1
|
Xiaoyang C, Yijun C, Chenguang Z, Wanying D, Zijun C, Jun W, Xuegong X, Wei W, Chun L. Resibufogenin protects against atherosclerosis in ApoE -/- mice through blocking NLRP3 inflammasome assembly. J Adv Res 2025:S2090-1232(25)00272-3. [PMID: 40258472 DOI: 10.1016/j.jare.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/01/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025] Open
Abstract
INTRODUCTION Atherosclerosis (AS), a major cause of cardiovascular diseases, is characterized by lipid accumulation and chronic inflammation within arterial walls. Traditional treatments, such as statins, are often ineffective for many patients, highlighting the need for novel therapeutic strategies. OBJECTIVE This study explores the potential of Resibufogenin (RBG) as an NLRP3 inflammasome inhibitor for treating AS in ApoE-/- mice. METHODS We performed experiments encompassing cellular studies, animal model assessments, molecular simulations, and binding assays to assess RBG's impact on the NLRP3 inflammasome, inflammatory cytokine release, and foam cell formation. RESULTS RBG treatment alleviated AS in ApoE-/- mice, evidenced by reduced body weight, smaller atherosclerotic plaques, and improved serum lipid profiles. Transcriptomics and molecular biology demonstrated that RBG suppressed the expression of key inflammatory markers such as NLRP3. RBG also reduced macrophage infiltration and promoted polarization toward the anti-inflammatory M2 phenotype. Molecular docking, SPR, Pull-down studies identified a non-covalent interaction between RBG and the CYS-279 residue of NLRP3, confirming its role as a potent NLRP3 inhibitor. CONCLUSION RBG effectively inhibits NLRP3 inflammasome activation, reduces pro-inflammatory cytokine release, and decreases formation of foamy macrophages, thereby slowing the progression of AS. Although these findings highlight RBG as a promising therapeutic approach for cardiovascular diseases, further research is necessary to assess its safety and effectiveness in humans and to investigate possible synergistic effects with other treatments.
Collapse
Affiliation(s)
- Chen Xiaoyang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Prescription and Syndrome Research, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chen Yijun
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhai Chenguang
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Prescription and Syndrome Research, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Du Wanying
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Prescription and Syndrome Research, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Chen Zijun
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510080, China
| | - Wang Jun
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Prescription and Syndrome Research, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xu Xuegong
- Zhengzhou Hospital of Traditional Chinese Medicine, Zhengzhou 450007, China; Institute of Geriatric Diseases, Henan Academy of Chinese Medical Sciences, Zhengzhou 451100, China.
| | - Wang Wei
- School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Prescription and Syndrome Research, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Li Chun
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Institute of Prescription and Syndrome Research, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
2
|
Yu Z, Wang Y, Fan L, Xiang J, Wang H, Xu L, Jia R. A Lipidomic Study to Identify the Serum Lipid Markers for Obstetric Antiphospholipid Syndrome. J Proteome Res 2025; 24:491-498. [PMID: 39761265 DOI: 10.1021/acs.jproteome.4c00623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2025]
Abstract
This study used untargeted lipidomics to analyze the characteristics of lipid metabolism in the serum of women with antiphospholipid syndrome. Twenty female patients with APS and 20 healthy controls were recruited to this study. Untargeted lipidomics with liquid chromatography-tandem mass spectrometry was used to profile serum lipids. Correlation analyses were performed on the significant differential lipids. Least absolute shrinkage and selection operator (LASSO) regression and binary logistic regression analyses were performed to find possible biomarkers. Finally, receiver operating characteristic (ROC) curve analysis was used to evaluate the diagnostic performance of biomarkers. The serum lipid profile of women with APS was significantly different from that of healthy controls. After statistical analysis and database matching, 12 lipids were found to be significantly different between the two groups, with the most obvious differences being triacylglycerols and phosphatidylcholines. LASSO regression and binary logistic regression models revealed that PC (17:0/22:6) and ACar (17:3) may be serum biomarkers of APS. ROC curve analysis shows that PC (17:0/22:6) and ACar (17:3) combined gave an area under the curve of 0.865 for the diagnosis of APS. PC (17:0/22:6) and ACar (17:3) could be used as biomarkers of APS.
Collapse
Affiliation(s)
- Zhaoer Yu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361000, China
| | - Yixiao Wang
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, Jiangsu, China
| | - Lili Fan
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, Jiangsu, China
| | - Jinfeng Xiang
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Hanwei Wang
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Linjie Xu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Ruizhe Jia
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, Jiangsu, China
| |
Collapse
|
3
|
Geremew GW, Alemayehu TT, Bekalu AF, Mengistu ME, Anberbr SS, Getahun AD, Fentahun S, Tadesse G, Wassie YA. Optimal medical therapy, clinical outcome and its predictors in patients with acute coronary syndrome after discharge with secondary prevention medications at University of Gondar Comprehensive Specialized Hospital, North West Ethiopia, 2023: A retrospective follow-up study. BMC Cardiovasc Disord 2024; 24:533. [PMID: 39363265 PMCID: PMC11448055 DOI: 10.1186/s12872-024-04199-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: 07/26/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND The life-threatening diseases known as ACS (acute coronary syndrome) continue to produce considerable rates of morbidity and mortality despite breakthroughs in therapy. The study determined clinical outcome and its predictors in patients at the University of Gondar Comprehensive and Specialized Hospital (UOGCSH), North West Ethiopia. DESIGN, SETTING, AND PARTICIPANTS A retrospective cohort study design was employed at UOGCSH from January 31, 2018 to February 1, 2023. The hospital used a systematic random sampling procedure to select study participants from the medical records of patients in chronic cardiac follow-up clinics. MAIN OUTCOME MEASURES Exposures were optimal medical therapy (OMT) versus non-optimal medical therapy collected from May to August 2023. Descriptive and analytical statistics were employed to compare study groups. A binary logistic regression model was employed to identify candidate variables for further analysis. Cox's proportional hazard model and log-rank test were employed, with a P-value < 0.05 used to evaluate statistical significance. A five-year all-cause mortality after discharge estimate was displayed by using Kaplan-Meier curves. RESULTS Among 422 patients with ACS [mean age, 61.56 (SD = 9.686) years; 54.7% male], of whom only 59.2% (250) received optimal medical therapy at discharge. Age ≥ 65, atrial fibrillation, chronic kidney diseases, and cardiogenic shock were negative independent predictors of optimal medical therapy. On the other hand, male sex was independently associated with the use of optimal medical therapy. All-cause mortality occurred in 16.6% (n = 70) and major adverse cardiac events occurred in 30.8% (n = 130) of patients with a 95% CI of 0.132-0.205 and 0.264-0.355, respectively. Multivariate analyses indicated that OMT was significantly associated with reduced all-cause mortality (aHR: 0.431, 95% CI: 0.222-0.835; P = 0.013). CONCLUSION This study revealed that the use of preventive OMT in patients discharged with acute coronary syndrome was associated with a reduction in all-cause mortality. However, the use of this OMT is suboptimal.
Collapse
Affiliation(s)
- Gebremariam Wulie Geremew
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tekletsadik Tekleslassie Alemayehu
- Departement of Social and Administrative Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Abaynesh Fentahun Bekalu
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Melak Erara Mengistu
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Sisay Sitotaw Anberbr
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Adane Desta Getahun
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Setegn Fentahun
- Departement of Psychiatry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gebresilassie Tadesse
- Departement of Psychiatry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yilkal Abebaw Wassie
- Department of Medical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
4
|
Zhu Y, Guo Y, Guo P, Zhang J, He Y, Xia Y, Wei Z, Dai Y. Estrogen receptor β activation alleviates inflammatory bowel disease by suppressing NLRP3-dependent IL-1β production in macrophages via downregulation of intracellular calcium level. J Adv Res 2024:S2090-1232(24)00228-5. [PMID: 38844124 DOI: 10.1016/j.jare.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024] Open
Abstract
INTRODUCTION Although several estrogen receptor β (ERβ) agonists have been reported to alleviate IBD, the pivotal mechanism remains obscure. OBJECTIVES To examine the effects and mechanisms of ERβ activation on cytokine/chemokine networks in colitis mice. METHODS Dextran sulfate sodium salt (DSS) and trinitro-benzene-sulfonic acid (TNBS) were used to induce mouse colitis model. Multiple molecular biological methods were employed to evaluate the severity of mouse colitis and the level of cytokine and/or chemokine. RESULTS Bioinformatics analysis, ELISA and immunofluorescence results showed that the targeted cytokines and/or chemokines associated with ERβ expression and activation is IL-1β, and the anti-colitis effect of ERβ activation was significantly attenuated by the overexpression of AAV9-IL-1β. Immunofluorescence analysis indicated that ERβ activation led to most evident downregulation of IL-1β expression in colonic macrophages as compared to monocytes and neutrophils. Given the pivotal roles of NLRP3, NLRC4, and AIM2 inflammasome activation in the production of IL-1β, we examined the influence of ERβ activation on inflammasome activity. ELISA and WB results showed that ERβ activation selectively blocked the NLRP3 inflammasome assembly-mediated IL-1β secretion. The calcium-sensing receptor (CaSR) and calcium signaling play crucial roles in the assembly of the NLRP3 inflammasome. WB and immunofluorescence results showed that ERβ activation reduced intracellular CaSR expression and calcium signaling in colonic macrophages. Combination with CaSR overexpression plasmid reversed the suppressive effect of ERβ activation on NLRP3 inflammasome assembly, and counteracting the downregulation of IL-1β secretion. CONCLUSION Our research uncovers that the anti-colitis effect of ERβ activation is accomplished through the reduction of IL-1β levels in colonic tissue, achieved by specifically decreasing CaSR expression in macrophages to lower intracellular calcium levels and inhibit NLRP3 inflammasome assembly-mediated IL-1β production.
Collapse
Affiliation(s)
- Yanrong Zhu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yilei Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Pengxiang Guo
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Jing Zhang
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yue He
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Yufeng Xia
- Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| | - Yue Dai
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Long Mian Avenue, Nanjing 211198, China.
| |
Collapse
|
5
|
Fang C, Zuo K, Liu Z, Xu L, Yang X. Disordered GPR43/NLRP3 expression in peripheral leukocytes of patients with atrial fibrillation is associated with intestinal short chain fatty acids levels. Eur J Med Res 2024; 29:233. [PMID: 38622672 PMCID: PMC11017637 DOI: 10.1186/s40001-024-01825-4] [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: 01/24/2024] [Accepted: 03/31/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with circulating inflammation. Short-chain fatty acids (SCFAs) derived from gut microbiota (GM) regulate leukocyte function and inhibit the release of inflammatory cytokines, which are partly mediated by the G-protein-coupled receptor 43 (GPR43) signaling. This study aimed to investigate the expression of GPR43/NOD-like receptors family pyrin domain containing 3 (NLRP3) in leukocytes and the interaction with intestinal SCFAs levels in AF patients. METHODS Expressions of GPR43 and NLRP3 mRNA in peripheral blood leukocytes from 23 AF patients and 25 non-AF controls were detected by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Expressions of leukocyte GPR43 and NLRP3 protein were evaluated by western blot analysis. The levels of plasma IL-1β were measured by enzyme-linked immunosorbent assay (ELISA). The fecal SCFAs levels based on GC/MS metabolome of corresponding 21 controls and 14 AF patients were acquired from our published dataset. To evaluate the expression of NLRP3 and GPR43 and the release of IL-1β, human THP-1 cells were stimulated with or without SCFAs (acetate, propionate, and butyrate), lipopolysaccharide (LPS), and nigericin in vitro, respectively. RESULTS Compared to the controls, the mRNA expression in peripheral leukocytes was significantly reduced in AF patients (P = 0.011) coupled with the increase in downstream leukocyte NLRP3 mRNA expression (P = 0.007) and plasma IL-1β levels (P < 0.001), consistent with changes in GPR43 and NLRP3 protein expression. Furthermore, leukocyte GPR43 mRNA levels were positively correlated with fecal GM-derived acetic acid (P = 0.046) and negatively correlated with NLRP3 mRNA expression (P = 0.024). In contrast to the negative correlation between left atrial diameter (LAD) and GPR43 (P = 0.008), LAD was positively correlated with the leukocyte NLRP3 mRNA levels (P = 0.024). Subsequent mediation analysis showed that 68.88% of the total effect of intestinal acetic acid on AF might be mediated by leukocyte GPR43/NLRP3. The constructed GPR43-NLRP3 score might have a predictive potential for AF detection (AUC = 0.81, P < 0.001). Moreover, SCFAs treatment increased GPR43 expression and remarkably reduced LPS/nigericin-induced NLRP3 expression and IL-1β release in human THP-1 cells in vitro. CONCLUSIONS Disrupted interactions between GPR43 and NLRP3 expression in peripheral blood leukocytes, associated with reduced intestinal GM-derived SCFAs, especially acetic acid, may be involved in AF development and left atrial enlargement by enhancing circulating inflammation.
Collapse
Affiliation(s)
- Chen Fang
- Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Kun Zuo
- Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Heart Center, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China
| | - Zheng Liu
- Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Heart Center, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China.
| | - Li Xu
- Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Heart Center, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China.
| | - Xinchun Yang
- Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Heart Center, Capital Medical University, 8th Gongtinanlu Rd, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
6
|
Hou XG, Wu TT, Zheng YY, Yang HT, Yang Y, Ma YT, Xie X. The Fibrinogen-to-Albumin Ratio Is Associated with Poor Prognosis in Patients with Coronary Artery Disease: Findings from a Large Cohort. J Cardiovasc Transl Res 2023; 16:1177-1183. [PMID: 37349658 DOI: 10.1007/s12265-023-10402-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/01/2023] [Indexed: 06/24/2023]
Abstract
We aimed to evaluate the association of the fibrinogen-to-albumin ratio (FAR) with the clinical outcomes of coronary artery disease (CAD). All 14,944 patients with CAD evaluated in the present study were from a prospective cohort that recruited 15,250 patients admitted in the First Affiliated Hospital of Xinjiang Medical University between December 2016 and October 2021. The all-cause mortality (ACM) and cardiac mortality (CM) were selected as the primary endpoints. The secondary endpoints were major adverse cardiovascular events (MACEs), major adverse cardiac and cerebrovascular events (MACCEs), and non-fatal myocardial infarction (NFMI). The optimal FAR cutoff value was determined by using a receiver operating characteristic (ROC) curve analysis. Using 0.1 as the cutoff value, all the patients were divided into two groups: a low-FAR group (FAR < 0.1, n = 10,076) and a high-FAR group (FAR ≥ 0.1, n = 4918). The incidence of outcomes between the two groups was compared. The high-FAR group exhibited a higher incidence of ACM (5.3% vs. 1.9%), CM (3.9% vs. 1.4%), MACEs (9.8% vs. 6.7%), MACCEs (10.4% vs. 7.6%), and NFMI (2.3% vs. 1.3%) than the low-FAR group. To adjust the confounders, multivariate Cox regression analyses showed that the risk in the high-FAR group was increased 2.182 fold in ACM (HR = 2.182, 95% CI: 1.761 ~ 2.704, P < 0.001), 2.116 fold in CM (HR = 2.116, 95% CI: 1.761 ~ 2.704, P < 0.001), 1.327 fold in MACEs (HR = 1.327, 95% CI: 1.166 ~ 1.510, P < 0.001), 1.280 fold in MACCEs (HR = 1.280, 95% CI: 1.131 ~ 1.448, P < 0.001), and 1.791 fold in NFMI (HR = 1.791, 95% CI:1.331 ~ 2.411, P < 0.001), compared to the low-FAR group. The present study suggested that the high-FAR group was an independent and powerful predictor of adverse outcomes in CAD patients.
Collapse
Affiliation(s)
- Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Hai-Tao Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China.
| |
Collapse
|
7
|
Zhao L, Kong X, Zhao J, Zhou J, Zhang S, Xu D, Tian X, Zeng X, Zhang F. Identifying risk and prognostic factors in polyarteritis nodosa patients with digital gangrene. Int J Rheum Dis 2023; 26:236-241. [PMID: 36261880 DOI: 10.1111/1756-185x.14467] [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: 04/21/2022] [Revised: 08/19/2022] [Accepted: 09/19/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Many patients with polyarteritis nodosa (PAN) complicated by digital gangrene have poor outcomes and related research information is limited. Our aim is to identify the associated risk and prognostic factors in PAN patients with digital gangrene. PATIENTS AND METHODS We conducted a retrospective study of 148 PAN patients admitted to Peking Union Medical College Hospital from Octorber 2001 to December 2018. Forty-seven (31.8%) PAN patients had digital gangrene. The average age was 40.4 ± 17.9 years. RESULTS The presence of digital gangrene was correlated with current smoking (P = .008, odds ratio [OR] 2.99, 95% CI, 1.33-6.73), eosinophil elevation (P = .003, OR 4.21, 95% CI, 1.62-10.91) and elevated leukocytes (P = .001, OR 4.26, 95% CI, 1.86-9.78). Thirty-two (68.1%) gangrene patients received methylprednisolone pulse therapy and all of these patients were treated with cyclophosphamide. Nine patients suffered irreversible organ injury and 2 died. Survival analysis showed higher serum C-reactive protein (CRP) was associated with poor prognosis in patients with gangrene (log-rank P = 0.042 and generalized Wilcoxon P = .020). CONCLUSIONS PAN patients with current smoking and eosinophil elevation were more prone to digital gangrene and a high serum CRP level predicted poor outcomes. The CRP level should be efficiently controlled to ensure a good prognosis.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China.,The Department of Rheumatology, 2nd Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaodan Kong
- The Department of Rheumatology, 2nd Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiuliang Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Shangzhu Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Dong Xu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| |
Collapse
|
8
|
Moore J, Lakshmanan S, Manubolu VS, Kinninger A, Stojan G, Goldman DW, Petri M, Budoff M, Karpouzas GA. Coronary plaque progression is greater in systemic lupus erythematosus than rheumatoid arthritis. Coron Artery Dis 2023; 34:52-58. [PMID: 36421035 DOI: 10.1097/mca.0000000000001205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) are associated with a high incidence of cardiovascular disease. Coronary atherosclerosis, particularly total plaque and noncalcified plaque on coronary computed tomography angiography (CCTA) has been correlated with cardiovascular events. We compared baseline coronary plaque burden and progression by serial CCTA in SLE and RA patients. METHODS We prospectively evaluated 44 patients who underwent serial CCTA examinations to quantify coronary plaque progression, 22 SLE patients, and 22 age- and sex-matched RA patients. Semiautomated plaque software was used for quantitative plaque assessment. Linear regression examined the effect of SLE diagnosis (versus RA) on annualized change in natural log-transformed total normalized atheroma volume (ln-TAV norm ) for low-attenuation, fibrofatty, fibrous, total noncalcified, densely calcified, and total plaque. RESULTS No quantitative differences for any plaque types were observed at baseline between SLE and RA patients ( P = 0.330-0.990). After adjustment for baseline plaque and cardiovascular risk factors, the increase in ln-TAV norm was higher in SLE than RA patients for fibrous [Exp-β: 0.202 (0.398), P = 0.0003], total noncalcified [Exp-β: 0.179 (0.393), P = 0.0001], and total plaque volume [Exp-β: 0.154 (0.501), P = 0.0007], but not for low-attenuation, fibrofatty, or densely calcified plaque ( P = 0.103-0.489). Patients with SLE had 80% more fibrous, 82% more noncalcified, and 85% more total plaque increase than those with RA. CONCLUSION Coronary plaque volume was similar in RA and SLE at baseline. Progression was greater in SLE, which may explain the greater cardiovascular risk in this disease. Further research to evaluate screening and management strategies for cardiovascular disease in these high-risk patients is warranted.
Collapse
Affiliation(s)
- Jeff Moore
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, California
| | - Suvasini Lakshmanan
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - Venkat Sanjay Manubolu
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - April Kinninger
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - George Stojan
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel W Goldman
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew Budoff
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| | - George A Karpouzas
- Chronic Disease Clinical Research Center, The Lundquist Institute for Biomedical Innovation at the Harbor University of California Los Angeles Medical Center, Torrance
| |
Collapse
|
9
|
There is urgent need to treat atherosclerotic cardiovascular disease risk earlier, more intensively, and with greater precision: A review of current practice and recommendations for improved effectiveness. Am J Prev Cardiol 2022; 12:100371. [PMID: 36124049 PMCID: PMC9482082 DOI: 10.1016/j.ajpc.2022.100371] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/10/2022] [Accepted: 08/05/2022] [Indexed: 12/12/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is epidemic throughout the world and is etiologic for such acute cardiovascular events as myocardial infarction, ischemic stroke, unstable angina, and death. ASCVD also impacts risk for dementia, chronic kidney disease peripheral arterial disease and mobility, impaired sexual response, and a host of other visceral impairments that adversely impact the quality and rate of progression of aging. The relationship between low-density lipoprotein cholesterol (LDL-C) and risk for ASCVD is one of the most highly established and investigated issues in the entirety of modern medicine. Elevated LDL-C is a necessary condition for atherogenesis induction. Basic scientific investigation, prospective longitudinal cohorts, and randomized clinical trials have all validated this association. Yet despite the enormous number of clinical trials which support the need for reducing the burden of atherogenic lipoprotein in blood, the percentage of high and very high-risk patients who achieve risk stratified LDL-C target reductions is low and has remained low for the last thirty years. Atherosclerosis is a preventable disease. As clinicians, the time has come for us to take primordial and primary prevention more serously. Despite a plethora of therapeutic approaches, the large majority of patients at risk for ASCVD are poorly or inadequately treated, leaving them vulnerable to disease progression, acute cardiovascular events, and poor aging due to loss of function in multiple visceral organs. Herein we discuss the need to greatly intensify efforts to reduce risk, decrease disease burden, and provide more comprehensive and earlier risk assessment to optimally prevent ASCVD and its complications. Evidence is presented to support that treatment should aim for far lower goals in cholesterol management, should take into account many more factors than commonly employed today and should begin significantly earlier in life.
Collapse
|
10
|
Correa TL, Quitete MAC, de Azevedo LT, Fraga IAC, Teixeira LC. Infarctus du myocarde, accident vasculaire cérébral et psoriasis: une revue systématique des études observationnelles. Ann Cardiol Angeiol (Paris) 2022; 72:101574. [PMID: 36529545 DOI: 10.1016/j.ancard.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022]
Abstract
Patients with psoriasis can have higher rates of hypertension, diabetes mellitus 2, and dyslipidemia. This study aimed to assess the association between psoriasis, myocardial infarction (MI), and stroke. A systematic review was carried out using PubMed/Medline, EMBASE, and LILACS databases searching for observational studies that assessed stroke and/or MI in patients diagnosed with psoriasis vulgaris compared to non-psoriatic patients. A total of 649 articles were identified but only 15 were included in the study. Although there were studies that have observed a significant positive association between psoriasis and MI or stroke, an almost equal number of studies showed no statistically significant association. The relationship between psoriasis and ischemic cardiovascular events is still controversial and it is difficult to establish psoriasis as the etiology of these events.
Collapse
Affiliation(s)
- Tulio Loyola Correa
- Faculty of Medicine, Federal University of Pelotas, Av. Duque de Caxias 250, Pelotas, RS, 96030-000, Brazil..
| | | | | | | | - Luísa Coutinho Teixeira
- Department of Dermatology, Santa Casa de Misericórdia de Belo Horizonte, Belo Horizonte, Brazil
| |
Collapse
|
11
|
Gao Y, Xie D, Wang Y, Niu L, Jiang H. Short-Chain Fatty Acids Reduce Oligodendrocyte Precursor Cells Loss by Inhibiting the Activation of Astrocytes via the SGK1/IL-6 Signalling Pathway. Neurochem Res 2022; 47:3476-3489. [PMID: 36098889 PMCID: PMC9546972 DOI: 10.1007/s11064-022-03710-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/09/2022]
Abstract
Short-chain fatty acids (SCFAs) are known to be actively involved in neurological diseases, but their roles in hypoxic-ischaemic brain injury (HIBI) are unclear. In this study, a rat model of HIBI was established, and this study measured the changes in IL-6 and NOD-like receptor thermal protein domain associated protein 3 (NLRP3), in addition to proliferation and apoptosis indicators of oligodendrocyte precursor cells (OPCs). The mechanism of action of SCFA on astrocytes was also investigated. Astrocytes were subjected to hypoxia in vitro, and OPCs were treated with IL-6. The results showed that SCFAs significantly alleviated HIBI-induced activation of astrocytes and loss of OPCs. SCFA pretreatment (1) downregulated the expression of NLRP3, IL-6, CCL2, and IP-10; (2) had no effect on the proliferation of OPCs; (3) ameliorated the abnormal expression of Bax and Bcl-2; and (4) regulated IL-6 expression via the SGK1-related pathway in astrocytes. Our findings revealed that SCFAs alleviated the loss of OPCs by regulating astrocyte activation through the SGK1/IL-6 signalling pathway.
Collapse
Affiliation(s)
- Yanmin Gao
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Department of General Practice, Kongjiang Community Health Service Center, No. 100, Yanji West Road, Yangpu District, Shanghai, 200093, China
| | - Di Xie
- Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Yang Wang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Lei Niu
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Hua Jiang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China. .,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
12
|
Zhou R, Stouffer GA, Frishman WH. Cholesterol Paradigm and Beyond in Atherosclerotic Cardiovascular Disease: Cholesterol, Sterol Regulatory Element-Binding Protein, Inflammation, and Vascular Cell Mobilization in Vasculopathy. Cardiol Rev 2022; 30:267-273. [PMID: 34224448 PMCID: PMC9351696 DOI: 10.1097/crd.0000000000000406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hypercholesterolemia is a well-established risk factor for atherosclerotic cardiovascular disease (ASCVD). How cholesterol and its carrier lipoproteins are involved in ASCVD is still under extensive investigation. Satins are thus far the best-proven class of cholesterol-lowering medications to improve the clinical outcomes of ASCVD. Statins specifically inhibit the rate-limiting enzyme 3-hydroxy-3-methylglutaryl-CoA reductase of the mevalonate pathway for cholesterol biosynthesis. The widely accepted theory is that statins inhibit the hepatic cholesterol synthesis causing upregulation of hepatocyte low-density lipoprotein (LDL) receptor; receptor-mediated LDL uptake and metabolism in the liver results in reduction of circulating LDL cholesterol, which subsequently reduces vascular deposition and retention of cholesterol or LDL in atherogenesis. Nevertheless, cholesterol biosynthesis is ubiquitous, also in extrahepatic cells including those in vascular wall, under tight regulation by sterol regulatory element-binding protein (SREBP), the master gene transcription factor governing cholesterol biosynthesis. Studies have shown that SREBP can be upregulated in vascular wall subject to injury or stent implantation. SREBP can be activated by proinflammatory and mitogenic factors in vascular cells, leading to hyperactive mevalonate pathway, which promotes vascular cell mobilization, further proinflammatory and mitogenic factor release from vascular cells, and vascular inflammation. In this article, we review the cellular cholesterol homeostasis regulation by SREBP and SREBP-mediated vascular hyperactive cholesterol biosynthesis, we term vascular hypercholesterolism, in the pathogenesis of ASCVD and vasculopathy. SREBP functions as a platform bridging cholesterol, inflammation, and vascular cell mobilization in ASCVD pathogenesis. Targeting vascular hypercholesterolism could open a new avenue in fighting against ASCVD.
Collapse
Affiliation(s)
- Ruihai Zhou
- From the Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - George A. Stouffer
- From the Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | |
Collapse
|
13
|
Analysis of Epidemiological Characteristics of New Cardiovascular Diseases in Cancer Patients with Cardiovascular Disease. JOURNAL OF ONCOLOGY 2022; 2022:5157398. [PMID: 36090898 PMCID: PMC9452938 DOI: 10.1155/2022/5157398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022]
Abstract
In cancer patients, a cardiovascular disease (CVD) is a prevalent occurrence. When a patient has both heart disease and cancer, the treatment can be complicated because treatment for one condition can have an adverse effect on the outcome of the other. A cardiovascular disease that involves heart failures, coronary artery disease (CAD), stroke, pericardial diseases, arrhythmias, and valve and vascular dysfunction is a serious worry for long-term cancer patients. Because preclinical research is limited, it is critical to comprehend the pathophysiology of CVD as a consequence of anticancerous therapies while taking into account the developing and expanding heart. As a result, in this research, we look at the epidemiological characteristics of cancer patients who also have cardiovascular illness. Low-dose chest computed tomography, cardiac CT, and cardiac magnetic resonance imaging (MRI) are used to acquire the data and perform the screening. Chemotherapeutic drugs such as anthracyclines and trastuzumab are used to treat the condition. Univariate analysis is used to examine risk factors and predict cardiovascular damage. Sensitivity, specificity, positive predictive value, negative predictive value, life expectancy, left ventricular ejection fraction (LVEF), and longitudinal strain are among the metrics examined.
Collapse
|
14
|
Lu Y, Zhang Y, Zhao X, Shang C, Xiang M, Li L, Cui X. Microbiota-derived short-chain fatty acids: Implications for cardiovascular and metabolic disease. Front Cardiovasc Med 2022; 9:900381. [PMID: 36035928 PMCID: PMC9403138 DOI: 10.3389/fcvm.2022.900381] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) have been on the rise around the globe in the past few decades despite the existing guidelines for prevention and treatment. Short-chain fatty acids (SCFAs) are the main metabolites of certain colonic anaerobic bacterial fermentation in the gastrointestinal tract and have been found to be the key metabolites in the host of CVDs. Accumulating evidence suggest that the end-products of SCFAs (including acetate, propionate, and butyrate) interact with CVDs through maintaining intestinal integrity, anti-inflammation, modulating glucolipid metabolism, blood pressure, and activating gut-brain axis. Recent advances suggest a promising way to prevent and treat CVDs by controlling SCFAs. Hence, this review tends to summarize the functional roles carried out by SCFAs that are reported in CVDs studies. This review also highlights several novel therapeutic interventions for SCFAs to prevent and treat CVDs.
Collapse
Affiliation(s)
- Yingdong Lu
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Zhang
- First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Zhao
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chang Shang
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Xiang
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Li
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Li Li,
| | - Xiangning Cui
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Xiangning Cui,
| |
Collapse
|
15
|
Shi H, Wu H, Winkler MA, Belin de Chantemèle EJ, Lee R, Kim HW, Weintraub NL. Perivascular adipose tissue in autoimmune rheumatic diseases. Pharmacol Res 2022; 182:106354. [PMID: 35842184 PMCID: PMC10184774 DOI: 10.1016/j.phrs.2022.106354] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/27/2022] [Accepted: 07/11/2022] [Indexed: 01/14/2023]
Abstract
Perivascular adipose tissue (PVAT) resides at the outermost boundary of the vascular wall, surrounding most conduit blood vessels, except for the cerebral vessels, in humans. A growing body of evidence suggests that inflammation localized within PVAT may contribute to the pathogenesis of cardiovascular disease (CVD). Patients with autoimmune rheumatic diseases (ARDs), e.g., systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriasis, etc., exhibit heightened systemic inflammation and are at increased risk for CVD. Data from clinical studies in patients with ARDs support a linkage between dysfunctional adipose tissue, and PVAT in particular, in disease pathogenesis. Here, we review the data linking PVAT to the pathogenesis of CVD in patients with ARDs, focusing on the role of novel PVAT imaging techniques in defining disease risk and responses to biological therapies.
Collapse
Affiliation(s)
- Hong Shi
- Division of Rheumatology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Hanping Wu
- Department of Radiology and Imaging, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Michael A Winkler
- Department of Radiology and Imaging, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric J Belin de Chantemèle
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Richard Lee
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ha Won Kim
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Neal L Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
16
|
Cho EB, Yeo Y, Jung JH, Jeong SM, Han K, Yang JH, Shin DW, Min JH. Acute myocardial infarction risk in multiple sclerosis and neuromyelitis optica spectrum disorder: A nationwide cohort study in South Korea. Mult Scler 2022; 28:1849-1858. [PMID: 35695204 DOI: 10.1177/13524585221096964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The link between neuromyelitis optica spectrum disorder (NMOSD) and cardiovascular disease is currently unclear. OBJECTIVE To determine the acute myocardial infarction (MI) risk in patients with MS and NMOSD. METHODS This study analyzed the Korean National Health Insurance Service database between January 2010 and December 2017. The included patients comprised 1503/1675 adults with MS/NMOSD who had not experienced ischemic heart disease or ischemic stroke at the index date. Matched controls were selected based on age, sex, and the presence of hypertension, diabetes mellitus (DM), and dyslipidemia. RESULTS The risks of developing MI were 2.61 (hazard ratio (HR), 95% confidence interval (CI) 1.73-3.95) and 1.95 (95% CI = 1.18-3.22) times higher in MS and NMOSD compared with the control populations. Patients with NMOSD had a similar MI risk compared with patients with MS, after adjusting for age, sex, income, hypertension, DM, and dyslipidemia (HR = 0.59, 95% CI = 0.34-1.02, p = 0.059). Among each patient group, the MI risk did not differ significantly with age (20-39, 40-64 or ⩾65 years), sex, or the presence of hypertension, DM, or dyslipidemia. CONCLUSION The MI risk increased in MS and NMOSD and seemed to be comparable between NMOSD and MS.
Collapse
Affiliation(s)
- Eun Bin Cho
- Department of Neurology, College of Medicine, Gyeongsang Institute of Health Science, Gyeongsang National University, Jinju, South Korea/Department of Neurology, Gyeongsang National University Changwon Hospital, Changwon, South Korea
| | - Yohwan Yeo
- Department of Family Medicine, College of Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, South Korea
| | - Jin-Hyung Jung
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Su-Min Jeong
- Department of Family Medicine & Supportive Care Center, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, South Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Jeong Hoon Yang
- Division of Cardiology, Department of Medicine, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, South Korea
| | - Dong Wook Shin
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, South Korea/Department of Clinical Research Design & Evaluation and Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea/Center for Wireless and Population Health Systems, University of California San Diego, San Diego, CA, USA
| | - Ju-Hong Min
- Department of Neurology, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, South Korea/Department of Neurology, Neuroscience Center, Samsung Medical Center, Seoul, South Korea/Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
17
|
Liu H, Yang Y, Liu Y, Cui L, Fu L, Li B. Various bioactive peptides in collagen hydrolysate from Salmo salar skin and the combined inhibitory effects on atherosclerosis in vitro and in vivo. Food Res Int 2022; 157:111281. [DOI: 10.1016/j.foodres.2022.111281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 11/04/2022]
|
18
|
Liu H, Li B. Separation and identification of collagen peptides derived from enzymatic hydrolysate of Salmo salar skin and their anti-inflammatory activity in lipopolysaccharide (LPS)-induced RAW264.7 inflammatory model. J Food Biochem 2022; 46:e14122. [PMID: 35332533 DOI: 10.1111/jfbc.14122] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/12/2022] [Accepted: 02/23/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is considered as a major risk for the pathogenesis of chronic diseases. Due to the adverse events caused by the long-term use of anti-inflammatory drugs, it is necessary to develop alternative and safe dietary supplements from natural products against inflammation. In this study, flavourzyme hydrolysate (for 0.5 hr) presented the strongest anti-inflammatory activity, which was further separated by ultrafiltration and column chromatography, followed by LC-MS/MS identification. Peptide APD, QA, KA, and WG were identified as anti-inflammatory peptides, which significantly reduced secretion of NO, IL-6, IL-1β, and TNF-α in inflammatory macrophages. Among them, peptide QA showed the best overall anti-inflammatory effect, with the IC50 value against NO production of 849.3 μM. Most of the identified anti-inflammatory peptides were stably against digestion, and they had abundant frequencies in the α (I/II) chain of Salmo salar collagen. Our findings indicated the potential of S. salar skin hydrolysates as functional food to prevent inflammation. PRACTICAL APPLICATIONS: Long-term use of anti-inflammatory drugs causes adverse events like gastrorrhagia, and it is necessary to develop alternative and safe dietary supplements from natural products against inflammation. Salmo salar skin, as a major byproduct of total fish, has not been effectively utilized during processing. In this study, novel anti-inflammatory oligopeptides with high activities were separated and identified from S. salar skin gelatin hydrolysate, which were stably against digestion, and presented a high bioavailability and abundant frequencies in collagen. Our study highlighted the added value of aquatic by-products and suggested that S. salar skin collagen hydrolysate could be used as a promising dietary supplement against inflammatory diseases.
Collapse
Affiliation(s)
- Hui Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Bo Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.,Key Laboratory of Functional Dairy, Ministry of Education, Beijing, China
| |
Collapse
|
19
|
Tektonidou MG. Cardiovascular disease risk in antiphospholipid syndrome: Thrombo-inflammation and atherothrombosis. J Autoimmun 2022; 128:102813. [PMID: 35247655 DOI: 10.1016/j.jaut.2022.102813] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/23/2022] [Accepted: 02/27/2022] [Indexed: 12/11/2022]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder characterized by the presence of antiphospholipid antibodies (aPL) (lupus anticoagulant, anticardiolipin antibodies and anti-beta2glycoprotein I (anti-β2GPI) antibodies) and a plethora of macro- and micro-vascular manifestations, affecting predominantly young adults. Cardiovascular events are the leading causes of morbidity and mortality in APS. APL-mediated thrombo-inflammation and atherothrombosis are emerging pathogenetic mechanisms of cardiovascular disease (CVD) in APS, involving endothelial cell and monocyte activation, cytokines and adhesion molecules expression, complement and neutrophils activation, neutrophil extracellular traps formation, platelet cell activation and aggregation, and subsequent thrombin generation, in parallel with an oxidized low-density lipoprotein (oxLDL)-β2GPI complex induced macrophage differentiation to foam cells. High risk aPL profile, especially the presence of lupus anticoagulant and triple aPL positivity (all three aPL subtypes), co-existence with Systemic Lupus Erythematosus (SLE), as well as traditional risk factors such as smoking, hypertension, hyperlipemia and obesity are associated with both subclinical atherosclerosis and cardiovascular events in APS. Increased awareness of CVD risk by the physicians and patients, regular assessment and strict control of traditional risk factors, and lifestyle modifications are recommended. Use of low-dose aspirin should be considered for cardiovascular prevention in asymptomatic aPL carriers or SLE patients with high-risk aPL profile. The role of older agents such as hydroxychloroquine and statins or new potential targeted treatments against immuno- and athero-thrombosis has been demonstrated by experimental and some clinical studies and needs to be further evaluated by randomized controlled studies. This review summarizes the available evidence about the pathogenetic mechanisms and prevalence of cardiovascular events and subclinical atherosclerosis, the interrelationship between traditional and disease-related CVD risk factors, and the cardiovascular risk assessment and management in APS.
Collapse
Affiliation(s)
- Maria G Tektonidou
- First Department of Propaedeutic Internal Medicine, Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| |
Collapse
|
20
|
Simões MHS, Salles BCC, Duarte SMDS, Silva MAD, Viana ALM, Moraes GDOID, Figueiredo SA, Ferreira EB, Rodrigues MR, Paula FBDA. Leaf extract of Coffea arabica L. reduces lipid peroxidation and has anti-platelet effect in a rat dyslipidemia model. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
21
|
Ding T, Li B, Su R, Su R, Wang Y, Gao C, Li X, Wang C. Elevated Th17 cells are associated with cardiovascular complications in ankylosing spondylitis. Rheumatology (Oxford) 2021; 61:3481-3490. [PMID: 34894210 DOI: 10.1093/rheumatology/keab888] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/22/2021] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE Patients with ankylosing spondylitis (AS) carry an increased burden of cardiovascular diseases (CVD), but features denoting the development of CVD in AS are unclear. This study aimed to evaluate the percentage and absolute number of lymphocytes and CD4+T cells in AS patients complicated with CVD (AS-CVD) and determine whether circulating Th17 cells are associated with the development of CVD in AS. METHOD A total of 117 AS patients (46 had CVD and 71 had no CVD) were enrolled in this retrospective study. The percentage and absolute number of lymphocytes and CD4+T cells were determined by Flow cytometry. Associations between CVD and clinical markers were analyzed using logistic regression. RESULTS The ratio of Th17/Treg cells (0.30 vs 0.19, p = 0.014) and the absolute number of Th17 cells (7.27 cells/μL vs 4.34 cells/μL, p < 0.001) was significantly elevated in AS-CVD group compared with AS-no-CVD group. Multivariate logistic regression revealed that elevated Th17 cells (OR = 1.20, p = 0.016) were associated with CVD complications in AS. Receiver operating characteristic (ROC) curves showed a contribution of Th17 cell for distinguishing AS patients with CVD, with the areas under the ROC curve (AUCs) of 0.729 (95%CI: 0.632-0.825, p < 0.001). CONCLUSION Our findings provide evidence for the association between Th17 cells and increased cardiovascular risk in AS. Th17 cells may contribute to accelerated atherogenesis and increased cardiovascular burden in AS and be valuable for early assessment and management of AS-CVD.
Collapse
Affiliation(s)
- Tingting Ding
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ronghui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanyan Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
22
|
Pan G, Yang S, Han X, Xie J, Li C, Wang X, Kou L. Therapeutic Effect of Platinum Nanoparticles on Atherosclerosis Research Model Based on Microfluidics and Determination of Injury. J Biomed Nanotechnol 2021; 17:2477-2484. [PMID: 34974870 DOI: 10.1166/jbn.2021.3210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The atherosclerosis (AS) microenvironment plays an important role in pathogenicity, including blood flow and blood pressure, high cholesterol, high blood sugar, angiotensin II, tumor necrosis factor, and the like. The AS microfluidic model was established, and the fluid shear stress and cyclic stretching were set to 5.07 Pa and 1.17 Hz to simulate normal blood flow, respectively. The effects of different biochemical environments on endothelial cells (ECs) and cardiomyocytes were analyzed. The results confirmed that different biochemical environments had significant damage to ECs and cardiomyocytes. Subsequently, the further effect of ECs on cardiomyocytes in AS microenvironment was studied, and the results proved that ECs caused further damage to cardiomyocytes under AS biochemical factors. We used Pt nanoparticles (Pt NPs) to study the anti-AS efficiency. The results showed that the addition of Pt NPs played a particular role in the AS treatment of ECs in the AS microenvironment, and the protection for myocardial cells was achieved.
Collapse
Affiliation(s)
- Guozhong Pan
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, PR China
| | - Shiwei Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 101100, PR China
| | - Xiaowan Han
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, PR China
| | - Jing Xie
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, PR China
| | - Chunyan Li
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, PR China
| | - Xian Wang
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, PR China
| | - Lanjun Kou
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 101100, PR China
| |
Collapse
|
23
|
Ma CY, Shi XY, Wu YR, Zhang Y, Yao YH, Qu HL, Zhang W, Guo YL, Xu RX, Li JJ. Berberine attenuates atherosclerotic lesions and hepatic steatosis in ApoE -/- mice by down-regulating PCSK9 via ERK1/2 pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1517. [PMID: 34790723 PMCID: PMC8576642 DOI: 10.21037/atm-20-8106] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/28/2021] [Indexed: 01/07/2023]
Abstract
Background It has been demonstrated that berberine (BBR), a kind of alkaloid derived from Chinese herbal medicine, has multiple pharmacological effects on human’s diseases including anti-atherosclerosis action. However, although the previous studies showed that the beneficial impact of BBR on atherosclerosis might be associated with proprotein convertase subtilisin/kexin type 9 (PCSK9), the exact underlying mechanism are not fully determined. The present study aimed to investigate potential mechanisms of anti-atherosclerosis by BBR using ApoE-/- mice. Methods The eight-week mice were divided into five groups: group 1 (wild type C57BL/6J mice with normal diet), group 2 (ApoE-/- mice with normal diet), group 3 [ApoE-/- mice with high-fat diet (HFD)], group 4 (ApoE-/- mice with HFD, and treatment with low dose BBR of 50 mg/kg/d), and group 5 (ApoE-/- mice with HFD, and treatment with high dose BBR of 100 mg/kg/d). After a 16-week treatment, the blood sample, aorta and liver were collected for lipid analysis, hematoxylin-eosin (HE) or oil red O staining, and Western blotting respectively. Besides, HepG2 Cells were cultured and treated with different concentrations of BBR (0, 5, 25 and 50 µg/mL) for 24 hours. Subsequently, cells were collected for real-time PCR or western blotting assays. Finally, the expression levels of PCSK9, LDL receptor (LDLR), ATP-binding cassette transporter A1 (ABCA1), ATP-binding cassette transporter G1 (ABCG1), and scavenger receptor class B type I (SR-BI) were examined. Results Fifty mg/kg/d and 100 mg/kg/d of BBR decreased total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL) cholesterol (LDL-C), and increased high-density lipoprotein cholesterol (HDL-C) level. Moreover, BBR reduced aorta atherosclerotic plaque, and ameliorated lipid deposition in ApoE-/- mice fed with HFD. Finally, in vitro study showed that BBR promoted intracellular cholesterol efflux, up-regulated LDLR and down-regulated PCSK9 expression via the ERK1/2 pathway in cultured HepG2 cells. Conclusions Data indicated that BBR significantly attenuated lipid disorder, reduced aortic plaque formation, and alleviated hepatic lipid accumulation in ApoE-/- mice fed with HFD, which was associated with down-regulation of PCSK9 through ERK1/2 pathway.
Collapse
Affiliation(s)
- Chun-Yan Ma
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Yun Shi
- Division of Endocrinology, Beijing Chaoyang Integrative Medicine Emergency Medical Center, Beijing, China
| | - Ya-Ru Wu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Zhang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Hong Yao
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui-Lin Qu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Zhang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Lin Guo
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui-Xia Xu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Jun Li
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Disease, Fu Wai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Hou J, Deng Q, Deng X, Zhong W, Liu S, Zhong Z. MicroRNA-146a-5p alleviates lipopolysaccharide-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production via the regulation of TRAF6 and IRAK1 in human umbilical vein endothelial cells (HUVECs). ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1433. [PMID: 34733985 PMCID: PMC8506750 DOI: 10.21037/atm-21-3903] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 01/02/2023]
Abstract
Background Microribonucleic acids (miRNAs) have an evident role in regulating endothelial inflammation and dysfunction, which characterizes the early stages of atherosclerosis. The NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome has been reported to contribute to the endothelial inflammatory response that promotes atherosclerosis development and progression. This study sought to investigate the effects of miR-146a-5p on lipopolysaccharide (LPS)-induced NLRP3 inflammasome injury and pro-inflammatory cytokine production in human umbilical vein endothelial cells (HUVECs). Methods HUVECs were transfected with a miR-146a-5p mimic, small-interfering RNA (siRNA) (si-TRAF6, and si-IRAK1), and were then stimulated with LPS for 24 h. The messenger (mRNA) and the protein levels of p-NF-κB/NF-κB, NLRP3, Caspase-1, pro-inflammatory cytokine [interleukin (IL)-6, IL-1β and tumor necrosis factor alpha (TNF-α)] in the HUVECs were analyzed by quantitative real-time polymerase chain reactions (PCRs) and western blot assays, respectively. The secretion of IL-6 from the cells was detected by enzyme-linked immunoassay (ELISA). Bioinformatic and dual-luciferase reporter assays were performed to identify the targets of miR-146a-5p. Results LPS promoted pro-inflammatory cytokine expression in a dose-dependent manner and significantly increased the expression levels of p-NF-κB/NF-κB p65, NLRP3, and Caspase-1. After transfection with a miR-146a-5p mimic, or si-TRAF6 or si-IRAK1, we observed that the mRNA and protein levels of NF-κB/p-NF-κB, NLRP3, Caspase-1, and pro-inflammatory cytokine in the HUVECs were all down-regulated, and the secretion of IL-6 from cells declined significantly. After transfection with a miR-146-5p mimic, the expression of TRAF6 and IRAK1 in HUVECs were both down-regulated. Dual-luciferase reporter assays confirmed that miR-146-5p directly targets the 3'-untranslated region (3'-UTR) of TRAF6 and IRAK1 to regulate their expression. Conclusions As a modulator of TRAF6 and IRAK1, miR-146a-5p negatively regulated LPS-induced NF-κB activation and the NLRP3 inflammasome signaling pathway in HUVECs. Thus, miRNA-146a-5p may serve as a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Jingyuan Hou
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China
| | - Qiaoting Deng
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Xunwei Deng
- Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China.,Guangdong Provincial Engineering and Technology Research Center for Molecular Diagnostics of Cardiovascular Diseases, Meizhou, China.,Guangdong Provincial Engineering and Technological Research Center for Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, China
| | - Wei Zhong
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
| | - Sudong Liu
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China.,Guangdong Provincial Key Laboratory of Precision Medicine and Clinical Translational Research of Hakka Population, Meizhou, China
| | - Zhixiong Zhong
- Meizhou Academy of Medical Sciences Cardiovascular Disease Research Institute, Meizhou People's Hospital, Meizhou, China
| |
Collapse
|
25
|
Martínez-Ceballos MA, Sinning Rey JC, Alzate-Granados JP, Mendoza-Pinto C, García-Carrasco M, Montes-Zabala L, Vargas-Vergara D, Munguia-Realpozo P, Etchegaray-Morales I, Rojas-Villarraga A. Coronary calcium in autoimmune diseases: A systematic literature review and meta-analysis. Atherosclerosis 2021; 335:68-76. [PMID: 34592584 DOI: 10.1016/j.atherosclerosis.2021.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/29/2021] [Accepted: 09/16/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND AIMS Autoimmune diseases (AID) share various clinical signs and symptoms and pathophysiological mechanisms including the increased risk of cardiovascular disease. The coronary artery calcium score (CACS) is potentially useful in improving the cardiovascular risk assessment. The aim of this study was to evaluate CACS in six AIDs analyzed as a group compared with controls through a systematic literature review (SLR) and meta-analysis. METHODS A literature search (Medline/OVID, Lilacs, Embase, and Cochrane/OVID) up to January 6, 2021 was made (PROSPERO CRD42020197182). Observational studies (patients with six AIDs: rheumatoid arthritis [RA], systemic lupus erythematosus [SLE], Sjögren's syndrome, systemic sclerosis, dermatopolymyositis, and antiphospholipid syndrome) compared with controls were included. CACS, reported in Agatston units, was the primary outcome in both groups. Mean differences and a random-effects model (DerSimonian and Laird) were calculated. RESULTS Nineteen articles were meta-analyzed (4568 subjects: 2142 AID and 2426 controls). Mean age was 48.1 and 44.2 years, respectively and 75.6% and 84.9% were women, respectively. Of cases, 52.9% had RA, 44.4% SLE and 2.7% had systemic sclerosis. The pooled analysis showed a higher CACS in patients with AIDs (7.42; 95% CI 1.79 to 13.05; chi2-p = 0.01) compared with controls. Meta-regression models showed that age in cases and controls reduced the difference in CACS between groups (p < 0.05), HDL had an inverse relationship (p = 0.04), and CRP levels had a directly proportional relationship with CACS in cases (p = 0.036). CONCLUSIONS The quantitative results of this meta-analysis suggest that CACS is higher in patients with AID, possibly due to chronic exposure to pro-inflammatory molecules. These results have clinical implications since the finding of highly elevated CACS in patients with AID will enable physicians and researchers to develop a risk stratification model that includes CACS as one of the screening tools for detecting coronary atherosclerosis in these patients.
Collapse
Affiliation(s)
| | - Jhoan Camilo Sinning Rey
- Department of Cardiology, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | | | - Claudia Mendoza-Pinto
- Systemic Autoimmune Diseases Research Unit, High Specialized Medical Unit, UMAE CMNMAC - CIBIOR, Mexican Social Security Institute, Puebla, Mexico; Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Mario García-Carrasco
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Lorena Montes-Zabala
- Department of Cardiology, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Diana Vargas-Vergara
- Department of Cardiology, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Pamela Munguia-Realpozo
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Ivet Etchegaray-Morales
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | | |
Collapse
|
26
|
Cheng ZB, Huang L, Xiao X, Sun JX, Zou ZK, Jiang JF, Lu C, Zhang HY, Zhang C. Irisin in atherosclerosis. Clin Chim Acta 2021; 522:158-166. [PMID: 34425103 DOI: 10.1016/j.cca.2021.08.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Irisin, a novel exercise-induced myokine, has been shown to play important roles in increasing white adipose tissue browning, regulating energy metabolism and improving insulin resistance. Growing evidence suggests a direct role for irisin in preventing atherosclerosis (AS) by inhibiting oxidative stress, improving dyslipidemia, facilitating anti-inflammation, reducing cellular damage and recovering endothelial function. In addition, some studies have noted that serum irisin levels play an essential role in cardiovascular diseases (CVDs) risk prediction, highlighting that irisin has the potential to be a useful predictive marker and therapeutic target of AS, especially in monitoring therapeutic efficacy. This review summarizes the understanding of irisin-mediated regulation in essential biological pathways and functions in atherosclerosis and prompts further exploitation of the biological properties of irisin in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Zhe-Bin Cheng
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuan Xiao
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Jia-Xiang Sun
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Zi-Kai Zou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jie-Feng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Lu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hai-Ya Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
27
|
Samuelsson I, Parodis I, Gunnarsson I, Zickert A, Hofman-Bang C, Wallén H, Svenungsson E. Myocardial infarctions, subtypes and coronary atherosclerosis in SLE: a case-control study. Lupus Sci Med 2021; 8:8/1/e000515. [PMID: 34290127 PMCID: PMC8296778 DOI: 10.1136/lupus-2021-000515] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022]
Abstract
Objective Patients with SLE have increased risk of myocardial infarction (MI). Few studies have investigated the characteristics of SLE-related MIs. We compared characteristics of and risk factors for MI between SLE patients with MI (MI-SLE), MI patients without SLE (MI-non-SLE) and SLE patients without MI (non-MI-SLE) to understand underlying mechanisms. Methods We identified patients with a first-time MI in the Karolinska SLE cohort. These patients were individually matched for age and gender with MI-non-SLE and non-MI-SLE controls in a ratio of 1:1:1. Retrospective medical file review was performed. Paired statistics were used as appropriate. Results Thirty-four MI-SLE patients (88% females) with a median age of 61 years were included. These patients had increased number of coronary arteries involved (p=0.04), and ≥50% coronary atherosclerosis/occlusion was numerically more common compared with MI-non-SLE controls (88% vs 66%; p=0.07). The left anterior descending artery was most commonly involved (73% vs 59%; p=0.11) and decreased (<50%) left ventricular ejection fraction occurred with similar frequency in MI-SLE and MI-non-SLE patients (45% vs 36%; p=0.79). Cardiovascular disease (44%, 5.9%, 12%; p<0.001) and coronary artery disease (32%, 2.9%, 0%; p<0.001), excluding MI, preceded MI/inclusion more commonly in MI-SLE than in MI-non-SLE and non-MI-SLE patients, respectively. MI-SLE patients had lower plasma albumin levels than non-MI-SLE patients (35 (29–37) vs 40 (37–42) g/L; p=0.002). Conclusion In the great majority of cases, MIs in SLE are associated with coronary atherosclerosis. Furthermore, MIs in SLE are commonly preceded by symptomatic vascular disease, calling for attentive surveillance of cardiovascular disease and its risk factors and early atheroprotective treatment.
Collapse
Affiliation(s)
- Isak Samuelsson
- Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden .,Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.,Unit of Cardiology, Danderyd Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Iva Gunnarsson
- Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Agneta Zickert
- Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| | - Claes Hofman-Bang
- Unit of Cardiology, Danderyd Hospital, Stockholm, Sweden.,Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Wallén
- Unit of Cardiology, Danderyd Hospital, Stockholm, Sweden.,Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Elisabet Svenungsson
- Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
28
|
Wu TT, Zheng YY, Xiu WJ, Wang WR, Xun YL, Ma YY, Kadir P, Pan Y, Ma YT, Xie X. White Blood Cell Counts to High-Density Lipoprotein Cholesterol Ratio, as a Novel Predictor of Long-Term Adverse Outcomes in Patients After Percutaneous Coronary Intervention: A Retrospective Cohort Study. Front Cardiovasc Med 2021; 8:616896. [PMID: 34307487 PMCID: PMC8295559 DOI: 10.3389/fcvm.2021.616896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/30/2021] [Indexed: 02/05/2023] Open
Abstract
Background: White blood cell (WBC) counts and high-density lipoprotein cholesterol (HDL-C) are widely available in clinical practice. However, the predictive value for cardiovascular disease (CVD) is uncertain. In the present study, we firstly assessed the prognostic value of WBC to HDL-C ratio (WHR) in patients with coronary artery disease (CAD) who underwent percutaneous coronary intervention (PCI). Methods: Six thousand and fifty patients with CAD after PCI from a retrospective cohort study (identifier: ChiCTR-INR-16010153) were evaluated initially. Three hundred and seventy-one patients were excluded due to HDL cholesterol data not available, malignancy, dementia, psoriasis or eczema, systemic connective tissue disorders, multiple sclerosis, chronic liver disease, and chronic obstructive pulmonary disorder. Finally, 5,679 patients were included in the study. The primary outcome was long-term mortality. Secondary endpoints were mainly major adverse cardiovascular and cerebrovascular events (MACCEs), defined as a combination of stroke, cardiac death, stent thrombosis, recurrent myocardial infarction, and target vessel revascularization. The mean follow-up time of this study was 35.9 ± 22.5 months. We defined the best cutoff value of MHR according to the receiver operating curve (ROC), and then patients were divided into high and low WHR groups according to the cutoff value. We analyzed the data in both an acute coronary syndrome group (ACS) and a stable CAD subgroup, respectively. Results: Overall, there were 293 cases of long-term mortality during the follow-up period. According to the cutoff value (WHR = 8.25), 1,901 ACS patients were divided into high WHR group (n = 724) and low WHR group (n = 1,177). Compared to low WHR group, the incidence of all-cause mortality (ACM, 5.5 vs. 3.6%, p = 0.048) and cardiac death (4.7vs. 2.9%, p = 0.042) were significantly higher in the high WHR group. In stable CAD group, we also found the incidence of ACM and cardiac death were significantly higher in the high group compared to that in the low group. We did not find significant difference between the high and the low WHR group in the incidence of MACCEs. The multivariate Cox proportional hazards model showed that increased WHR level was independently correlated with the mortality. In the high WHR group, the risk of ACM increased two times in ACS [adjusted HR = 2.036 (1.258–3.296), p = 0.004] and 1.5 times in stable CAD [adjusted HR = 1.586 (1.178–2.136), p = 0.002]. Conclusion: The present study indicated that an increased WBC count to HDL-C ratio was independently associated with long-term mortality in CAD patients who underwent PCI.
Collapse
Affiliation(s)
- Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wen-Juan Xiu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wan-Rong Wang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi-Li Xun
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yan-Yan Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Patigvl Kadir
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ying Pan
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
29
|
Tabares-Guevara JH, Jaramillo JC, Ospina-Quintero L, Piedrahíta-Ochoa CA, García-Valencia N, Bautista-Erazo DE, Caro-Gómez E, Covián C, Retamal-Díaz A, Duarte LF, González PA, Bueno SM, Riedel CA, Kalergis AM, Ramírez-Pineda JR. IL-10-Dependent Amelioration of Chronic Inflammatory Disease by Microdose Subcutaneous Delivery of a Prototypic Immunoregulatory Small Molecule. Front Immunol 2021; 12:708955. [PMID: 34305950 PMCID: PMC8297659 DOI: 10.3389/fimmu.2021.708955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
One of the interventional strategies to reestablish the immune effector/regulatory balance, that is typically altered in chronic inflammatory diseases (CID), is the reinforcement of endogenous immunomodulatory pathways as the one triggered by interleukin (IL)-10. In a recent work, we demonstrated that the subcutaneous (sc) administration of an IL-10/Treg-inducing small molecule-based formulation, using a repetitive microdose (REMID) treatment strategy to preferentially direct the effects to the regional immune system, delays the progression of atherosclerosis. Here we investigated whether the same approach using other IL-10-inducing small molecule, such as the safe, inexpensive, and widely available polyphenol curcumin, could induce a similar protective effect in two different CID models. We found that, in apolipoprotein E deficient mice, sc treatment with curcumin following the REMID strategy induced atheroprotection that was not consequence of its direct systemic lipid-modifying or antioxidant activity, but instead paralleled immunomodulatory effects, such as reduced proatherogenic IFNγ/TNFα-producing cells and increased atheroprotective FOXP3+ Tregs and IL-10-producing dendritic and B cells. Remarkably, when a similar strategy was used in the neuroinflammatory model of experimental autoimmune encephalomyelitis (EAE), significant clinical and histopathological protective effects were evidenced, and these were related to an improved effector/regulatory cytokine balance in restimulated splenocytes. The essential role of curcumin-induced IL-10 for neuroprotection was confirmed by the complete abrogation of the clinical effects in IL-10-deficient mice. Finally, the translational therapeutic prospection of this strategy was evidenced by the neuroprotection observed in mice starting the treatment one week after disease triggering. Collectively, results demonstrate the power of a simple natural IL-10-inducing small molecule to tackle chronic inflammation, when its classical systemic and direct pharmacological view is shifted towards the targeting of regional immune cells, in order to rationally harness its immunopharmacological potential. This shift implies that many well-known IL-10-inducing small molecules could be easily reformulated and repurposed to develop safe, innovative, and accessible immune-based interventions for CID.
Collapse
Affiliation(s)
- Jorge H Tabares-Guevara
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Julio C Jaramillo
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Laura Ospina-Quintero
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Christian A Piedrahíta-Ochoa
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Natalia García-Valencia
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - David E Bautista-Erazo
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Erika Caro-Gómez
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| | - Camila Covián
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angello Retamal-Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José R Ramírez-Pineda
- Grupo Inmunomodulación (GIM), Instituto de Investigaciones Médicas, Facultad de Medicina, Corporación Académica para el Estudio de Patologías Tropicales (CAEPT), Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
30
|
Smoking and Neuropsychiatric Disease-Associations and Underlying Mechanisms. Int J Mol Sci 2021; 22:ijms22147272. [PMID: 34298890 PMCID: PMC8304236 DOI: 10.3390/ijms22147272] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/28/2021] [Accepted: 07/03/2021] [Indexed: 01/02/2023] Open
Abstract
Despite extensive efforts to combat cigarette smoking/tobacco use, it still remains a leading cause of global morbidity and mortality, killing more than eight million people each year. While tobacco smoking is a major risk factor for non-communicable diseases related to the four main groups—cardiovascular disease, cancer, chronic lung disease, and diabetes—its impact on neuropsychiatric risk is rather elusive. The aim of this review article is to emphasize the importance of smoking as a potential risk factor for neuropsychiatric disease and to identify central pathophysiological mechanisms that may contribute to this relationship. There is strong evidence from epidemiological and experimental studies indicating that smoking may increase the risk of various neuropsychiatric diseases, such as dementia/cognitive decline, schizophrenia/psychosis, depression, anxiety disorder, and suicidal behavior induced by structural and functional alterations of the central nervous system, mainly centered on inflammatory and oxidative stress pathways. From a public health perspective, preventive measures and policies designed to counteract the global epidemic of smoking should necessarily include warnings and actions that address the risk of neuropsychiatric disease.
Collapse
|
31
|
The impact of antimalarial agents on traditional and non-traditional subclinical atherosclerosis biomarkers in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2021; 20:102887. [PMID: 34237422 DOI: 10.1016/j.autrev.2021.102887] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Cardiovascular (CV) morbidity is a well-established problem in systemic lupus erythematosus (SLE). Antimalarial (AM) therapy has been seen as a potential atheroprotective agent. The aim was to assess the impact of AM therapy on traditional and novel atherosclerosis (AT) biomarkers in patients with SLE. METHODS A search of MEDLINE, EMbase, and Cochrane library for studies evaluating the impact of AM on AT biomarkers in SLE was conducted. Data extraction included serum, functional and structural traditional and novel biomarkers. A narrative synthesis of the findings and a meta-analysis with random effects was conducted estimating mean differences (MD), OR, HR and 95% CIs. RESULTS The search strategy produced 148 articles, of which 64 were extracted for analysis. The MD in VLDL-cholesterol (-10.29, 95% CI -15.35, 5.24), triglycerides (-15.68, 95% CI -27.51, -3.86), and diastolic BP (-3.42, 95% CI -5.62, -1.23) differed significantly in patients on AM therapy compared with those without AM therapy. Patients on AM had a lower prevalence and incidence of diabetes mellitus than patients not on AM (HR: 0.39, 95% CI 0.17, 0.88). HCQ use was associated with lower blood pressure (BP) variability. Structural markers like carotid intima-media thickness (IMT), carotid plaque (CP) and coronary artery calcification (CAC) were not influenced by AM. For functional markers like endothelial and arterial stiffness the benefit was unclear. The GRADE approach showed a very low-to-low quality of evidence (QoE) per outcome. CONCLUSIONS There is some evidence on the associations between AM therapy and some AT markers. However, the data on which this conclusion was based was of low to very low evidence.
Collapse
|
32
|
Arterial stiffness, the hidden face of cardiovascular risk in autoimmune and chronic inflammatory rheumatic diseases. Autoimmun Rev 2021; 20:102891. [PMID: 34229047 DOI: 10.1016/j.autrev.2021.102891] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Cardiovascular diseases (CVD) are the leading causes of death in chronic inflammatory rheumatic diseases and are not solely explained by the increased prevalence of cardiovascular (CV) risk factors in this population. Arterial stiffness, assessed primarily by pulse wave velocity (PWV) and more indirectly by augmentation index (AIx), is a surrogate marker of CVD that should be considered. The objective of this review was to investigate the relationship between arterial stiffness and chronic inflammatory and/or autoimmune diseases. METHODS We performed a systemic literature review of articles published in Medline from January 2012 to April 2020 restricted to English languages and to human adults. We selected relevant articles about the relationship between arterial stiffness and rheumatoid arthritis, systemic lupus erythematosus, psoriasis, Sjogren's syndrome and ankylosing spondylitis. For each selected article, data on PWV and AIx were extracted and factors that may have an impact on arterial stiffness were identified. RESULTS A total of 214 references were identified through database searching and 82 of them were retained for analysis. Arterial stiffness is increased in chronic inflammatory and autoimmune diseases. Traditional CV risk factors such as hypertension and dyslipidemia accentuate this relationship. Current data are insufficient to determine whether disease activity significantly influences arterial stiffness, whereas disease duration seems rather critical. TNF-alpha inhibitors and cardiorespiratory fitness tend to decrease arterial stiffness. Finally, increased arterial stiffness leads to diastolic dysfunction, which is the main mechanism of heart failure in chronic inflammatory rheumatic diseases. CONCLUSION CV risk assessment in chronic inflammatory and autoimmune diseases should also rely on PWV and AIx.
Collapse
|
33
|
Zhou R, Stouffer GA, Smith SC. Targeting the Cholesterol Paradigm in the Risk Reduction for Atherosclerotic Cardiovascular Disease: Does the Mechanism of Action of Pharmacotherapy Matter for Clinical Outcomes? J Cardiovasc Pharmacol Ther 2021; 26:533-549. [PMID: 34138676 DOI: 10.1177/10742484211023632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hypercholesterolemia is a well-established risk factor for atherosclerotic cardiovascular disease (ASCVD). Low-density lipoprotein cholesterol (LDL-C) has been labeled as "bad" cholesterol and high-density lipoprotein cholesterol (HDL-C) as "good" cholesterol. The prevailing hypothesis is that lowering blood cholesterol levels, especially LDL-C, reduces vascular deposition and retention of cholesterol or apolipoprotein B (apoB)-containing lipoproteins which are atherogenic. We review herein the clinical trial data on different pharmacological approaches to lowering blood cholesterol and propose that the mechanism of action of cholesterol lowering, as well as the amplitude of cholesterol reduction, are critically important in leading to improved clinical outcomes in ASCVD. The effects of bile acid sequestrants, fibrates, niacin, cholesteryl ester transfer protein (CETP) inhibitors, apolipoprotein A-I and HDL mimetics, apoB regulators, acyl coenzyme A: cholesterol acyltransferase (ACAT) inhibitors, cholesterol absorption inhibitors, statins, and proprotein convertase subtilisin kexin 9 (PCSK9) inhibitors, among other strategies are reviewed. Clinical evidence supports that different classes of cholesterol lowering or lipoprotein regulating approaches yielded variable effects on ASCVD outcomes, especially in cardiovascular and all-cause mortality. Statins are the most widely used cholesterol lowering agents and have the best proven cardiovascular event and survival benefits. Manipulating cholesterol levels by specific targeting of apoproteins or lipoproteins has not yielded clinical benefit. Understanding why lowering LDL-C by different approaches varies in clinical outcomes of ASCVD, especially in survival benefit, may shed further light on our evolving understanding of how cholesterol and its carrier lipoproteins are involved in ASCVD and aid in developing effective pharmacological strategies to improve the clinical outcomes of ASCVD.
Collapse
Affiliation(s)
- Ruihai Zhou
- Division of Cardiology, Department of Medicine, 2332University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - George A Stouffer
- Division of Cardiology, Department of Medicine, 2332University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sidney C Smith
- Division of Cardiology, Department of Medicine, 2332University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, Weng J. Metformin, Macrophage Dysfunction and Atherosclerosis. Front Immunol 2021; 12:682853. [PMID: 34163481 PMCID: PMC8215340 DOI: 10.3389/fimmu.2021.682853] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Metformin is one of the most widely prescribed hypoglycemic drugs and has the potential to treat many diseases. More and more evidence shows that metformin can regulate the function of macrophages in atherosclerosis, including reducing the differentiation of monocytes and inhibiting the inflammation, oxidative stress, polarization, foam cell formation and apoptosis of macrophages. The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-κB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. On the basis of summarizing these studies, we further discussed the future research directions of metformin: single-cell RNA sequencing, neutrophil extracellular traps (NETs), epigenetic modification, and metformin-based combination drugs. In short, macrophages play an important role in a variety of diseases, and improving macrophage dysfunction may be an important mechanism for metformin to expand its pleiotropic pharmacological profile. In addition, the combination of metformin with other drugs that improve the function of macrophages (such as SGLT2 inhibitors, statins and IL-1β inhibitors/monoclonal antibodies) may further enhance the pleiotropic therapeutic potential of metformin in conditions such as atherosclerosis, obesity, cancer, dementia and aging.
Collapse
Affiliation(s)
- Xiaojun Feng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Wenxu Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Peter J. Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| |
Collapse
|
35
|
Townsend EM, Kelly L, Muscatt G, Box JD, Hargraves N, Lilley D, Jameson E. The Human Gut Phageome: Origins and Roles in the Human Gut Microbiome. Front Cell Infect Microbiol 2021; 11:643214. [PMID: 34150671 PMCID: PMC8213399 DOI: 10.3389/fcimb.2021.643214] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/19/2021] [Indexed: 12/14/2022] Open
Abstract
The investigation of the microbial populations of the human body, known as the microbiome, has led to a revolutionary field of science, and understanding of its impacts on human development and health. The majority of microbiome research to date has focussed on bacteria and other kingdoms of life, such as fungi. Trailing behind these is the interrogation of the gut viruses, specifically the phageome. Bacteriophages, viruses that infect bacterial hosts, are known to dictate the dynamics and diversity of bacterial populations in a number of ecosystems. However, the phageome of the human gut, while of apparent importance, remains an area of many unknowns. In this paper we discuss the role of bacteriophages within the human gut microbiome. We examine the methods used to study bacteriophage populations, how this evolved over time and what we now understand about the phageome. We review the phageome development in infancy, and factors that may influence phage populations in adult life. The role and action of the phageome is then discussed at both a biological-level, and in the broader context of human health and disease.
Collapse
Affiliation(s)
- Eleanor M Townsend
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
| | - Lucy Kelly
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
| | - George Muscatt
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
| | - Joshua D Box
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
| | - Nicole Hargraves
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
| | - Daniel Lilley
- Warwick Medical School, The University of Warwick, Coventry, United Kingdom
| | - Eleanor Jameson
- School of Life Sciences, The University of Warwick, Coventry, United Kingdom
| |
Collapse
|
36
|
Lai JH, Hung LF, Huang CY, Wu DW, Wu CH, Ho LJ. Mitochondrial protein CMPK2 regulates IFN alpha-enhanced foam cell formation, potentially contributing to premature atherosclerosis in SLE. Arthritis Res Ther 2021; 23:120. [PMID: 33874983 PMCID: PMC8054390 DOI: 10.1186/s13075-021-02470-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 03/02/2021] [Indexed: 12/22/2022] Open
Abstract
Background Premature atherosclerosis occurs in patients with SLE; however, the mechanisms remain unclear. Both mitochondrial machinery and proinflammatory cytokine interferon alpha (IFN-α) potentially contribute to atherogenic processes in SLE. Here, we explore the roles of the mitochondrial protein cytidine/uridine monophosphate kinase 2 (CMPK2) in IFN-α-mediated pro-atherogenic events. Methods Foam cell measurements were performed by oil red O staining, Dil-oxLDL uptake and the BODIPY approach. The mRNA and protein levels were measured by qPCR and Western blotting, respectively. Isolation of CD4+ T cells and monocytes was performed with monoclonal antibodies conjugated with microbeads. Manipulation of protein expression was conducted by either small interference RNA (siRNA) knockdown or CRISPR/Cas9 knockout. The expression of mitochondrial reactive oxygen species (mtROS) was determined by flow cytometry and confocal microscopy. Results IFN-α enhanced oxLDL-induced foam cell formation and Dil-oxLDL uptake by macrophages. In addition to IFN-α, several triggers of atherosclerosis, including thrombin and IFN-γ, can induce CMPK2 expression, which was elevated in CD4+ T cells and CD14+ monocytes isolated from SLE patients compared to those isolated from controls. The analysis of cellular subfractions revealed that CMPK2 was present in both mitochondrial and cytosolic fractions. IFN-α-induced CMPK2 expression was inhibited by Janus kinase (JAK)1/2 and tyrosine kinase 2 (Tyk2) inhibitors. Both the knockdown and knockout of CMPK2 attenuated IFN-α-mediated foam cell formation, which involved the reduction of scavenger receptor class A (SR-A) expression. CMPK2 also regulated IFN-α-enhanced mtROS production and inflammasome activation. Conclusions The study suggests that CMPK2 plays contributing roles in the pro-atherogenic effects of IFN-α. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02470-6.
Collapse
Affiliation(s)
- Jenn-Haung Lai
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Lin-Kou, Tao-Yuan, Taiwan, Republic of China.,Graduate Institute of Clinical Research, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Li-Feng Hung
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, Republic of China
| | - Chuan-Yueh Huang
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, Republic of China
| | - De-Wei Wu
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Lin-Kou, Tao-Yuan, Taiwan, Republic of China
| | - Chien-Hsiang Wu
- Department of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital, Lin-Kou, Tao-Yuan, Taiwan, Republic of China
| | - Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, Republic of China.
| |
Collapse
|
37
|
Gülcan HO, Orhan IE. General Perspectives for the Treatment of Atherosclerosis. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999201016154400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Atherosclerosis, a cardiovascular disease, is at the top of the list among the diseases leading
to death. Although the biochemical and pathophysiological cascades involved within the development
of atherosclerosis have been identified clearly, its nature is quite complex to be treated with
a single agent targeting a pathway. Therefore, many natural and synthetic compounds have been
suggested for the treatment of the disease. The majority of the drugs employed target one of the
single components of the pathological outcomes, resulting in many times less effective and longterm
treatments. In most cases, treatment options prevent further worsening of the symptoms rather
than a radical treatment. Consequently, the current review has been prepared to focus on the validated
and non-validated targets of atherosclerosis as well as the alternative treatment options such
as hydroxymethyl glutaryl coenzyme A (HMG-CoA) reductase inhibitors, acyl-CoA cholesterol
acyl transferase (ACAT) inhibitors, lipoprotein lipase stimulants, bile acid sequestrants, and some
antioxidants. Related to the topic, both synthetic compounds designed employing medicinal chemistry
skills and natural molecules becoming more popular in drug development are scrutinized in this
mini review.
Collapse
Affiliation(s)
- Hayrettin Ozan Gülcan
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, TR. North Cyprus, via Mersin 10,Turkey
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara- 06300,Turkey
| |
Collapse
|
38
|
The revisited role of interleukin-1 alpha and beta in autoimmune and inflammatory disorders and in comorbidities. Autoimmun Rev 2021; 20:102785. [PMID: 33621698 DOI: 10.1016/j.autrev.2021.102785] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023]
Abstract
The interleukin (IL) 1 family of cytokines is noteworthy to have pleiotropic functions in inflammation and acquired immunity. Over the last decades, several progresses have been made in understanding the function and regulation of the prototypical inflammatory cytokine (IL-1) in human diseases. IL-1α and IL-1β deregulated signaling causes devastating diseases manifested by severe acute or chronic inflammation. In this review, we examine and compare the key aspects of IL-1α and IL-1β biology and regulation and discuss their importance in the initiation and maintenance of inflammation that underlie the pathology of many human diseases. We also report the current and ongoing inhibitors of IL-1 signaling, targeting IL-1α, IL-1β, their receptor or other molecular compounds as effective strategies to prevent or treat the onset and progression of various inflammatory disorders.
Collapse
|
39
|
He X, Fan X, Bai B, Lu N, Zhang S, Zhang L. Pyroptosis is a critical immune-inflammatory response involved in atherosclerosis. Pharmacol Res 2021; 165:105447. [PMID: 33516832 DOI: 10.1016/j.phrs.2021.105447] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/28/2020] [Accepted: 01/17/2021] [Indexed: 02/07/2023]
Abstract
Pyroptosis is a form of programmed cell death activated by various stimuli and is characterized by inflammasome assembly, membrane pore formation, and the secretion of inflammatory cytokines (IL-1β and IL-18). Atherosclerosis-related risk factors, including oxidized low-density lipoprotein (ox-LDL) and cholesterol crystals, have been shown to promote pyroptosis through several mechanisms that involve ion flux, ROS, endoplasmic reticulum stress, mitochondrial dysfunction, lysosomal rupture, Golgi function, autophagy, noncoding RNAs, post-translational modifications, and the expression of related molecules. Pyroptosis of endothelial cells, macrophages, and smooth muscle cells in the vascular wall can induce plaque instability and accelerate atherosclerosis progression. In this review, we focus on the pathogenesis, influence, and therapy of pyroptosis in atherosclerosis and provide novel ideas for suppressing pyroptosis and the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xiao He
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilongjiang Province, China.
| | - Xuehui Fan
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilongjiang Province, China.
| | - Bing Bai
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilongjiang Province, China.
| | - Nanjuan Lu
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilongjiang Province, China.
| | - Shuang Zhang
- General Surgery, Harbin Changzheng Hospital, 363 Xuan Hua Street, Harbin 150001, Heilongjiang Province, China.
| | - Liming Zhang
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, 23 You Zheng Street, Harbin 150001, Heilongjiang Province, China.
| |
Collapse
|
40
|
Wafriy CI, Kamsani YS, Nor-Ashikin MNK, Nasir NAA, Hanafiah M. Ovalbumin causes impairment of preimplantation embryonic growth in asthma-induced mice. J Reprod Immunol 2020; 143:103240. [PMID: 33166807 DOI: 10.1016/j.jri.2020.103240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/04/2020] [Accepted: 10/21/2020] [Indexed: 10/23/2022]
Abstract
Insufficient experimental studies have reported the effect of ovalbumin (OVA) as an allergen towards embryonic growth in asthma mouse model. The impact of 10 μg/200 μL OVA on maternal inflammatory and oxidative stress (OS) responses, and preimplantation embryonic development was investigated in this study. We first established OVA-induced asthma mouse model, and following superovulation, mated the females and challenged them with Methacholine (Mch) test. Upon embryo retrieval, only those with the highest implantation potential were cultured in vitro. Significant reduction in the number of embryos at each preimplantation stage was noted in the treated group. Uneven sized blastomeres at 2-, 4- and 8-cell stages were also evident in this group. Embryo fragmentation was significant at only 2-, 4- and 8-cell stages. We also found that OVA tended to raise maternal inflammatory and OS biomarker levels as well as to cause inappropriate levels of pregnancy hormones progesterone (P4) and estrogen (E2) although insignificant. The combined results indicate that 10 μg/200 μL OVA had altered both quality and quantity of the embryos in asthma mouse model although its effect on pregnancy hormones, inflammatory and OS responses were non-pathological.
Collapse
Affiliation(s)
- Che Ismail Wafriy
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia.
| | - Yuhaniza Shafinie Kamsani
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia; Maternofetal and Embryo (MatE) Research Group, Universiti Teknologi Mara, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia.
| | - Mohamed Noor Khan Nor-Ashikin
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia; Maternofetal and Embryo (MatE) Research Group, Universiti Teknologi Mara, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia.
| | - Nurul Alimah Abdul Nasir
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia; Centre for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia.
| | - Mohammad Hanafiah
- Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Selangor, 47000, Malaysia; Assunta Hospital, Jalan Templer, Pjs 4, 46050 Petaling Jaya, Selangor, Malaysia.
| |
Collapse
|
41
|
Xie W, Huang H, Xiao S, Yang X, Zhang Z. Effect of statin use on cardiovascular events and all-cause mortality in immune-mediated inflammatory diseases: A systematic review and meta-analysis involving 148,722 participants. Pharmacol Res 2020; 160:105057. [DOI: 10.1016/j.phrs.2020.105057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/15/2020] [Accepted: 06/28/2020] [Indexed: 02/07/2023]
|
42
|
Castañeda S, Vicente-Rabaneda EF, Valero C, Remuzgo-Martínez S, López-Mejías R, González-Gay MA. Effect of cardiovascular disease on chronic inflammatory joint disease: reverse causality? Expert Rev Clin Immunol 2020; 16:855-858. [PMID: 32867563 DOI: 10.1080/1744666x.2020.1814146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Santos Castañeda
- Department of Rheumatology, Hospital Universitario De La Princesa, IIS-IP , Madrid, Spain.,Cátedra UAM-ROCHE, EPID-Future, Universidad Autónoma De Madrid (UAM) , Madrid, Spain
| | | | - Cristina Valero
- Department of Rheumatology, Hospital Universitario De La Princesa, IIS-IP , Madrid, Spain
| | - Sara Remuzgo-Martínez
- Division and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marqués De Valdecilla, IDIVAL , Santander, Spain
| | - Raquel López-Mejías
- Division and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marqués De Valdecilla, IDIVAL , Santander, Spain
| | - Miguel A González-Gay
- Division and Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Rheumatology Division, Hospital Universitario Marqués De Valdecilla, IDIVAL , Santander, Spain.,Department of Medicine, University of Cantabria , Santander, Spain.,Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand , Johannesburg, South Africa
| |
Collapse
|
43
|
Komici K, Faris P, Negri S, Rosti V, García-Carrasco M, Mendoza-Pinto C, Berra-Romani R, Cervera R, Guerra G, Moccia F. Systemic lupus erythematosus, endothelial progenitor cells and intracellular Ca2+ signaling: A novel approach for an old disease. J Autoimmun 2020; 112:102486. [DOI: 10.1016/j.jaut.2020.102486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 02/07/2023]
|
44
|
Escárcega RO, García-Carrasco M, Mendoza-Pinto C. The Cardio-Rheumatology Approach to Atherosclerotic Cardiovascular Disease. ACTA ACUST UNITED AC 2020; 16:311-312. [PMID: 32718860 DOI: 10.1016/j.reuma.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/03/2020] [Indexed: 02/02/2023]
Affiliation(s)
- Ricardo O Escárcega
- Heart and Vascular Institute, Lee Health System and Florida Heart Associates, Fort Myers, FL, USA
| | - Mario García-Carrasco
- Systemic Autoimmune Diseases Research Unit and Rheumatology/Immunology Department, BUAP School of Medicine, Puebla, Mexico
| | - Claudia Mendoza-Pinto
- Systemic Autoimmune Diseases Research Unit and Rheumatology/Immunology Department, BUAP School of Medicine, Puebla, Mexico.
| |
Collapse
|
45
|
Migliorini P, Italiani P, Pratesi F, Puxeddu I, Boraschi D. The IL-1 family cytokines and receptors in autoimmune diseases. Autoimmun Rev 2020; 19:102617. [PMID: 32663626 DOI: 10.1016/j.autrev.2020.102617] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 03/13/2020] [Indexed: 12/12/2022]
Abstract
The role of the cytokines and receptors of the IL-1 family in inflammation is well known. Several cytokines of the family have a powerful inflammatory activity, with IL-1β being the best-characterized factor. The inflammatory activity of IL-1 cytokines is regulated by other factors of the family, including receptor antagonists, soluble receptors and anti-inflammatory cytokines. The causative role of IL-1β is well-established in autoinflammatory diseases, mainly due to gain-of-function mutations in genes encoding the IL-1β-maturing inflammasome. Exaggerated production of IL-1β and IL-18 correlates with disease and disease severity also in several autoimmune and chronic inflammatory and degenerative pathologies, although it is not clear whether they have a causative role or are only involved in the downstream disease symptoms. A better understanding of the pathological role of IL-1 family cytokines in autoimmunity involves a deeper evaluation, in the pathological situations, of the possible anomalies in the feed-back anti-inflammatory mechanisms that in physiological reactions control and dump IL-1-mediated inflammation. Thus, we expect that IL-1 cytokines may be pathogenic only when, in addition to enhanced production, there is a concomitant failure of their control mechanisms. In this review we will examine the current knowledge on the role of IL-1 family cytokines in autoimmune and chronic inflammatory and degenerative diseases, with a particular focus on their endogenous control mechanisms, mainly based on soluble receptors/inhibitors and receptor antagonists. This will allow us to formulate a knowledge-based hypothesis on the involvement of IL-1 cytokines in the pathogenesis vs. the clinical features of these diseases.
Collapse
Affiliation(s)
- Paola Migliorini
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Paola Italiani
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Federico Pratesi
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Ilaria Puxeddu
- Clinical Immunology and Allergy Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Diana Boraschi
- Institute of Biochemistry and Cell Biology, National Research Council, Via Pietro Castellino 111, 80131 Napoli, Italy
| |
Collapse
|
46
|
Geng J, Fu W, Yu X, Lu Z, Liu Y, Sun M, Yu P, Li X, Fu L, Xu H, Sui D. Ginsenoside Rg3 Alleviates ox-LDL Induced Endothelial Dysfunction and Prevents Atherosclerosis in ApoE -/- Mice by Regulating PPARγ/FAK Signaling Pathway. Front Pharmacol 2020; 11:500. [PMID: 32390845 PMCID: PMC7188907 DOI: 10.3389/fphar.2020.00500] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
The initiation of atherosclerosis (AS) induced by dyslipidemia is accompanied by endothelial dysfunction, including decreased healing ability and increased recruitment of monocytes. Studies showed ginsenoside Rg3 has potential to treat diseases associated with endothelial dysfunction which can protects against antineoplastic drugs induced cardiotoxicity by repairing endothelial function, while the effect and mechanism of Rg3 on dyslipidemia induced endothelial dysfunction and AS are not clear. Therefore, we investigated the effects of Rg3 on oxidized low-density lipoprotein (ox-LDL) induced human umbilical vein endothelial cells (HUVECs) dysfunction and high-fat diets (HFD) induced atherosclerosis in ApoE−/− mice, as well as the mechanism. For in vitro assay, Rg3 enhanced healing of HUVECs and inhibited human monocytes (THP-1) adhesion to HUVECs disturbed by ox-LDL, down-regulated focal adhesion kinase (FAK)-mediated expression of vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1); restrained the FAK-mediated non-adherent dependent pathway containing matrix metalloproteinase (MMP)-2/9 expression, activation of nuclear factor-kappa B (NF-κB), high mRNA levels of monocyte chemotactic protein 1 (MCP-1) and interleukin 6 (IL-6), besides Rg3 up-regulated peroxisome proliferators-activated receptor γ (PPARγ) in ox-LDL-stimulated HUVECs. GW9662, the PPARγ-specific inhibitor, can repressed the effects of Rg3 on ox-LDL-stimulated HUVECs. For in vivo assay, Rg3 significantly reduced atherosclerotic pathological changes in ApoE−/− mice fed with HFD, up-regulated PPARγ, and inhibited activation FAK in aorta, thus inhibited expression of VCAM-1, ICAM-1 in intima. We conclude that Rg3 may protect endothelial cells and inhibit atherosclerosis by up-regulating PPARγ via repressing FAK-mediated pathways, indicating that Rg3 have good potential in preventing dyslipidemia induced atherosclerosis.
Collapse
Affiliation(s)
- Jianan Geng
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Wenwen Fu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xiaofeng Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zeyuan Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yanzhe Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Mingyang Sun
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Ping Yu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xin Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Li Fu
- Institute of Traditional Chinese Medicine Innovation, Jilin Yatai Pharmaceutical Co., Ltd., Changchun, China.,Institute of Dalian Fusheng Natural Medicine, Dalian Fusheng Pharmaceutical Co., Ltd., Dalian, China
| | - Huali Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dayun Sui
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| |
Collapse
|
47
|
Scherlinger M, Mertz P, Sagez F, Meyer A, Felten R, Chatelus E, Javier RM, Sordet C, Martin T, Korganow AS, Guffroy A, Poindron V, Richez C, Truchetet ME, Blanco P, Schaeverbeke T, Sibilia J, Devillers H, Arnaud L. Worldwide trends in all-cause mortality of auto-immune systemic diseases between 2001 and 2014. Autoimmun Rev 2020; 19:102531. [PMID: 32234406 DOI: 10.1016/j.autrev.2020.102531] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023]
Abstract
AIM To describe changes in the 2001-2014 mortality of 6 autoimmune systemic diseases (AISDs), namely Systemic Lupus Erythematosus (SLE), Systemic Sclerosis (SSc), Idiopathic Inflammatory Myopathies (IIM), Sjögren's Syndrome (SS), Mixed Connective Tissue Disease (MCTD) and ANCA-associated vasculitis (AAV) at the country-, continent-, and world-levels. METHODS Mortality data were retrieved from the World Health Organization (WHO) mortality database for each disease, based on ICD-10 codes. We computed age-standardized mortality rate (ASMR) as the estimated number of deaths per million inhabitants and its 95% confidence interval (95%CI). The association between gender, geographical areas and disease-specific mortality was analyzed using multivariate Poisson regression. The 2001-2014 temporal trends were analyzed using Jointpoint software. RESULTS In 2014, the worldwide ASMR for SLE was 2.68 (95%CI: 2.62-2.75) deaths/millions inhabitants, 1.46 (1.42-1.51) for SSc, 0.47 (0.44-0.49) for IIM, 0.17 (0.15-0.18) for SS, 0.11 (0.10-0.13) for MCTD and 0.53 (0.50-0.56) for AAV, with ASMRs generally lower in Europe than in North America, Latin America and Asia. Between 2001 and 2014, the worldwide ASMR decreased significantly for SSc (-0.71%/year), IIM (-1.65%/year) and AAV (-1.01%/year; p < .001 for all) and increased for SS (+1.53%/year, p = .01). The worldwide ASMR of SLE decreased significantly between 2001 and 2003 (-6.37%, p < .05) before increasing slightly between 2004 and 2014 (+0.58%, p < .01). CONCLUSIONS We observed a strong heterogeneity of standardized mortality rates across all countries analyzed for 6 autoimmune diseases. Those results further highlight the impact of world-wide inequities and major gaps in access to care and strategies for diagnosis and management of rare diseases, a crucial finding for world-wide physicians, patient associations and policy makers.
Collapse
Affiliation(s)
- Marc Scherlinger
- Centre Hospitalier Universitaire de Bordeaux, FHU ACRONIM, Place Amélie Raba Léon, 33076 Bordeaux, France; CNRS-UMR 5164 ImmunoConcEpT, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France
| | - Philippe Mertz
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Flora Sagez
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Alain Meyer
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Renaud Felten
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Emmanuel Chatelus
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Rose-Marie Javier
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Christelle Sordet
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France
| | - Thierry Martin
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service d'immunologie clinique, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France; Immuno-rhumatologie moléculaire, INSERM UMR-S 1109, Strasbourg, France
| | - Anne-Sophie Korganow
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service d'immunologie clinique, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France; Immuno-rhumatologie moléculaire, INSERM UMR-S 1109, Strasbourg, France
| | - Aurélien Guffroy
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service d'immunologie clinique, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France; Immuno-rhumatologie moléculaire, INSERM UMR-S 1109, Strasbourg, France
| | - Vincent Poindron
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service d'immunologie clinique, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France
| | - Christophe Richez
- Centre Hospitalier Universitaire de Bordeaux, FHU ACRONIM, Place Amélie Raba Léon, 33076 Bordeaux, France; CNRS-UMR 5164 ImmunoConcEpT, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France
| | - Marie-Elise Truchetet
- Centre Hospitalier Universitaire de Bordeaux, FHU ACRONIM, Place Amélie Raba Léon, 33076 Bordeaux, France; CNRS-UMR 5164 ImmunoConcEpT, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France
| | - Patrick Blanco
- CNRS-UMR 5164 ImmunoConcEpT, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France
| | - Thierry Schaeverbeke
- Centre Hospitalier Universitaire de Bordeaux, FHU ACRONIM, Place Amélie Raba Léon, 33076 Bordeaux, France; CNRS-UMR 5164 ImmunoConcEpT, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - Jean Sibilia
- CNRS-UMR 5164 ImmunoConcEpT, 146 rue Léo Saignat, 33076 Bordeaux, France; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France
| | - Hervé Devillers
- Service De Médecine Interne et Maladies Systémiques (médecine interne 2), Centre Hospitalier Universitaire de Dijon, Hôpital François-Mitterrand, Dijon, France; Centre d'investigation clinique - Epidémiologie Clinique, INSERM CIC 1432, Hôpital François-Mitterrand, Dijon, France
| | - Laurent Arnaud
- Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France; Immuno-rhumatologie moléculaire, INSERM UMR-S 1109, Strasbourg, France.
| |
Collapse
|
48
|
Han W, Wei Z, Zhang H, Geng C, Dang R, Yang M, Zhang J, Wang C, Jiang P. The Association Between Sortilin and Inflammation in Patients with Coronary Heart Disease. J Inflamm Res 2020; 13:71-79. [PMID: 32104044 PMCID: PMC7020934 DOI: 10.2147/jir.s240421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/21/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose Inflammation is a key contributor to coronary heart disease (CHD). Sortilin is a sorting receptor and has been identified as a critical regulator of inflammatory response. Therefore, our study aimed to determine the link between circulating sortilin levels, proinflammatory cytokine levels, and the occurrence of CHD. Patients and Methods Our study included 227 CHD patients and 101 matched healthy individuals. Circulating serum levels of sortilin and proinflammatory cytokines, including IL-1β, IL-6 and TNF-α, were assessed by a double-antibody sandwich enzyme-linked immunosorbent assay (DAS-ELISA). Linear regression and correlation analyses were used to estimate the associations between sortilin and proinflammatory cytokines. Moreover, six single-nucleotide polymorphisms (SNPs) spanning the sortilin and SORL1 genes were genotyped. Results Elevated levels of sortilin (P=0.027) and proinflammatory cytokines IL-1β (P=0.013), IL-6 (P=0.000) and TNF-α (P=0.010) were observed in CHD patients compared to those in healthy controls. Furthermore, sortilin levels were significantly positively correlated with IL-1β (r=0.252, P=0.0001), IL-6 (r=0.250, P=0.0001) and TNF-α (r=0.180, P=0.0064) levels. Notably, sortilin polymorphisms were revealed to be associated with the occurrence of CHD and varying sortilin levels. Subjects with the rs599839 AA risk genotype for CHD had significantly higher sortilin levels than those with the GG and GA genotypes (P=0.000); the same tendency was also observed in the levels of the proinflammatory cytokines IL-1β (P=0.003) and TNF-α (P=0.000). Similarly, GG carriers of rs464218 with increased sortilin levels were found to be at increased risk for CHD (P=0.014). The levels of IL-1β (P=0.025) and IL-6 (P=0.015) were also increased in these patients. Conclusion Our findings reveal that high sortilin levels may interact with inflammatory response to contribute to the occurrence of CHD. Considering that our clinical evidence suggests for the first time that sortilin involves in inflammatory response in CHD, the mechanistic role of sortilin in the progression of CHD deserves detailed investigation.
Collapse
Affiliation(s)
- Wenxiu Han
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Zhijie Wei
- Department of Medical Administration, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Hailiang Zhang
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Chunmei Geng
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Ruili Dang
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Mengqi Yang
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Jun Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, People's Republic of China
| | - Changshui Wang
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| | - Pei Jiang
- Department of Pharmacy, Jining First People's Hospital, Jining Medical University, Jining 272011, People's Republic of China
| |
Collapse
|
49
|
Głowinska-Olszewska B, Borysewicz-Sańczyk H, Sawicka B, Klonowska B, Charemska D, Żelazowska-Rutkowska B, Bossowski A. Does Hashimoto's Thyroiditis Increase the Risk of Cardiovascular Disease in Young Type 1 Diabetic Patients? Front Endocrinol (Lausanne) 2020; 11:431. [PMID: 32793113 PMCID: PMC7393727 DOI: 10.3389/fendo.2020.00431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Immunological and hormonal disorders have undoubted influence on the development of atherosclerotic process. Autoimmune diseases accompanying type 1 diabetes (T1D) may additionally accelerate atherosclerosis progression and increase the risk of cardiovascular events in the future. The influence of subclinical hypothyroidism on the cardiovascular system, in particular, has recently aroused great interest. The aim of our study was to assess intima-media thickness (cIMT) of common carotid arteries and the occurrence of classical atherosclerosis risk factors together with selected new biomarkers of cardiovascular diseases in young patients with type 1 diabetes mellitus coexisting with Hashimoto's disease (HD). Patients and Methods: The study included 50 adolescents and young adults with T1D with mean age 17.1 ± 3 years, with mean diabetes duration of 10.5 ± 3.3 years, including 20 patients with diagnosed HD: T1D and HD(+), and 30 patients with no additional diseases: T1D and HD(-). Twenty-two healthy, age-matched volunteers formed control group (C). We analyzed mean HbA1c value from all years of disease, BMI, blood pressure, lipids, new biomarkers of atherosclerosis (hsCRP, adiponectin, myeloperoxidase, NT-proBNP peptide, vitamin D), and cIMT of common carotid arteries. Results: In the group of patients with T1D and HD(+), significantly higher BMI was found: 23.3 ± 4.4 vs. 21.28 ± 2.9 in group HD(-) and 19.65 ± 2.4 kg/m2 in group C (p = 0.003), and higher waist circumference: 79 ± 10.9 vs. 75.10 ± 7.6 in group HD(-) vs. 69.0 ± 7.4 cm in group C (p < 0.001). The mean value of HbA1c was higher in group T1D and HD(+): 8.8% than in group HD(-): 8.1% (p = 0.04). Significantly higher concentration of hsCRP and lower vitamin D were observed in T1D and HD(+) in comparison to T1D and HD(-) and the control group. The IMT index in the HD(+) group was 0.46 ± 0.05 mm and was comparable to the HD(-) group but significantly higher than in healthy controls: 0.41 ± 0.03 mm (P < 0.05). Conclusions: Young patients with type 1 diabetes mellitus and with coexisting Hashimoto's thyroiditis have a higher BMI, a higher waist circumference, and a higher HbA1c value, which altogether may cause faster development of macroangiopathy in the near future. Additional risk for cardiovascular disease may result from low vitamin D and increased hsCRP concentration in this group of patients. Coexistence of Hashimoto's thyroiditis did not significantly affect the cIMT value in the studied population.
Collapse
Affiliation(s)
- Barbara Głowinska-Olszewska
- Department of Pediatrics, Endocrinology, Diabetology With Cardiology Division, Medical University of Białystok, Białystok, Poland
- *Correspondence: Barbara Głowinska-Olszewska
| | - Hanna Borysewicz-Sańczyk
- Department of Pediatrics, Endocrinology, Diabetology With Cardiology Division, Medical University of Białystok, Białystok, Poland
| | - Beata Sawicka
- Department of Pediatrics, Endocrinology, Diabetology With Cardiology Division, Medical University of Białystok, Białystok, Poland
| | - Bożenna Klonowska
- Department of Clinical Pediatrics, Faculty of Medical Sciences, Specialist Children's Hospital, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Dorota Charemska
- Department of Clinical Pediatrics, Faculty of Medical Sciences, Specialist Children's Hospital, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | | | - Artur Bossowski
- Department of Pediatrics, Endocrinology, Diabetology With Cardiology Division, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
50
|
Granule of BU-XIN RUAN-MAI Attenuates the Patients' Angina Pectoris of Coronary Heart Disease via Regulating miR-542-3p/GABARAP Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1808419. [PMID: 31949464 PMCID: PMC6948311 DOI: 10.1155/2019/1808419] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 12/22/2022]
Abstract
Objective Coronary heart disease (CHD) has been regarded as a serious and common disease in the modern society. This study aims to investigate the effect of Granule of BU-XIN RUAN-MAI (BXRM) on angina pectoris of coronary heart disease and to explore the molecular mechanisms underlying Granule of BU-XIN RUAN-MAI-mediated protective activity against this disease. Methods The effects of Granule of BU-XIN RUAN-MAI on clinical symptoms of patients' angina were indicated by hemorheology indicators including high shear of blood viscosity, low shear of blood viscosity, plasma viscosity, erythrocyte rigidity index, D-D dimer, fibrinogen content, and lipid content. The effects of Granule of BU-XIN RUAN-MAI on isoprenaline-induced myocardial cell injury were determined by conducting H&E staining and by performing ELISA to examine the serum content of MDA, SOD, Na+/K+-ATPase, cAMP, and the content of inflammatory factors in isoprenaline-induced rats. Meanwhile, western blot and real time PCR were used to determine the expression of genes involved in oxidation and energy metabolism, and real time PCR was also used for determination of miR-542-3p expression. Luciferase reporter assay was conducted to test the binding sites of miR-542-3p on GABARAP 3'UTR. The chemical compositions of Granule of BU-XIN RUAN-MAI were determined by liquid LC-QTOF-MS. Results Granule of BU-XIN RUAN-MAI significantly attenuated the clinical symptoms of patients' angina by improving the patients' heart rate and by decreasing the level of hemorheology indicators and also by reducing the serum content of TC, TG, LDL, and elevated HDL content. H&E staining demonstrated that Granule of BU-XIN RUAN-MAI ameliorated the myocardial ischemia in a dose-dependent manner. Besides, Granule of BU-XIN RUAN-MAI downregulated serum MDA content and upregulated the content of SOD, Na+/K+-ATPase, and cAMP in isoprenaline-induced rats. Granule of BU-XIN RUAN-MAI significantly improved oxidation stress by increasing PPARα expression, and it inhibited inflammation by downregulating expression and contents of IL-6, IL-1β, and TNF-α. Then, Granule of BU-XIN RUAN-MAI-containing serum increased the SOD content, and reduced the MDA content in angiotensin II-stimulated HUVEC cells. The granule of BU-XIN RUAN-MAI-containing serum obviously downregulated protein expressions of P40phox, P47phox, and P67phox in plasma membrane, and it significantly increased protein levels of P40phox, P47phox, and P67phox in the cytoplasm of HUVEC cells. Furthermore, GABARAP was reduced in heart tissues of ISO-induced rats and in angiotensin II-stimulated cell lines, and GABARAP was required for the inhibitory activity of Granule of BU-XIN RUAN-MAI on oxidation and inflammation in vivo and in vivo. GABARAP could be upregulated by Granule of BU-XIN RUAN-MAI by inhibiting the expression of miR-542-3p, which may significantly enhance oxidation and inflammation by targeting GABARAP in cardiomyocytes. Moreover, the silencing of GABARAP could obviously reverse the granule of BU-XIN RUAN-MAI-mediated protective activity against coronary heart disease, and interfering GABARAP expression also could partly block the anti-miR-542-3p-controlled oxidation and inflammation in cardiomyocytes. Besides, salidroside, loganin, and polydatin were the main compounds of granules of BU-XIN RUAN-MAI. Conclusion Granule of BU-XIN RUAN-MAI is an excellent prescription for treatment of coronary heart disease by suppressing inflammation and NAPDH-mediated oxidative stress. The miR-542-3p/GABARAP axis is required for Granule of BU-XIN RUAN-MAI, exhibiting its protective activity against the pectoris of coronary heart disease.
Collapse
|