharmacol 2025; 16:1571276. [PMID: 40356977 PMCID: PMC12066669 DOI: 10.3389/fphar.2025.1571276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and disability worldwide, driven by genetic predispositions and environmental interactions, with epigenetics playing a pivotal role in mediating these processes. Specific modifying enzymes that regulate epigenetic changes have emerged as promising targets for IS treatment. DNA methyltransferases (DNMTs), ten-eleven translocation (TET) dioxygenases, histone acetyltransferases (HATs), and histone deacetylases (HDACs) are central to epigenetic regulation. These enzymes maintain a dynamic balance between DNA methylation/demethylation and histone acetylation/deacetylation, which critically influences gene expression and neuronal survival in IS. This review is based on both in vivo and in vitro experimental studies, exploring the roles of DNMT/TET and HAT/HDAC in IS, evaluating their potential as therapeutic targets, and discussing the use of natural compounds as modulators of these enzymes to develop novel treatment strategies.
Collapse
Affiliation(s)
- Yurou Guo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaodan Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Huang Ding
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| | - Wei Zhang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Key Laboratory of Hunan Provincial for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, China
| |
Collapse
|
215
|
Gotelli E, Campitiello R, Pizzorni C, Sammorì S, Aitella E, Ginaldi L, De Martinis M, Carubbi F, Di Ruscio E, Cuomo G, Martinelli E, Marrone S, De Angelis R, Giuggioli D, Guiducci S, Ingegnoli F, Riccieri V, Sebastiani M, Sulli A, Smith V, Cutolo M. Multicentre retrospective detection of nailfold videocapillaroscopy abnormalities in long covid patients. RMD Open 2025; 11:e005469. [PMID: 40295119 PMCID: PMC12039021 DOI: 10.1136/rmdopen-2025-005469] [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: 01/17/2025] [Accepted: 04/06/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND SARS-CoV-2 induces acute non-specific endothelial/microvascular alterations that have been identified by nailfold videocapillaroscopy (NVC). Details on NVC abnormalities in long covid (LC) patients (pts) are unknown. METHODS LC pts without and with systemic sclerosis (non-SSc-LC and SSc-LC), recovered COVID-19 (RC) pts that did not develop LC and healthy matched control subjects (CNT) that underwent NVC examinations were evaluated in a multicentre national study from the Capillaroscopy and Microcirculation in Rheumatic Diseases Study Group of the Italian Society of Rheumatology. Retrospective collection was performed for demographic data, course of SARS-CoV-2 infection, comorbidities, concomitant drugs. NVC alterations were quantified by validated scores. Pre-COVID-19 and post-COVID-19 microvascular status was analysed by NVC. RESULTS 62 non-SSc-LC pts (49 female/13 male, 51±16 years old), 24 SSc-LC pts (21 female/3 male, 59±17 years old), 23 RC pts (18 female/5 male, 51±18 years old) and 84 CNT (68 female/16 male, 52±12 years old) were analysed. Non-SSc-LC pts showed significantly more dilated capillaries (p<0.01, p multivariate<0.01), microhaemorrhages (p=0.01, p multivariate<0.05), abnormal shapes (p<0.05, p multivariate<0.05) than CNT and of note, lower mean capillary number per linear millimetre (p<0.01, p multivariate<0.01) than both RC pts and CTN (p<0.01, p multivariate<0.05).Of highest interest, 16 non-SSc-LC pts showed statistically significantly more dilated capillaries (p<0.05) and microhaemorrhages (p<0.05) in NVC examinations after COVID-19, compared with pre-COVID-19 status.Similarly, SSc-LC pts (24) showed significantly lower capillary density (p=0.01) and more dilated capillaries (p<0.01) in NVC examinations after COVID-19, compared with pre-COVID-19 status. CONCLUSIONS LC pts show more microvascular alterations at NVC as compared with RC patients and CNT, which may contribute to the pathogenesis of persistent organ/systems dysfunction.
Collapse
Affiliation(s)
- Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Sammorì
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Ernesto Aitella
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Allergy and Clinical Immunology Unit, G. Mazzini Hospital, Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
- Long-Term Care Unit, Maria SS dello Splendore Hospital, Giulianova, Italy
| | - Francesco Carubbi
- Internal Medicine and Nephrology Division, Department of Clinical Medicine, Life, Health and Environmental Sciences, San Salvatore Hospital, L'Aquila, Italy
| | - Evy Di Ruscio
- Internal Medicine and Nephrology Division, Department of Clinical Medicine, Life, Health and Environmental Sciences, San Salvatore Hospital, L'Aquila, Italy
| | - Giovanna Cuomo
- Department of Precision Medicine, University Hospital Luigi Vanvitelli, Naples, Italy
| | - Emanuela Martinelli
- Department of Precision Medicine, University Hospital Luigi Vanvitelli, Naples, Italy
| | - Sabrina Marrone
- Department of Precision Medicine, University Hospital Luigi Vanvitelli, Naples, Italy
| | - Rossella De Angelis
- Rheumatology Unit, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Carlo Urbani Hospital, Jesi, Italy
- IRCCS INRCA, Ancona, Italy
| | - Dilia Giuggioli
- Rheumatology Unit, Department of Medical and Surgical Sciences for Children and Adults, UNIMORE, Modena, Italy
| | - Serena Guiducci
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesca Ingegnoli
- Clinical Rheumatology Unit, Department of Clinical Science and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Valeria Riccieri
- Department of Internal Medicine, Anesthesiology and Cardiovascular Sciences, University of Rome La Sapienza, Rome, Italy
| | - Marco Sebastiani
- Rheumatology Unit, AUSL Piacenza, Piacenza, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Alberto Sulli
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Vanessa Smith
- Department of Rheumatology, Ghent University, Ghent, Belgium
- Department of Internal Medicine, Ghent University, Ghent, Belgium
- Unit for Molecular Immunology and Inflammation, Flemish Institute for Biotechnology (VIB), Ghent, Belgium
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
216
|
Foster MW, McMahon TJ, Ngocho JS, Kipengele AH, Violette M, Chen Y, Madut DB, Plumb RS, Wong AI, Chen L, Lee GM, Sakasaka PA, Mmbaga BT, Crump JA, McClain MT, Woods CW, Maro VP, Rubach MP. Mass spectrometry-based quantification of proteins and post-translational modifications in dried blood: longitudinal sampling of patients with sepsis in Tanzania. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.22.650109. [PMID: 40492198 PMCID: PMC12148059 DOI: 10.1101/2025.04.22.650109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The proteomic analysis of blood is routine for disease phenotyping and biomarker development. Whole blood is commonly separated into soluble and cellular fractions. However, this can introduce pre-analytical variability; and analysis of a single component (which is common) may ignore important pathophysiology. We have recently developed methods for the facile processing of dried blood for mass spectrometry-based quantification of the proteome, N-glycoproteome and phosphoproteome. Here, we applied this approach to 38 patients in Tanzania who presented to the hospital with sepsis. Blood was collected on Mitra devices at presentation and 1, 3 and 28-42 days post-enrollment. Processing of 96 total samples was performed in plate-based formats and completed within two days. Approximately 2,000 protein groups and 8,000 post-translational modifications were quantified in 3 LC-MS/MS runs at ∼1.5 hours per sample. Analysis of differential abundance revealed blood proteome signatures of acute phase response and neutrophilic inflammation that partially resolved at the 28-42 day timepoint. Numerous analytes correlated with clinical laboratory values for c-reactive protein and white blood cell counts, as well as the Universal Vital Assessment illness severity score. These datasets serve as proof-of-concept for large scale MS-based (sub)phenotyping of disease using dried blood and are available via the ProteomeXchange consortium (PXD060377).
Collapse
|
217
|
Oberther TJ, Moore AR, Kohler AA, Holland-Winkler AM. Relationship Between Systemic Inflammation and Glycemic Control in Firefighters. J Funct Morphol Kinesiol 2025; 10:148. [PMID: 40407432 PMCID: PMC12101145 DOI: 10.3390/jfmk10020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Abstract
Background: Firefighters are at risk for developing metabolic diseases such as type 2 diabetes due to occupational-related stress and poor health behaviors. Firefighters often experience chronic inflammatory responses that may contribute to the development of insulin resistance. This study examined the relationship between systemic inflammation markers and glycemic control markers in firefighters. Methods: Blood samples were collected from twenty full-time male firefighters to assess HbA1c, fasting glucose, and insulin to estimate the Homeostatic Model of Assessment of Insulin Resistance (HOMA-IR), C-reactive protein (CRP), and homocysteine. Body composition and cardiovascular metrics were also recorded. Pearson partial correlation analyses were performed to evaluate relationships between homocysteine and CRP and the variables HOMA-IR and HbA1c while controlling for age and body fat percentage (BF%). SPSS version 29 was used for all analyses (α = 0.05). Data transformation was used where appropriate to ensure the normal distribution of each variable. Results: A significant positive correlation was found between homocysteine and HbA1c before (p = 0.006, r = 0.605) and after controlling for age and BF% (ppartial = 0.013, rpartial = 0.588), indicating that homocysteine levels are associated with impaired glycemic control in firefighters. No other relationships were found to be significant. Conclusions: The findings support a potential link between systemic inflammation and poor glycemic control in firefighters. Due to the occupational hazards that contribute to chronic inflammation, targeted interventions such as dietary modifications may help decrease the risk of diabetes and cardiovascular disease in this high-risk population.
Collapse
|
218
|
Modiga A, Butiurca VO, Boeriu CM, Truta TS, Turucz E, Halațiu VB, Rodean IP, Russu PC, Gherghinescu MC, Molnar C. Pathophysiological Mechanisms Linking COVID-19 and Acute Surgical Abdomen: A Literature Review. Life (Basel) 2025; 15:707. [PMID: 40430138 PMCID: PMC12113513 DOI: 10.3390/life15050707] [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: 03/18/2025] [Revised: 04/19/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Acute surgical abdomen is characterized by intense, sudden abdominal pain due to intra-abdominal conditions requiring prompt surgical intervention. The coronavirus disease 2019 (COVID-19) pandemic has led to various complications related to the disease's complex pathophysiological mechanisms, hence the hypothesis of COVID-19-induced acute abdominal surgical pathologies. The connection between acute surgical abdomen and COVID-19 involves two primary mechanisms. First, there is the presence of angiotensin-converting enzyme 2 (ACE2) receptors in multiple abdominal organs. This facilitates the cytokine storm through direct viral injury and inflammation. Second, the hypercoagulable state induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) increases the thrombotic risk within abdominal vessels, which can subsequently lead to ischemia. ACE2 receptors are notably expressed in the gastric, duodenal, and rectal epithelium, with SARS-CoV-2 viral RNA and nucleocapsid proteins detected in these tissues. The inflammatory response results in significant endothelial damage, activating coagulation pathways that cause monocellular infiltration, lymphocytic inflammation, and uncontrolled coagulation. These findings highlight the need for further research to clarify how COVID-19 leads to acute abdominal pathologies. Understanding these mechanisms is vital for improving clinical management and patient outcomes during future health crises and in the aftermath of the pandemic.
Collapse
Affiliation(s)
- Andrei Modiga
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Vlad-Olimpiu Butiurca
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Cristian Marius Boeriu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Teodora Sorana Truta
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Emilia Turucz
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- Clinical Emergency Department (UCPU-SMURD), County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Vasile-Bogdan Halațiu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
| | - Ioana-Patricia Rodean
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
| | - Paul Cristian Russu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Mircea Constantin Gherghinescu
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| | - Călin Molnar
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540139 Targu Mures, Romania; (C.M.B.); (T.S.T.); (E.T.); (V.-B.H.); (I.-P.R.); (P.C.R.); (M.C.G.); (C.M.)
- General Surgery Clinic No. 1, County Emergency Clinical Hospital of Targu-Mures, 540136 Targu Mures, Romania
| |
Collapse
|
219
|
Li Q, Cui T, Ding H, Shi X, Zhang Y, Jiang P, Han J, Li J, Liu J. Exploring the correlation between high-risk coronary plaque and hepatic fat fraction in non-alcoholic fatty liver disease using spectral computed tomography (CT). Clin Radiol 2025; 86:106943. [PMID: 40403341 DOI: 10.1016/j.crad.2025.106943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/24/2025]
Abstract
AIM To quantitatively assess the fat volume fraction (FVF) in nonalcoholic fatty liver disease (NAFLD) using the spectral computed tomography (CT) multimaterial decomposition (MMD) algorithm and to investigate its association with high-risk coronary plaques (HRP). MATERIALS AND METHODS This retrospective study included patients diagnosed with coronary artery disease (CAD) from August 2023 to August 2024 who underwent coronary CT angiography and abdominal enhanced spectral CT imaging. Patients were categorised into three groups based on HRP imaging features (positive remodelling, low-density plaques, spotty calcification, and napkin ring sign): no plaque (n = 57), non-HRP (n = 54), and HRP (n = 48) groups. FVF was measured using the spectral CT MMD algorithm to quantify liver fat content. Clinical characteristics, biochemical markers, and imaging differences among the groups were analysed. Univariate and multivariate logistic regression analyses were performed to determine the association between FVF and HRP. RESULTS FVF values were significantly higher in the HRP group (13.2%) compared to the non-HRP group (9.2%) and the no plaque group (6.5%) (P<0.001). Multivariate binary logistic regression analysis identified FVF as an independent risk factor for HRP (odds ratio [OR]: 2.55, P<0.001), along with high-sensitivity C-reactive protein (hs-CRP) (OR: 1.94, P=0.025) and diabetes mellitus (OR: 9.83, P=0.002). Additionally, FVF correlated epicardial and pericoronary adipose tissue (PCAT) volume and CT attenuation (P<0.001). CONCLUSION The spectral CT MMD algorithm enables quantitative assessment of FVF, which is independently associated with coronary HRP formation in NAFLD patients. Elevated FVF serves as a risk factor for CAD in patients with NAFLD.
Collapse
Affiliation(s)
- Q Li
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - T Cui
- Graduate School of Hebei North University, China
| | - H Ding
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - X Shi
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - Y Zhang
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - P Jiang
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - J Han
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - J Li
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China
| | - J Liu
- Department of Medical Imaging, Kailuan General Hospital Affiliated to North China University of Science and Technology, China.
| |
Collapse
|
220
|
Mohammadzadeh M, Amirpour M, Ahmadirad H, Abdi F, Khalesi S, Naghshi N, Bahrami A, Hejazi E. Impact of Fasting Mimicking Diet (FMD) on cardiovascular risk factors: a systematic review and meta-analysis of randomized control trials. Diabetol Metab Syndr 2025; 17:137. [PMID: 40287774 PMCID: PMC12032729 DOI: 10.1186/s13098-025-01709-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION The Fasting Mimicking Diet (FMD) has gained significant attention as a potential intervention for reducing cardiovascular risk factors. While studies have investigated its effectiveness, findings have been inconsistent. Therefore, this systematic review and meta-analysis aimed to clarify evidence on the impact of FMD on cardiovascular risk factors. METHOD PubMed, Web of Science, Scopus, and Google Scholar were searched for eligible Randomized controlled trials (RCTs) published up to July 2024. Weighted mean differences (WMD) were calculated for the net changes in risk factors using random-effects models. RESULTS Eleven RCTs (with twelve treatment arms) were included. FMD significantly reduced glycated hemoglobin (HbA1c) (WMD = -8.589 mmol/mol, 95% CI: -12.389, -4.769), insulin-like growth factor 1 (IGF-1) (WMD= -19.211 ng/ml, 95% CI: -32.986, -5.437), systolic blood pressure (SBP) (WMD = -4.148 mmHg, 95% CI: -7.584, -0.711), and diastolic blood pressure (DBP) (WMD = -2.263 mmHg, 95% CI: -4.162, -0.364) levels. No significant effects were observed on other cardiovascular risk factors. CONCLUSION This meta-analysis suggests that FMD can significantly reduce HbA1c, IGF-1, SBP, and DBP levels. Further research is warranted to investigate the long-term and potential clinical implications of FMD on cardiovascular health. PROSPERO REGISTRATION The protocol of the study was registered in the International Prospective Register of Systematic Reviews (PROSPERO registration no: CRD42024569426).
Collapse
Affiliation(s)
- Milad Mohammadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Amirpour
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahmadirad
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Abdi
- Department of Nutrition, Faculty of Nutrition and food science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saman Khalesi
- Appleton Institute, School of Health, Medical and Applied Sciences, Central Queensland University, Brisbane, Australia
| | - Niayesh Naghshi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Bahrami
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Hejazi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
221
|
Jin X, Zhu Y, Xing L, Ding X, Liu Z. PANoptosis: a potential target of atherosclerotic cardiovascular disease. Apoptosis 2025:10.1007/s10495-025-02089-x. [PMID: 40285923 DOI: 10.1007/s10495-025-02089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 04/29/2025]
Abstract
PANoptosis is a newly discovered cell death pathway triggered by the innate immunizer, which in turn promotes the assembly of the PANoptosome and activates downstream effectors. As a special cell death mode, it is characterized by apoptosis, pyroptosis, and necroptosis at the same time; therefore, it is not feasible to inhibit PANoptosis by suppressing a single cell death pathway. However, active ingredients targeting the PANoptosome can effectively inhibit PANoptosis.Given the importance of cell death in disease, targeting PANoptosis would be an important therapeutic tool. Previous studies have focused more on infectious diseases and cancer, and the role of PANoptosis in the cardiovascular field has not been comprehensively addressed. While ASCVD is the number one killer of cardiovascular diseases, it is important to explore new targets to determine future research directions. Therefore, this review focuses on the assembly of PANoptosome, the molecular mechanism of PANoptosis, and the related mechanisms of PANoptosis leading to ASCVD such as myocardial infarction, ischemic cardiomyopathy and ischemic stroke, in order to provide a new perspective for the prevention and treatment of ASCVD.
Collapse
Affiliation(s)
- Xiao Jin
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yanan Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Lina Xing
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Xinyue Ding
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Zongjun Liu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
- , No. 164, Lanxi Road, Putuo District, Shanghai, China.
| |
Collapse
|
222
|
Joy S, Prasannan A, Venkatachalam K, Binesh A. Molecular Mechanism of Notch Signaling and Macrophages in Deep Vein Thrombosis: A Comprehensive Review. Cell Biochem Biophys 2025:10.1007/s12013-025-01761-y. [PMID: 40279070 DOI: 10.1007/s12013-025-01761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Deep vein thrombosis is an acute medical condition, and the molecular basis of this etiology will be crucial in the discovery of more advanced therapies. This review has focused at the Notch signaling pathway, which plays a significant role in different physiological activities such as homeostasis, development, and disease. Also, reveal macrophage function in inflammation and thrombosis in depth, with a focus on their polarization and interaction with the endothelium during thrombosis. In this context, some essential cellular and molecular mechanisms relevant to thrombus pathogenesis, DVT aetiology and risk factors, as well as elements and composition of the Notch pathway, are covered in the end, with a focus on elements that distinguish canonical from non-canonical signaling pathways and their biological relevance to macrophages. Notch signaling has been shown to influence macrophage activation and polarization, influencing their function in thrombosis breakdown and resolution. This interplay between Notch signaling and macrophages may reveal possible treatment targets for DVT. Discuss the physiological role of Notch signaling in vascular biology, as well as how it contributes to thrombosis. The difficulties in implementing these discoveries in clinical practice are discussed, along with the status of ongoing clinical trials and experimental investigations focussing on macrophage-directed treatments and Notch inhibitors. These molecular insights synthesis provides a basis for the creation of novel strategies for the efficient management of DVT.
Collapse
Affiliation(s)
- Sisira Joy
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India
| | - Anusha Prasannan
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India
| | - Kaliyamurthi Venkatachalam
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India
| | - Ambika Binesh
- Department of Basic Sciences, Institute of Fisheries Post Graduate Studies, Tamil Nadu Dr. J. Jayalalithaa Fisheries University (TNJFU), OMR Campus, Vaniyanchavadi, Chennai, Tamil Nadu, India.
| |
Collapse
|
223
|
Jing M, Liu Q, Xi H, Zhu H, Sun Q, Liu X, Zhang Y, Ren W, Deng L, Han T, Zhang B, Zhou J. Relationship between left atrium, epicardial fat and severity of atrial fibrillation. THE INTERNATIONAL JOURNAL OF CARDIOVASCULAR IMAGING 2025:10.1007/s10554-025-03405-x. [PMID: 40281367 DOI: 10.1007/s10554-025-03405-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
To investigate the relationship between left atrium (LA) and epicardial adipose tissue (EAT) parameters and different disease severities (paroxysmal and persistent) in patients with atrial fibrillation (AF). A total of 115 patients with AF (58 paroxysmal and 57 persistent) who underwent cardiac computed tomography angiography (CTA) at our institution between October 2021 and May 2022 were included. The left atrium volume index (LAVI) and left atrium fractal dimension (LAFD) were measured for each patient. EAT volumes and attenuation values for total heart and LA in early and delayed enhancement phases were calculated using semi-automated software. LA and EAT parameters were compared with patients with paroxysmal and persistent AF. Compared with paroxysmal AF, persistent AF had significantly greater LAVI (33.60 ml/m2 vs. 26.65 ml/m2, P < 0.001) and LAFD (1.31 vs. 1.22, P = 0.001). At both early and late enhancement, the total EAT volume (136.29 cm3 vs. 88.68 cm3, 152.30 cm3 vs. 88.96 cm3; all P < 0.001) and attenuation values (-84.00 HU vs. -87.50 HU, -83.00 HU vs. -86.00 HU; all P < 0.05) were significantly higher in persistent AF than in paroxysmal AF. Additionally, LA EAT volumes (15.53 cm3 vs. 8.19 cm3, 18.57 cm3 vs. 9.26 cm3; all P < 0.001) and attenuation values (-74.00 HU vs. -77.00 HU, -75.00 HU vs. -77.00 HU; all P < 0.05) were significantly larger in persistent AF compared with paroxysmal AF, in both early and late enhancement phases. Correlation analysis showed that both LA (r = 0.381, 0.310; P < 0.05) and EAT parameters (r = 0.524, 0.334, 0.665, 0.208, 0.537, 0.223, 0.606, 0.276; P < 0.05) were positively associated with AF severity. Both EAT (volume and attenuation values) and parameters for assessing LA size and morphology, including LAVI and LAFD, were related to the severity of AF.
Collapse
Affiliation(s)
- Mengyuan Jing
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qing Liu
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Huaze Xi
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Hao Zhu
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qiu Sun
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Xuehui Liu
- Ultrasound Medical Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Yuting Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Wei Ren
- Computed Tomography Research Center, GE Healthcare, Beijing, P. R. China
| | - Liangna Deng
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Tao Han
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China
- Second Clinical School, Lanzhou University, Lanzhou, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No.82, Chengguan District, Lanzhou, 730030, PR China.
- Second Clinical School, Lanzhou University, Lanzhou, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
224
|
Liu B, Jin Q, Sun YK, Yang ZM, Meng P, Zhang X, Chen Q, Gan P, Zhao T, He JJ, He GP, Xue Q. From bench to bedside: targeting ferroptosis and mitochondrial damage in the treatment of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2025; 16:1563362. [PMID: 40352456 PMCID: PMC12061709 DOI: 10.3389/fendo.2025.1563362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common and fatal cardiac complication caused by diabetes, with its pathogenesis involving various forms of cell death and mitochondrial dysfunction, particularly ferroptosis and mitochondrial injury. Recent studies have indicated that ferroptosis and mitochondrial damage play crucial roles in the onset and progression of DCM, though their precise regulatory mechanisms remain unclear. Of particular interest is the interaction between ferroptosis and mitochondrial damage, as well as their synergistic effects, which are not fully understood. This review summarizes the roles of ferroptosis and mitochondrial injury in the progression of DCM and explores the molecular mechanisms involved, with an emphasis on the interplay between these two processes. Additionally, the article offers an overview of targeted drugs shown to be effective in cellular experiments, animal models, and clinical trials, analyzing their mechanisms of action and potential side effects. The goal is to provide insights for future drug development and clinical applications. Moreover, the review explores the challenges and prospects of multi-target combination therapies and personalized medicine interventions in clinical practice to offer strategic guidance for the comprehensive prevention and management of DCM.
Collapse
Affiliation(s)
- Bin Liu
- Department of Cardiology, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu People’s Hospital, Gejiu, Yunnan, China
| | - Qing Jin
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Yi Kang Sun
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Zhi Ming Yang
- Department of Cardiology, The Fifth Affiliated Hospital of Kunming Medical University, Gejiu People’s Hospital, Gejiu, Yunnan, China
| | - Ping Meng
- Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Xi Zhang
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Qiu Chen
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
- Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Pin Gan
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Tao Zhao
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Jia Ji He
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Gui Ping He
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| | - Qiang Xue
- Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Key Laboratory of Cardiovascular Disease of Yunnan Province, Kun Min, Yunnan, China
| |
Collapse
|
225
|
Smadja DM, Massonnaud CR, Philippe A, Rosa M, Luneau S, Rauch A, Peiffer-Smadja N, Gagneux-Brunon A, Poissy J, Gruest M, Ung A, Pourcher V, Raffi F, Piroth L, Bouiller K, Esperou H, Delmas C, Belhadi D, Diallo A, Saillard J, Dechanet A, Mercier N, Dupont A, Lescure FX, Goehringer F, Jaureguiberry S, Danion F, Tolsma V, Cabie A, Courjon J, Leroy S, Mootien J, Mourvillier B, Gallien S, Lanoix JP, Botelho-Nevers E, Wallet F, Richard JC, Reuter J, Gaymard A, Greil R, Martin-Blondel G, Andrejak C, Yazdanpanah Y, Burdet C, Diehl JL, Hites M, Ader F, Susen S, Mentré F, Dupont A. sST2 is a key outcome biomarker in COVID-19: insights from discovery randomized trial. Sci Rep 2025; 15:14348. [PMID: 40274842 PMCID: PMC12022249 DOI: 10.1038/s41598-025-95122-7] [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: 08/27/2024] [Accepted: 03/19/2025] [Indexed: 04/26/2025] Open
Abstract
We investigated whether baseline levels of biomarkers related to endotheliopathy, thromboinflammation, and fibrosis were associated with clinical outcomes in hospitalized COVID-19 patients. We analyzed the associations between baseline levels of 21 biomarkers and time to hospital discharge and change in NEWS-2 score in patients from DisCoVeRy trial. We fitted multivariate models adjusted for baseline ISARIC 4C score, disease severity, D-dimer values, and treatment regimen. Between March 22 and June 29, 2020, 603 participants were randomized; 454 had a sample collected at baseline and analyzed. The backward selection of multivariate models showed that higher baseline levels of soluble suppressor of tumorigenicity 2 (sST2) and nucleosomes were statistically associated with a lower chance of hospital discharge before day 29 (sST2: aHR 0.24, 95% CI [0.15-0.38], p < 10-9; nucleosomes: aHR 0.62, 95% CI [0.48-0.81], p < 10-3). Likewise, higher levels of baseline sST2 were statistically associated with lower changes in the NEWS-2 score between baseline and day 15 (adjusted beta 4.47, 95% CI [2.65-6.28], p < 10-5). Moreover, we evaluated sST2 involvement in a confirmation cohort (SARCODO study, 103 patients) and found that elevated baseline sST2 levels were significantly associated with lower rates of hospital discharge before day 29 and a higher model performance (AUC at day 29 of 92%) compared to models without sST2. sST2 emerged as an independent predictor of clinical outcomes in two large cohort of hospitalized COVID-19 patients, warranting further investigation to elucidate its role in disease progression and potential as a therapeutic target.
Collapse
Affiliation(s)
- David M Smadja
- Innovative Therapies in Hemostasis, INSERM, University Paris Cité, 75006, Paris, France.
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France.
| | - Clément R Massonnaud
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Aurélien Philippe
- Innovative Therapies in Hemostasis, INSERM, University Paris Cité, 75006, Paris, France
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France
| | - Mickael Rosa
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Sophie Luneau
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France
| | - Antoine Rauch
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Nathan Peiffer-Smadja
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - Amandine Gagneux-Brunon
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC-INSERM 1408, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Julien Poissy
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Maxime Gruest
- Hematology Department, AP-HP Centre, Georges Pompidou European Hospital, Université Paris Cité, 20 Rue Leblanc, 75015, Paris, France
| | - Alexandre Ung
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - Valérie Pourcher
- Assistance Publique - Hôpitaux de Paris, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Service de Maladies Infectieuses et Tropicales, INSERM 1136, Institut Pierre Louis d'Épidémiologie et de Santé Publique, Sorbonne Université, 75013, Paris, France
| | - François Raffi
- Department of Infectious Diseases, CIC 1413, INSERM, CHU Nantes, Nantes Université, Nantes, France
| | - Lionel Piroth
- Infectious Diseases Department, CHU, Dijon, and INSERM CIC 1432, Université de Bourgogne-Franche Comté, Dijon, France
| | - Kévin Bouiller
- UMR-CNRS 6249 Chrono-Environnement, Department of Infectious and Tropical Diseases, CHU Besançon, Université de Franche-Comté, 25000, Besançon, France
| | - Hélène Esperou
- Institut de Santé Publique, INSERM, Pôle Recherche Clinique, Paris, France
| | | | - Drifa Belhadi
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Alpha Diallo
- ANRS, Maladies Infectieuses Émergentes, Paris, France
| | | | - Aline Dechanet
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | | | - Axelle Dupont
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - François-Xavier Lescure
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - François Goehringer
- Service de Maladies Infectieuses et Tropicales, CHRU-Nancy, Université de Lorraine, 54000, Nancy, France
| | - Stéphane Jaureguiberry
- AP-HP, Service Des Maladies Infectieuses, Hôpital Bicêtre, 94270, Le Kremlin Bicêtre, France
- AP-HP, Centre National de Référence du Paludisme, Paris, France
| | - François Danion
- Service des Maladies Infectieuses et Tropicales, Hôpitaux Universitaires de Strasbourg, 67091, Strasbourg, France
| | - Violaine Tolsma
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Annecy Genevois, 74374, Annecy, France
| | - André Cabie
- PCCEI, Inserm, EFS, Univ Montpellier, Univ Antilles, Montpellier, France
- Service des Maladies Infectieuses et Tropicales, Inserm CIC1424, CHU de Martinique, Martinique, France
| | - Johan Courjon
- Service des Maladies Infectieuses et Tropicales, CHU de Nice, Nice, France
- U1065, Centre Méditerranéen de Médecine Moléculaire, C3M, Virulence Microbienne et Signalisation Inflammatoire, INSERM, Université Côte d'Azur, Nice, France
| | - Sylvie Leroy
- Fédération Hospitalo-Universitaire OncoAge, Nice, France
- Département de Pneumologie et d'Oncologie, CHU de Nice, 06000, Nice, France
- CNRS UMR 7275, IPMC, Université Côte d'Azur, Sophia Antipolis, France
| | - Joy Mootien
- Service de Réanimation Médicale, Groupe Hospitalier de la Région Mulhouse Sud-Alsace, Mulhouse, France
| | - Bruno Mourvillier
- CHU de Reims, Service de Réanimation Médicale, Université de Reims Champagne-Ardenne, Reims, France
| | - Sébastien Gallien
- Service d'Immunologie et Maladies Infectieuses, AP-HP, Hôpital Henri Mondor, 94000, Créteil, France
- INSERM U955, Université Paris-Est Créteil, 94000, Créteil, France
| | - Jean-Philippe Lanoix
- Service de Maladies Infectieuses et Tropicales, CHU Amiens-Picardie, 80000, Amiens, France
- AGIR UR UPJV 4294, CURS, Université Picardie Jules Verne, 80000, Amiens, France
| | - Elisabeth Botelho-Nevers
- Department of Infectious Diseases, University Hospital of Saint-Etienne, Saint-Etienne, France
- CIC-INSERM 1408, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Florent Wallet
- Département de Soins Intensifs, Hospices Civils de Lyon, Hôpital Lyon-Sud Pierre-Bénite, 69000, Lyon, France
| | - Jean-Christophe Richard
- Service de Réanimation Médicale, Hospices Civils de Lyon, Hôpital de la Croix-Rousse, 69000, Lyon, France
- CREATIS, CNRS UMR5220, INSERM U1044, INSA, Université Lyon I, 69000, Lyon, France
| | - Jean Reuter
- Service de Réanimation-Soins Intensifs, Centre Hospitalier de Luxembourg, 1210, Luxembourg, Luxembourg
| | - Alexandre Gaymard
- Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des Virus Respiratoires France Sud, Hospices Civils de Lyon, 69317, Lyon, France
- Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, 69372, Lyon, France
| | - Richard Greil
- Department of Internal Medicine III With Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University Salzburg, 5020, Salzburg, Austria
| | - Guillaume Martin-Blondel
- Service des Maladies Infectieuses et Tropicales, CHU de Toulouse, 31320, Toulouse, France
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM UMR1291 - CNRS UMR5051, Université Toulouse III, 31320, Toulouse, France
| | - Claire Andrejak
- Département de Pneumologie, CHU d'Amiens, 80000, Amiens, France
| | - Yazdan Yazdanpanah
- IAME, INSERM, Université de Paris, 75018, Paris, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat, 75018, Paris, France
| | - Charles Burdet
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Jean-Luc Diehl
- Innovative Therapies in Hemostasis, INSERM, University Paris Cité, 75006, Paris, France
- Intensive Care Unit, AP-HP Centre Université Paris Cité, Georges Pompidou European Hospital, 75015, Paris, France
| | - Maya Hites
- Clinic of Infectious Diseases, Hôpital Universitaire de Bruxelles (H.U.B), 1070, Brussels, Belgium
| | - Florence Ader
- Département des Maladies Infectieuses et Tropicales, Hospices Civils de Lyon, Lyon, France
- Département des Maladies Infectieuses et Tropicales, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Sophie Susen
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| | - France Mentré
- Département d'Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, 75018, Paris, France
- IAME, INSERM, Université de Paris, 75018, Paris, France
| | - Annabelle Dupont
- Inserm, CHU Lille, Institut Pasteur de Lille, U1011- EGID, Université de Lille, 59000, Lille, France
| |
Collapse
|
226
|
Wang Y, Bi L, Li Q, Wang Q, Lv T, Zhang P. Remnant cholesterol inflammatory index and its association with all-cause and cause-specific mortality in middle-aged and elderly populations: evidence from US and Chinese national population surveys. Lipids Health Dis 2025; 24:155. [PMID: 40275392 PMCID: PMC12020154 DOI: 10.1186/s12944-025-02580-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND The remnant cholesterol inflammatory index (RCII) is a novel metric that combines remnant cholesterol and high-sensitivity C-reactive protein, reflecting the metabolic and inflammatory risk. This study investigates the association between RCII and long-term risks of all-cause and cause-specific mortality in middle-aged and elderly populations in the US and China. METHOD We analyzed data from the National Health and Nutrition Examination Survey (NHANES) and the China Health and Retirement Longitudinal Study (CHARLS), including 7,565 and 12,932 participants aged 45 years and older, respectively. The participants were categorized into quartiles based on natural log-transformed RCII (lnRCII) values. Kaplan-Meier survival analysis, Cox proportional hazards models, restricted cubic splines (RCS) and mediation analysis were used to examine the relationship between lnRCII and mortality outcomes, adjusting for potential covariates. RESULT The mean age of the participants was 59.90 ± 10.44 years (NHANES) and 58.64 ± 9.78 years (CHARLS), with 53.28% and 52.50% female, respectively. Kaplan-Meier survival analysis showed that higher lnRCII quartiles (≥ 0.79 in NHANES, ≥ -0.13 in CHARLS) were significantly associated with increased all-cause mortality risk (p < 0.001). Each standard deviation (SD) increase in lnRCII corresponded to a higher risk of all-cause mortality, and the hazard ratios (HRs) and 95% confidence interval (CI) were 1.29 (95% CI: 1.21-1.36) in NHANES and 1.26 (95% CI: 1.15-1.38) in CHARLS. In NHANES, lnRCII was also associated with elevated risks of cardiovascular mortality (HR = 1.21, 95% CI: 1.08-1.35) and cancer mortality (HR = 1.30, 95% CI: 1.09-1.55). RCS analysis indicated a J-shaped relationship between lnRCII and both all-cause and cardiovascular mortality, and a linear association with cancer mortality. Mediation analysis showed that systolic blood pressure and fasting plasma glucose partially mediated these associations. Subgroup analyses suggested a stronger association between lnRCII and all-cause mortality in middle-aged US participants (p for interaction = 0.010). CONCLUSIONS Elevated RCII levels are significantly associated with increased all-cause mortality risk middle-aged and elderly populations in both the US and China. In the US population, RCII is also associated with increased risks of cardiovascular and cancer mortality. By integrating metabolic and inflammatory risk factors, RCII may serve as a valuable tool for mortality risk stratification and clinical decision-making.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Lei Bi
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Qing Li
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Qiuyu Wang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Tingting Lv
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China.
| | - Ping Zhang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China.
| |
Collapse
|
227
|
Tang Y, Guo T, Wang X, Li C, Zhang X, Zhang J. Cyclodextrin-Derived Macromolecular Therapies for Inflammatory Diseases. Macromol Biosci 2025:e2400637. [PMID: 40271896 DOI: 10.1002/mabi.202400637] [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: 12/21/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Inflammation is an essential physiological defense mechanism against harmful stimuli, yet dysregulated inflammatory responses are closely associated with the pathogenesis of numerous acute and chronic diseases. Recent advances highlight the remarkable anti-inflammatory potential of bioactive macromolecules, particularly cyclodextrins (CDs) and their engineered derivatives, which are emerging as promising therapeutic agents. This review systematically introduces different CDs and CD-derived macromolecules that demonstrate anti-inflammatory properties, with emphasis on their molecular mechanisms of action. Native CDs exhibit direct therapeutic effects through host-guest interactions, enabling selective sequestration of pathogenic components such as cholesterol crystals and proteins that drive inflammatory cascades. Moreover, chemically modified CD derivatives incorporating functional groups demonstrate enhanced capabilities in neutralizing inflammatory mediators and modulating immune cell responses. This work further discusses the expanding therapeutic applications of these macromolecules across diverse inflammatory conditions, ranging from acute tissue injuries to chronic autoimmune disorders. Finally, this work critically analyzes the crucial challenges and emerging opportunities in translating CD-based macromolecular therapies into clinical practice, addressing key considerations in biocompatibility, targeted delivery, and therapeutic efficacy optimization.
Collapse
Affiliation(s)
- Yige Tang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xuanran Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China
| |
Collapse
|
228
|
Bonaventura A, Liberale L, Carbone F, Montecucco F. Cancer and cardiovascular diseases: A deadly combination. Eur J Clin Invest 2025:e70060. [PMID: 40270272 DOI: 10.1111/eci.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025]
Affiliation(s)
- Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Department of Internal Medicine, Ospedale di Circolo and Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
229
|
Xiong Z, Hu L, Peng H, Jiang X, Wang X, Tu L, Xu S. UCN expresses differently in left-sided and right-sided colon cancer contributing to distinct immune microenvironment via regulating CCL23. Hum Immunol 2025:111308. [PMID: 40274491 DOI: 10.1016/j.humimm.2025.111308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/28/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025]
Abstract
PURPOSE Urocortin (UCN), is found to be overexpressed in colorectal cancer (CRC) but its role in tumor immune microenvironment (TIME) remains unclear. METHODS UCN expression was analyzed using RNA sequencing data from TCGA and detected in tumor samples from 18 patients with LC and 27 patients with RC. Tumor infiltrated T cells (TILs) were isolated from tumors and the exhaustion markers including PD-1, TIGIT, TIM-3 and LAG3 on CD8 + TILs were detected by flow cytometry. Correlations between UCN level and immunoregulatory factors were analyzed using Spearman correlation analysis. UCN was overexpressed in CRC cell lines and the CCL23 levels were detected by quantitative RT-PCR and enzyme-linked immunosorbent assay. RESULTS UCN was mainly overexpressed in Right-sided colon cancer (RC) and to be related to poor prognosis. Higher UCN level in tumors related to increased abundance of regulatory T cells and upregulated exhaustion markers in CD8 + T cells. UCN is positively correlated with CCL23 level in CRC and mainly upregulated in RC samples. Overexpression of UCN in HCT116 and HT-29 cells upregulated CCL23 expression and promoted CCL23 secretion. CD8 + T cells cultured with medium from UCN overexpressed CRC cells exhibited exhaustion phenotype with increased expression of CTLA-4, PD-1, TIGIT, TIM-3 and LAG-3, which was restored by CCL23 antibody. CONCLUSION This study successfully constructed the correlation between UCN overexpression and immunosuppressive TIME formation in CRC, providing a candidate target for new immunotherapy against CRC development.
Collapse
Affiliation(s)
- Zhenfang Xiong
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Long Hu
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Haiyan Peng
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyue Jiang
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Xu Wang
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Luxia Tu
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory for Precision Pathology and Intelligent Diagnosis, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
230
|
Tangianu F, Nicolini E, Dentali F, Bonaventura A. SGLT-2 inhibitors for cardiac amyloidosis: Hype or hope? Eur J Clin Invest 2025:e70063. [PMID: 40264435 DOI: 10.1111/eci.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Affiliation(s)
- Flavio Tangianu
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Varese, Italy
| | - Eleonora Nicolini
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Varese, Italy
| | - Francesco Dentali
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Aldo Bonaventura
- Medical Center, S.C. Medicina Generale 1, Ospedale di Circolo and Fondazione Macchi, Department of Internal Medicine, ASST Sette Laghi Varese, Varese, Italy
| |
Collapse
|
231
|
Morrissey SM, Kirkland LG, Phillips TK, Levit RD, Hopke A, Jensen BC. Multifaceted roles of neutrophils in cardiac disease. J Leukoc Biol 2025; 117:qiaf017. [PMID: 39936506 DOI: 10.1093/jleuko/qiaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/15/2025] [Accepted: 02/11/2025] [Indexed: 02/13/2025] Open
Abstract
Neutrophils, the most abundant leukocytes in human blood, have long been recognized as critical first responders in the innate immune system's defense against pathogens. Some of the more notable innate antimicrobial properties of neutrophils include generation of superoxide free radicals like myeloperoxidase, production of proteases that reshape the extracellular matrix allowing for easier access to infected tissues, and release of neutrophil extracellular traps, extruded pieces of DNA that ensnare bacterial and fungi. These mechanisms developed to provide neutrophils with a vast array of specialized functions to provide the host defense against infection in an acute setting. However, emerging evidence over the past few decades has revealed a far more complex and nuanced role for these neutrophil-driven processes in various chronic conditions, particularly in cardiovascular diseases. The pathophysiology of cardiac diseases involves a complex interplay of hemodynamic, neurohumoral, and inflammatory factors. Neutrophils, as key mediators of inflammation, contribute significantly to this intricate network. Their involvement extends far beyond their classical role in pathogen clearance, encompassing diverse functions that can both exacerbate tissue damage and contribute to repair processes. Here, we consider the contributions of neutrophils to myocardial infarction, heart failure, cardiac arrhythmias, and nonischemic cardiomyopathies. Understanding these complex interactions is crucial for developing novel therapeutic strategies aimed at modulating neutrophil functions in these highly morbid cardiac diseases.
Collapse
Affiliation(s)
- Samantha M Morrissey
- Department of Medicine, University of North Carolina School of Medicine, 125 MacNider Hall, Chapel Hill, NC 27599-7005, United States
| | - Logan G Kirkland
- McAllister Heart Institute, University of North Carolina School of Medicine, 111 Mason Farm Rd., Chapel Hill, NC 27599-7126, United States
| | - Tasha K Phillips
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, United States
| | - Rebecca D Levit
- Division of Cardiology, Department of Medicine, Emory University, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Alex Hopke
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, PO Box 70577, Johnson City, TN 37614, United States
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, PO Box 70300, Johnson City, TN 37614, United States
| | - Brian C Jensen
- Department of Medicine, University of North Carolina School of Medicine, 125 MacNider Hall, Chapel Hill, NC 27599-7005, United States
- McAllister Heart Institute, University of North Carolina School of Medicine, 111 Mason Farm Rd., Chapel Hill, NC 27599-7126, United States
- Department of Pharmacology, University of North Carolina School of Medicine, 120 Mason Farm Rd., Chapel Hill, NC 27599-7365, United States
| |
Collapse
|
232
|
Li M, Sun X, Zeng L, Sun A, Ge J. Metabolic Homeostasis of Immune Cells Modulates Cardiovascular Diseases. RESEARCH (WASHINGTON, D.C.) 2025; 8:0679. [PMID: 40270694 PMCID: PMC12015101 DOI: 10.34133/research.0679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025]
Abstract
Recent investigations into the mechanisms underlying inflammation have highlighted the pivotal role of immune cells in regulating cardiac pathophysiology. Notably, these immune cells modulate cardiac processes through alternations in intracellular metabolism, including glycolysis and oxidative phosphorylation, whereas the extracellular metabolic environment is changed during cardiovascular disease, influencing function of immune cells. This dynamic interaction between immune cells and their metabolic environment has given rise to the novel concept of "immune metabolism". Consequently, both the extracellular and intracellular metabolic environment modulate the equilibrium between anti- and pro-inflammatory responses. This regulatory mechanism subsequently influences the processes of myocardial ischemia, cardiac fibrosis, and cardiac remodeling, ultimately leading to a series of cardiovascular events. This review examines how local microenvironmental and systemic environmental changes induce metabolic reprogramming in immune cells and explores the subsequent effects of aberrant activation or polarization of immune cells in the progression of cardiovascular disease. Finally, we discuss potential therapeutic strategies targeting metabolism to counteract abnormal immune activation.
Collapse
Affiliation(s)
- Mohan Li
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Linqi Zeng
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| |
Collapse
|
233
|
Wu H, Huang J, Wu H, Xu W, Zhong Q, Song J, Linghu X, Gao B, Wa Q. Enhancement of in vitro and in vivo bone repair performance of decalcified bone/gelma by desferrioxamine. Sci Rep 2025; 15:14092. [PMID: 40269226 PMCID: PMC12019368 DOI: 10.1038/s41598-025-99101-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: 10/14/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Autologous and allogeneic bone grafting is currently the clinical gold standard for the treatment of bone defects; however, it is limited by the scarcity of autologous sources and the risk of secondary trauma, as well as the complications of disease transmission and immune rejection associated with allogeneic grafts. The clinical management of bone defects remains a significant challenge. In this study, we prepared a demineralized bone matrix/gelatin methacrylate composite hydrogel loaded with deferoxamine (GelMA/DBM/DFO) using a freeze-drying method and investigated its properties. Assessments using CCK-8, live-dead fluorescence staining, alkaline phosphatase staining, and Alizarin Red staining indicated that the GelMA/DBM/DFO composite hydrogel demonstrated superior biocompatibility and in vitro osteogenic differentiation capacity compared with the GelMA/DBM composite hydrogel. We established a cranial defect model in Sprague-Dawley (SD) rats and examined peripheral blood indices, micro-computed tomography (Micro-CT), hematoxylin and eosin (HE) staining, Masson's trichrome staining, and immunohistochemical staining for bone morphogenetic protein-2 (BMP-2) and collagen type I (COL-1). Both hydrogels exhibited good biosafety and the GelMA/DBM/DFO hydrogel showed more effective repair of cranial defects in SD rats. This study provides a novel material for bone-defect repair.
Collapse
Affiliation(s)
- Honghan Wu
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Jun Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510632, China
| | - Hengpeng Wu
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Weikang Xu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510632, China
| | - Qian Zhong
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Jiaxiang Song
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Xitao Linghu
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510632, China.
| | - Qingde Wa
- Departament of Orthopadic Surgery, The Second Affiliated Hospital of Zunyi Medical University, zunyi, 563000, China.
| |
Collapse
|
234
|
Kress TC, Barris CT, Kovacs L, Khakina BN, Jordan CR, Bruder-Nascimento T, Stepp DW, MacArthur R, Patel VS, Chen J, Pacholczyk R, Kennard S, Belin de Chantemèle EJ. CD4 + T Cells Expressing Viral Proteins Induce HIV-Associated Endothelial Dysfunction and Hypertension Through Interleukin 1α-Mediated Increases in Endothelial NADPH Oxidase 1. Circulation 2025; 151:1187-1203. [PMID: 39907014 PMCID: PMC12011537 DOI: 10.1161/circulationaha.124.070538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 01/13/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Although combination antiretroviral therapy has increased life expectancy in people living with HIV, it has led to a marked increase in the prevalence of hypertension, the cause of which is unknown. Despite combination antiretroviral therapy, HIV-derived proteins remain expressed and produced by CD4+ T lymphocytes in people living with HIV. However, their contribution to HIV-associated hypertension and impaired endothelium-dependent relaxation remains ill defined. METHODS Here, we tested the hypothesis that CD4+ T cells expressing viral proteins contribute to endothelial dysfunction and hypertension using the Tg26 mouse model of HIV that expresses 7 of the 9 HIV proteins under the long terminal repeat promoter. We used male and female mice, bone marrow transplantation (BMT), adoptive transfer of CD4+ T cells, and aorta specimen discarded from people living with HIV. RESULTS We reported that intact Tg26 mice and mice receiving BMT (Tg26→WT) or CD4+ T cells from Tg26 mice display impaired endothelium-dependent relaxation and hypertension. Conversely, BMT from WT mice into Tg26 mice, inhibition of T cell activation, and CD4+ T cell depletion restored endothelial function and blood pressure in Tg26 mice. Cytokine profiling revealed that Tg26 mice, Tg26→WT, and Tg26 CD4+ T cells consistently exhibit high interleukin 1α (IL-1α) levels with no significant increase in other cytokines, whereas BMT from WT mice into Tg26 mice reduced IL-1α levels. IL-1α neutralization reduced blood pressure and restored endothelial function in Tg26 mice. To investigate the role of CD4+ T cells and IL-1α in endothelial dysfunction, we developed an aorta-immune cell coculture system. Exposure of WT aortas to Tg26 CD4+ T cells impaired endothelium-dependent relaxation, which was blocked by IL-1α-neutralizing antibody. While investigating the mechanisms of endothelial dysfunction, we reported that Tg26 mice, Tg26→WT aorta exhibit high NADPH oxidase (NOX) 1 expression. IL-1α exposure increased NOX1 in human microvascular endothelial cells, and NOX1 blockade restored endothelial function in Tg26 and Tg26→WT arteries, whereas NOX1 deficiency protected against Tg26 BMT-induced impaired endothelium-dependent relaxation and hypertension. Aortas from people living with HIV exhibit high NOX1 levels, and exposure of human aorta to Tg26 T cells increased NOX1 expression. CONCLUSIONS We provide the first evidence that CD4+ T cells expressing HIV viral proteins induced hypertension through IL-1α-mediated increases in vascular NOX1, which impairs endothelial function in males and females.
Collapse
Affiliation(s)
- Taylor C Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Laszlo Kovacs
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Beryl N. Khakina
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Coleton R. Jordan
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Thiago Bruder-Nascimento
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL 36688, USA
| | - David W. Stepp
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Rodger MacArthur
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Vijay S. Patel
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Jie Chen
- Department of Biostatistics, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Rafal Pacholczyk
- Department of Biochemistry and Molecular Biology, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| | - Eric J. Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia 30912
| |
Collapse
|
235
|
Franco-da-Silva MM, Aubin MR, de Vasconcellos AA, Sirena DH, Marchaki GB, Ruggeri LR, Hennigen AF, Muradás T, da Silveira ABT, Braganhol E, Schuh RS, Baldo G, Araújo AB, Paz AH. Effects of chorionic mesenchymal stromal cells, their conditioned medium, and membrane particles on neutrophil functionality. Cell Tissue Res 2025:10.1007/s00441-025-03970-6. [PMID: 40261417 DOI: 10.1007/s00441-025-03970-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Mesenchymal stromal cells (MSC) are multipotent cells that can modulate immune cells, affecting macrophages, monocytes, and lymphocytes. Neutrophils are circulating leucocytes responsible for the first line of defense and can assume different phenotypes depending on their environment: N0, the naïve form, N1 (inflammatory), N2 (anti-inflammatory). This study explores the potentially protective roles of chorionic membrane MSCs and their products-conditioned medium and pre-conditioned cMSC-derived membrane microparticles (MP-cMSC)-on neutrophils. Conditioned medium treatment reduced the rate of apoptosis and enhanced the immunosuppressive potential consistent with an anti-inflammatory profile. MP-cMSC are a noteworthy cell-free therapy, consisting of artificially generated circular lipid bilayer structures with no cargo and approximately 200 nm in size. When added to neutrophil culture, MPs increased neutral red uptake, suggesting an enhanced phagocytic activity. In the MSC co-culture group, a reduced rate of apoptosis, increased neutral red uptake, and elevated programed death-ligand 1 (PD-L1) expression were observed. These findings suggest that the distinct effects elicited by conditioned media, microparticles, and co-culture are likely influenced by the specific nature of the interactions involved-whether purely paracrine, mediated through direct cell-to-cell contact, or a combination of both.
Collapse
Affiliation(s)
- Monique Maria Franco-da-Silva
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Mariana Rauback Aubin
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Alessandra Amaral de Vasconcellos
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Dienifer Hermann Sirena
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Giovana Bangel Marchaki
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Laíza Rief Ruggeri
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - André Ferreira Hennigen
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Thaís Muradás
- Pharmaceutical Sciences - Graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Ana Beatriz Tittoni da Silveira
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Elizandra Braganhol
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Roselena Silvestri Schuh
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Pharmaceutical Sciences - Graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Guilherme Baldo
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Anelise Bergmann Araújo
- Cell Processing Center, Hemotherapy Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Ana Helena Paz
- Cells, Tissues and Genes Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
- Graduate Program in Physiology, Basic Health Sciences Institute, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| |
Collapse
|
236
|
Crea F. Focus on heart failure: sex-related differences, new light shed on SGLT2i mechanism of action, and heart failure after myocardial infarction. Eur Heart J 2025; 46:1457-1461. [PMID: 40259437 DOI: 10.1093/eurheartj/ehaf185] [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: 04/23/2025] Open
Affiliation(s)
- Filippo Crea
- Center of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
- Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
237
|
Padmavathi V, Ganesan K. Metaheuristic optimizers integrated with vision transformer model for severity detection and classification via multimodal COVID-19 images. Sci Rep 2025; 15:13941. [PMID: 40263404 PMCID: PMC12015488 DOI: 10.1038/s41598-025-98593-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/14/2025] [Indexed: 04/24/2025] Open
Abstract
This study introduces a novel hybrid framework for classifying COVID-19 severity using chest X-rays (CXR) and computed tomography (CT) scans by integrating Vision Transformers (ViT) with metaheuristic optimization techniques. The framework employs the Grey Wolf Optimizer (GWO) for hyperparameter tuning and Particle Swarm Optimization (PSO) for feature selection, leveraging the ViT model's self-attention mechanism to extract global and local image features crucial for severity classification. A multi-phase classification strategy refines predictions by progressively distinguishing normal, mild, moderate, and severe COVID-19 cases. The proposed GWO_ViT_PSO_MLP model achieves outstanding accuracy, with 99.14% for 2-class CXR classification and 98.89% for 2-class CT classification, outperforming traditional CNN-based approaches such as ResNet34 (84.22%) and VGG19 (93.24%). Furthermore, it demonstrates superior performance in multi-class severity classification, especially in differentiating challenging cases like mild and moderate infections. Compared to existing studies, this framework significantly improves accuracy and computational efficiency, highlighting its potential as a scalable and reliable solution for automated COVID-19 severity detection in clinical applications.
Collapse
Affiliation(s)
- V Padmavathi
- Department of Biomedical Engineering, CEG Campus, Anna University, Chennai, 600025, India
| | - Kavitha Ganesan
- Department of Biomedical Engineering, CEG Campus, Anna University, Chennai, 600025, India.
| |
Collapse
|
238
|
Ji XM, Dong XX, Li JP, Tai GJ, Qiu S, Wei W, Silumbwe CW, Damdinjav D, Otieno JN, Li XX, Xu M. Fisetin Clears Senescent Cells Through the Pi3k-Akt-Bcl-2/Bcl-xl Pathway to Alleviate Diabetic Aortic Aging. Phytother Res 2025. [PMID: 40259678 DOI: 10.1002/ptr.8507] [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: 12/10/2024] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 04/23/2025]
Abstract
Vascular aging is a major contributor to age-related cardiovascular diseases (CVDs) and type 2 diabetes mellitus (T2DM) induced early arterial aging and excessive senescent cells (SCs) burden in vessels. Inhibiting cellular senescence or eliminating SCs could effectively improve aging-related CVDs. Fisetin, a flavonoid extracted from cotinus coggygria scop, has shown potential in alleviating aging by clearing SCs. This study investigated the unexplored mechanisms and efficacy of fisetin in alleviating T2DM-related aortic aging. The T2DM mouse model was induced using a high-fat diet and low-dose streptozotocin injection. Chronic fisetin treatment's protective effects against aortic aging were assessed via senescence-associated beta-galactosidase (SA-β-Gal) staining, histopathology, and vasomotor function. RNA-sequencing and western blotting identified relevant signaling pathways and protein expression. Fisetin's effects on SCs and senescence-associated secretory phenotype (SASP) factors were evaluated through cell viability, apoptosis, and co-culture assays. Docking simulations suggested fisetin as a potential Phosphoinositide 3-kinase (Pi3k) inhibitor. In vivo, chronic fisetin treatment reduced aortic SCs burden, alleviating T2DM-related and natural aortic aging. In vitro, fisetin selectively induced apoptosis of senescent endothelial cells via regulating the Pi3k-Protein Kinase B (Akt)-B-cell lymphoma (Bcl)-2/Bcl-xl pathway and suppressed SASP and its detrimental effects. Furthermore, fisetin combined with metformin therapy showed superior anti-aging effects on T2DM-related aortic aging compared to metformin monotherapy. In conclusion, chronic fisetin treatment alleviates T2DM-related aortic aging via clearing the SCs burden and abrogating the SASP factors. Fisetin combined with metformin therapy might be a potential therapeutic strategy for T2DM-related CVDs.
Collapse
Affiliation(s)
- Xiao-Man Ji
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin-Xin Dong
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jia-Peng Li
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Guang-Jie Tai
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shu Qiu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wei Wei
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ceaser Wankumbu Silumbwe
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Joseph Nicolao Otieno
- Institute of Traditional Medicine, Muhimbili University of Health and Allied Sciencea, Dar es Salaam, Tanzania
| | - Xiao-Xue Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ming Xu
- Department of Clinical Pharmacy, School of Preclinical Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
239
|
Marcos-Ríos D, Rochano-Ortiz A, San Sebastián-Jaraba I, Fernández-Gómez MJ, Méndez-Barbero N, Oller J. Mitochondrial Dysfunction: A New Hallmark in Hereditable Thoracic Aortic Aneurysm Development. Cells 2025; 14:618. [PMID: 40277943 PMCID: PMC12026022 DOI: 10.3390/cells14080618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/11/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
Thoracic aortic aneurysms (TAAs) pose a significant health burden due to their asymptomatic progression, often culminating in life-threatening aortic rupture, and due to the lack of effective pharmacological treatments. Risk factors include elevated hemodynamic stress on the ascending aorta, frequently associated with hypertension and hereditary genetic mutations. Among the hereditary causes, Marfan syndrome is the most prevalent, characterized as a connective tissue disorder driven by FBN1 mutations that lead to life-threatening thoracic aortic ruptures. Similarly, mutations affecting the TGF-β pathway underlie Loeys-Dietz syndrome, while mutations in genes encoding extracellular or contractile apparatus proteins, such as ACTA2, are linked to non-syndromic familial TAA. Despite differences in genetic origin, these hereditary conditions share central pathophysiological features, including aortic medial degeneration, smooth muscle cell dysfunction, and extracellular remodeling, which collectively weaken the aortic wall. Recent evidence highlights mitochondrial dysfunction as a crucial contributor to aneurysm formation in Marfan syndrome. Disruption of the extracellular matrix-mitochondrial homeostasis axis exacerbates aortic wall remodeling, further promoting aneurysm development. Beyond its structural role in maintaining vascular integrity, the ECM plays a pivotal role in supporting mitochondrial function. This intricate relationship between extracellular matrix integrity and mitochondrial homeostasis reveals a novel dimension of TAA pathophysiology, extending beyond established paradigms of extracellular matrix remodeling and smooth muscle cell dysfunction. This review summarizes mitochondrial dysfunction as a potential unifying mechanism in hereditary TAA and explores how understanding mitochondrial dysfunction, in conjunction with established mechanisms of TAA pathogenesis, opens new avenues for developing targeted treatments to address these life-threatening conditions. Mitochondrial boosters could represent a new clinical opportunity for patients with hereditary TAA.
Collapse
Affiliation(s)
- Daniel Marcos-Ríos
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
| | - Antonio Rochano-Ortiz
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
| | - Irene San Sebastián-Jaraba
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - María José Fernández-Gómez
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
| | - Nerea Méndez-Barbero
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio (UAX), Villanueva de la Cañada, 28691 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; (D.M.-R.); (A.R.-O.); (I.S.S.-J.); (M.J.F.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio (UAX), Villanueva de la Cañada, 28691 Madrid, Spain
| |
Collapse
|
240
|
Bruns B, Elsous N, Burghaus I, Steyrer K, Joos M, Krämer T, Scheffel M, Blankenberg S, Eitel I, Massberg S, Thiele H, Meder B, Backs J, Frey N. Rationale and design of the cyclosporine in Takotsubo syndrome (CIT) trial. Am Heart J 2025:S0002-8703(25)00133-4. [PMID: 40268179 DOI: 10.1016/j.ahj.2025.04.020] [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: 01/06/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Takotsubo syndrome (TTS) is associated with substantial morbidity and mortality, even though ejection fraction frequently recovers spontaneously. TTS has been suggested to be caused by catecholamine excess leading to myocardial inflammation as an additional driver of cardiac damage and impaired outcome. Currently, no evidence-based treatment exists. In a preclinical model of catecholamine-driven TTS, cyclosporine A (CsA) bolus therapy significantly improved outcome, likely mediated via suppression of calcineurin-driven inflammation. The Cyclosporine In Takotsubo syndrome (CIT) trial is a pilot multicenter double-blinded randomized placebo-controlled trial (RCT) to investigate the impact of CsA bolus therapy in patients suffering from acute TTS. STUDY DESIGN AND OBJECTIVES This RCT is designed to investigate the impact of repetitive CsA bolus therapy vs. placebo in acute high-risk TTS patients with an increased risk of intrahospital complications and long-term mortality. The main goal is to reduce myocardial damage quantified by AUC of a centrally measured high-sensitive cardiac Troponin T (hs-cTnT) over 72 hours (primary endpoint). Therefore, patients with TTS will be randomized 1:1 after angiography and receive an intravenous bolus of 2.5 mg/kg CsA or an equivalent amount of placebo immediately after baseline measurements. At 12 and 24 hours additional doses of the study drug will be applied accumulating to 7.5 mg/kg in the intervention group. After baseline laboratory measurements (including hs-cTnT) and echocardiography (TTE), serum parameters will be measured again at 3 hours and every 12 hours from baseline. TTE imaging will be performed at 24, 48 and 72 hours, and cardiac magnetic resonance imaging (CMR) at 24 to 96 hours. Left ventricular function recovery, myocardial edema (CMR), in-hospital complications, length of hospital stay, 30-day and 1-year composite cardiovascular outcome, as well as Kansas City Cardiomyopathy Questionnaire self-assessment are included as secondary endpoints. CONCLUSIONS The CIT trial is designed to assess the safety and potential benefit of CsA on hs-cTnT release as an established marker of myocardial injury in high-risk TTS patients. The results of this trial may reveal CsA as a first pathophysiology-driven treatment option of TTS and enable a phase III follow-up trial powered for clinical outcome parameters as primary endpoint. CLINICAL TRIALS IDENTIFIER NCT05946772.
Collapse
Affiliation(s)
- Bastian Bruns
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany; Department of General Internal Medicine and Psychosomatics, Heidelberg University Hospital, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Nesrin Elsous
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Ina Burghaus
- Coordinating Center for Clinical Trials (KKS) Heidelberg, Heidelberg, Germany
| | - Kathleen Steyrer
- Coordinating Center for Clinical Trials (KKS) Heidelberg, Heidelberg, Germany
| | - Maximilian Joos
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Tobias Krämer
- Coordinating Center for Clinical Trials (KKS) Heidelberg, Heidelberg, Germany
| | - Marina Scheffel
- Coordinating Center for Clinical Trials (KKS) Heidelberg, Heidelberg, Germany
| | - Stefan Blankenberg
- University Heart Center Hamburg, Clinic of general and interventional Cardiology, German Center for Cardiovascular Research (DZHK), University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Eitel
- University Medical Center Schleswig-Holstein/Campus Lübeck, German Center for Cardiovascular Research (DZHK), Lübeck, Germany
| | - Steffen Massberg
- Medical Clinic and Polyclinic I, German Center for Cardiovascular Research (DZHK), Partner site Munich, Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Benjamin Meder
- Heart Center Heidelberg, Institute for Cardiomyopathies Heidelberg, Heidelberg University Hospital, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Johannes Backs
- Institute of Experimental Cardiology, Heidelberg University Hospital, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany.
| |
Collapse
|
241
|
Mai A, Yasarlar Y, Awad M, Terasaki Y, Zehr K, Soubani AO, Esber Z. The Role of Venoarterial Ecmo in the Management of Sepsis-Induced Cardiomyopathy and Refractory Shock: An Analytic Review. J Intensive Care Med 2025:8850666251321788. [PMID: 40255084 DOI: 10.1177/08850666251321788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Sepsis-induced cardiomyopathy is a severe complication seen in patients with refractory septic shock, characterized by cardiac dysfunction due to an overwhelming inflammatory response secondary to infection. Despite its reversible nature, sepsis-induced cardiomyopathy (SIC) can significantly increase mortality in septic shock patients despite treating the underlying infection. Prompt recognition and aggressive intervention can potentially support the recovery of cellular injuries caused by inflammation and improve outcomes. Recently, venoarterial-extracorporeal membrane oxygenation (VA-ECMO) has gained attention as a potential mechanical support for managing these patients with SIC and cardiogenic shock. However, the lack of consensus in defining SIC and the variation in inclusion criteria for VA-ECMO implementation in acute sepsis-induced cardiogenic shock made it challenging to interpret the results of related studies. A more specific definition of SIC and careful selection of patients who may benefit from ECMO support could improve outcomes in this population. This analytic review discusses the mechanisms leading to cardiomyocyte injury and SIC, and summarizes recent studies including the efficacy of VA-ECMO in managing refractory septic shock due to SIC.
Collapse
Affiliation(s)
- AnhTuan Mai
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, DMC Sinai-Grace Hospital, Detroit, Michigan, USA
| | - Yusuf Yasarlar
- Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mohammed Awad
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yusuke Terasaki
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Michael & Marian Ilitch Department of Surgery, Division of Cardiothoracic Surgery, Detroit, Michigan, USA
| | - Kenton Zehr
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Michael & Marian Ilitch Department of Surgery, Division of Cardiothoracic Surgery, Detroit, Michigan, USA
| | - Ayman O Soubani
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Zahia Esber
- Wayne State University School of Medicine, Detroit, Michigan, USA
- Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
242
|
Xiong Y, Wang J, Shang X, Chen T, Fraser DD, Fonseca GJ, Rousseau S, Ding J. Efficient and scalable construction of clinical variable networks for complex diseases with RAMEN. CELL REPORTS METHODS 2025; 5:101022. [PMID: 40215965 DOI: 10.1016/j.crmeth.2025.101022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 12/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Understanding the interplay among clinical variables-such as demographics, symptoms, and laboratory results-and their relationships with disease outcomes is critical for advancing diagnostics and understanding mechanisms in complex diseases. Existing methods fail to capture indirect or directional relationships, while existing Bayesian network learning methods are computationally expensive and only infer general associations without focusing on disease outcomes. Here we introduce random walk- and genetic algorithm-based network inference (RAMEN), a method for Bayesian network inference that uses absorbing random walks to prioritize outcome-relevant variables and a genetic algorithm for efficient network refinement. Applied to COVID-19 (Biobanque québécoise de la COVID-19), intensive care unit (ICU) septicemia (MIMIC-III), and COPD (CanCOLD) datasets, RAMEN reconstructs networks linking clinical markers to disease outcomes, such as elevated lactate levels in ICU patients. RAMEN demonstrates advantages in computational efficiency and scalability compared to existing methods. By modeling outcome-specific relationships, RAMEN provides a robust tool for uncovering critical disease mechanisms, advancing diagnostics, and enabling personalized treatment strategies.
Collapse
Affiliation(s)
- Yiwei Xiong
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Jingtao Wang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Xiaoxiao Shang
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Mathematics and Statistics, McGill University, 805 Sherbrooke Street West, Montreal, QC H3A 0B9, Canada
| | - Tingting Chen
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Hematology Department, Beijing Luhe Hospital, Capital Medical University, Xinhua South Road No. 82, Tongzhou District, Beijing 101149, China
| | - Douglas D Fraser
- Children's Health Research Institute, Victoria Research Laboratories, 800 Commissioners Road East, London, ON N6C 2V5, Canada; Lawson Health Research Institute, London, ON N6C 2R5, Canada; Department of Pediatrics, Western University, London, ON N6A 5C1, Canada; Department of Physiology & Pharmacology, Western University, London, ON N6A 5C1, Canada; Department of Clinical Neurological Sciences, Western University, London, ON N6A 5C1, Canada
| | - Gregory J Fonseca
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| | - Simon Rousseau
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| | - Jun Ding
- Meakins-Christe Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Medicine, Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; School of Computer Science, McGill University, 3480 Rue University, Montreal, QC H3A 2A7, Canada; Mila-Quebec AI Institute, 6666 Rue Saint-Urbain, Montreal, QC H2S 3H1, Canada.
| |
Collapse
|
243
|
Lozano-López DA, Hernández-Ortega LD, González-Mariscal L, Díaz-Coránguez M, Pinto-Dueñas DC, Castañeda-Arellano R. Preserving Blood-Brain Barrier Integrity in Ischemic Stroke: a Review on MSCs-sEVs Content and Potential Molecular Targets. Mol Neurobiol 2025:10.1007/s12035-025-04956-9. [PMID: 40259172 DOI: 10.1007/s12035-025-04956-9] [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: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/23/2025]
Abstract
Ischemic stroke (IS) is a life-threatening condition that constitutes the second leading cause of death globally. Despite its high impact on public health, there is a shortage of treatments due to the complexity of the cellular and molecular mechanisms implicated. One main limiting factor for successful IS therapeutic intervention is stroke-induced blood-brain barrier (BBB) damage, particularly over tight junction proteins (TJs). BBB disruption is a well-established feature of IS, accelerating ischemic tissue damage and worsening prognosis. In recent years, mesenchymal stem cells (MSCs) and their small extracellular vesicles (MSCs-sEVs) have emerged as promising therapeutic interventions for several neurological disorders, including IS. However, its effects on BBB repair after IS are not completely understood. In this review, we will discuss novel experimental evidence of MSCs-sEVs effects in BBB protection and highlight the relevance of molecules reported in MSCs-sEVs, their potential cellular and molecular targets, and putative mechanisms implicated in BBB repair, providing a promising research avenue that may translate into effective therapeutic strategies for IS.
Collapse
Affiliation(s)
- David Arturo Lozano-López
- Pharmacology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México
| | - Luis Daniel Hernández-Ortega
- Molecular Biology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México
| | - Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Mónica Díaz-Coránguez
- Department of Pharmacobiology, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Diana Cristina Pinto-Dueñas
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies of the National Polytechnic Institute (Cinvestav-IPN), Mexico City, México
| | - Rolando Castañeda-Arellano
- Pharmacology Laboratory, Center for Multidisciplinary Health Research, University Center of Tonalá, University of Guadalajara, Guadalajara, México.
| |
Collapse
|
244
|
Lim WL, Hong MGY, Iijima Y, Yan Y, Zhao X, Qian J, Ge J. Navigating Diagnosis and Management for Takotsubo Syndrome Following Esophageal Cancer Surgery. Am J Cardiol 2025; 249:5-8. [PMID: 40258458 DOI: 10.1016/j.amjcard.2025.04.015] [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: 02/17/2025] [Revised: 03/25/2025] [Accepted: 04/13/2025] [Indexed: 04/23/2025]
Abstract
Takotsubo syndrome (TTS), despite its low incidence, often remains underrecognized or misdiagnosed due to its clinical similarity to acute coronary syndromes and atypical cardiac manifestations in critically ill patients. This applied clinical evidence presents a case of a 72-year-old male patient who developed TTS following esophagectomy. The discussion focuses on the most appropriate diagnostic approach for critically ill patients presenting with chest pain in the intensive care unit (ICU). In addition, it explores the optimal management strategy for TTS in the context of rapid atrial fibrillation and pulmonary complications, emphasizing the importance of timely recognition and targeted intervention.
Collapse
Affiliation(s)
- Woon Ling Lim
- Shanghai Medical College, Fudan University, Shanghai, China; Department of Internal Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Madeleine Goh Yee Hong
- Shanghai Medical College, Fudan University, Shanghai, China; Department of Internal Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuka Iijima
- Shanghai Medical College, Fudan University, Shanghai, China; Department of Internal Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Yan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xin Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China; National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; NHC Key Laboratory of Ischemic Heart Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
245
|
Nguyen KH, Wasielewski ML, Yalavarthi S, Qu X, Knight JS, Takayama S. A Mimetic Assay of Neutrophil Extracellular Trap Degradation Using YOYO-1-Stained DNA-Histone Surface Webs. Cells 2025; 14:615. [PMID: 40277940 PMCID: PMC12025948 DOI: 10.3390/cells14080615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/06/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025] Open
Abstract
Neutrophil extracellular traps (NETs) are not only promising biomarkers of disease, but also potential therapeutic targets. Overproduction or the improper clearance of NETs has been linked to disease severity. In vitro NET degradation assays can reveal mechanisms and degradation efficiency differences in diseased serum samples. There is a need for more convenient assays to increase the speed of NET degradation studies. This paper describes a simplified, lower variability mimetic assay with DNA-histone structures, referred to as surface webs, that performs functionally similarly to traditional NET degradation assays with increased scalability, ease of use, shorter preparation time, and lowered costs. The surface webs are created and dehydrated in a 96-well microplate that is shelf-stable, transportable, and viable for 30 days of storage at room temperature. The surface webs, compared to NETs, have similar shapes and distribution but lower intraplate variability while degrading with healthy serum and DNase I within the same timeframe. The assay can identify patient serum with reduced degradation capabilities. This assay opens new opportunities for NET-targeted drug discovery and studies on the role of NETs as modulators of disease.
Collapse
Affiliation(s)
- Katherine H. Nguyen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (K.H.N.); (M.L.W.)
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Midori L. Wasielewski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (K.H.N.); (M.L.W.)
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Xianggui Qu
- Department of Mathematics and Statistics, Oakland University, Rochester, MI 48309, USA;
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (K.H.N.); (M.L.W.)
- The Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
246
|
Li M, Yang L, Wang Y, Zhang L. Comprehensive analysis of diagnostic biomarkers related to histone acetylation in acute myocardial infarction. BMC Med Genomics 2025; 18:75. [PMID: 40251588 DOI: 10.1186/s12920-025-02135-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/27/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) has become a serious disease that endangers human health, with high morbidity and mortality. Numerous studies have reported histone acetylation can result in the occurrence of cardiovascular diseases. This article aims to explore the potential biomarkers of histone acetylation regulatory genes (ARGs) in AMI patients. METHODS Five AMI datasets were downloaded from the Gene Expression Omnibus (GEO) database. Next, ARG-related genes were gathered by gene set variation analysis (GSVA) and Spearman's correlation analysis. Subsequently, weighted gene co-expression network analysis (WGCNA) was performed to identify the module genes related to histone acetylation regulation. In the GSE60993 and GSE48060 datasets, the common differentially expressed genes (DEGs) between AMI and control samples were screened. Importantly, the intersecting genes were obtained by overlapping ARGs-related genes, common DEGs, and module genes. Then, the biomarkers in AMI were determined by machine learning, receiver operating characteristic (ROC) curves, and quantitative PCR (qPCR). In addition, immune analysis, drug prediction, molecular docking, and the lncRNA-miRNA-mRNA regulatory network targeting the biomarkers were analyzed, respectively. RESULTS Here, a total of 18 intersecting genes were identified by overlapping 7,349 ARGs-related genes, 5,565 module genes, and 25 common DEGs. Further, five biomarkers (AQP9, HLA-DQA1, MCEMP1, NKG7, and S100A12) were obtained, and a nomogram was constructed and verified based on these biomarkers. Notably, the biomarkers were significantly associated with CD8 T cells and neutrophils. In addition, the drugs related to biomarkers were predicted, and ATOGEPANT with the molecular target (S100A12) had a high binding affinity (docking score = -10 kcal/mol). CONCLUSION AQP9, HLA-DQA1, MCEMP1, NKG7, and S100A12 were identified as biomarkers related to ARGs in AMI, which provides a new perspective to study the relationship between ARGs and AMI.
Collapse
Affiliation(s)
- Man Li
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China
- Tongji Shanxi Hospital, Taiyuan, China
- Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lifeng Yang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China
- Tongji Shanxi Hospital, Taiyuan, China
- Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Wang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China
- Tongji Shanxi Hospital, Taiyuan, China
- Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Zhang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China.
- Tongji Shanxi Hospital, Taiyuan, China.
- Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
247
|
Putthanbut N, Lee JY, Borlongan CV. Commentary on: Lipoprotein(a), remote ischemic conditioning, and stroke recurrence in patients with symptomatic intracranial atherosclerotic stenosis. Neurotherapeutics 2025:e00590. [PMID: 40253243 DOI: 10.1016/j.neurot.2025.e00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025] Open
Affiliation(s)
- Napasiri Putthanbut
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, USA; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jea-Young Lee
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, USA
| | - Cesario V Borlongan
- Center of Aging and Brain Repair, Department of Neurosurgery, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
248
|
Tan D, Yang X, Yang J, Fan G, Xiong G. PCSK9 in Vascular Aging and Age-Related Diseases. Aging Dis 2025:AD.2024.1713. [PMID: 40354375 DOI: 10.14336/ad.2024.1713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/27/2025] [Indexed: 05/14/2025] Open
Abstract
The aging process significantly contributes to human disease, and as worldwide life expectancy increases, addressing the challenges of aging and age-related cardiovascular diseases is becoming increasingly urgent. Vascular aging is a key link between aging and the development of age-related diseases. Recent studies indicate that proprotein convertase subtilisin/kexin type 9 (PCSK9), a type of protein involved in the metabolism of lipids, is crucial in modulating vascular aging by affecting the physiological functioning of vascular cells. PCSK9 is linked to lipid metabolism and chronic inflammation and is involved in regulating senescence-related activities, including migration, proliferation, apoptosis, and differentiation. These factors contribute to the aging of vascular cells and age-related vascular diseases, including atherosclerosis, hypertension, coronary artery disease, and cerebrovascular diseases. Given its involvement in these processes, this article provides a comprehensive summary of PCSK9's regulatory functions in vascular aging, highlighting potential therapeutic targets for combating age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Dong Tan
- Department of Vascular Surgery, the Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
- Pan-Vascular Research Group, Shenzhen University Affiliated Sixth Hospital, Shenzhen, Guangdong, China
| | - Xin Yang
- Pan-Vascular Research Group, Shenzhen University Affiliated Sixth Hospital, Shenzhen, Guangdong, China
- Department of Metabolism and Endocrinology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- Pan-Vascular Research Group, Shenzhen University Affiliated Sixth Hospital, Shenzhen, Guangdong, China
- Department of Metabolism and Endocrinology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Gang Fan
- Pan-Vascular Research Group, Shenzhen University Affiliated Sixth Hospital, Shenzhen, Guangdong, China
- Department of Urology, Shenzhen University Affiliated Sixth Hospital, Shenzhen, Guangdong Province, China
| | - Guozuo Xiong
- Department of Vascular Surgery, the Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
- Hunan Province Thrombotic Disease Prevention and Treatment Clinical Medical Research Center, The Third Affiliated Hospital of University of South China, Hengyang, Hunan, China
- Hunan Province Thrombotic Disease Prevention and Treatment Clinical Medical Research Center, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, China
| |
Collapse
|
249
|
Hu F, Guo R, Zhi Y, Hu H, Tang T, Wang P, Xue L. Exploring the key target molecules of angiogenesis in diabetic cardiomyopathy based on bioinformatics analysis. Front Endocrinol (Lausanne) 2025; 16:1561142. [PMID: 40313483 PMCID: PMC12043471 DOI: 10.3389/fendo.2025.1561142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Backgrounds Diabetic cardiomyopathy has a very high incidence and serious clinical consequences, making it an urgent clinical problem to be solved. Angiogenesis is a significant phenotype in the occurrence and development of diabetic cardiomyopathy, especially the damage to angiogenesis of cardiac microvessels, which is inextricably linked to the cardiac risk of diabetic patients. In the current basic and clinical research, there is still a lack of treatment methods that directly target the angiogenesis of diabetic cardiomyopathy. This study hopes to discover the key molecules related to diabetic cardiomyopathy and angiogenesis damage, to provide ideas for possible interventions. Methods Sequencing data of animals and cells were obtained from the GEO database, and differentially expressed genes were analyzed. Subsequently, the angiogenesis-related genes were clustered for functional and pathway analysis. Then, the microangiogenesis of the diabetic mice and the angiogenesis changes of high glucose-stimulated HUVECs were verified, and the top three genes related to diabetic cardiomyopathy and angiogenesis were verified using western blot. Results 24 differentially expressed genes associated with angiogenesis were found in GSE241565(human) and GSE215979(mice). Among them, 11 genes showed the same trend in the two databases. Then CD31 staining of diabetic mice hearts showed that microvascular angiogenesis was impaired, high glucose-stimulated HUVECs decreased tube formation, and wound healing migration was weakened. Finally, the top 3 genes most associated with diabetic cardiomyopathy were verified, and there was no significant difference between the changes of Edn1 and Lepr. At the same time, Efnb2 was significantly increased under high glucose stimulation. Conclusion Combined with the sequencing data of animal and cell models of diabetic cardiomyopathy, the differential genes associated with angiogenesis were screened. These findings not only elucidate a novel molecular axis linking angiogenesis damage to diabetic cardiomyopathy but also highlight Efnb2 as a potential therapeutic target.
Collapse
Affiliation(s)
- Fengli Hu
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ruixue Guo
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Yaxin Zhi
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Haijuan Hu
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ting Tang
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Pengfei Wang
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ling Xue
- Department of Cardiology, Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
250
|
Sun SW, Wang XJ, Yan RC, Huang L, Hou M. Oxidative Stress-related Gene Signature: A Prognostic Tool for Predicting Survival in ST-elevation MI. Eur Cardiol 2025; 20:e11. [PMID: 40343143 PMCID: PMC12060176 DOI: 10.15420/ecr.2024.58] [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: 12/07/2024] [Accepted: 01/26/2025] [Indexed: 05/11/2025] Open
Abstract
Background This study aimed to identify differentially expressed oxidative stress-related genes (DEOSRGs) in ST-elevation MI (STEMI) patients and examine their connection to clinical outcomes. Methods We conducted a systematic review of Gene Expression Omnibus datasets, selecting GSE49925, GSE60993 and GSE61144 for analysis. DEOSRGs were identified using GEO2R2, overlapping across the selected datasets. Functional enrichment analysis was performed to understand the biological roles of the DEOSRGs. An optimal model was constructed using Least Absolute Shrinkage and Selection Operator penalised Cox proportional hazards regression. The clinical utility of the signature was assessed through survival analysis, receiver operating characteristic (ROC) curve and decision curve analysis. A prognostic nomogram was developed to predict survival risk, with the signature being externally validated using our own plasma samples. Results A prognostic signature was formulated, incorporating three upregulated DEOSRGs (matrix metalloproteinase-9, arginase 1, interleukin 18 receptor accessory protein) and three clinical variables (age, serum creatinine level, Gensini score). This signature successfully stratified patients into low- and high-risk groups. Survival analysis, ROC curve analysis and decision curve analysis demonstrated the signature's robust predictive performance and clinical utility within 2 years post-disease onset. External validation confirmed significant outcome differences between the risk groups. Conclusion This study identified DEOSRGs in STEMI patients and developed a prognostic signature integrating gene expression levels and clinical variables. While the signature showed promising predictive performance and clinical utility, the findings should be interpreted considering the limitations of small sample size and control group selection.
Collapse
Affiliation(s)
- Shuo-wen Sun
- Department of Laboratory Medicine, Tianjin Chest HospitalTianjin, China
| | - Xing-jie Wang
- Department of Laboratory Medicine, Tianjin Chest HospitalTianjin, China
| | - Ruo-chen Yan
- Department of Laboratory Medicine, Tianjin Chest HospitalTianjin, China
| | - Lei Huang
- Heart Center, Tianjin Third Central HospitalTianjin, China
| | - Min Hou
- Department of Laboratory Medicine, Tianjin Chest HospitalTianjin, China
| |
Collapse
|