1
|
Xu W, Wang Z, Yao H, Zeng Z, Lan X. Distribution of Arteriosclerotic Vessels in Patients with Arteriosclerosis and the Differences of Serum Lipid Levels Classified by Different Sites. Int J Gen Med 2024; 17:4733-4744. [PMID: 39429964 PMCID: PMC11491091 DOI: 10.2147/ijgm.s483324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024] Open
Abstract
Objective To investigate the distribution of arteriosclerotic vessels of arteriosclerosis, differential serum lipid profiles, and differences in the proportion of dyslipidaemia between patients with single-site arteriosclerosis and multi-site arteriosclerosis (significant hardening of ≥2 arteries). Methods The data of 6581 single-site arteriosclerosis patients and 5940 multi-site arteriosclerosis patients were extracted from the hospital medical record system. Serum total cholesterol (TC), triglycerides (TGs), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), apolipoprotein (Apo) A1, ApoB concentrations and C-reactive protein (CRP) between patients with single-site arteriosclerosis and multi-site arteriosclerosis were collected and analyzed. Results The most diseased arteries were coronary arteries (n=7099, 33.7%), limb arteries (n=6546, 31.1%), and carotid arteries (n=5279, 25.1%). TC, LDL-C, TC/HDL-C, and LDL-C/HDL-C levels were higher and CRP level was lower in multi-site arteriosclerosis patients than those in single-site arteriosclerosis patients. The TC, LDL-C levels in non-elderly (<65 years old) female patients were higher and TG/HDL-C, TC/HDL-C, LDL-C/HDL-C levels were lower than those in non-elderly male patients, while the TG, TC, LDL-C, and TG/HDL-C levels in elderly (≥65 years old) female patients were higher and LDL-C/HDL-C level was lower than those in elderly male patients. The proportion of dyslipidemia in descending order was as follows: low HDL-C (31.9%), elevated TG (16.9%), elevated TC (9.0%), and elevated LDL-C (4.2%). The levels of TC, LDL-C, TC/HDL-C, and LDL-C/HDL-C in patients with peripheral arteriosclerosis were higher than those in patients with cardio-cerebrovascular arteriosclerosis. Conclusion There were differences in serum lipid levels in patients with arteriosclerosis with different age, gender and distribution of arteriosclerotic vessels.
Collapse
Affiliation(s)
- Weiyong Xu
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Zhenchang Wang
- Department of Emergency Medicine, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Huaqing Yao
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Zifeng Zeng
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| | - Xinping Lan
- Center for Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
- Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou People’s Hospital, Meizhou, People’s Republic of China
| |
Collapse
|
2
|
Huo L, Zhao W, Ji X, Chen K, Liu T. The Combination Effect of the Red Blood Cell Distribution Width and Prognostic Nutrition Index on the Prognosis in Patients Undergoing PCI. Nutrients 2024; 16:3176. [PMID: 39339776 PMCID: PMC11434894 DOI: 10.3390/nu16183176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Inflammation and malnutrition are related to adverse clinical outcomes in patients with coronary artery disease (CAD). However, it is unclear whether there is a relationship between the PNI (prognostic nutritional index) and RDW (red blood cell distribution width) regarding the impact on the prognosis in patients with CAD undergoing percutaneous coronary intervention (PCI). METHODS A total of 5605 consecutive CAD patients undergoing PCI were selected retrospectively. The patients were stratified into four groups according to the PNI [high PNI (H-PNI) and low PNI (L-PNI)] and RDW [high RDW (H-RDW) and low RDW (L-RDW)]. The cutoff values of RDW and PNI were calculated using receiver-operating characteristic curve analysis. The primary endpoint was 1-year all-cause mortality (ACM). The secondary endpoint was major adverse cardiac cerebrovascular events (MACCEs), the composite of cardiac death (CD), the recurrence of MI, target lesion revascularization (TLR), and stroke. A Cox proportional hazards model was used to evaluate the association between the PNI, RDW, and clinical endpoints. RESULTS During 1-year follow-up, 235 (4.19%) patients died. In multivariate regression analysis, the L-PNI/H-RDW group was found to have the highest risk of 1-year ACM [hazard ratio (HR) = 8.85, 95% confidence interval (CI): 5.96-13.15, p = 0.020] with the H-PNI/L-RDW group as a reference, followed by the L-PNI/L-RDW (HR = 3.96, 95% CI: 2.60-6.00, p < 0.001) and H-RDW/H-PNI groups (HR = 3.00, 95% CI: 1.99-4.50, p < 0.001). Nomograms were developed to predict the probability of 1-year ACM and MACCEs. CONCLUSIONS CAD patients with L-PNI and H-RDW experienced the worst prognosis. The combination of PNI and RDW was a strong predictor of 1-year ACM. The coexistence of PNI and RDW appears to have a synergistic effect, providing further information for the risk stratification of CAD patients.
Collapse
Affiliation(s)
- Likun Huo
- Department of Emergency, Tianjin Huanhu Hospital, Tianjin 300222, China
- Tianjin Key Laboratory of Ions and Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wenjuan Zhao
- Department of Emergency, Tianjin Huanhu Hospital, Tianjin 300222, China
| | - Xiang Ji
- Department of Emergency, Tianjin Huanhu Hospital, Tianjin 300222, China
| | - Kangyin Chen
- Tianjin Key Laboratory of Ions and Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tong Liu
- Tianjin Key Laboratory of Ions and Molecular Function of Cardiovascular Diseases, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
3
|
Chen Z, Wang Z, Cui Y, Xie H, Yi L, Zhu Z, Ni J, Du R, Wang X, Zhu J, Ding F, Quan W, Zhang R, Wang Y, Yan X. Serum BAFF level is associated with the presence and severity of coronary artery disease and acute myocardial infarction. BMC Cardiovasc Disord 2024; 24:471. [PMID: 39227771 PMCID: PMC11370111 DOI: 10.1186/s12872-024-04146-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: 06/12/2023] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between circulating levels of B cell activating factor (BAFF) and the presence and severity of coronary artery disease (CAD) and acute myocardial infarction (AMI) in humans, as its biological functions in this context remain unclear. METHODS Serum BAFF levels were measured in a cohort of 723 patients undergoing angiography, including 204 patients without CAD (control group), 220 patients with stable CAD (CAD group), and 299 patients with AMI (AMI group). Logistic regression analyses were used to assess the association between BAFF and CAD or AMI. RESULTS Significantly elevated levels of BAFF were observed in patients with CAD and AMI compared to the control group. Furthermore, BAFF levels exhibited a positive correlation with the SYNTAX score (r = 0.3002, P < 0.0001) and the GRACE score (r = 0.5684, P < 0.0001). Logistic regression analysis demonstrated that increased BAFF levels were an independent risk factor for CAD (adjusted OR 1.305, 95% CI 1.078-1.580) and AMI (adjusted OR 2.874, 95% CI 1.708-4.838) after adjusting for confounding variables. Additionally, elevated BAFF levels were significantly associated with a high GRACE score (GRACE score 155 to 319, adjusted OR 4.297, 95% CI 1.841-10.030). BAFF exhibited a sensitivity of 75.0% and specificity of 71.4% in differentiating CAD patients with a high SYNTAX score, and a sensitivity of 75.5% and specificity of 72.8% in identifying AMI patients with a high GRACE score. CONCLUSION Circulating BAFF levels serve as a valuable diagnostic marker for CAD and AMI. Elevated BAFF levels are associated with the presence and severity of these conditions, suggesting its potential as a clinically relevant biomarker in cardiovascular disease.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ziyang Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yuke Cui
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hongyang Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Lei Yi
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Zhengbin Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Jingwei Ni
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Run Du
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Xiaoqun Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Jinzhou Zhu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Fenghua Ding
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Weiwei Quan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
| | - Yueying Wang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
- Institute of Cardiovascular Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
4
|
Ait-Oufella H, Libby P. Inflammation and Atherosclerosis: Prospects for Clinical Trials. Arterioscler Thromb Vasc Biol 2024; 44:1899-1905. [PMID: 39167675 PMCID: PMC11343092 DOI: 10.1161/atvbaha.124.320155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Affiliation(s)
- Hafid Ait-Oufella
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Sorbonne Université, Paris, France
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
5
|
Baragetti A, Grigore L, Olmastroni E, Mattavelli E, Catapano AL. Plasma proteins associate with carotid plaques and predict incident atherosclerotic cardiovascular events. Vascul Pharmacol 2024; 156:107394. [PMID: 38866119 DOI: 10.1016/j.vph.2024.107394] [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: 02/17/2024] [Revised: 05/10/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Performing non-invasive carotid imaging is challenging, owing inter-operator variability and organizational barriers, but plasma proteomics can offer an alternative. We sought plasma proteins that associate with the presence of carotid plaques, their number and predict the incidence of clinically overt atherosclerotic cardiovascular events (ASCVD) above currently recognized risk factors in "apparently healthy" subjects. METHODS We studied the plasma levels of 368 proteins in 664 subjects from the PLIC study, who underwent an ultrasound imaging screening of the carotids to check for the presence of plaques. We clustered, by artificial intelligence (A.I.), the proteins that associate with the presence, the number of plaques and that predict incident ASCVDs over 22 years (198 events were registered). FINDINGS 299/664 subjects had at least 1 carotid plaque (1+) (77 with only one plaque, 101 with 2 plaques, 121 with ≥3 plaques (3+)). The remaining 365 subjects with no plaques acted as controls. 106 proteins were associated with 1+ plaques, but 97 proteins significantly predicted 3+ plaques only (AUC = 0.683 (0.601-0.785), p < 0.001), when considered alone. A.I. underscored 87 proteins that improved the performance of the classical risk factors both in detecting 3+ plaques (AUC = 0.918 (0.887-0.943) versus risk factors alone, AUC = 0.760 (0.716-0.801), p < 0.001) and in predicting the incident ASCVD (AUC = 0.739 (0.704-0.773) vs risk factors alone AUC = 0.559 (0.521-0.598), p < 0.001). The chemotaxis/migration of leukocytes and interleukins/cytokines signaling were biological pathways mostly represented by these proteins. DISCUSSION AND CONCLUSIONS Plasma proteomics marks the number of carotid plaques and improve the prediction of incidence ASCVDs in apparently healthy subjects.
Collapse
Affiliation(s)
- Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | | | - Elena Olmastroni
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy
| | - Elisa Mattavelli
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy; Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy; IRCCS Multimedica Hospital, Milan, Italy
| |
Collapse
|
6
|
Tramujas L, Nogueira A, Felix N, de Barros E Silva PGM, Abizaid A, Cavalcanti AB. Association of colchicine use with cardiovascular and limb events in peripheral artery disease: Insights from a retrospective cohort study. Atherosclerosis 2024:118563. [PMID: 39299823 DOI: 10.1016/j.atherosclerosis.2024.118563] [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: 04/16/2024] [Revised: 07/06/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND AIMS Colchicine has demonstrated efficacy in treating coronary artery disease, but its efficacy in peripheral artery disease (PAD) remains uncertain. This study aims to address this gap in knowledge. METHODS A retrospective cohort study was conducted using the TriNetX Network, selecting patients with lower limb PAD between January 1, 2011, and January 1, 2024. Colchicine users were matched 1:1 with non-users through propensity score matching, considering demographics, medical conditions, medications, and psychosocial factors. The primary outcome was a composite of major adverse cardiovascular and limb events (MACLE) - including lower limb amputation, revascularization for lower limb ischemia, acute myocardial infarction, ischemic stroke, and all-cause mortality - over a ten-year follow-up. RESULTS From 53,568 colchicine-treated and 1,499,969 untreated patients with lower limb PAD, 52,350 pairs were successfully matched. Over ten years, colchicine was associated with a significant reduction in MACLE (hazard ratio, [HR] 0.90, 95% CI 0.88-0.92, p < 0.001), any lower limb amputation (HR 0.84, 95% CI 0.75-0.94, p = 0.002), revascularization for lower limb ischemia (HR 0.85, 95% CI 0.82-0.88, p < 0.001), major adverse cardiovascular events (HR 0.93, 95% CI 0.91-0.95, p < 0.001), and all-cause mortality (HR 0.90, 95% CI 0.87-0.92, p < 0.001). It also result in a reduced risk of ischemic stroke (HR 0.95, 95% CI 0.92-0.98, p = 0.001), but not of acute myocardial infarction (HR 0.98, 95% CI 0.95-1.01, p = 0.24). CONCLUSIONS Colchicine significantly reduced major adverse cardiovascular and limb events in patients with lower limb PAD, supporting the need for further investigation.
Collapse
Affiliation(s)
| | - Alleh Nogueira
- Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
| | - Nicole Felix
- Federal University of Campina Grande, Campina Grande, Brazil
| | | | - Alexandre Abizaid
- Hcor Research Institute, São Paulo, Brazil; Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
7
|
Wang Y, Yan K, Duan H, Tao N, Zhu S, Zhang Y, You Y, Zhang Z, Wang H, Hu S. High-fat-diet-induced obesity promotes simultaneous progression of lung cancer and atherosclerosis in apolipoprotein E-knockout mice. CANCER INNOVATION 2024; 3:e127. [PMID: 38948249 PMCID: PMC11212317 DOI: 10.1002/cai2.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/15/2023] [Accepted: 01/19/2024] [Indexed: 07/02/2024]
Abstract
Background Clinical studies have shown that atherosclerotic cardiovascular disease and cancer often co-exist in the same individual. The present study aimed to investigate the role of high-fat-diet (HFD)-induced obesity in the coexistence of the two diseases and the underlying mechanism in apolipoprotein E-knockout (ApoE-/-) mice. Methods Male ApoE-/- mice were fed with a HFD or a normal diet (ND) for 15 weeks. On the first day of Week 13, the mice were inoculated subcutaneously in the right axilla with Lewis lung cancer cells. At Weeks 12 and 15, serum lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) and vascular endothelial growth factor levels were measured by enzyme-linked immunosorbent assay, and blood monocytes and macrophages were measured by fluorescence-activated cell sorting. At Week 15, the volume and weight of the local subcutaneous lung cancer and metastatic lung cancer and the amount of aortic atherosclerosis were measured. Results At Week 15, compared with mice in the ND group, those in the HFD group had a larger volume of local subcutaneous cancer (p = 0.0004), heavier tumors (p = 0.0235), more metastatic cancer in the lungs (p < 0.0001), a larger area of lung involved in metastatic cancer (p = 0.0031), and larger areas of atherosclerosis in the aorta (p < 0.0001). At Week 12, serum LOX-1, serum vascular endothelial growth factor, and proportions of blood monocytes and macrophages were significantly higher in the HFD group than those in the ND group (p = 0.0002, p = 0.0029, p = 0.0480, and p = 0.0106, respectively); this trend persisted until Week 15 (p = 0.0014, p = 0.0012, p = 0.0001, and p = 0.0204). Conclusions In this study, HFD-induced obesity could simultaneously promote progression of lung cancer and atherosclerosis in the same mouse. HFD-induced upregulation of LOX-1 may play an important role in the simultaneous progression of these two conditions via the inflammatory response and VEGF.
Collapse
Affiliation(s)
- Yihao Wang
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Kaixin Yan
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Han Duan
- Beijing Institute of Radiation MedicineBeijingChina
| | - Ning Tao
- Beijing Institute of Radiation MedicineBeijingChina
| | - Shaoning Zhu
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Yuning Zhang
- Beijing Institute of Radiation MedicineBeijingChina
| | - Yonggang You
- Department of OrthopaedicsChinese PLA General HospitalBeijingChina
| | - Zhen Zhang
- Department of OrthopaedicsChinese PLA General HospitalBeijingChina
| | - Hua Wang
- Beijing Institute of Radiation MedicineBeijingChina
| | - Shunying Hu
- Department of CardiologyChinese PLA General HospitalBeijingChina
| |
Collapse
|
8
|
Sepehrinia M, Pourmontaseri H, Naghizadeh MM, Vahid F, Hebert JR, Homayounfar R, Alkamel A. The association between energy-adjusted dietary inflammatory index and 10-year cardiovascular risk: Fasa adult cohort study. Food Sci Nutr 2024; 12:5530-5537. [PMID: 39139971 PMCID: PMC11317652 DOI: 10.1002/fsn3.4181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/27/2024] [Accepted: 04/09/2024] [Indexed: 08/15/2024] Open
Abstract
A healthy diet is dominant in cardiovascular disease (CVD) prevention. Inflammation is pivotal for CVD development. This study aimed to evaluate the association between the pro-inflammatory diet and the CVD risk. This cross-sectional study involved 10,138 Fasa adult cohort study participants. After excluding participants with missing data, the Energy-Adjusted Dietary Inflammatory Index (E-DII) was calculated to assess the inflammatory potential of diet using the recorded Food Frequency Questionnaire. Framingham risk score (FRS) was used to predict the 10-year risk of CVD. The association between E-DII and high risk for CVD was investigated using multinominal regression. After exclusion, the mean age of studied individuals (n = 10,030) was 48.6 ± 9.6 years, including 4522 men. Most participants were low risk (FRS <10%) for CVD (87.6%), while 2.7% of them were high risk (FRS ≥20%). The median FRS was 2.80 (1.70, 6.30). The E-DII ranged from -4.22 to 4.49 (mean E-DII = 0.880 ± 1.127). E-DII was significantly associated with FRS. This result persisted after adjusting for confounding factors and in both genders. This study revealed that the pro-inflammatory diet significantly increases the CVD risk. Consequently, reducing the inflammatory potential of diet should be considered an effective dietary intervention in CVD prevention.
Collapse
Affiliation(s)
- Matin Sepehrinia
- Student Research CommitteeFasa University of Medical SciencesFasaIran
- Non‐Communicable Diseases Research CenterFasa University of Medical SciencesFasaIran
| | | | | | - Farhad Vahid
- Department of Precision Health, Nutrition and Health Research GroupLuxembourg Institute of HealthStrassenLuxembourg
| | - James R. Hebert
- Department of Epidemiology and Biostatistics, Arnold School of Public HealthUniversity of South CarolinaColumbiaSouth CarolinaUSA
- South Carolina Statewide Cancer Prevention and Control ProgramUniversity of South CarolinaColumbiaSouth CarolinaUSA
| | - Reza Homayounfar
- National Nutrition and Food Technology Research Institute (WHO Collaborating Center), Faculty of Nutrition Sciences and Food TechnologyShahid Beheshti University of Medical SciencesTehranIran
| | - Abdulhakim Alkamel
- Non‐Communicable Diseases Research CenterFasa University of Medical SciencesFasaIran
- Department of Cardiovascular Disease, Faculty of MedicineFasa University of Medical SciencesFasaIran
| |
Collapse
|
9
|
Guo YS, Yang N, Wang Z, Wei YM. Research Progress on the Pathogenesis and Treatment of Neoatherosclerosis. Curr Med Sci 2024; 44:680-685. [PMID: 39096479 DOI: 10.1007/s11596-024-2915-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/19/2024] [Indexed: 08/05/2024]
Abstract
Neoatherosclerosis (NA) within stents has become an important clinical problem after coronary artery stent implantation. In-stent restenosis and in-stent thrombosis are the two major complications following coronary stent placement and seriously affect patient prognosis. As the common pathological basis of these two complications, NA plaques, unlike native atherosclerotic plaques, often grow around residual oxidized lipids and stent struts. The main components are foam cells formed by vascular smooth muscle cells (VSMCs) engulfing oxidized lipids at lipid residue sites. Current research mainly focuses on optical coherence tomography (OCT) and intravascular ultrasound (IVUS), but the specific pathogenesis of NA is still unclear. A thorough understanding of the pathogenesis and pathological features of NA provides a theoretical basis for clinical treatment. This article reviews the previous research of our research group and the current situation of domestic and foreign research.
Collapse
MESH Headings
- Humans
- Tomography, Optical Coherence
- Coronary Restenosis/etiology
- Coronary Restenosis/diagnostic imaging
- Coronary Restenosis/therapy
- Coronary Restenosis/pathology
- Atherosclerosis/therapy
- Atherosclerosis/diagnostic imaging
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/therapy
- Plaque, Atherosclerotic/diagnostic imaging
- Stents/adverse effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Ultrasonography, Interventional/methods
- Coronary Artery Disease/therapy
- Coronary Artery Disease/diagnostic imaging
- Coronary Artery Disease/etiology
- Coronary Artery Disease/pathology
- Foam Cells/pathology
- Foam Cells/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
Collapse
Affiliation(s)
- Yi-Shan Guo
- Department of Cardiology, Hubei Key Laboratory of Biological Targeted Therapy, Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, 256600, China
| | - Ning Yang
- Department of Gynecology, Binzhou Medical University Hospital, Binzhou, 256600, China
| | - Zhen Wang
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, 256600, China.
| | - Yu-Miao Wei
- Department of Cardiology, Hubei Key Laboratory of Biological Targeted Therapy, Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
10
|
Zhao G, Tang W, Yang C, Liu X, Huang J. The Prognostic Value of Advanced Lung Cancer Inflammation Index in Elderly Patients with Acute Coronary Syndrome Undergoing Percutaneous Coronary Intervention. Int Heart J 2024; 65:621-629. [PMID: 39010222 DOI: 10.1536/ihj.24-046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
This study aimed to investigate the predictive value of advanced lung cancer inflammation index (ALI) for major adverse cardiovascular events (MACEs) in elderly patients with acute coronary syndrome (ACS).A total of 586 ACS patients undergoing percutaneous coronary intervention (PCI) over 65 years old between January 2017 and December 2018 were retrospectively collected. The patients were divided into two groups by the optimal cutoff value of ALI. Spearman rank correlation coefficient was used to evaluate the correlation between ALI and the Global Registry of Acute Coronary Events (GRACE). Time-dependent receiver operating characteristic (ROC) curves, Cox survival analysis, and Kaplan Meier curves were used to assess the predictive value of ALI for MACEs.Spearman's nonparametric test revealed a moderate correlation between ALI and the GRACE (r: -0.417, P < 0.001). Time-dependent ROC curves showed that the area under the curve for ALI was 0.751 (95% CI, 0.699-0.798) in predicting MACEs, higher than Geriatric Nutritional Risk Index (0.531, 95% CI 0.435-0.627) and Prognostic Nutritional Index (0.590, 95% CI 0.505-0.676), and for combined diagnostic models (ALI + GRACE) was 0.913, (95% CI 0.875 - 0.942, P < 0.001). Multivariate Cox analysis demonstrated that ALI (HR: 0.974, 95% CI: 0.952-0.996, P = 0.017) was an independent risk factor for MACEs. Kaplan Meier survival analysis showed that the cumulative incidence of MACEs was significantly higher in elderly ACS patients with lower ALI (log-rank test, P < 0.001).ALI could be a nutrition-inflammation indicator with independent predictive value for long-term MACEs of elderly ACS patients after PCI.
Collapse
Affiliation(s)
- Guoying Zhao
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University
| | - Wenbin Tang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University
| | - Chao Yang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University
| | - Xiao Liu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University
| | - Jinyu Huang
- Department of Cardiology, Hangzhou First People's Hospital
| |
Collapse
|
11
|
Xue Q, Ma Y, Shao H. Bacillus amyloliquefaciens Protect Against Atherosclerosis Through Alleviating Foam Cell Formation and Macrophage Polarization. Curr Microbiol 2024; 81:263. [PMID: 38997545 DOI: 10.1007/s00284-024-03775-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/01/2023] [Indexed: 07/14/2024]
Abstract
This study was to investigate the therapeutic effect of Bacillus amyloliquefaciens (Ba) on atherosclerosis (AS). THP-1 monocyte was differentiated to THP-1 macrophage (THP-M) through phorbol 12-myristate 13-acetate. After pre-treatment by 108 cfu/ml Ba lasting 6 h, THP-M was induced with 100 mg/l ox-LDL lasting 48 h to form macrophage foam cell (THP-F). RT-qPCR and flow cytometry were employed to determine the polarization of THP-M and THP-F. ApoE-/- mice with high-fat and high-cholesterol diet were used for constructing an AS model to evaluate the effect of Ba on AS. Our in vitro results showed that Ba vegetative cells pre-treatment distinctly inhibited the levels of iNOS and CD16/CD32 (M1 macrophage markers), and increased the levels of FIZZ1, Ym1, Arg1, CD163, and CD206 (M2 macrophage markers), indicating that Ba pre-treatment promoted anti-inflammatory M2-like polarization both in THP-M and THP-F. Meanwhile, it also suppressed cholesterol uptake, esterification, and hydrolysis, and efflux by THP-M and THP-F. Additionally, our animal experiments demonstrated that Ba vegetative cells treatment suppressed high cholesterol, hyperglycemia, hyperlipidemia, and the release of inflammatory factors (TNF-α, IL-6 and IL-1β) in ApoE-/- AS mice. In a word, our results indicated that Ba may protect against AS through alleviating foam cell formation and macrophage polarization through targeting certain stages of AS.
Collapse
Affiliation(s)
- Qi Xue
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Yuan Ma
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China
| | - Hong Shao
- Department of Cardiology, Zhejiang Provincial People's Hospital, No. 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
12
|
Jeong Y, Lee BJ, Hur W, Lee M, Han SH. Associations of Insulin Resistance and High-Sensitivity C-Reactive Protein with Metabolic Abnormalities in Korean Patients with Type 2 Diabetes Mellitus: A Preliminary Study. Metabolites 2024; 14:371. [PMID: 39057694 PMCID: PMC11279201 DOI: 10.3390/metabo14070371] [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: 04/10/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
We conducted this single-center, retrospective, cohort study to examine whether insulin resistance (IR) and high-sensitivity C-reactive protein (hsCRP) have a relationship with metabolic abnormalities in patients with type 2 diabetes mellitus (T2DM). In a total of 3758 patients (n = 3758) with T2DM, we analyzed medical records and thereby evaluated their baseline characteristics such as age, sex, duration of T2DM, systolic blood pressure (SBP), diastolic blood pressure (DBP), waist circumference, body mass index (BMI), visceral fat thickness (VFT), fasting plasma insulin levels, C-peptide levels, glycated hemoglobin (HbA1c), fasting plasma glucose (FPG), postprandial plasma glucose (PPG), homeostatic model assessment of insulin resistance (HOMA-IR), homeostatic model assessment of β-cell function (HOMA-β), aspartate aminotransferase (AST), alanine aminotransferase (ALT), total cholesterol (TC), triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), albuminuria, intima-media thickness (IMT) and hsCRP. The patients were stratified according to the tertile of the K index of the insulin tolerance test (KITT) or hsCRP. Thus, they were divided into the lowest (≥2.37), middle (1.54-2.36) and highest tertile (0-1.53) of KITT and the lowest (0.00-0.49), middle (0.50-1.21) and highest tertile (≥1.22) of hsCRP. Moreover, associations of KITT and hsCRP with metabolic abnormalities, such as steatotic liver disease (SLD), metabolic syndrome (MetS), albuminuria, diabetic retinopathy and carotid atherosclerosis, were also analyzed. There was a significant positive correlation between the prevalence of SLD, MetS, albuminuria and diabetic retinopathy and KITT (p < 0.001). Moreover, there was a significant positive association between the prevalence of SLD, MetS and albuminuria and hsCRP (p < 0.001). In conclusion, our results indicate that clinicians should consider the relationships of IR and hsCRP with metabolic abnormalities in the management of patients with T2DM. However, further large-scale, prospective, multi-center studies are warranted to confirm our results.
Collapse
Affiliation(s)
- Yuchul Jeong
- Department of Internal Medicine, Chungna Good Hospital, Incheon 22738, Republic of Korea
| | - Beom Jun Lee
- St. Mary’s Best ENT Clinic, Seoul 08849, Republic of Korea
| | - Wonjai Hur
- Department of Internal Medicine, Sejong General Hospital, Bucheon 14754, Republic of Korea
| | - Minjoon Lee
- Department of Internal Medicine, BS General Hospital, Incheon 23037, Republic of Korea
| | - Se-Hyeon Han
- Department of Companion Animal Industry, College of Health Science, Honam University, Gwangju 62399, Republic of Korea
| |
Collapse
|
13
|
Bulnes JF, González L, Velásquez L, Orellana MP, Venturelli PM, Martínez G. Role of inflammation and evidence for the use of colchicine in patients with acute coronary syndrome. Front Cardiovasc Med 2024; 11:1356023. [PMID: 38993522 PMCID: PMC11236697 DOI: 10.3389/fcvm.2024.1356023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/29/2024] [Indexed: 07/13/2024] Open
Abstract
Acute Coronary Syndrome (ACS) significantly contributes to cardiovascular death worldwide. ACS may arise from the disruption of an atherosclerotic plaque, ultimately leading to acute ischemia and myocardial infarction. In the pathogenesis of atherosclerosis, inflammation assumes a pivotal role, not solely in the initiation and complications of atherosclerotic plaque formation, but also in the myocardial response to ischemic insult. Acute inflammatory processes, coupled with time to reperfusion, orchestrate ischemic and reperfusion injuries, dictating infarct magnitude and acute left ventricular (LV) remodeling. Conversely, chronic inflammation, alongside neurohumoral activation, governs persistent LV remodeling. The interplay between chronic LV remodeling and recurrent ischemic episodes delineates the progression of the disease toward heart failure and cardiovascular death. Colchicine exerts anti-inflammatory properties affecting both the myocardium and atherosclerotic plaque by modulating the activity of monocyte/macrophages, neutrophils, and platelets. This modulation can potentially result in a more favorable LV remodeling and forestalls the recurrence of ACS. This narrative review aims to delineate the role of inflammation across the different phases of ACS pathophysiology and describe the mechanistic underpinnings of colchicine, exploring its purported role in modulating each of these stages.
Collapse
Affiliation(s)
- Juan Francisco Bulnes
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leticia González
- Centro de Imágenes Biomédicas, Departamento de Radiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leonardo Velásquez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Paz Orellana
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paula Muñoz Venturelli
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Gonzalo Martínez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
- Heart Research Institute, Sydney, NSW, Australia
| |
Collapse
|
14
|
Li B, Hussain W, Jiang ZL, Wang JY, Hussain S, Yasoob TB, Zhai YK, Ji XY, Dang YL. Nuclear proteins and diabetic retinopathy: a review. Biomed Eng Online 2024; 23:62. [PMID: 38918766 PMCID: PMC11197269 DOI: 10.1186/s12938-024-01258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/23/2024] [Indexed: 06/27/2024] Open
Abstract
Diabetic retinopathy (DR) is an eye disease that causes blindness and vision loss in diabetic. Risk factors for DR include high blood glucose levels and some environmental factors. The pathogenesis is based on inflammation caused by interferon and other nuclear proteins. This review article provides an overview of DR and discusses the role of nuclear proteins in the pathogenesis of the disease. Some core proteins such as MAPK, transcription co-factors, transcription co-activators, and others are part of this review. In addition, some current advanced treatment resulting from the role of nuclear proteins will be analyzes, including epigenetic modifications, the use of methylation, acetylation, and histone modifications. Stem cell technology and the use of nanobiotechnology are proposed as promising approaches for a more effective treatment of DR.
Collapse
Affiliation(s)
- Bin Li
- Department of Ophthalmology, The First Affiliated Hospital, Henan University, Kaifeng, 475004, Henan, China
| | - Wahab Hussain
- School of Stomatology, Henan University, Kaifeng, 475000, China
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medicine Science, Henan University, Kaifeng, 475000, China
| | - Zhi-Liang Jiang
- School of Clinical Medicine, Henan University, Kaifeng, 475004, Henan, China
| | - Jia-Yi Wang
- San-Quan College, XinXiang Medical University, No. 688 Xiangyang Road, Hongmen Town, Hongqi District, Xinxiang City, Henan, 453003, China
| | - Sarfraz Hussain
- College of Environment, Hohai University, Nanjing, 210098, China
| | - Talat Bilal Yasoob
- Department of Animal Sciences, Ghazi University, Dera Ghazi Khan, 32200, Pakistan
| | - Yuan-Kun Zhai
- School of Stomatology, Henan University, Kaifeng, 475000, China.
- Kaifeng Key Laboratory of Periodontal Tissue Engineering, Kaifeng, 475000, China.
| | - Xin-Ying Ji
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medicine Science, Henan University, Kaifeng, 475000, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China.
| | - Ya-Long Dang
- Department of Ophthalmology, Sanmenxia Central Hospital, Henan University of Science and Technology, Sanmenxia, Henan, China.
- Department of Ophthalmology, Sanmenxia Eye Hospital, Sanmenxia, Henan, China.
- Department of Ophthalmology, Henan University of Science and Technology School of Medicine, Luoyang, Henan, China.
| |
Collapse
|
15
|
Xu B, Wu Q, La R, Lu L, Abdu FA, Yin G, Zhang W, Ding W, Ling Y, He Z, Che W. Is systemic inflammation a missing link between cardiometabolic index with mortality? Evidence from a large population-based study. Cardiovasc Diabetol 2024; 23:212. [PMID: 38902748 PMCID: PMC11191290 DOI: 10.1186/s12933-024-02251-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 04/26/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND This study sought to elucidate the associations of cardiometabolic index (CMI), as a metabolism-related index, with all-cause and cardiovascular mortality among the older population. Utilizing data from the National Health and Nutrition Examination Survey (NHANES), we further explored the potential mediating effect of inflammation within these associations. METHODS A cohort of 3029 participants aged over 65 years old, spanning six NHANES cycles from 2005 to 2016, was enrolled and assessed. The primary endpoints of the study included all-cause mortality and cardiovascular mortality utilizing data from National Center for Health Statistics (NCHS). Cox regression model and subgroup analysis were conducted to assess the associations of CMI with all-cause and cardiovascular mortality. The mediating effect of inflammation-related indicators including leukocyte, neutrophil, lymphocyte, systemic immune-inflammation index (SII), neutrophil to lymphocyte ratio (NLR) were evaluated to investigate the potential mechanism of the associations between CMI and mortality through mediation package in R 4.2.2. RESULTS The mean CMI among the enrolled participants was 0.74±0.66, with an average age of 73.28±5.50 years. After an average follow-up period of 89.20 months, there were 1,015 instances of all-cause deaths and 348 cardiovascular deaths documented. In the multivariable-adjusted model, CMI was positively related to all-cause mortality (Hazard Ratio (HR)=1.11, 95% CI=1.01-1.21). Mediation analysis indicated that leukocytes and neutrophils mediated 6.6% and 13.9% of the association of CMI with all-cause mortality. CONCLUSION Elevated CMI is positively associated with all-cause mortality in the older adults. The association appeared to be partially mediated through inflammatory pathways, indicating that CMI may serve as a valuable indicator for poor prognosis among the older population.
Collapse
Affiliation(s)
- Bin Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
- Department of Cardiology, Zhongshan-Xuhui Hospital, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Qian Wu
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China.
- Research Institute of Clinical Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea.
| | - Rui La
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Lingchen Lu
- Department of Pediatric Surgery and Rehabilitation, Kunshan Maternity and Children's Health Care Hospital, Kunshan, Jiangsu, China
| | - Fuad A Abdu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Guoqing Yin
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Wen Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Wenquan Ding
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Yicheng Ling
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Zhiyuan He
- Department of Orthopedic Surgery, Orthopedic Institute, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Wenliang Che
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China.
| |
Collapse
|
16
|
Yamada Y, Furukawa K, Haruki K, Okui N, Shirai Y, Tsunematsu M, Yanagaki M, Yasuda J, Onda S, Ikegami T. Abdominal aortic calcification volume as a preoperative prognostic predictor for pancreatic cancer. Surg Today 2024:10.1007/s00595-024-02882-2. [PMID: 38880804 DOI: 10.1007/s00595-024-02882-2] [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: 03/22/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE Atherosclerosis and cancer may progress through common pathological factors. This study was performed to investigate the association between the abdominal aortic calcification (AAC) volume and outcomes following surgical treatment for pancreatic cancer. METHODS The subjects of this retrospective study were 194 patients who underwent pancreatic cancer surgery between 2007 and 2020. The AAC volume was assessed through routine preoperative computed tomography. Univariate and multivariate analyses were performed to evaluate the impact of the AAC volume on oncological outcomes. RESULTS A higher AAC volume (≥ 312 mm3) was identified in 66 (34%) patients, who were significantly older and had a higher prevalence of diabetes and sarcopenia. Univariate analysis revealed several risk factors for overall survival (OS), including male sex, an AAC volume ≥ 312 mm3, elevated carbohydrate antigen 19-9, prolonged operation time, increased intraoperative bleeding, lymph node metastasis, poor differentiation, and absence of adjuvant chemotherapy. Multivariate analysis identified an AAC volume ≥ 312 mm3, prolonged operation time, lymph node metastasis, poor differentiation, and absence of adjuvant chemotherapy as independent OS risk factors. The OS rate was significantly lower in the high AAC group than in the low AAC group. CONCLUSION The AAC volume may serve as a preoperative prognostic indicator for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yuta Yamada
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Kenei Furukawa
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| | - Koichiro Haruki
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Norimitsu Okui
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yoshihiro Shirai
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Masashi Tsunematsu
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Mitsuru Yanagaki
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Jungo Yasuda
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Shinji Onda
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Toru Ikegami
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| |
Collapse
|
17
|
Cignarella A, Bolego C, Barton M. Sex and sex steroids as determinants of cardiovascular risk. Steroids 2024; 206:109423. [PMID: 38631602 DOI: 10.1016/j.steroids.2024.109423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/08/2024] [Accepted: 04/14/2024] [Indexed: 04/19/2024]
Abstract
There are considerable sex differences regarding the risk of cardiovascular disease (CVD), including arterial hypertension, coronary artery disease (CAD) and stroke, as well as chronic renal disease. Women are largely protected from these conditions prior to menopause, and the risk increases following cessation of endogenous estrogen production or after surgical menopause. Cardiovascular diseases in women generally begin to occur at a later age than in men (on average with a delay of 10 years). Cessation of estrogen production also impacts metabolism, increasing the risk of developing obesity and diabetes. In middle-aged individuals, hypertension develops earlier and faster in women than in men, and smoking increases cardiovascular risk to a greater degree in women than it does in men. It is not only estrogen that affects female cardiovascular health and plays a protective role until menopause: other sex hormones such as progesterone and androgen hormones generate a complex balance that differentiates heart and blood vessel function in women compared to men. Estrogens improve vasodilation of epicardial coronary arteries and the coronary microvasculature by augmenting the release of vasodilating factors such as nitric oxide and prostacyclin, which are mechanisms of coronary vasodilatation that are more pronounced in women compared to men. Estrogens are also powerful inhibitors of inflammation, which in part explains their protective effects on CVD and chronic renal disease. Emerging evidence suggests that sex chromosomes also play a significant role in shaping cardiovascular risk. The cardiovascular protection conferred by endogenous estrogens may be extended by hormone therapy, especially using bioidentical hormones and starting treatment early after menopause.
Collapse
Affiliation(s)
| | - Chiara Bolego
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland; Andreas Grüntzig Foundation, Zürich, Switzerland.
| |
Collapse
|
18
|
Kirimi MT, Hoare D, Holsgrove M, Czyzewski J, Mirzai N, Mercer JR, Neale SL. Detection of Blood Clots Using a Whole Stent as an Active Implantable Biosensor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304748. [PMID: 38342628 DOI: 10.1002/advs.202304748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/28/2023] [Indexed: 02/13/2024]
Abstract
Many cardiovascular problems stem from blockages that form within the vasculature and often treatment includes fitting a stent through percutaneous coronary intervention. This offers a minimally invasive therapy but re-occlusion through restenosis or thrombosis formation often occurs post-deployment. Research is ongoing into the creation of smart stents that can detect the occurrence of further problems. In this study, it is shown that selectively metalizing a non-conductive stent can create a set of electrodes that are capable of detecting a build-up of material around the stent. The associated increase in electrical impedance across the electrodes is measured, testing the stent with blood clot to mimic thrombosis. It is shown that the device is capable of sensing different amounts of occlusion. The stent can reproducibly sense the presence of clot showing a 16% +/-3% increase in impedance which is sufficient to reliably detect the clot when surrounded by explanted aorta (one sample t-test, p = 0.009, n = 9). It is demonstrated that this approach can be extended beyond the 3D printed prototypes by showing that it can be applied to a commercially available stent and it is believed that it can be further utilized by other types of medical implants.
Collapse
Affiliation(s)
- Mahmut Talha Kirimi
- Centre for Medical and Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Daniel Hoare
- Institute of Cardiovascular and Medical Sciences/British Heart Foundation, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Michael Holsgrove
- BioElectronics Unit, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Jakup Czyzewski
- BioElectronics Unit, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Nosrat Mirzai
- BioElectronics Unit, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - John R Mercer
- Institute of Cardiovascular and Medical Sciences/British Heart Foundation, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Steve L Neale
- Centre for Medical and Industrial Ultrasonics, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
19
|
Bonacina F, Della-Morte D. Editorial: Exploiting cellular immunometabolism as a strategy for innovative cardiovascular therapies. Front Cardiovasc Med 2024; 11:1435850. [PMID: 38883983 PMCID: PMC11176533 DOI: 10.3389/fcvm.2024.1435850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/18/2024] Open
Affiliation(s)
- F Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - D Della-Morte
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
20
|
Zhou ZY, Wu L, Liu YF, Tang MY, Tang JY, Deng YQ, Liu L, Nie BB, Zou ZK, Huang L. IRE1α: from the function to the potential therapeutic target in atherosclerosis. Mol Cell Biochem 2024; 479:1079-1092. [PMID: 37310588 DOI: 10.1007/s11010-023-04780-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/03/2023] [Indexed: 06/14/2023]
Abstract
Inositol requiring enzyme 1 (IRE1) is generally thought to control the most conserved pathway in the unfolded protein response (UPR). Two isoforms of IRE1, IRE1α and IRE1β, have been reported in mammals. IRE1α is a ubiquitously expressed protein whose knockout shows marked lethality. In contrast, the expression of IRE1β is exclusively restricted in the epithelial cells of the respiratory and gastrointestinal tracts, and IRE1β-knockout mice are phenotypically normal. As research continues to deepen, IRE1α was showed to be tightly linked to inflammation, lipid metabolism regulation, cell death and so on. Growing evidence also suggests an important role for IRE1α in promoting atherosclerosis (AS) progression and acute cardiovascular events through disrupting lipid metabolism balance, facilitating cells apoptosis, accelerating inflammatory responses and promoting foam cell formation. In addition, IRE1α was recognized as novel potential therapeutic target in AS prevention. This review provides some clues about the relationship between IRE1α and AS, hoping to contribute to further understanding roles of IRE1α in atherogenesis and to be helpful for the design of novel efficacious therapeutics agents targeting IRE1α-related pathways.
Collapse
Affiliation(s)
- Zheng-Yang Zhou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Li Wu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Yi-Fan Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Mu-Yao Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Jing-Yi Tang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Anaesthesiology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Ya-Qian Deng
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Lei Liu
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Bin-Bin Nie
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zi-Kai Zou
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
- Department of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Liang Huang
- The Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
21
|
Wei C, Fan W, Zhang Y, Sun Q, Liu Y, Wang X, Liu J, Sun L. Albumin combined with neutrophil-to-lymphocyte ratio score and outcomes in patients with acute coronary syndrome treated with percutaneous coronary intervention. Coron Artery Dis 2024; 35:221-230. [PMID: 38299258 DOI: 10.1097/mca.0000000000001333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
BACKGROUND Evidence about the association between albumin combined with neutrophil-to-lymphocyte ratio score (ANS) and survival outcomes in patients with acute coronary syndrome (ACS) undergoing percutaneous coronary intervention (PCI) is rare. This study aimed to evaluate the prognostic value of ANS in patients with ACS undergoing PCI by propensity score matching (PSM) analysis. PATIENTS AND METHODS Patients with ACS undergoing PCI were consecutively enrolled in this prospective cohort study from January 2016 to December 2018. The albumin and neutrophil-to-lymphocyte ratio cutoff values for predicting major adverse cardiovascular events (MACEs) were calculated using receiver operating characteristic curves. Survival analysis was performed using Kaplan-Meier estimates, the Cox proportional hazard regression models and PSM. The study endpoint was the occurrence of a MACE, which included all-cause mortality and rehospitalization for severe heart failure during follow-up. RESULTS Overall, 1549 patients with adequate specimens were identified and assigned into different groups for comparison. Before and after PSM, the Kaplan-Meier curves showed that a higher ANS value was associated with a higher risk of MACEs (all P < 0.001). The multivariate Cox proportional hazard regression model showed that the ANS (per 1 score increase) [hazard ratio (HR), 2.016; 95% confidence interval (CI), 1.329-3.057; P = 0.001 vs. HR, 2.166; 95% CI, 1.344-3.492; P = 0.002] was an independent predictor for MACEs. CONCLUSION This study tentatively confirms that ANS may be a valuable clinical indicator to identify high-risk ACS patients after PCI. More high-quality prospective studies are needed in the future.
Collapse
Affiliation(s)
- Chen Wei
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Wenjun Fan
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Ying Zhang
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Qiyu Sun
- Department of Clinical Laboratory, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yixiang Liu
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Xinchen Wang
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Jingyi Liu
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
22
|
Traughber CA, Timinski K, Prince A, Bhandari N, Neupane K, Khan MR, Opoku E, Opoku E, Brubaker G, Shin J, Hong J, Kanuri B, Ertugral EG, Nagareddy PR, Kothapalli CR, Cherepanova O, Smith JD, Gulshan K. Disulfiram Reduces Atherosclerosis and Enhances Efferocytosis, Autophagy, and Atheroprotective Gut Microbiota in Hyperlipidemic Mice. J Am Heart Assoc 2024; 13:e033881. [PMID: 38563369 PMCID: PMC11262521 DOI: 10.1161/jaha.123.033881] [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: 12/07/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Pyroptosis executor GsdmD (gasdermin D) promotes atherosclerosis in mice and humans. Disulfiram was recently shown to potently inhibit GsdmD, but the in vivo efficacy and mechanism of disulfiram's antiatherosclerotic activity is yet to be explored. METHODS AND RESULTS We used human/mouse macrophages, endothelial cells, and smooth muscle cells and a hyperlipidemic mouse model of atherosclerosis to determine disulfiram antiatherosclerotic efficacy and mechanism. The effects of disulfiram on several atheroprotective pathways such as autophagy, efferocytosis, phagocytosis, and gut microbiota were determined. Atomic force microscopy was used to determine the effects of disulfiram on the biophysical properties of the plasma membrane of macrophages. Disulfiram-fed hyperlipidemic apolipoprotein E-/- mice showed significantly reduced interleukin-1β release upon in vivo Nlrp3 (NLR family pyrin domain containing 3) inflammasome activation. Disulfiram-fed mice showed smaller atherosclerotic lesions (~27% and 29% reduction in males and females, respectively) and necrotic core areas (~50% and 46% reduction in males and females, respectively). Disulfiram induced autophagy in macrophages, smooth muscle cells, endothelial cells, hepatocytes/liver, and atherosclerotic plaques. Disulfiram modulated other atheroprotective pathways (eg, efferocytosis, phagocytosis) and gut microbiota. Disulfiram-treated macrophages showed enhanced phagocytosis/efferocytosis, with the mechanism being a marked increase in cell-surface expression of efferocytic receptor MerTK. Atomic force microscopy analysis revealed altered biophysical properties of disulfiram-treated macrophages, showing increased order-state of plasma membrane and increased adhesion strength. Furthermore, 16sRNA sequencing of disulfiram-fed hyperlipidemic mice showed highly significant enrichment in atheroprotective gut microbiota Akkermansia and a reduction in atherogenic Romboutsia species. CONCLUSIONS Taken together, our data show that disulfiram can simultaneously modulate several atheroprotective pathways in a GsdmD-dependent as well as GsdmD-independent manner.
Collapse
Affiliation(s)
- C. Alicia Traughber
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Kara Timinski
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Ashutosh Prince
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Nilam Bhandari
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Kalash Neupane
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Mariam R. Khan
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Esther Opoku
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
| | - Emmanuel Opoku
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Gregory Brubaker
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Junchul Shin
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Babunageswararao Kanuri
- Department of Internal Medicine, Cardiovascular SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | - Elif G. Ertugral
- Department of Chemical & Biomedical EngineeringCleveland State UniversityClevelandOHUSA
| | - Prabhakara R. Nagareddy
- Department of Internal Medicine, Cardiovascular SectionUniversity of Oklahoma Health Sciences Center (OUHSC)Oklahoma CityOKUSA
| | | | - Olga Cherepanova
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Jonathan D. Smith
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| | - Kailash Gulshan
- Center for Gene Regulation in Health and DiseaseCleveland State UniversityClevelandOHUSA
- Department of Biology, Geology, and Environmental SciencesCleveland State UniversityClevelandOHUSA
- Department of Cardiovascular and Metabolic SciencesLerner Research Institute, Cleveland ClinicClevelandOHUSA
| |
Collapse
|
23
|
Salloum Z, Dauner K, Li YF, Verma N, Valdivieso-González D, Almendro-Vedia V, Zhang JD, Nakka K, Chen MX, McDonald J, Corley CD, Sorisky A, Song BL, López-Montero I, Luo J, Dilworth JF, Zha X. Statin-mediated reduction in mitochondrial cholesterol primes an anti-inflammatory response in macrophages by upregulating Jmjd3. eLife 2024; 13:e85964. [PMID: 38602170 PMCID: PMC11186637 DOI: 10.7554/elife.85964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/13/2024] [Indexed: 04/12/2024] Open
Abstract
Statins are known to be anti-inflammatory, but the mechanism remains poorly understood. Here, we show that macrophages, either treated with statin in vitro or from statin-treated mice, have reduced cholesterol levels and higher expression of Jmjd3, a H3K27me3 demethylase. We provide evidence that lowering cholesterol levels in macrophages suppresses the adenosine triphosphate (ATP) synthase in the inner mitochondrial membrane and changes the proton gradient in the mitochondria. This activates nuclear factor kappa-B (NF-κB) and Jmjd3 expression, which removes the repressive marker H3K27me3. Accordingly, the epigenome is altered by the cholesterol reduction. When subsequently challenged by the inflammatory stimulus lipopolysaccharide (M1), macrophages, either treated with statins in vitro or isolated from statin-fed mice, express lower levels proinflammatory cytokines than controls, while augmenting anti-inflammatory Il10 expression. On the other hand, when macrophages are alternatively activated by IL-4 (M2), statins promote the expression of Arg1, Ym1, and Mrc1. The enhanced expression is correlated with the statin-induced removal of H3K27me3 from these genes prior to activation. In addition, Jmjd3 and its demethylase activity are necessary for cholesterol to modulate both M1 and M2 activation. We conclude that upregulation of Jmjd3 is a key event for the anti-inflammatory function of statins on macrophages.
Collapse
Affiliation(s)
- Zeina Salloum
- Chronic Disease Program, Ottawa Hospital Research InstituteOttawaCanada
| | - Kristin Dauner
- Chronic Disease Program, Ottawa Hospital Research InstituteOttawaCanada
| | - Yun-feng Li
- College of Life Sciences, Wuhan UniversityWuhanChina
| | - Neha Verma
- Chronic Disease Program, Ottawa Hospital Research InstituteOttawaCanada
| | - David Valdivieso-González
- Departamento Química Física, Universidad Complutense de Madrid, AvdaMadridSpain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12)MadridSpain
| | - Víctor Almendro-Vedia
- Departamento Química Física, Universidad Complutense de Madrid, AvdaMadridSpain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12)MadridSpain
| | - John D Zhang
- Chronic Disease Program, Ottawa Hospital Research InstituteOttawaCanada
| | - Kiran Nakka
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
| | - Mei Xi Chen
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
- Department of Cell and Regenerative Biology, University of WisconsinMadisonUnited States
| | - Jeffrey McDonald
- Department of Molecular Genetics, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Chase D Corley
- Department of Molecular Genetics, The University of Texas Southwestern Medical CenterDallasUnited States
| | - Alexander Sorisky
- Chronic Disease Program, Ottawa Hospital Research InstituteOttawaCanada
- Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | | | - Iván López-Montero
- Departamento Química Física, Universidad Complutense de Madrid, AvdaMadridSpain
- Instituto de Investigación Biomédica Hospital Doce de Octubre (imas12)MadridSpain
| | - Jie Luo
- College of Life Sciences, Wuhan UniversityWuhanChina
| | - Jeffrey F Dilworth
- Sprott Center for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research InstituteOttawaCanada
- Department of Cell and Regenerative Biology, University of WisconsinMadisonUnited States
- Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Xiaohui Zha
- Chronic Disease Program, Ottawa Hospital Research InstituteOttawaCanada
- Departments of Medicine and of Biochemistry, Microbiology & Immunology, University of OttawaOttawaCanada
| |
Collapse
|
24
|
Wang X, Huang M, Li Z, Liu Y, Ma M, He Y, Yang R, Li L, Gao S, Yu C. Fibrinogen/albumin ratio and carotid artery plaques in coronary heart disease patients with different glucose metabolic states: a RCSCD-TCM study. Endocrine 2024; 84:100-108. [PMID: 37824044 DOI: 10.1007/s12020-023-03558-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023]
Abstract
AIM The relationship between fibrinogen/albumin ratio (FAR) and carotid artery plaques (CAPs) was investigated in patients with coronary heart disease (CHD). METHODS A total of 11,624 patients with CHD were enrolled and divided into quartiles based on the FAR (Q1: FAR index ≤ 0.0663; Q2: 0.0664 ≤ FAR index ≤ 0.0790; Q3: 0.0791 ≤ FAR index ≤ 0.0944; Q4: FAR index > 0.0944). Patients were classified into three groups according to their blood glucose levels: normal glucose regulation (NGR), prediabetes mellitus (pre-DM), and diabetes mellitus (DM) groups. Carotid ultrasonography was performed to detect CAPs. The relationship between FAR and CAPs was evaluated using logistic and subgroup analyses. RESULTS Among 11,624 participants, 8738 (75.14%) had CAPs. Compared with Q1, the odds ratio (OR) of Q4 in patients with CHD was 2.00 (95% confidence interval [CI]: 1.71-2.34) after multivariate adjustment. Taking Q1 as a reference, a higher OR was observed in Q4 of FAR for CAPs in men [OR: 2.26; 95% CI: 1.73-2.95] in the multi-adjusted models. Moreover, multivariate adjustment indicated that the highest OR was observed in patients with CHD and DM (OR: 2.36; 95% CI: 1.80-3.10). CONCLUSIONS A significant association between FAR and CAPs was observed in patients with CHD, regardless of sex or blood glucose levels. Therefore, FAR may be used as an effective indicator to identify patients at a high risk of CAPs among patients with CHD.
Collapse
Affiliation(s)
- Xu Wang
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Mengnan Huang
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Zhu Li
- Zhejiang Chinese Medical University, No. 548, Binwen Road, Binjiang District Hangzhou City, Hangzhou, Zhejiang, China
| | - Yijia Liu
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Mei Ma
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Yuanyuan He
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Rongrong Yang
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China.
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China.
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, No. 10, Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, China.
| |
Collapse
|
25
|
Biwer LA, Man JJ, Camarda ND, Carvajal BV, Karumanchi SA, Jaffe IZ. Prior Exposure to Experimental Preeclampsia Increases Atherosclerotic Plaque Inflammation in Atherogenic Mice-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:946-953. [PMID: 38450510 PMCID: PMC10978246 DOI: 10.1161/atvbaha.123.320474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND Women with a history of preeclampsia have evidence of premature atherosclerosis and increased risk of myocardial infarction and stroke compared with women who had a normotensive pregnancy. Whether this is due to common risk factors or a direct impact of prior preeclampsia exposure has never been tested in a mouse atherosclerosis model. METHODS Pregnant LDLR-KO (low-density lipoprotein receptor knockout; n=35) female mice were randomized in midgestation to sFlt1 (soluble fms-like tyrosine kinase 1)-expressing adenovirus or identical control adenovirus. Postpartum, mice were fed high-fat diet for 8 weeks to induce atherogenesis. Comparison between the control and preeclampsia models was made for metabolic parameters, atherosclerosis burden and composition by histology, plaque inflammation by flow cytometry, and aortic cytokines and inflammatory markers using a cytokine array. RESULTS In pregnant LDLR-KO mice, sFlt1 adenovirus significantly induced serum sFlt1, blood pressure, renal endotheliosis, and decreased pup viability. After 8 weeks of postpartum high fat feeding, body weight, fasting glucose, plasma cholesterol, HDL (high-density lipoprotein), and LDL (low-density lipoprotein) were not significantly different between groups with no change in aortic root plaque size, lipid content, or necrotic core area. Flow cytometry demonstrated significantly increased CD45+ aortic arch leukocytes and CD3+T cells and aortic lysate contained more CCL (CC motif chemokine ligand) 22 and fetuin A and decreased expression of IGFBP6 (insulin-like growth factor-binding protein 6) and CCL21 in preeclampsia-exposed mice compared with controls. CONCLUSIONS In atherogenic LDLR-KO mice, exposure to sFlt1-induced preeclampsia during pregnancy increases future atherosclerotic plaque inflammation, supporting the concept that preeclampsia directly exacerbates atherosclerotic inflammation independent of preexisting risk factors. This mechanism may contribute to ischemic vascular disease in women after preeclampsia pregnancy.
Collapse
Affiliation(s)
- Lauren A. Biwer
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
- Department of Comparative Medicine, Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT
| | - Joshua J. Man
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
| | | | | | | | - Iris Z. Jaffe
- Molecular Cardiology Research Center, Tufts Medical Center, Boston MA
| |
Collapse
|
26
|
Liu S, Han Y, Kong L, Wang G, Ye Z. Atomic force microscopy in disease-related studies: Exploring tissue and cell mechanics. Microsc Res Tech 2024; 87:660-684. [PMID: 38063315 DOI: 10.1002/jemt.24471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/22/2023] [Accepted: 11/26/2023] [Indexed: 03/02/2024]
Abstract
Despite significant progress in human medicine, certain diseases remain challenging to promptly diagnose and treat. Hence, the imperative lies in the development of more exhaustive criteria and tools. Tissue and cellular mechanics exhibit distinctive traits in both normal and pathological states, suggesting that "force" represents a promising and distinctive target for disease diagnosis and treatment. Atomic force microscopy (AFM) holds great promise as a prospective clinical medical device due to its capability to concurrently assess surface morphology and mechanical characteristics of biological specimens within a physiological setting. This review presents a comprehensive examination of the operational principles of AFM and diverse mechanical models, focusing on its applications in investigating tissue and cellular mechanics associated with prevalent diseases. The findings from these studies lay a solid groundwork for potential clinical implementations of AFM. RESEARCH HIGHLIGHTS: By examining the surface morphology and assessing tissue and cellular mechanics of biological specimens in a physiological setting, AFM shows promise as a clinical device to diagnose and treat challenging diseases.
Collapse
Affiliation(s)
- Shuaiyuan Liu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Yibo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Central Hospital of Chongqing University, Chongqing Emergency Medical Center, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratory, Chongqing, China
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
- JinFeng Laboratory, Chongqing, China
| |
Collapse
|
27
|
Hong S, He H, Fang P, Liu S, Chen C. Association of neutrophil-to-lymphocyte ratio and risk of cardiovascular and all-cause mortality in hypertension patients. Heliyon 2024; 10:e27517. [PMID: 38496832 PMCID: PMC10944217 DOI: 10.1016/j.heliyon.2024.e27517] [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: 11/28/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
Background and objective Hypertension affects over a billion people worldwide and is often associated with poor prognoses. The neutrophil-to-lymphocyte ratio (NLR) has become a significant marker, showing a connection to adverse outcomes in cardiovascular diseases (CVDs). The objective of this study is to examine the relationship between the NLR and outcomes in patients with hypertension. Methods The study included hypertensive individuals who were surveyed in the National Health and Nutrition Examination Survey (NHANES) from 2009 to 2018. Mortality status was determined using the data from National Death Index (NDI). To investigate the dose-response relationship, restricted cubic spline (RCS) models were used. This study employed adjusted cox proportional hazards regression models to compute hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs) for all-cause and cardiovascular mortality. The predictive accuracy of the NLR for survival outcomes was assessed utilizing time-dependent receiver operating characteristic (ROC) curve analysis. Results A total of 13,724 participants were included in the final analysis, including 7073 males and 6651 females. The cohort was stratified into higher (>2.0) and lower (≤2.0) NLR groups according to the median value. Over a median follow-up of 64 months, there were 1619 all-cause deaths and 522 cardiovascular deaths among participants. The RCS analysis indicated a non-linear relationship between NLR and the risk of mortality. The adjusted model showed that the group with a higher NLR had a significantly higher risk of all-cause (HR 1.47, 95% CI 1.22-1.77) and cardiovascular mortality (HR 2.08, 95% CI 1.52-2.86). ROC analysis showed that the area under the curves (AUCs) of 0.692, 0.662, 0.644, and 0.625 for predicting all-cause mortality, and 0.712, 0.692, 0.687, and 0.660 for cardiovascular mortality at 1, 3, 5, and 10 years. Conclusion Elevated NLR is associated with increased risk of cardiovascular and all-cause mortality, and NLR may independently predict outcomes in individuals with hypertension.
Collapse
Affiliation(s)
- Shaoqing Hong
- Corresponding author. Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.98 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China.
| | | | - Peng Fang
- Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.33 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China
| | - Shuai Liu
- Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.33 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China
| | - Changyi Chen
- Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.33 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China
| |
Collapse
|
28
|
Volpatti LR, de Matos SN, Borjas G, Reda J, Watkins EA, Zhou Z, Nguyen M, Solanki A, Fang Y, Hubbell JA. LDL-Binding IL-10 Reduces Vascular Inflammation in Atherosclerotic Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.582839. [PMID: 38496521 PMCID: PMC10942346 DOI: 10.1101/2024.03.04.582839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease associated with the accumulation of low-density lipoprotein (LDL) in arterial walls. Higher levels of the anti-inflammatory cytokine IL-10 in serum are correlated with reduced plaque burden. However, cytokine therapies have not translated well to the clinic, partially due to their rapid clearance and pleiotropic nature. Here, we engineered IL-10 to overcome these challenges by hitchhiking on LDL to atherosclerotic plaques. Specifically, we constructed fusion proteins in which one domain is IL-10 and the other is an antibody fragment (Fab) that binds to protein epitopes of LDL. In murine models of atherosclerosis, we show that systemically administered Fab-IL-10 constructs bind circulating LDL and traffic to atherosclerotic plaques. One such construct, 2D03-IL-10, significantly reduces aortic immune cell infiltration to levels comparable to healthy mice, whereas non-targeted IL-10 has no therapeutic effect. Mechanistically, we demonstrate that 2D03-IL-10 preferentially associates with foamy macrophages and reduces pro-inflammatory activation markers. This platform technology can be applied to a variety of therapeutics and shows promise as a potential targeted anti-inflammatory therapy in atherosclerosis.
Collapse
Affiliation(s)
- Lisa R. Volpatti
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, United States
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208, United States
| | - Salvador Norton de Matos
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Medical Scientist Training Program, Pritzker School of Medicine, University of Chicago, IL, 60637, United States
| | - Gustavo Borjas
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Joseph Reda
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Elyse A. Watkins
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
| | - Zhengjie Zhou
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
| | - Mindy Nguyen
- Animal Resources Center, University of Chicago, Chicago, IL 60637, United States
| | - Ani Solanki
- Animal Resources Center, University of Chicago, Chicago, IL 60637, United States
| | - Yun Fang
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL 60637, United States
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, USA
- Committee on Molecular Medicine, University of Chicago, Chicago, IL, 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, United States
- Committee on Immunology, University of Chicago, Chicago, IL 60637, United States
- Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, United States
| |
Collapse
|
29
|
Saiioum Z, Dauner K, Li YF, Verma N, Almendro-Vedia V, Valdivieso Gonzalez D, Zhang DJ, Nakka K, McDonald J, Sorisky A, Song BL, Lopez Montero I, Luo J, Dilworth J, Zha X. Statin-mediated reduction in mitochondrial cholesterol primes an anti-inflammatory response in macrophages by upregulating JMJD3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.09.523264. [PMID: 36711703 PMCID: PMC9881925 DOI: 10.1101/2023.01.09.523264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Stains are known to be anti-inflammatory, but the mechanism remains poorly understood. Here we show that macrophages, either treated with statin in vitro or from statin-treated mice, have reduced cholesterol levels and higher expression of Jmjd3, a H3K27me3 demethylase. We provide evidence that lowering cholesterol levels in macrophages suppresses the ATP synthase in the inner mitochondrial membrane (IMM) and changes the proton gradient in the mitochondria. This activates NFkB and Jmjd3 expression to remove the repressive marker H3K27me3. Accordingly, the epigenome is altered by the cholesterol reduction. When subsequently challenged by the inflammatory stimulus LPS (M1), both macrophages treated with statins in vitro or isolated from statin-treated mice in vivo, express lower levels pro-inflammatory cytokines than controls, while augmenting anti-inflammatory Il10 expression. On the other hand, when macrophages are alternatively activated by IL4 (M2), statins promote the expression of Arg1, Ym1, and Mrc1. The enhanced expression is correlated with the statin-induced removal of H3K27me3 from these genes prior to activation. In addition, Jmjd3 and its demethylase activity are necessary for cholesterol to modulate both M1 and M2 activation. We conclude that upregulation of Jmjd3 is a key event for the anti-inflammatory function of statins on macrophages.
Collapse
|
30
|
Yu Y, Bian S, Jiang Y, Li B, Cui X, Ding S, Dai Z, Chen R, Zhong W, Yuan W. An Ex Vivo Aorta Culture Model to Study Vascular Cellular Senescence. Adv Biol (Weinh) 2024; 8:e2300140. [PMID: 38051940 DOI: 10.1002/adbi.202300140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 10/29/2023] [Indexed: 12/07/2023]
Abstract
Animal studies on vascular aging pose a few limitations. One of the most important reasons for this is the absence of a fast and efficient model of vascular tissue aging. In this study, ex vivo aortic culture and Matrigel subcutaneous implantation are combined to develop a new model for studying vascular cellular senescence. Eight-week-old C57BL/6J mice are used to obtain aortas. Bleomycin is used to induce aortas senescence in vitro. Then, aortas are transplanted to the acceptor mice with Matrigel. Senescence is evaluated using western blotting, quantitative polymerase chain reaction, and senescence-associated beta-galactosidase activity. Inflammatory cytokines are detected using Luminex Liquid Suspension Chip. RNA levels are analyzed by transcriptome sequencing. The results revealed that vessels in the bleomycin group exhibited significant senescence than those in the control group that can be enhanced by stripping vessel adventitia. The levels of cytokines such as interleukin (IL-2, IL-1β, and IL-6 increased significantly in the ex vivo model. Furthermore, transcriptome sequencing revealed 56 significantly differentially expressed genes (DEGs) in ex vivo model vessels compared with those in naturally aging aortas. In conclusion, this study introduces a cost-effective and time-saving vessel senescence model for vascular cellular senescence.
Collapse
Affiliation(s)
- Yijie Yu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Shihui Bian
- Department of Geriatrics, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Yu Jiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Bo Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Xinggang Cui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Shu Ding
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Rui Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Wei Zhong
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, P. R. China
| |
Collapse
|
31
|
Li Y, Zhang M, Li Y, Shen Y, Wang X, Li X, Wang Y, Yu T, Lv J, Qin Y. Flagellar hook protein FlgE promotes macrophage activation and atherosclerosis by targeting ATP5B. Atherosclerosis 2024; 390:117429. [PMID: 38278062 DOI: 10.1016/j.atherosclerosis.2023.117429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/28/2024]
Abstract
BACKGROUND AND AIMS Pseudomonas aeruginosa (P. aeruginosa) infections are strongly linked to the development of cardiovascular disease and atherosclerosis; however, the underlying mechanisms remain unclear. We previously confirmed that the flagellar hook protein FlgE in P. aeruginosa has immunostimulatory effects. This study investigated the effects and mechanisms of action of FlgE on atherogenesis. METHODS ApoE-/- mice were intravenously challenged with FlgE or FlgEM recombinant proteins for eight weeks. A murine model of chronic lung colonization was established using beads containing either mutable- or wild-type bacteria. Aortic sinus sections were stained to assess atherosclerosis progression. THP-1 macrophages exposed to FlgE or FlgEM were evaluated for their effects on lipid uptake and inflammation in vitro. Western blotting and pull-down assays were used to identify the binding proteins and signaling pathways involved, and specific blocking experiments were performed to confirm these effects. RESULTS FlgE accelerated atherosclerosis progression by triggering lipid deposition and inflammatory responses in high-fat diet (HFD)-fed ApoE-/- mice. In comparison to infection with wild-type PAO1, infection with PAO1/flgEΔBmF resulted in reduced atherosclerosis. Mechanistic analysis indicated that FlgE exacerbated lipoprotein uptake and foam cell formation by upregulating SR-A1 expression. Moreover, FlgE activated NF-κB and MAPK signaling, which subsequently led to inflammatory responses in THP-1-derived macrophages. Pull-down assays revealed that FlgE directly interacted with ATP5B, whereas blocking ATP5B attenuated FlgE-induced responses in macrophages. CONCLUSIONS FlgE induces macrophage lipid uptake and pro-inflammatory responses mediated by ATP5B/NF-kB/AP-1 signaling, which eventually results in atherosclerosis. These findings support the development of therapeutic strategies for P. aeruginosa infection-induced atherosclerosis.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Laboratory Examination, People's Hospital of Rizhao City, The Affiliated Hospital of Jining Medical College, Rizhao, China
| | - Min Zhang
- Department of Laboratory Examination, People's Hospital of Rizhao City, The Affiliated Hospital of Jining Medical College, Rizhao, China
| | - Yanmeng Li
- Department of Laboratory Examination, People's Hospital of Rizhao City, The Affiliated Hospital of Jining Medical College, Rizhao, China
| | - Ying Shen
- National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Suchow University, Suzhou, 215006, China
| | - Xiaoping Wang
- Department of Laboratory Examination, People's Hospital of Rizhao City, The Affiliated Hospital of Jining Medical College, Rizhao, China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, 266000, China
| | - Yiqiang Wang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000, China.
| | - Jie Lv
- Department of Laboratory Examination, People's Hospital of Rizhao City, The Affiliated Hospital of Jining Medical College, Rizhao, China.
| | - Yan Qin
- Department of Laboratory Examination, People's Hospital of Rizhao City, The Affiliated Hospital of Jining Medical College, Rizhao, China.
| |
Collapse
|
32
|
Nègre-Salvayre A, Salvayre R. Reactive Carbonyl Species and Protein Lipoxidation in Atherogenesis. Antioxidants (Basel) 2024; 13:232. [PMID: 38397830 PMCID: PMC10886358 DOI: 10.3390/antiox13020232] [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: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a multifactorial disease of medium and large arteries, characterized by the presence of lipid-rich plaques lining the intima over time. It is the main cause of cardiovascular diseases and death worldwide. Redox imbalance and lipid peroxidation could play key roles in atherosclerosis by promoting a bundle of responses, including endothelial activation, inflammation, and foam cell formation. The oxidation of polyunsaturated fatty acids generates various lipid oxidation products such as reactive carbonyl species (RCS), including 4-hydroxy alkenals, malondialdehyde, and acrolein. RCS covalently bind to nucleophilic groups of nucleic acids, phospholipids, and proteins, modifying their structure and activity and leading to their progressive dysfunction. Protein lipoxidation is the non-enzymatic post-translational modification of proteins by RCS. Low-density lipoprotein (LDL) oxidation and apolipoprotein B (apoB) modification by RCS play a major role in foam cell formation. Moreover, oxidized LDLs are a source of RCS, which form adducts on a huge number of proteins, depending on oxidative stress intensity, the nature of targets, and the availability of detoxifying systems. Many systems are affected by lipoxidation, including extracellular matrix components, membranes, cytoplasmic and cytoskeletal proteins, transcription factors, and other components. The mechanisms involved in lipoxidation-induced vascular dysfunction are not fully elucidated. In this review, we focus on protein lipoxidation during atherogenesis.
Collapse
Affiliation(s)
- Anne Nègre-Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| | - Robert Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| |
Collapse
|
33
|
Mustafa K, Han Y, He D, Wang Y, Niu N, Jose PA, Jiang Y, Kopp JB, Lee H, Qu P. Poly-(ADP-ribose) polymerases inhibition by olaparib attenuates activities of the NLRP3 inflammasome and of NF-κB in THP-1 monocytes. PLoS One 2024; 19:e0295837. [PMID: 38335214 PMCID: PMC10857571 DOI: 10.1371/journal.pone.0295837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 12/01/2023] [Indexed: 02/12/2024] Open
Abstract
Poly-(ADP-ribose) polymerases (PARPs) are a protein family that make ADP-ribose modifications on target genes and proteins. PARP family members contribute to the pathogenesis of chronic inflammatory diseases, including atherosclerosis, in which monocytes/macrophages play important roles. PARP inhibition is protective against atherosclerosis. However, the mechanisms by which PARP inhibition exerts this beneficial effect are not well understood. Here we show that in THP-1 monocytes, inhibition of PARP by olaparib attenuated oxidized low-density lipoprotein (oxLDL)-induced protein expressions of nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing-3 (NLRP3) inflammasome components: NLRP3, apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), and caspase-1. Consistent with this effect, olaparib decreased oxLDL-enhanced interleukin (IL)-1β and IL-18 protein expression. Olaparib also decreased the oxLDL-mediated increase in mitochondrial reactive oxygen species. Similar to the effects of the NLRP3 inhibitor, MCC950, olaparib attenuated oxLDL-induced adhesion of monocytes to cultured human umbilical vein endothelial cells and reduced foam cell formation. Furthermore, olaparib attenuated the oxLDL-mediated activation of nuclear factor (NF)-κB through the oxLDL-mediated increase in IκBα phosphorylation and assembly of NF-κB subunits, demonstrated by co-immunoprecipitation of IκBα with RelA/p50 and RelB/p52 subunits. Moreover, PARP inhibition decreased oxLDL-mediated protein expression of a NF-κB target gene, VCAM1, encoding vascular cell adhesion molecule-1. This finding indicates an important role for NF-κB activity in PARP-mediated activation of the NLRP3 inflammasome. Thus, PARP inhibition by olaparib attenuates NF-κB and NLRP3 inflammasome activities, lessening monocyte cell adhesion and macrophage foam cell formation. These inhibitory effects of olaparib on NLRP3 activity potentially protect against atherosclerosis.
Collapse
Affiliation(s)
- Khamis Mustafa
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ying Han
- Department of Cardiology, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Dan He
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ying Wang
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Nan Niu
- Department of Cardiology, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Pedro A. Jose
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, United States of America
- Department of Physiology/Pharmacology, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, United States of America
| | - Yinong Jiang
- Department of Cardiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jeffrey B. Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hewang Lee
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
- Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, District of Columbia, United States of America
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peng Qu
- Institute of Heart and Vessel Diseases, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
- Department of Cardiology, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
- Faculty of Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
34
|
Asenjo-Lobos C, González L, Bulnes JF, Roque M, Muñoz Venturelli P, Rodríguez GM. Cardiovascular events risk in patients with systemic autoimmune diseases: a prognostic systematic review and meta-analysis. Clin Res Cardiol 2024; 113:246-259. [PMID: 37650912 DOI: 10.1007/s00392-023-02291-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Chronic inflammation is considered a risk factor for the development of atherosclerosis and cardiovascular (CV) events. We seek to assess the risk of CV events in patients with Systemic autoimmune diseases (SAD), such as Systemic Lupus Erythematosus (SLE), Rheumatoid Arthritis (RA), Psoriasis (Ps) and Ankylosing Spondylitis (AS), compared with the general population. METHODS AND RESULTS A systematic search of MEDLINE from inception up to May 2021 was performed. Observational studies including individuals with and without autoimmune diseases (SLE, RA, Ps, AS), which reported a measure of association and variability for the effect of SAD on CV events, were included. The random effects meta-analysis was performed using the Hartung-Knapp-Sidik-Jonkman approach to obtain the pooled estimates. Cardiovascular Events including CV mortality, non-fatal myocardial infarction (MI), non-fatal stroke and coronary revascularization were the main outcomes evaluated. Fifty-four studies were selected, with a total of 24,107,072 participants. The presence of SAD was associated with an increased risk of CV mortality (HR 1.49 [95% CI 1.10-2.03]), non-fatal MI (HR 1.42 [95% CI 1.23-1.62]), and non-fatal stroke (HR 1.47 [95% CI 1.28-1.70]). RA, SLE, and Ps (particularly with arthritis) were significantly associated with a higher risk of MI and stroke. SAD was also associated with an increased risk of Major Adverse Cardiovascular Events (MACE) (HR 1.45 [95% CI 1.16-1.83]). CONCLUSION Patients with SAD present an increased risk of CV morbidity and mortality, which should be considered when establishing therapeutic strategies. These findings support the role of systemic inflammation in the development of atherosclerosis-driven disease.
Collapse
Affiliation(s)
- Claudia Asenjo-Lobos
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad de Desarrollo, Santiago, Chile
| | - Leticia González
- Centro de Imágenes Biomédicas, Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto Milenio de Ingeniería e Inteligencia Artificial para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Francisco Bulnes
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marta Roque
- Iberoamerican Cochrane Centre, Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain
| | - Paula Muñoz Venturelli
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana Universidad de Desarrollo, Santiago, Chile
- Faculty of Medicine, The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | | |
Collapse
|
35
|
Patel KK, Shaw LJ. Perivascular Adipose Tissue Inflammation: Does It Explain Sex-Based Differences in Atherosclerotic Heart Disease? Circ Cardiovasc Imaging 2024; 17:e016559. [PMID: 38377231 PMCID: PMC10883611 DOI: 10.1161/circimaging.124.016559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Affiliation(s)
- Krishna K. Patel
- The Zena and Michael A. Wiener Cardiovascular Institute, Blavatnik Family Women’s Health Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Leslee J. Shaw
- The Zena and Michael A. Wiener Cardiovascular Institute, Blavatnik Family Women’s Health Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
36
|
Chen G, Li X, Li X, Liu S, Xie J. Mucosal membrane pressure injury in intensive care units: A scoping review. Intensive Crit Care Nurs 2024; 80:103560. [PMID: 37918080 DOI: 10.1016/j.iccn.2023.103560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/11/2023] [Accepted: 09/29/2023] [Indexed: 11/04/2023]
Abstract
AIM To describe published work on the current situation of mucosal membrane pressure injury of patients in the intensive care unit. BACKGROUND Device-related pressure injuries in critically ill patients are mostly focused on skin surface injuries, and less attention is paid to mucosal membrane pressure injury. METHODS We searched PubMed, Embase, Web of Science, CINAHL, and Cochrane Library from database construction until March 1, 2023. Two researchers independently screened and extracted data. RESULTS Eighteen articles met our criteria and were published in 2014-2023. The included studies showed that the incidence of mucosal membrane pressure injury in critically ill patients ranged from 0.83% to 88.2%, and the prevalence ranged from 0.16% to 55.6%. The most frequently reported site of mucosal injury is the oral mucosa, followed by the nasal mucosa. Ten studies used Braden to assess the risk of mucosal membrane pressure injury, and only six studies reported specific stages of mucosal membrane pressure injury. Thirteen studies described 30 risk factors for mucosal membrane pressure injury, with albumin being the most frequently reported risk factor, followed by the vasoconstrictive drugs use. Thirty risk factors were summarized in six aspects: medical device-related factors, disease-related factors, treatment- related factors, physiological and biochemical parameters, demographic-related factors, and microbial colonisation. CONCLUSIONS The incidence or prevalence of mucosal membrane pressure injury varies widely, and specific risk assessment tools and standardized staging criteria need to be further determined. The risk factors of mucosal membrane pressure injury involve multiple aspects, and some risk factors have only been explored in few studies and need to be further verified, in order to detect the risk group of mucosal membrane pressure injury early and take targeted preventive measures. IMPLICATIONS FOR CLINICAL PRACTICE We synthesized the current research status of mucosal membrane pressure injury in critically ill patients, which can provide a valuable reference for the clinical staff to develop preventive and management measures for such patients.
Collapse
Affiliation(s)
- Guanjie Chen
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaoqing Li
- Department of Nursing, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Xuezhu Li
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu, China
| | - Songqiao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu, China
| | - Jianfeng Xie
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
37
|
Wang SQ, Xiang J, Zhang GQ, Fu LY, Xu YN, Chen Y, Tao L, Hu XX, Shen XC. Essential oil from Fructus Alpinia zerumbet ameliorates atherosclerosis by activating PPARγ-LXRα-ABCA1/G1 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155227. [PMID: 38128398 DOI: 10.1016/j.phymed.2023.155227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/11/2023] [Accepted: 11/18/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Atherosclerosis (AS) is a progressive chronic disease. Currently, cardiovascular diseases (CVDs) caused by AS is responsible for the global increased mortality. Yanshanjiang as miao herb in Guizhou of China is the dried and ripe fruit of Fructus Alpinia zerumbet. Accumulated evidences have confirmed that Yanshanjiang could ameliorate CVDs, including AS. Nevertheless, its effect and mechanism on AS are still largely unknown. PURPOSE To investigate the role of essential oil from Fructus Alpinia zerumbet (EOFAZ) on AS, and the potential mechanism. METHODS A high-fat diet (HFD) ApoE-/- mice model of AS and a oxLDL-induced model of macrophage-derived foam cells (MFCs) were reproduced to investigate the pharmacological properties of EOFAZ on AS in vivo and foam cell formation in vitro, respectively. The underlying mechanisms of EOFAZ were investigated using Network pharmacology and molecular docking. EOFAZ effect on PPARγ protein stability was measured using a cellular thermal shift assay (CETSA). Pharmacological agonists and inhibitors and gene interventions were employed for clarifying EOFAZ's potential mechanism. RESULTS EOFAZ attenuated AS progression in HFD ApoE-/- mice. This attenuation was manifested by the reduced aortic intima plaque development, increased collagen content in aortic plaques, notable improvement in lipid profiles, and decreased levels of inflammatory factors. Moreover, EOFAZ inhibited the formation of MFCs by enhancing cholesterol efflux through activiting the PPARγ-LXRα-ABCA1/G1 pathway. Interestingly, the pharmacological knockdown of PPARγ impaired the beneficial effects of EOFAZ on MFCs. Additionally, our results indicated that EOFAZ reduced the ubiquitination degradation of PPARγ, and the chemical composition of EOFAZ directly bound to the PPARγ protein, thereby increasing its stability. Finally, PPARγ knockdown mitigated the protective effects of EOFAZ on AS in HFD ApoE-/- mice. CONCLUSION These findings represent the first confirmation of EOFAZ's in vivo anti-atherosclerotic effects in ApoE-/- mice. Mechanistically, its chemical constituents can directly bind to PPARγ protein, enhancing its stability, while reducing PPARγ ubiquitination degradation, thereby inhibiting foam cell formation via activation of the PPARγ-LXRα-ABCA1/G1 pathway. Simultaneously, EOFAZ could ameliorates blood lipid metabolism and inflammatory microenvironment, thus synergistically exerting its anti-atherosclerotic effects.
Collapse
Affiliation(s)
- Sheng-Quan Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Jun Xiang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Guang-Qiong Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling-Yun Fu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yi-Ni Xu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Yan Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
| | - Xiao-Xia Hu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, China.
| | - Xiang-Chun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Yunmanhu Campus, Guizhou Medical University, Guian New District, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025,China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China; The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
38
|
Zhang Z, Zou Y, Song C, Cao K, Cai K, Chen S, Wu Y, Geng D, Sun G, Zhang N, Zhang X, Zhang Y, Sun Y, Zhang Y. Advances in the study of exosomes in cardiovascular diseases. J Adv Res 2023:S2090-1232(23)00402-2. [PMID: 38123019 DOI: 10.1016/j.jare.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Cardiovascular disease (CVD) has been the leading cause of death worldwide for many years. In recent years, exosomes have gained extensive attention in the cardiovascular system due to their excellent biocompatibility. Studies have extensively researched miRNAs in exosomes and found that they play critical roles in various physiological and pathological processes in the cardiovascular system. These processes include promoting or inhibiting inflammatory responses, promoting angiogenesis, participating in cell proliferation and migration, and promoting pathological progression such as fibrosis. AIM OF REVIEW This systematic review examines the role of exosomes in various cardiovascular diseases such as atherosclerosis, myocardial infarction, ischemia-reperfusion injury, heart failure and cardiomyopathy. It also presents the latest treatment and prevention methods utilizing exosomes. The study aims to provide new insights and approaches for preventing and treating cardiovascular diseases by exploring the relationship between exosomes and these conditions. Furthermore, the review emphasizes the potential clinical use of exosomes as biomarkers for diagnosing cardiovascular diseases. KEY SCIENTIFIC CONCEPTS OF REVIEW Exosomes are nanoscale vesicles surrounded by lipid bilayers that are secreted by most cells in the body. They are heterogeneous, varying in size and composition, with a diameter typically ranging from 40 to 160 nm. Exosomes serve as a means of information communication between cells, carrying various biologically active substances, including lipids, proteins, and small RNAs such as miRNAs and lncRNAs. As a result, they participate in both physiological and pathological processes within the body.
Collapse
Affiliation(s)
- Zhaobo Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Kexin Cai
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Guozhe Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine, China Medical University, National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Xingang Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning Province, People's Republic of China; Institute of Health Sciences, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
39
|
Cheng T, You Y, Jia B, Wang H, Lv M, Zhu X, Hu Y. Knowledge mapping of B cell and atherosclerosis over the past 20 years: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2277567. [PMID: 37953301 PMCID: PMC10760366 DOI: 10.1080/21645515.2023.2277567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023] Open
Abstract
Atherosclerosis (AS) is the main underlying cause of cardiovascular disease, and B cells are considered a key immune cell type to regulate AS. So far, there is no bibliometric study on B cell and AS. This study aims to comprehensively analyze the scientific output about B cell and AS, summarize the literature characteristics, explore research hotspots, and point out emerging trends. We searched the literature from 2003 to 2022 from the Web of Science Core Collection (WoSCC) database. CiteSpace, VOSviewer, and the R package "Bibliometrix" were used for literature analysis and visualization. A total of 1,062 articles and reviews were identified. The number of annual publications generally showed an upward trend. The United States and China were the most productive countries. Medical University of Vienna was the most productive research institution, and Binder Christoph J. was the most productive author, who was also from Medical University of Vienna. "Arteriosclerosis Thrombosis and Vascular Biology" was the most published journal and the most frequently cited journal. The most cited reference was written by Caligiuri G (2002) in "Journal of Clinical Investigation." The most frequent keywords were "inflammation," "macrophages," "cardiovascular disease," "T cells," "apoptosis," "immunity," "cytokines," "lymphocytes," etc. The trend topics were mainly focused on "immune infiltration," "immunoglobulins," and "biomarkers." The complex role of B cell subtypes and a variety of B cell mediators is the main research direction at present. In-depth analysis of B cell-specific targets can provide new ideas and methods for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yaping You
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Bochao Jia
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Wang
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Meng Lv
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xueping Zhu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
40
|
Ridker PM. Targeting residual inflammatory risk: The next frontier for atherosclerosis treatment and prevention. Vascul Pharmacol 2023; 153:107238. [PMID: 37871757 DOI: 10.1016/j.vph.2023.107238] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
Inflammation and hyperlipidemia act synergistically to drive atherosclerotic progression. Multiple randomized trials now demonstrate that "lower is better" not only for LDLC, but also for hsCRP. Recent data among statin treated patients indicates that residual inflammatory risk is a stronger determinant of recurrent events than residual cholesterol risk. Based on trial data demonstrating a 31% reduction in events with minimal side effects, low-dose colchicine (0.5 mg daily) has been approved by the United States Food and Drug Administration to lower rates of myocardial infarction, stroke, and cardiovascular death as an adjunct to statin therapy. Physicians can anticipate novel anti-inflammatory agents in the future.
Collapse
Affiliation(s)
- Paul M Ridker
- From the Center for Cardiovascular Disease Prevention, Division of Preventive Medicine and the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
41
|
Pink C, Holtfreter B, Völzke H, Nauck M, Dörr M, Kocher T. Periodontitis and systemic inflammation as independent and interacting risk factors for mortality: evidence from a prospective cohort study. BMC Med 2023; 21:430. [PMID: 37953258 PMCID: PMC10642059 DOI: 10.1186/s12916-023-03139-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Recent studies have highlighted the role of low-grade systemic inflammation in linking periodontitis to cardiovascular disease (CVD) outcomes, but many aspects remain unclear. This study examines the independent and reciprocal associations of periodontitis and low-grade systemic inflammation with all-cause and CVD mortality in a large-scale cohort. METHODS A total of 3047 participants from the prospective, population-based Study of Health in Pomerania (SHIP-START) were followed for a period of 13.0 ± 2.4 years. For the association between various inflammation/periodontitis measures and mortality, hazard ratios (HRs) were obtained from covariate-adjusted Cox proportional hazards models. Interactions were analysed in joint models: on the multiplicative scale, HRs were reported and on the additive scale, relative excess risks due to interaction (RERI) were calculated. Subject and variable-specific interval records were used to account for time-varying exposures and covariates. RESULTS During the observation period, 380 (12.5%) individuals died from CVD (n = 125) or other causes (n = 255). All markers of periodontitis and inflammation showed apparent associations with all-cause mortality (HRs per SD-increase: mean PPD: 1.068 (95% confidence interval (CI): 0.988-1.155), mean CAL: 1.205 (95% CI: 1.097-1.323), missing teeth: 1.180 (95% CI: 1.065-1.307), periodontitis score: 1.394 (95% CI: 1.202-1.616), leukocytes: 1.264 (95% CI: 1.163-1.374), fibrinogen: 1.120 (95% CI: 1.030-1.218), CRP: 1.231 (95% CI: 1.109-1.366), inflammation score: 1.358 (95% CI: 1.210-1.523)). For CVD mortality, all PPD related variables showed significant associations. Interaction modelling revealed some variation with respect to mortality type and exposure combinations. On the additive scale, RERIs for periodontitis score and inflammation score implied 18.9% and 27.8% excess mortality risk for all-cause and CVD mortality, respectively. On the multiplicative scale, the HRs for interaction were marginal. CONCLUSIONS Both periodontitis and inflammation were significantly associated with all-cause mortality and CVD mortality. On the additive scale, a substantial excess risk was observed due to the interaction of periodontitis and inflammation, suggesting that the greatest treatment benefit may be achieved in patients with both periodontitis and high systemic inflammation. As periodontal therapy has been reported to also reduce systemic inflammation, the possibility of a reduction in CVD mortality risk by anti-inflammatory treatments, including periodontal interventions, seems worthy of further investigation.
Collapse
Affiliation(s)
- Christiane Pink
- Department of Restorative Dentistry, Periodontology, Endodontology and Preventive and Pediatric Dentistry, University Medicine Greifswald, Fleischmannstr. 42, 17475, Greifswald, Germany.
- Department of Orthodontics, University Medicine Greifswald, Greifswald, Germany.
| | - Birte Holtfreter
- Department of Restorative Dentistry, Periodontology, Endodontology and Preventive and Pediatric Dentistry, University Medicine Greifswald, Fleischmannstr. 42, 17475, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, SHIP/Clinical-Epidemiological Research, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology, Endodontology and Preventive and Pediatric Dentistry, University Medicine Greifswald, Fleischmannstr. 42, 17475, Greifswald, Germany
| |
Collapse
|
42
|
Skov V, Thomassen M, Kjaer L, Larsen MK, Knudsen TA, Ellervik C, Kruse TA, Hasselbalch HC. Whole blood transcriptional profiling reveals highly deregulated atherosclerosis genes in Philadelphia-chromosome negative myeloproliferative neoplasms. Eur J Haematol 2023; 111:805-814. [PMID: 37640394 DOI: 10.1111/ejh.14081] [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: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND The Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are associated with a huge comorbidity burden, including an increased risk of cardiovascular diseases. Recently, chronic inflammation has been suggested to be the driving force for clonal evolution and disease progression in MPN but also potentially having an impact upon the development of accelerated (premature) atherosclerosis. OBJECTIVES Since chronic inflammation, atherosclerosis, and atherothrombosis are prevalent in MPNs and we have previously shown oxidative stress genes to be markedly upregulated in MPNs, we hypothesized that genes linked to development of atherosclerosis might be highly deregulated as well. METHODS Using whole blood gene expression profiling in patients with essential thrombocythemia (ET; n = 19), polycythemia vera (PV; n = 41), or primary myelofibrosis (PMF; n = 9), we herein for the first time report aberrant expression of several atherosclerosis genes. RESULTS Of 84 atherosclerosis genes, 45, 56, and 46 genes were deregulated in patients with ET, PV, or PMF, respectively. Furthermore, BCL2L1, MMP1, PDGFA, PTGS1, and THBS4 were progressively significantly upregulated and BCL2 progressively significantly downregulated from ET over PV to PMF (all FDR <0.05). CONCLUSIONS We have for the first time shown massive deregulation of atherosclerosis genes in MPNs, likely reflecting the inflammatory state in MPNs in association with in vivo activation of leukocytes, platelets, and endothelial cells being deeply involved in the atherosclerotic process.
Collapse
Affiliation(s)
- Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Lasse Kjaer
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | | | - Trine A Knudsen
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Torben A Kruse
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|
43
|
Charles-Schoeman C, Choy E, McInnes IB, Mysler E, Nash P, Yamaoka K, Lippe R, Khan N, Shmagel AK, Palac H, Suboticki J, Curtis JR. MACE and VTE across upadacitinib clinical trial programmes in rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis. RMD Open 2023; 9:e003392. [PMID: 37945286 PMCID: PMC10649869 DOI: 10.1136/rmdopen-2023-003392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/26/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVES To provide an integrated analysis of major adverse cardiovascular events (MACEs) and events of venous thromboembolism (VTE) and associated risk factors across rheumatoid arthritis (RA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS) phase 2b/3 upadacitinib clinical programmes. METHODS Data were analysed and summarised from clinical trials of RA, PsA and AS treated with upadacitinib 15 mg once daily (QD) and 30 mg QD (as of 30 June 2021). Data from adalimumab (RA and PsA) and methotrexate (RA) arms were included as comparators. Adjudicated MACEs and VTE events were presented as exposure-adjusted rates per 100 patient-years (E/100 PY). Univariable Cox proportional hazard regression analyses assessed potential associations of risk factors for MACE and VTE. RESULTS In total, 4298 patients received upadacitinib 15 mg (RA n=3209, PsA n=907 and AS n=182) and 2125 patients received upadacitinib 30 mg (RA n=1204 and PsA n=921). In patients with RA and PsA, rates of MACE (0.3-0.6 E/100 PY) and VTE (0.2-0.4 E/100 PY) were similar across upadacitinib doses; in patients with AS, no MACEs and one VTE event occurred. Most patients experiencing MACEs or VTE events had two or more baseline cardiovascular risk factors. Across RA and PsA groups, rates of MACEs and VTE events were similar. CONCLUSIONS Rates of MACEs and VTE events with upadacitinib were consistent with previously reported data for patients receiving conventional synthetic and biologic disease-modifying anti-rheumatic drugs and comparable with active comparators adalimumab and methotrexate. Associated patient characteristics are known risk factors for MACEs and VTE events. TRIAL REGISTRATION NUMBERS RA (SELECT-NEXT: NCT02675426; SELECT-MONOTHERAPY: NCT02706951; SELECT-BEYOND: NCT02706847; SELECT-COMPARE: NCT02629159; SELECT-EARLY: NCT02706873, SELECT-CHOICE: NCT03086343), PsA (SELECT-PsA 2: NCT03104374; SELECT-PsA 1: NCT03104400), and AS (SELECT-AXIS 1: NCT03178487).
Collapse
Affiliation(s)
| | - Ernest Choy
- Division of Infection and Immunity, CREATE Centre, Cardiff University, Cardiff, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Eduardo Mysler
- Department of Rheumatology, OMI (Medical Research Organization), Buenos Aires, Argentina
| | - Peter Nash
- Department of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Kunihiro Yamaoka
- Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ralph Lippe
- AbbVie Deutschland GmbH & Co KG, Wiesbaden, Germany
| | | | | | | | | | - Jeffrey R Curtis
- Department of Rheumatology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
44
|
Song M, Zhang G, Shi H, Zhu E, Deng L, Shen H. Serum YKL-40 in coronary heart disease: linkage with inflammatory cytokines, artery stenosis, and optimal cut-off value for estimating major adverse cardiovascular events. Front Cardiovasc Med 2023; 10:1242339. [PMID: 38028459 PMCID: PMC10644235 DOI: 10.3389/fcvm.2023.1242339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Objective YKL-40, previously known as chitinase-3-like protein 1 (CHI3L1), is an inflammation-related glycoprotein that promotes atherosclerosis, but its application and optimal cut-off value as a prognostic biomarker in coronary heart disease (CHD) require more clinical evidence. Thus, this prospective study aimed to evaluate the linkage of serum YKL-40 with disease features, inflammatory cytokines, and major adverse cardiovascular events (MACEs) in CHD patients. Methods A total of 410 CHD patients were enrolled for serum YKL-40 determination via enzyme-linked immunosorbent assay. Meanwhile, serum YKL-40 levels in 100 healthy controls (HCs) were also quantified. Results YKL-40 level was higher in CHD patients compared with that in HCs (P < 0.001). YKL-40 was positively linked with hyperlipidemia (P = 0.014), diabetes mellitus (P = 0.001), fasting blood glucose (P = 0.045), C-reactive protein (P < 0.001), the Gensini score (P < 0.001), and stenosis degree (graded by the Gensini score) (P < 0.001) in CHD patients. In addition, an elevated YKL-40 level was associated with increased levels of tumor necrosis factor alpha (P = 0.001), interleukin (IL)-1β (P = 0.001), IL-6 (P < 0.001), and IL-17A (P = 0.002) in CHD patients. The 1-/2-/3-year cumulative MACE rates of CHD patients were 5.5%, 14.4%, and 25.0%, respectively. Regarding the prognostic capability, YKL-40 ≥100 ng/ml (the median cut-off value) (P = 0.003) and YKL-40 ≥150 ng/ml (the third interquartile cut-off value) (P = 0.021) reflected an elevated accumulating MACE rate, whereas accumulating MACE was not different between CHD patients with YKL-40 ≥80 and <80 ng/ml (the first interquartile cut-off value) (P = 0.083). Conclusion Serum YKL-40 is positively linked with inflammatory cytokines and the Gensini score, whose high expression cut-off by 100 and 150 ng/ml estimates a higher MACE risk in CHD patients.
Collapse
Affiliation(s)
- Mowei Song
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guofu Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hang Shi
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Erjun Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Li Deng
- Department of Extracorporeal Life Support, The People’s Hospital of Gaozhou, Gaozhou, China
| | - Hongtao Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
Rentz T, Dorighello GG, dos Santos RR, Barreto LM, Freitas IN, Lazaro CM, Razolli DS, Cazita PM, Oliveira HCF. CETP Expression in Bone-Marrow-Derived Cells Reduces the Inflammatory Features of Atherosclerosis in Hypercholesterolemic Mice. Biomolecules 2023; 13:1556. [PMID: 37892238 PMCID: PMC10605246 DOI: 10.3390/biom13101556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
CETP activity reduces plasma HDL-cholesterol concentrations, a correlate of an increased risk of atherosclerotic events. However, our recent findings suggest that CETP expression in macrophages promotes an intracellular antioxidant state, reduces free cholesterol accumulation and phagocytosis, and attenuates pro-inflammatory gene expression. To determine whether CETP expression in macrophages affects atherosclerosis development, we transplanted bone marrow from transgenic mice expressing simian CETP or non-expressing littermates into hypercholesterolemic LDL-receptor-deficient mice. The CETP expression did not change the lipid-stained lesion areas but decreased the macrophage content (CD68), neutrophil accumulation (LY6G), and TNF-α aorta content of young male transplanted mice and decreased LY6G, TNF-α, iNOS, and nitrotyrosine (3-NT) in aged female transplanted mice. These findings suggest that CETP expression in bone-marrow-derived cells reduces the inflammatory features of atherosclerosis. These novel mechanistic observations may help to explain the failure of CETP inhibitors in reducing atherosclerotic events in humans.
Collapse
Affiliation(s)
- Thiago Rentz
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas 13083-862, SP, Brazil; (T.R.); (G.G.D.); (L.M.B.); (I.N.F.); (C.M.L.)
| | - Gabriel G. Dorighello
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas 13083-862, SP, Brazil; (T.R.); (G.G.D.); (L.M.B.); (I.N.F.); (C.M.L.)
| | - Renata R. dos Santos
- Division of Radiotherapy, Medical School Hospital, Faculty of Medical Sciences, State University of Campinas, Campinas 13083-887, SP, Brazil;
| | - Lohanna M. Barreto
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas 13083-862, SP, Brazil; (T.R.); (G.G.D.); (L.M.B.); (I.N.F.); (C.M.L.)
| | - Israelle N. Freitas
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas 13083-862, SP, Brazil; (T.R.); (G.G.D.); (L.M.B.); (I.N.F.); (C.M.L.)
| | - Carolina M. Lazaro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas 13083-862, SP, Brazil; (T.R.); (G.G.D.); (L.M.B.); (I.N.F.); (C.M.L.)
| | - Daniela S. Razolli
- Obesity and Comorbidities Research Center, State University of Campinas, Campinas 13083-864, SP, Brazil;
| | - Patricia M. Cazita
- Laboratório de Lípides (LIM10), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 01246-903, SP, Brazil;
| | - Helena C. F. Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas 13083-862, SP, Brazil; (T.R.); (G.G.D.); (L.M.B.); (I.N.F.); (C.M.L.)
- Obesity and Comorbidities Research Center, State University of Campinas, Campinas 13083-864, SP, Brazil;
| |
Collapse
|
46
|
Traughber CA, Timinski K, Prince A, Bhandari N, Neupane K, Khan MR, Opoku E, Opoku E, Brubaker G, Nageshwar K, Ertugral EG, Naggareddy P, Kothapalli CR, Smith JD, Gulshan K. Disulfiram reduces atherosclerosis and enhances efferocytosis, autophagy, and atheroprotective gut microbiota in hyperlipidemic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562757. [PMID: 37905037 PMCID: PMC10614849 DOI: 10.1101/2023.10.17.562757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Pyroptosis executor Gasdermin (GsdmD) promotes atherosclerosis in mice and humans. Disulfiram (DSF) was recently shown to potently inhibit GsdmD, but the in-vivo efficacy and mechanism of DSF's anti-atherosclerotic activity is yet to be explored. We used human/mouse macrophages and a hyperlipidemic mouse model of atherosclerosis to determine DSF anti-atherosclerotic efficacy and mechanism. DSF-fed hyperlipidemic apoE -/- mice showed significantly reduced IL-1β release upon in-vivo Nlrp3 inflammasome assembly and showed smaller atherosclerotic lesions (∼27% and 29% reduction in males and females, respectively). The necrotic core area was also smaller (∼50% and 46% reduction in DSF-fed males and females, respectively). DSF induced autophagy in macrophages, hepatocytes/liver, and in atherosclerotic plaques. DSF modulated other atheroprotective pathways such as efferocytosis, phagocytosis, and gut microbiota. DSF-treated macrophages showed enhanced phagocytosis/efferocytosis, with a mechanism being a marked increase in cell-surface expression of efferocytic receptor MerTK. Atomic-force microscopy analysis revealed altered biophysical membrane properties of DSF treated macrophages, showing increased ordered-state of the plasma membrane and increased adhesion strength. Furthermore, the 16sRNA sequencing of DSF-fed hyperlipidemic mice showed highly significant enrichment in atheroprotective gut microbiota Akkermansia and a reduction in atherogenic Romboutsia species. Taken together, our data shows that DSF can simultaneously modulate multiple atheroprotective pathways, and thus may serve as novel adjuvant therapeutic to treat atherosclerosis.
Collapse
|
47
|
Catană MG, Popențiu IA, Văleanu M, Roman-Filip C, Mihăilă RG. IL-1 Beta-A Biomarker for Ischemic Stroke Prognosis and Atherosclerotic Lesions of the Internal Carotid Artery. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1790. [PMID: 37893508 PMCID: PMC10608497 DOI: 10.3390/medicina59101790] [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: 08/26/2023] [Revised: 09/26/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Stroke is a leading cause of mortality and morbidity worldwide. Treatment of this pathology is still under development and its risk factors remain to be determined. Therefore, we aim to determine the role of interleukin-1 beta in atherosclerotic lesions of the internal carotid artery as a risk factor for stroke and the role of this biomarker in stroke prognosis. Materials and Methods: This study enrolled 56 patients diagnosed with ischemic stroke in the anterior vascular territory (AVT) and posterior vascular territory (PVT). All the patients had venous blood collected at admission and 7 days after the onset of the cerebral ischemia in order to determine the plasma concentration of interleukin-1 beta. At the same time, an extracranial carotid ultrasound was performed. Results: The interleukin-1 beta collected at admission was positively correlated with the NIHSS at admission (Pearson index 0.424), and both measurements were correlated with carotid stenosis (Spearmen correlation index of 0.529 and 0.653, respectively). Conclusions: Interleukin-1 beta could be a reliable biomarker for stroke prognosis and the development of atherosclerotic lesions of the internal carotid.
Collapse
Affiliation(s)
- Maria-Gabriela Catană
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Neurology Department, Emergency County Clinical Hospital Sibiu, Corneliu Coposu bvd, 550245 Sibiu, Romania
| | - Ioan-Adrian Popențiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Department of General Surgery, “Alexandru Augustin” Military Emergency Hospital, Victoriei bvd, 550024 Sibiu, Romania
| | - Mădălina Văleanu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj-Napoca, 7 Horea Street, 400174 Cluj-Napoca, Romania
| | - Corina Roman-Filip
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Neurology Department, Emergency County Clinical Hospital Sibiu, Corneliu Coposu bvd, 550245 Sibiu, Romania
| | - Romeo-Gabriel Mihăilă
- Faculty of Medicine, Lucian Blaga University of Sibiu, Izvorului Street, 550169 Sibiu, Romania
- Hematology Department, Emergency County Clinical Hospital Sibiu, Sibiu Corneliu Coposu bvd, 550245 Sibiu, Romania
| |
Collapse
|
48
|
Lu LQ, Li NS, Li MR, Peng JY, Tang LJ, Luo XJ, Peng J. DL-3-n-butylphthalide improves the endothelium-dependent vasodilation in high-fat diet-fed ApoE -/- mice via suppressing inflammation, endothelial necroptosis and apoptosis. Eur J Pharmacol 2023; 956:175938. [PMID: 37536623 DOI: 10.1016/j.ejphar.2023.175938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/03/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
Impaired endothelium-dependent vasodilation in atherosclerosis is a high-risk factor for myocardial infarction and ischemic stroke, and inflammation, necroptosis and apoptosis contribute to endothelial dysfunction in atherosclerosis. Although DL-3-n-butylphthalide (NBP) has been widely used in treating ischemic stroke, its effect on endothelium-dependent vasodilation remains unknown. This study aims to explore whether NBP is able to improve endothelium-dependent vasodilation in atherosclerosis and the underlying mechanisms. Male ApoE-/- mice were fed with a high-fat diet (HFD) for 9-16 weeks to establish a model of atherosclerosis. NBP were given to the mice after eating HFD for 6 weeks and atorvastatin served as a positive control. The endothelium-dependent vasodilation, the blood flow velocity, the atherosclerotic lesion area, the serum levels of lipids, inflammatory cytokines and necroptosis-relevant proteins (RIPK1, RIPK3 and MLKL), and the endothelial necroptosis and apoptosis within the aorta were measured. Human umbilical vein endothelial cells (HUVECs) were incubated with oxidized low-density lipoprotein (ox-LDL) for 48 h to mimic endothelial injury in atherosclerosis, lactate dehydrogenase release, the ratio of necroptosis and apoptosis and the expression of necroptosis-relevant proteins were examined. Similar to atorvastatin, NBP improves endothelium-dependent vasodilation, decreases aortic flow velocity and reduces atherosclerotic lesion area in HFD-fed ApoE-/- mice, concomitant with a reduction in serum lipids, inflammatory cytokines and necroptosis-relevant proteins, and endothelial necroptosis and apoptosis. Consistently, NBP inhibited necroptosis and apoptosis in ox-LDL-treated HUVECs. Based on these observations, we conclude that NBP exerts beneficial effects on improving the endothelium-dependent vasodilation in atherosclerosis via suppressing inflammation, endothelial necroptosis and apoptosis.
Collapse
Affiliation(s)
- Li-Qun Lu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Nian-Sheng Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jiao-Yang Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Li-Jing Tang
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
49
|
Zhang P, Zhang Z, Li D, Han R, Li H, Ma J, Xu P, Qi Z, Liu L, Zhang A. Association of remnant cholesterol with intracranial atherosclerosis in community-based population: The ARIC study. J Stroke Cerebrovasc Dis 2023; 32:107293. [PMID: 37604080 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107293] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
OBJECTIVE To evaluate the association between remnant cholesterol (remnant-C) and intracranial atherosclerotic disease (ICAD) in the Atherosclerosis Risk in Communities Neurocognitive Study (ARIC-NCS). METHODS We studied 1,564 participants with data on lipid profiles and high-resolution vessel wall MRI (VWMRI) from the ARIC-NCS. Remnant-C was computed as total cholesterol minus high-density lipoprotein cholesterol minus low-density lipoprotein cholesterol (LDL-C). The primary outcomes were the presence of intracranial plaques and luminal stenosis. Contributors were separated into four different groups based on remnant-C (22 mg/dL) and LDL-C (100 mg/dL) levels to investigate the function of remnant-C vs. LDL-C on ICAD. Multivariable logistic regression models were utilized to estimate the correlation among the discordant/concordant remnant-C and LDL-C, and ICAD. RESULTS A total of 1,564 participants were included (age 76.2 ± 5.3). After multivariable adjustment, log remnant-C was correlated with greater ICAD risk [odds ratio (OR) 1.36, 95% confidence interval (CI) 1.01 to 1.83]. The lower remnant-C/higher LDL-C group and the higher remnant-C/lower LDL-C group manifested a 1.53-fold (95% CI 1.06 to 2.20) and 1.52-fold (95% CI 1.08 to 2.14) greater risk of ICAD, relative to those having lower remnant-C/low LDL-C. Additionally, remnant-C ≥ 22 mg/dL distinguished participants at a greater risk of the presence of any stenosis compared to those at lower levels, even in participants with optimal levels of LDL-C. CONCLUSIONS Elevated levels of remnant-C were connected to ICAD independent of LDL-C and traditional risk factors. The mechanisms of remnant-C association with ICAD probably offer insight into preventive risk-factor of ischemic stroke.
Collapse
Affiliation(s)
- Peng Zhang
- Clinical Medical College, Jining Medical University, Jining, China
| | - Ziheng Zhang
- Clinical Medical College, Jining Medical University, Jining, China
| | - Daojing Li
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China
| | - Rongrong Han
- Clinical Medical College, Jining Medical University, Jining, China
| | - Hongfang Li
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China
| | - Jinfeng Ma
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China
| | - Peng Xu
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China
| | - Ziyou Qi
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China
| | - Lixia Liu
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China
| | - Aimei Zhang
- Department of Neurology, the Affiliated Hospital of Jining Medical University, Jining, China.
| |
Collapse
|
50
|
Shakya K, Ahirwar D, Nabeel PM, Roy Chowdhury S. Carotid hemodynamic response to external pressure and comparison with induced-stenosis progression: a fluid-structure interaction study. Comput Methods Biomech Biomed Engin 2023; 26:1595-1609. [PMID: 36200483 DOI: 10.1080/10255842.2022.2128785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/02/2022] [Accepted: 09/11/2022] [Indexed: 11/03/2022]
Abstract
Non-invasive stenosis detection has always been difficult. A new concept of applying external pressure over the artery was compared with stenosis growth in this computational study. When stenosis develops, the artery constricts, obstructing blood flow in that area. Under external pressure, the constricted artery behaves similarly. The current fluid-structure interaction study compares the hemodynamic parameters of a stenosed artery and an artery subjected to external pressure. Significant similarities were discovered when the velocity profile and arterial displacement for both scenarios were compared. This study can be used to characterise stenosis experimentally while remaining non-invasive.
Collapse
Affiliation(s)
- Kshitij Shakya
- School of Computing and Electrical Engineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Dalchand Ahirwar
- School of Computing and Electrical Engineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - P M Nabeel
- Healthcare Technology Innovation Centre, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Shubhajit Roy Chowdhury
- School of Computing and Electrical Engineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| |
Collapse
|