1
|
Kobayashi S, Ikuno Y, Yamaguchi A, Takahashi T, Arakawa A, Shioyama W, Fujimoto N. Vasculo-Behçet disease with venous thromboembolism successfully treated with direct oral anticoagulants: A literature review. J Dermatol 2024. [PMID: 39495001 DOI: 10.1111/1346-8138.17535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/08/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Souta Kobayashi
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Yasuaki Ikuno
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Akihiko Yamaguchi
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | | | - Akiko Arakawa
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
- Department of Dermatology, Ludwig-Maximilian-University, Munich, Germany
| | - Wataru Shioyama
- Department of Cardiovascular Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
2
|
Huang J, Huang Y, Feng X, Huang C, Huang M, Wen Z, Xu A, Ge R, Yuan H, Shi H, Ma G, Li C, Lin J, Yi R, Hu Y, Jin Y, Liang S, Bi Y, Su S, Zhang X, Li X, Duan C. Predictive value of the systemic immune-inflammation index for periprocedural complications in flow diverter treatment for patients with intracranial aneurysms. Neurosurg Rev 2024; 47:809. [PMID: 39436476 DOI: 10.1007/s10143-024-03053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/23/2024]
Abstract
Flow-diverter devices (FDs) are effective in treating intracranial aneurysms (IAs) but carry substantial periprocedural risks, particularly ischemic complications. This study aimed to determine if elevated Systemic Immune-Inflammation Index (SII) can independently predict these risks and assess the impact of age and dual antiplatelet therapy on this association. We conducted a retrospective analysis of patients treated with FDs between February 2016 and August 2023, using blood samples taken within six days before surgery to calculate SII. Logistic regression and decision tree analyses assessed the link between SII and periprocedural complications, with subgroups exploring influencing factors. Multivariable analysis identified high SII as an independent predictor of periprocedural complications (OR = 5.306, 95% CI: 1.367-18.455; P = 0.009). The decision tree model confirmed SII > 0.437 as a critical threshold. Subgroup analysis showed a pronounced association of SII with periprocedural complications in patients ≥ 65 years (OR = 36.979, 95% CI: 2.103-650.134; P = 0.014) and in those on clopidogrel plus aspirin therapy (OR = 16.921, 95% CI: 2.733-104.746; P = 0.002). An elevated Systemic Immune-Inflammation Index (SII) > 0.437 significantly correlates with increased periprocedural complications (6.5% vs. 1.8%, P = 0.017). Although not statistically significant, higher SII is associated with a greater rate of ischemic events (3.9% vs. 0.9%). Elevated preoperative SII independently predicts periprocedural complications, particularly ischemic events, in patients undergoing FDs treatment for intracranial aneurysms. This association is particularly pronounced in older patients (> 65 years) and those receiving dual therapy with clopidogrel plus aspirin. Trial Registration: ClinicalTrials.gov (NCT06446778). Registered on May 22, 2024.
Collapse
Affiliation(s)
- Jiwan Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yaxian Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xin Feng
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China.
| | - Chi Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Mengshi Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Zhuohua Wen
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Anqi Xu
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Runze Ge
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Hao Yuan
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Hongyu Shi
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Gengwu Ma
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Can Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Jiancheng Lin
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Ruizhe Yi
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yuqi Hu
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yuheng Jin
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Shuyin Liang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yiming Bi
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Shixing Su
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xin Zhang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Xifeng Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Chuanzhi Duan
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China.
| |
Collapse
|
3
|
Muric M, Nikolic M, Todorovic A, Jakovljevic V, Vucicevic K. Comparative Cardioprotective Effectiveness: NOACs vs. Nattokinase-Bridging Basic Research to Clinical Findings. Biomolecules 2024; 14:956. [PMID: 39199344 PMCID: PMC11352257 DOI: 10.3390/biom14080956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
The use of non-vitamin K antagonist oral anticoagulants (NOACs) has brought a significant progress in the management of cardiovascular diseases, considered clinically superior to vitamin K antagonists (VKAs) particularly in the prevention and treatment of thromboembolic events. In addition, numerous advantages such as fixed dosing, lack of laboratory monitoring, and fewer food and drug-to-drug interactions make the use of NOACs superior to VKAs. While NOACs are synthetic drugs prescribed for specific conditions, nattokinase (NK) is a natural enzyme derived from food that has potential health benefits. Various experimental and clinical studies reported the positive effects of NK on the circulatory system, including the thinning of blood and the dissolution of blood clots. This enzyme showed not only fibrinolytic activity due to its ability to degrade fibrin, but also an affinity as a substrate for plasmin. Recent studies have shown that NK has additional cardioprotective effects, such as antihypertensive and anti-atherosclerotic effects. In this narrative review, we presented the cardioprotective properties of two different approaches that go beyond anticoagulation: NOACs and NK. By combining evidence from basic research with clinical findings, we aim to elucidate the comparative cardioprotective efficacy of these interventions and highlight their respective roles in modern cardiovascular care.
Collapse
Affiliation(s)
- Maja Muric
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (V.J.)
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia;
| | - Marina Nikolic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (V.J.)
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia;
| | - Andreja Todorovic
- Department of Cardiology, General Hospital Ćuprija, 35230 Ćuprija, Serbia;
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (V.J.)
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia;
- Department of Human Pathology, First Moscow State Medical, University IM Sechenov, 119991 Moscow, Russia
| | - Ksenija Vucicevic
- Center of Excellence for Redox Balance Research in Cardiovascular and Metabolic Disorders, 34000 Kragujevac, Serbia;
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
4
|
Wang H, Li S, Yu J, Xu J, Xu Y. Role of leukocyte parameters in patients with ST-segment elevation myocardial infarction undergoing primary percutaneous coronary intervention with high thrombus burden. Front Cardiovasc Med 2024; 11:1397701. [PMID: 38962087 PMCID: PMC11221325 DOI: 10.3389/fcvm.2024.1397701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
Objective Leukocyte parameters are associated with cardiovascular diseases. The aim of the present study was to investigate the role of leukocyte parameters in patients with ST-segment elevation myocardial infarction (STEMI) undergoing primary percutaneous coronary intervention (PPCI) with high thrombus burden (HTB). Methods A total of 102 consecutive STEMI patients with HTB who underwent PPCI within 12 h from the onset of symptoms between June 2020 and September 2021 were enrolled in this study. In addition, 101 age- and sex-matched STEMI patients with low thrombus burden (LTB) who underwent PPCI within 12 h from the onset of symptoms were enrolled as controls. Leukocyte parameters, such as neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), and monocyte to lymphocyte ratio (MLR), were calculated at the time of admission. Results The value of NLR and MLR were significantly higher in the HTB group than in the LTB group (6.24 ± 4.87 vs. 4.65 ± 3.47, p = 0.008; 0.40 ± 0.27 vs. 0.33 ± 0.20, p = 0.038). A cutoff value of >5.38 for NLR had a sensitivity and specificity of 53.9% and 74.3%, respectively, and MLR >0.29 had a sensitivity and specificity of 60.8% and 55.4%, respectively, for determining the STEMI patients with HTB [area under the receiver operating characteristic curve (AUC): 0.603, 95% confidence interval (CI): 0.524-0.681, p = 0.012; AUC: 0.578, 95% CI: 0.499-0.656, p = 0.046]. There was no significant difference of all-cause mortality rate and major adverse cardiac events (MACEs) between the STEMI patients with HTB or with LTB (3.92% in HTB group vs. 2.97% in LTB group, p = 0.712; 10.78% in HTB group vs. 8.91% in LTB group, p = 0.215). Compared with the HTB patients in the low NLR group, C-reactive protein, baseline troponin I, baseline brain natriuretic peptide, and leukocyte parameters, such as white blood cell, neutrophil, lymphocyte, NLR, PLR, and MLR, were also significantly higher in the high NLR group in STEMI patients who underwent PPCI with HTB (18.94 ± 19.06 vs. 35.23 ± 52.83, p = 0.037; 10.99 ± 18.07 vs. 21.37 ± 19.64, p = 0.007; 199.39 ± 323.67 vs. 430.72 ± 683.59, p = 0.028; 11.55 ± 3.56 vs. 9.31 ± 2.54, p = 0.001; 9.77 ± 3.17 vs. 5.79 ± 1.97, p = 0.000; 1.16 ± 0.44 vs. 2.69 ± 1.23, p = 0.000; 9.37 ± 4.60 vs 1.31 ± 2.58, p = 0.000; 200.88 ± 89.90 vs. 97.47 ± 50.99, p = 0.000; 0.52 ± 0.29 vs. 0.26 ± 0.14, p = 0.000, respectively). MACEs and heart failure in the high NLR group were significantly higher than that in the low NLR group of STEMI patients who underwent PPCI with HTB (20.45% vs. 4.25%, p = 0.041; 10.91% vs. 2.13%, p = 0.038). Conclusion The value of NLR and MLR were higher in STEMI patients who underwent PPCI with HTB. In STEMI patients who underwent PPCI with HTB, a raised NLR could effectively predict the occurrence of MACEs and heart failure.
Collapse
Affiliation(s)
| | | | | | | | - Yan Xu
- Department of Cardiology, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
5
|
Rapetti F, Spallarossa A, Russo E, Caviglia D, Villa C, Tasso B, Signorello MG, Rosano C, Iervasi E, Ponassi M, Brullo C. Investigations of Antioxidant and Anti-Cancer Activities of 5-Aminopyrazole Derivatives. Molecules 2024; 29:2298. [PMID: 38792163 PMCID: PMC11124527 DOI: 10.3390/molecules29102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
To further extend the structure-activity relationships (SARs) of 5-aminopyrazoles (5APs) and identify novel compounds able to interfere with inflammation, oxidative stress, and tumorigenesis, 5APs 1-4 have been designed and prepared. Some chemical modifications have been inserted on cathecol function or in aminopyrazole central core; in detail: (i) smaller, bigger, and more lipophilic substituents were introduced in meta and para positions of catechol portion (5APs 1); (ii) a methyl group was inserted on C3 of the pyrazole scaffold (5APs 2); (iii) a more flexible alkyl chain was inserted on N1 position (5APs 3); (iv) the acylhydrazonic linker was moved from position 4 to position 3 of the pyrazole scaffold (5APs 4). All new derivatives 1-4 have been tested for radical scavenging (DPPH assay), anti-aggregating/antioxidant (in human platelets) and cell growth inhibitory activity (MTT assay) properties. In addition, in silico pharmacokinetics, drug-likeness properties, and toxicity have been calculated. 5APs 1 emerged to be promising anti-proliferative agents, able to suppress the growth of specific cancer cell lines. Furthermore, derivatives 3 remarkably inhibited ROS production in platelets and 5APs 4 showed interesting in vitro radical scavenging properties. Overall, the collected results further confirm the pharmaceutical potentials of this class of compounds and support future studies for the development of novel anti-proliferative and antioxidant agents.
Collapse
Affiliation(s)
- Federica Rapetti
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| | - Andrea Spallarossa
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| | - Eleonora Russo
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| | - Debora Caviglia
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| | - Carla Villa
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| | - Bruno Tasso
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| | - Maria Grazia Signorello
- Department of Pharmacy (DIFAR), Biochemistry Lab., University of Genoa, Viale Benedetto XV, 3, 16132 Genova, Italy;
| | - Camillo Rosano
- IRCCS Ospedale Policlinico San Martino, Proteomics and Mass Spectrometry Unit, L.go. R. Benzi, 10, 16132 Genova, Italy; (C.R.); (E.I.); (M.P.)
| | - Erika Iervasi
- IRCCS Ospedale Policlinico San Martino, Proteomics and Mass Spectrometry Unit, L.go. R. Benzi, 10, 16132 Genova, Italy; (C.R.); (E.I.); (M.P.)
| | - Marco Ponassi
- IRCCS Ospedale Policlinico San Martino, Proteomics and Mass Spectrometry Unit, L.go. R. Benzi, 10, 16132 Genova, Italy; (C.R.); (E.I.); (M.P.)
| | - Chiara Brullo
- Department of Pharmacy (DIFAR), University of Genoa, Viale Benedetto XV, 3, 16132 Genoa, Italy; (F.R.); (A.S.); (E.R.); (D.C.); (C.V.); (B.T.)
| |
Collapse
|
6
|
Liu Y, Zhao Y, Guo Z, Li M, Shan H, Zhang Y, Miao C, Gu Y. Pericarotid Fat Stranding at Computed Tomography Angiography: A Marker of the Short-Term Prognosis of Acute Ischemic Stroke. J Comput Assist Tomogr 2024; 48:311-316. [PMID: 37876252 DOI: 10.1097/rct.0000000000001555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
PURPOSE Perivascular epicardial fat stranding detected in the coronary computed tomography (CT) angiography is associated with culprit lesions and provides helpful information on the risk of acute coronary syndrome. This study aimed to evaluate the potential clinical significance of pericarotid fat stranding (PCFS) and investigate the association between PCFS and short-term prognosis in acute stroke using head and neck CT angiography (CTA). METHODS This study included 80 patients (mean age, 69.69 ± 11.03; 58 men) who underwent both head and neck CTA and magnetic resonance imaging within a 1-week period. Baseline characteristics, pericarotid adipose tissue attenuation, plaque characteristics, ischemic penumbra, infarct core volume, infarct core growth rate (CGR), and the grade of collateral status were recorded and compared between a PCFS group and a non-PCFS group. Data were compared using the 2-sample t test, Mann-Whitney U test, Fisher exact test, and Spearman rank correlation analysis. RESULTS We found that patients with PCFS had a significantly higher pericarotid adipose tissue density than patients without PCFS (-55.75 ± 5.53 vs -65.82 ± 9.65, P < 0.001). Patients with PCFS showed a larger infarct core volume (166.43 ± 73.07 vs 91.43 ± 55.03, P = 0.001) and faster CGR (39.57 ± 12.01 vs 19.83 ± 32.77; P < 0.001), and the frequency of adverse prognosis was more significant than in control participants (83.33% vs 19.11%). CONCLUSIONS Individuals with PCFS showed higher CGR, which was substantially related to worse outcomes in patients with acute stroke with ipsilateral carotid atherosclerosis. Recognition of PCFS may help predict stroke prognosis and allow doctors to take early action to improve patient prognosis.
Collapse
Affiliation(s)
- Ying Liu
- From the Department of Radiology, The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Wang H, Nie H, Bu G, Tong X, Bai X. Systemic immune-inflammation index (SII) and the risk of all-cause, cardiovascular, and cardio-cerebrovascular mortality in the general population. Eur J Med Res 2023; 28:575. [PMID: 38066657 PMCID: PMC10709886 DOI: 10.1186/s40001-023-01529-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND An elevated systemic immune-inflammation index (SII) is associated with higher mortality in patients with coronary artery disease and other diseases. However, the potential of SII for predicting mortality in the general population has been underexplored. Therefore, this study aimed to analyze the relationship between the SII and all-cause, cardiovascular disease, and cardiocerebrovascular disease mortality in the general population. METHODS This study involved 26,855 participants (≥ 18 years) from the National Health and Nutrition Examination Survey 1999-2014 who were grouped according to the SII tertiles. Survival differences between the groups were analyzed using log-rank tests and Kaplan-Meier plots. Furthermore, multivariate Cox regression and restricted cubic spline analyses were used to examine the relationship between the SII and all-cause, cardiovascular, and cardio-cerebrovascular mortality. RESULTS Overall, 1947 (7.425%) participants died following an average follow-up of 87.99 ± 54.04 months. Among these, 325 (1.210%) deaths were related to cardiovascular diseases and 392 (1.459%) to cardio-cerebrovascular mortality. Kaplan-Meier analysis revealed statistically significant differences in all-cause, cardiovascular, and cerebrovascular mortality between the SII tertiles (log-rank test: all P < 0.001). Multi-adjusted models showed that participants in the highest tertile of SII had a higher risk of death from all-cause (hazard ratio [HR] = 1.48, 95% confidence interval [CI] 1.48-1.48) and cardiovascular mortality (HR = 1.60, 95% CI 1.60-1.61) compared with those in the lowest tertile. In addition, the restricted cubic spline curve indicated a nonlinear association between SII and all-cause mortality (P < 0.001), with threshold value of SII at 18.284. There was a 15% decrease in the risk of all-cause mortality for each twofold change in SII on the left flank (HR = 0.85, 95% CI 0.69-1.05) and a 42% increase (HR = 1.42, 95% CI 1.23-1.64) on the right flank of the inflection point. In addition, the risk of cardiovascular mortality increased nonlinearly by 39% per twofold change in SII (HR = 1.39, 95% CI 1.07-1.81). There was also a nonlinear increase in the risk of cardio-cerebrovascular mortality per twofold change in SII (HR = 1.29, 95% CI 1.00-1.66). CONCLUSIONS In the general population, the SII was significantly associated with all-cause, cardiovascular, and cardio-cerebrovascular mortality, regardless of the established risk factors.
Collapse
Affiliation(s)
- Huan Wang
- Department of Pain Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an , 710061, China
| | - Huiyong Nie
- Department of Pain Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an , 710061, China
| | - Gang Bu
- Department of Pain Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an , 710061, China
| | - Xiaoning Tong
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaofang Bai
- The Department of Ultrasound Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
8
|
Leberzammer J, von Hundelshausen P. Chemokines, molecular drivers of thromboinflammation and immunothrombosis. Front Immunol 2023; 14:1276353. [PMID: 37954596 PMCID: PMC10637585 DOI: 10.3389/fimmu.2023.1276353] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023] Open
Abstract
Blood clotting is a finely regulated process that is essential for hemostasis. However, when dysregulated or spontaneous, it promotes thrombotic disorders. The fact that these are triggered, accompanied and amplified by inflammation is reflected in the term thromboinflammation that includes chemokines. The role of chemokines in thrombosis is therefore illuminated from a cellular perspective, where endothelial cells, platelets, red blood cells, and leukocytes may be both the source and target of chemokines. Chemokine-dependent prothrombotic processes may thereby occur independently of chemokine receptors or be mediated by chemokine receptors, although the binding and activation of classical G protein-coupled receptors and their signaling pathways differ from those of atypical chemokine receptors, which do not function via cell activation and recruitment. Regardless of binding to their receptors, chemokines can induce thrombosis by forming platelet-activating immune complexes with heparin or other polyanions that are pathognomonic for HIT and VITT. In addition, chemokines can bind to NETs and alter their structure. They also change the electrical charge of the cell surface of platelets and interact with coagulation factors, thereby modulating the balance of fibrinolysis and coagulation. Moreover, CXCL12 activates CXCR4 on platelets independently of classical migratory chemokine activity and causes aggregation and thrombosis via the PI3Kβ and Btk signaling pathways. In contrast, typical chemokine-chemokine receptor interactions are involved in the processes that contribute to the adhesiveness of the endothelium in the initial phase of venous thrombosis, where neutrophils and monocytes subsequently accumulate in massive numbers. Later, the reorganization and resolution of a thrombus require coordinated cell migration and invasion of the thrombus, and, as such, indeed, chemokines recruit leukocytes to existing thrombi. Therefore, chemokines contribute in many independent ways to thrombosis.
Collapse
Affiliation(s)
- Julian Leberzammer
- Institute of Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiology and Angiology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Philipp von Hundelshausen
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute for Cardiovascular Prevention, Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
9
|
Sohma R, Sakuma M, Obi S, Nishino S, Inoue KI, Kishimoto S, Lu T, Toyoda S, Inoue T. Effects of the factor Xa inhibitor rivaroxaban on the differentiation of endothelial progenitor cells. BMC Cardiovasc Disord 2023; 23:282. [PMID: 37268884 DOI: 10.1186/s12872-023-03318-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 05/24/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND We evaluated the efficacy of the factor Xa inhibitor rivaroxaban on the differentiation ability of vascular endothelial progenitor cells (EPCs), which play roles in vascular injury repair and atherogenesis. Antithrombotic treatment in patients with atrial fibrillation undergoing percutaneous coronary intervention (PCI) is challenging, and current guidelines recommend oral anticoagulant monotherapy 1 year or more after PCI. However, biological evidence of the pharmacological effects of anticoagulants is insufficient. METHODS EPC colony-forming assays were performed using peripheral blood-derived CD34-positive cells from healthy volunteers. Adhesion and tube formation of cultured EPCs were assessed in human umbilical cord-derived CD34-positive cells. Endothelial cell surface markers were assessed using flow cytometry, and Akt and endothelial nitric oxide synthase (eNOS) phosphorylation were examined using western blot analysis of EPCs. Adhesion, tube formation and endothelial cell surface marker expression was observed in EPCs transfected with small interfering RNA (siRNA) against protease-activated receptor (PAR)-2. Finally, EPC behaviors were assessed in patients with atrial fibrillation undergoing PCI in whom warfarin was changed to rivaroxaban. RESULTS Rivaroxaban increased the number of large EPC colonies and increased the bioactivities of EPCs, including adhesion and tube formation. Rivaroxaban also increased vascular endothelial growth factor receptor (VEGFR)-1, VEGFR-2, Tie-2, and E-selectin expression as well as Akt and eNOS phosphorylation. PAR-2 knockdown increased the bioactivities of EPCs and endothelial cell surface marker expression. Patients in whom the number of large colonies increased after switching to rivaroxaban showed better vascular repair. CONCLUSIONS Rivaroxaban increased the differentiation ability of EPCs, leading to potential advantages in the treatment of coronary artery disease.
Collapse
Affiliation(s)
- Ryoichi Sohma
- Center for Advanced Medical Science Research, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan.
| | - Syotaro Obi
- Center for Advanced Medical Science Research, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Setsu Nishino
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Ken-Ichi Inoue
- Center for Advanced Medical Science Research, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Satoko Kishimoto
- Center for Advanced Medical Science Research, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Tianyang Lu
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| | - Teruo Inoue
- Japan Red Cross Society, Nasu Red Cross Hospital, 1081-4 Nakadawara, Tochigi, 324-8686, Otawara, Japan
- Dokkyo Medical University, 880 Kitakobayashi, Mibu, Tochigi, 321-0293, Japan
| |
Collapse
|
10
|
Cimmino G, Muscoli S, De Rosa S, Cesaro A, Perrone MA, Selvaggio S, Selvaggio G, Aimo A, Pedrinelli R, Mercuro G, Romeo F, Perrone Filardi P, Indolfi C, Coronelli M. Evolving concepts in the pathophysiology of atherosclerosis: from endothelial dysfunction to thrombus formation through multiple shades of inflammation. J Cardiovasc Med (Hagerstown) 2023; 24:e156-e167. [PMID: 37186566 DOI: 10.2459/jcm.0000000000001450] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Atherosclerosis is the anatomo-pathological substrate of most cardio, cerebro and vascular diseases such as acute and chronic coronary syndromes, stroke and peripheral artery diseases. The pathophysiology of atherosclerotic plaque and its complications are under continuous investigation. In the last 2 decades our understanding on the formation, progression and complication of the atherosclerotic lesion has greatly improved and the role of immunity and inflammation is now well documented and accepted. The conventional risk factors modulate endothelial function determining the switch to a proatherosclerotic phenotype. From this point, lipid accumulation with an imbalance from cholesterol influx and efflux, foam cells formation, T-cell activation, cytokines release and matrix-degrading enzymes production occur. Lesions with high inflammatory rate become vulnerable and prone to rupture. Once complicated, the intraplaque thrombogenic material, such as the tissue factor, is exposed to the flowing blood, thus inducing coagulation cascade activation, platelets aggregation and finally intravascular thrombus formation that leads to clinical manifestations of this disease. Nonconventional risk factors, such as gut microbiome, are emerging novel markers of atherosclerosis. Several data indicate that gut microbiota may play a causative role in formation, progression and complication of atherosclerotic lesions. The gut dysbiosis-related inflammation and gut microbiota-derived metabolites have been proposed as the main working hypothesis in contributing to disease formation and progression. The current evidence suggest that the conventional and nonconventional risk factors may modulate the degree of inflammation of the atherosclerotic lesion, thus influencing its final fate. Based on this hypothesis, targeting inflammation seems to be a promising approach to further improve our management of atherosclerotic-related diseases.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
| | | | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro
| | - Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Naples
- Division of Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta
| | - Marco A Perrone
- Department of Cardiology and CardioLab, University of Rome Tor Vergata, Rome
| | | | | | - Alberto Aimo
- Fondazione Toscana Gabriele Monasterio
- Institute of Life Sciences, Scuola Superiore Sant'Anna
| | - Roberto Pedrinelli
- Critical Care Medicine-Cardiology Division, Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa
| | - Giuseppe Mercuro
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi, Cagliari
| | | | - Pasquale Perrone Filardi
- Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli 'Federico II', Napoli
| | - Ciro Indolfi
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Catanzaro
| | - Maurizio Coronelli
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
| |
Collapse
|
11
|
Ning F, Liu H, Yan Y. Clinical characteristics, prognostic factors, and outcomes of ventricular septal rupture in patients with acute myocardial infarction. J Investig Med 2023; 71:361-371. [PMID: 36692151 DOI: 10.1177/10815589221149185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ventricular septal rupture (VSR) is a rare but devastating complication in patients with acute myocardial infarction (AMI). This was a retrospective single-center observational study which aimed to assess the characteristics and outcomes of VSR patients and identify risk factors for in-hospital mortality of VSR patients in the era of percutaneous intervention. Patients with VSR after AMI at the First Affiliated Hospital of Xi'an Jiaotong University from January 2016 to December 2020 were enrolled. Among 5395 consecutive patients with AMI, 42 patients (0.78%) were diagnosed with VSR. Left anterior descending coronary artery was the culprit vessel in most cases (84.4%, 27/32). In the multivariate analysis, female sex (odds ratio (OR): 14.043, 95% confidence interval (CI): 1.396-141.283, p = 0.025) and lower platelet count on admission (OR: 0.979; 95% CI: 0.963-0.995; p = 0.009) were significant risk factors of in-hospital death in VSR patients. In all, 11 patients (26.2%) underwent surgical repair, and the rest were treated medically. The 1-year mortality was lower in the surgical group (36.4%, 4/11) than that in the conservative group (74.2%, 23/31) (p = 0.034). During the follow-up, VSR patients treated surgically tended to have a higher long-term survival rate than those treated medically (log rank χ2 = 5.005, p = 0.025). The prognosis of patients with VSR remained poor in this study. Female sex and lower platelet count were independent risk factors of in-hospital death in VSR patients. The long-term survival rate of patients treated with surgical repair was significantly better than that of patients treated conservatively.
Collapse
Affiliation(s)
- Feifei Ning
- Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Hui Liu
- The Biobank of the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| | - Yang Yan
- Department of Cardiovascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, P.R. China
| |
Collapse
|
12
|
Hara T, Sata M, Fukuda D. Emerging roles of protease-activated receptors in cardiometabolic disorders. J Cardiol 2023; 81:337-346. [PMID: 36195252 DOI: 10.1016/j.jjcc.2022.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Abstract
Cardiometabolic disorders, including obesity-related insulin resistance and atherosclerosis, share sterile chronic inflammation as a major cause; however, the precise underlying mechanisms of chronic inflammation in cardiometabolic disorders are not fully understood. Accumulating evidence suggests that several coagulation proteases, including thrombin and activated factor X (FXa), play an important role not only in the coagulation cascade but also in the proinflammatory responses through protease-activated receptors (PARs) in many cell types. Four members of the PAR family have been cloned (PAR 1-4). For instance, thrombin activates PAR-1, PAR-3, and PAR-4. FXa activates both PAR-1 and PAR-2, while it has no effect on PAR-3 or PAR-4. Previous studies demonstrated that PAR-1 and PAR-2 activated by thrombin or FXa promote gene expression of inflammatory molecules mainly via the NF-κB and ERK1/2 pathways. In obese adipose tissue and atherosclerotic vascular tissue, various stresses increase the expression of tissue factor and procoagulant activity. Recent studies indicated that the activation of PARs in adipocytes and vascular cells by coagulation proteases promotes inflammation in these tissues, which leads to the development of cardiometabolic diseases. This review briefly summarizes the role of PARs and coagulation proteases in the pathogenesis of inflammatory diseases and describes recent findings (including ours) on the potential participation of this system in the development of cardiometabolic disorders. New insights into PARs may ensure a better understanding of cardiometabolic disorders and suggest new therapeutic options for these major health threats.
Collapse
Affiliation(s)
- Tomoya Hara
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Daiju Fukuda
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan; Department of Cardiovascular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
13
|
Huseynov A, Reinhardt J, Chandra L, Dürschmied D, Langer HF. Novel Aspects Targeting Platelets in Atherosclerotic Cardiovascular Disease—A Translational Perspective. Int J Mol Sci 2023; 24:ijms24076280. [PMID: 37047253 PMCID: PMC10093962 DOI: 10.3390/ijms24076280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Platelets are important cellular targets in cardiovascular disease. Based on insights from basic science, translational approaches and clinical studies, a distinguished anti-platelet drug treatment regimen for cardiovascular patients could be established. Furthermore, platelets are increasingly considered as cells mediating effects “beyond thrombosis”, including vascular inflammation, tissue remodeling and healing of vascular and tissue lesions. This review has its focus on the functions and interactions of platelets with potential translational and clinical relevance. The role of platelets for the development of atherosclerosis and therapeutic modalities for primary and secondary prevention of atherosclerotic disease are addressed. Furthermore, novel therapeutic options for inhibiting platelet function and the use of platelets in regenerative medicine are considered.
Collapse
|
14
|
Tugrul Ersak D, Kara Ö, Yakut K, Tokmak A, Sanhal CY, Yücel A, Şahin D. The Association between Placental Abruption and Platelet Indices. Fetal Pediatr Pathol 2023; 42:367-375. [PMID: 36715068 DOI: 10.1080/15513815.2023.2166798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Objective: Placental abruption (PA) is an obstetric emergency. This study investigated the use of platelet indices in PA in its early stages to determine if it could aid in diagnosis. Materials and Methods: Sixty-two pregnant women with PA and 130 pregnant women who delivered due to idiopathic preterm delivery were included in this case-control study. Blood samples including platelet indices, biochemical, and coagulation parameters were obtained before cesarean section. Maternal and neonatal outcomes were recorded. Results: There was no significant difference between the groups as to hemoglobin, hematocrit, and white blood count. Platelet, mean platelet volume (MPV), and platelet to lymphocyte ratio (PLR) were significantly lower, platelet distribution width (PDW) was significantly higher in the PA patients. Conclusion: In the current study, MPV and PLR were lower and PDW was higher in PA patients. These parameters may be useful in assessment of PA.
Collapse
Affiliation(s)
- Duygu Tugrul Ersak
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| | - Özgür Kara
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| | - Kadriye Yakut
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| | - Aytekin Tokmak
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| | - Cem Yaşar Sanhal
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| | - Aykan Yücel
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| | - Dilek Şahin
- Ankara Dr. Zekai Tahir Burak Women's Health Training Research Hospital, Ankara, Turkey
| |
Collapse
|
15
|
Wang Y, Li C, Yuan M, Ren B, Liu C, Zheng J, Lin Z, Ren F, Gao D. Development of a complete blood count with differential-based prediction model for in-hospital mortality among patients with acute myocardial infarction in the coronary care unit. Front Cardiovasc Med 2022; 9:1001356. [PMID: 36277791 PMCID: PMC9581274 DOI: 10.3389/fcvm.2022.1001356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose In recent years, the complete blood count with differential (CBC w/diff) test has drawn strong interest because of its prognostic value in cardiovascular diseases. We aimed to develop a CBC w/diff-based prediction model for in-hospital mortality among patients with severe acute myocardial infarction (AMI) in the coronary care unit (CCU). Materials and methods This single-center retrospective study used data from a public database. The neural network method was applied. The performance of the model was assessed by discrimination and calibration. The discrimination performance of our model was compared to that of seven other classical machine learning models and five well-studied CBC w/diff clinical indicators. Finally, a permutation test was applied to evaluate the importance rank of the predictor variables. Results A total of 2,231 patient medical records were included. With a mean area under the curve (AUC) of 0.788 [95% confidence interval (CI), 0.736-0.838], our model outperformed all other models and indices. Furthermore, it performed well in calibration. Finally, the top three predictors were white blood cell count (WBC), red blood cell distribution width-coefficient of variation (RDW-CV), and neutrophil percentage. Surprisingly, after dropping seven variables with poor prediction values, the AUC of our model increased to 0.812 (95% CI, 0.762-0.859) (P < 0.05). Conclusion We used a neural network method to develop a risk prediction model for in-hospital mortality among patients with AMI in the CCU based on the CBC w/diff test, which performed well and would aid in early clinical decision-making. The top three important predictors were WBC, RDW-CV and neutrophil percentage.
Collapse
Affiliation(s)
- Yu Wang
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Changfu Li
- Department of Digestive Medicine, Daqing Longnan Hospital, Daqing, China
| | - Miao Yuan
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Bincheng Ren
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Chang Liu
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Jiawei Zheng
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Zehao Lin
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| | - Fuxian Ren
- Department of Cardiology, Meishan Branch of the Third Affiliated Hospital, Yanan University School of Medical, Meishan, China
| | - Dengfeng Gao
- Department of Cardiology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, China
| |
Collapse
|
16
|
Duca ȘT, Chetran A, Miftode RȘ, Mitu O, Costache AD, Nicolae A, Iliescu-Halițchi D, Halițchi-Iliescu CO, Mitu F, Costache II. Myocardial Ischemia in Patients with COVID-19 Infection: Between Pathophysiological Mechanisms and Electrocardiographic Findings. Life (Basel) 2022; 12:life12071015. [PMID: 35888103 PMCID: PMC9318430 DOI: 10.3390/life12071015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 12/29/2022] Open
Abstract
Given the possible pathophysiological links between myocardial ischemia and SARS-CoV-2 infection, several studies have focused attention on acute coronary syndromes in order to improve patients’ morbidity and mortality. Understanding the pathophysiological aspects of myocardial ischemia in patients infected with SARS-CoV-2 can open a broad perspective on the proper management for each patient. The electrocardiogram (ECG) remains the easiest assessment of cardiac involvement in COVID-19 patients, due to its non-invasive profile, accessibility, low cost, and lack of radiation. The ECG changes provide insight into the patient’s prognosis, indicating either the worsening of an underlying cardiac illnesses or the acute direct injury by the virus. This indicates that the ECG is an important prognostic tool that can affect the outcome of COVID-19 patients, which important to correlate its aspects with the clinical characteristics and patient’s medical history. The ECG changes in myocardial ischemia include a broad spectrum in patients with COVID-19 with different cases reported of ST-segment elevation, ST-segment depression, and T wave inversion, which are associated with severe COVID-19 disease.
Collapse
Affiliation(s)
- Ștefania Teodora Duca
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Adriana Chetran
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
- Correspondence: ; Tel.: +40-741089910
| | - Radu Ștefan Miftode
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Ovidiu Mitu
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ana Nicolae
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| | - Dan Iliescu-Halițchi
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, Arcadia Hospital, 700620 Iasi, Romania
| | - Codruța-Olimpiada Halițchi-Iliescu
- Department of Mother and Child Medicine-Pediatrics, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
- Department of Pedriatics, Arcadia Hospital, 700620 Iasi, Romania
| | - Florin Mitu
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Irina Iuliana Costache
- Department of Internal Medicine I, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700145 Iasi, Romania; (Ș.T.D.); (R.Ș.M.); (O.M.); (A.D.C.); (A.N.); (D.I.-H.); (F.M.); (I.I.C.)
- Department of Cardiology, “St. Spiridon” Emergency County Hospital, 700111 Iasi, Romania
| |
Collapse
|
17
|
Yang J, Li C, Zheng Y, Gao J, Liu YP, Wang JJ, Song JJ, Zhou Q, Meng X, Zhang K, Wang W, Shao C, Tang YD. The Association Between High-Sensitivity C-Reactive Protein/Albumin Ratio and Cardiovascular Prognosis in Patients Undergoing Percutaneous Coronary Intervention. Angiology 2022; 73:818-826. [PMID: 35748815 DOI: 10.1177/00033197221110715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Limited studies have focused on the impact of high-sensitivity C-reactive protein (hsCRP) to albumin ratio (CAR) on cardiovascular outcomes in patients undergoing percutaneous coronary intervention (PCI). Hence, the present study evaluates the association between CAR and cardiovascular outcomes in patients undergoing drug-eluting stent (DES) implantation. We consecutively enrolled 9375 CHD patients undergoing DES implantation. All patients were divided into 3 groups according to their CAR: tertile 1 (CAR ≤.02, n=3125), tertile 2 (.02<CAR≤.06, n = 3125), and tertile 3 (CAR >.06, n = 3125). The primary endpoint was major adverse cardiovascular and cerebrovascular events (MACCE). Kaplan-Meier analysis indicated that the incidences of MACCE and MI increased with high tertiles of the CAR (MACCE: 8.7 vs 10.5 vs 12.3%, log-rank P < .001; MI: 3.3 vs 4.0 vs 4.7%, long-rank P = .015). Cox regression analysis suggested that CAR was an independent risk factors for MACCE (HR per standard deviation (SD) increase: 1.07, 95% CI, 1.01-1.14, P = .024), and MI (HR per SD increase: 1.11, 95% CI, 1.01-1.22, P = .028). In conclusion, the CAR is an independent predictor of MACCE and MI in CHD patients.
Collapse
Affiliation(s)
- Jie Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Li
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yitian Zheng
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Gao
- Department of Cardiology and Institute of Vascular Medicine, 66482Peking University Third Hospital; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yu Peng Liu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Jia Wang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Jing Song
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Zhou
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangbin Meng
- Department of Cardiology and Institute of Vascular Medicine, 66482Peking University Third Hospital; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Kuo Zhang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenyao Wang
- Department of Cardiology and Institute of Vascular Medicine, 66482Peking University Third Hospital; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Chunli Shao
- Department of Cardiology and Institute of Vascular Medicine, 66482Peking University Third Hospital; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yi-Da Tang
- Department of Cardiology and Institute of Vascular Medicine, 66482Peking University Third Hospital; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
18
|
Hossain MA, Kim JH. Possibility as role of ginseng and ginsenosides on inhibiting the heart disease of COVID-19: A systematic review. J Ginseng Res 2022; 46:321-330. [PMID: 35068945 PMCID: PMC8767974 DOI: 10.1016/j.jgr.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/25/2022] Open
Abstract
Coronavirus has been spreading rapidly around the world since it broke out in China in 2019. Respiratory diseases caused by coronavirus infection cause various diseases ranging from asymptomatic subclinical infections to severe pneumonia and cardiovascular complications, leading to death. In this regard, natural products are being studied to prevent various diseases caused by COVID-19. In current review, we would like to present mechanisms related to the inhibition of heart disease in ginseng and ginsenoside against SARS-CoV-2. In many previous studies, ginseng and ginsenoside are known to have antioxidant, blood flow improvement, improvement of vascular and heart function, blood pressure control, suppression of myocardial infarction and heart failure, and antiarrhythmia. Therefore, ginseng and ginsenoside have a possibility to suppress cardiovascular complications caused by COVID-19. Many of research provide evidence for ginseng and ginsenoside as treatments for the risk of cardiovascular complications. However, in this review, more specific contents on the proposition of the efficacy of ginseng and ginsenoside for COVID-19 should be presented. Therefore, we hope that researches to reduce cardiovascular complications of ginseng and ginsenoside for COVID-19 should be presented to reduce mortality for COVID-19.
Collapse
Affiliation(s)
- Mohammad Amjad Hossain
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Biosafety Research Institute, Jeonbuk National University, Iksan-city, Republic of Korea
| |
Collapse
|
19
|
Custodero C, Ciavarella A, Panza F, Gnocchi D, Lenato GM, Lee J, Mazzocca A, Sabbà C, Solfrizzi V. Role of inflammatory markers in the diagnosis of vascular contributions to cognitive impairment and dementia: a systematic review and meta-analysis. GeroScience 2022; 44:1373-1392. [PMID: 35486344 PMCID: PMC9213626 DOI: 10.1007/s11357-022-00556-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 03/23/2022] [Indexed: 12/29/2022] Open
Abstract
Vascular contribution to cognitive impairment and dementia (VCID) is a clinical label encompassing a wide range of cognitive disorders progressing from mild to major vascular cognitive impairment (VCI), which is also defined as vascular dementia (VaD). VaD diagnosis is mainly based on clinical and imaging findings. Earlier biomarkers are needed to identify subjects at risk to develop mild VCI and VaD. In the present meta-analysis, we comprehensively evaluated the role of inflammatory biomarkers in differential diagnosis between VaD and Alzheimer’s disease (AD), and assessed their prognostic value on predicting VaD incidence. We collected literature until January 31, 2021, assessing three inflammatory markers [interleukin(IL)-6, C-reactive protein (CRP), tumor necrosis factor (TNF)-α] from blood or cerebrospinal fluid (CSF) samples. Thirteen cross-sectional and seven prospective studies were included. Blood IL-6 levels were cross-sectionally significantly higher in people with VaD compared to AD patients (SMD: 0.40, 95% CI: 0.18 to 0.62) with low heterogeneity (I2: 41%, p = 0.13). Higher IL-6 levels were also associated to higher risk of incident VaD (relative risk: 1.28, 95% CI: 1.03 to 1.59, I2: 0%). IL-6 in CSF was significantly higher in people with VaD compared to healthy subjects (SMD: 0.77, 95% CI: 0.17 to 1.37, I2: 70%), and not compared to AD patients, but due to limited evidence and high inconsistency across studies, we could not draw definite conclusion. Higher blood IL-6 levels might represent a useful biomarker able to differentiate people with VaD from those with AD and might be correlated with higher risk of future VaD.
Collapse
Affiliation(s)
- Carlo Custodero
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Ciavarella
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy.,Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, A. Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Francesco Panza
- Population Health Unit-"Salus In Apulia Study", National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy
| | - Davide Gnocchi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Gennaro M Lenato
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Juhan Lee
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Antonio Mazzocca
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Carlo Sabbà
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Solfrizzi
- Dipartimento Interdisciplinare di Medicina, Clinica Medica e Geriatria "Cesare Frugoni", University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
20
|
Sauter M, Langer HF. Targeting Cell-Specific Molecular Mechanisms of Innate Immunity in Atherosclerosis. Front Physiol 2022; 13:802990. [PMID: 35432000 PMCID: PMC9010538 DOI: 10.3389/fphys.2022.802990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanisms of innate immunity contribute to inflammation, one of the major underlying causes of atherogenesis and progression of atherosclerotic vessel disease. How immune cells exactly contribute to atherosclerosis and interact with molecules of cholesterol homeostasis is still a matter of intense research. Recent evidence has proposed a potential role of previously underappreciated cell types in this chronic disease including platelets and dendritic cells (DCs). The pathophysiology of atherosclerosis is studied in models with dysfunctional lipid homeostasis and several druggable molecular targets are derived from these models. Specific therapeutic approaches focussing on these immune mechanisms, however, have not been successfully introduced into everyday clinical practice, yet. This review highlights molecular insights into immune processes related to atherosclerosis and potential future translational approaches targeting these molecular mechanisms.
Collapse
Affiliation(s)
- M. Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - H. F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- Department of Cardiology, University Heart Center Luebeck, University Hospital, Luebeck, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
- *Correspondence: H. F. Langer,
| |
Collapse
|
21
|
Agnello F, Capodanno D. Anti-inflammatory strategies for atherosclerotic artery disease. Expert Opin Drug Saf 2022; 21:661-672. [DOI: 10.1080/14740338.2022.2036717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Federica Agnello
- Division of Cardiology, A.O.U. Policlinico “G. Rodolico-San Marco”, University of Catania, Catania, Italy
| | - Davide Capodanno
- Division of Cardiology, A.O.U. Policlinico “G. Rodolico-San Marco”, University of Catania, Catania, Italy
| |
Collapse
|
22
|
Janus Kinase Signaling Pathway and Its Role in COVID-19 Inflammatory, Vascular, and Thrombotic Manifestations. Cells 2022; 11:cells11020306. [PMID: 35053424 PMCID: PMC8773838 DOI: 10.3390/cells11020306] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection continues to be a worldwide public health crisis. Among the several severe manifestations of this disease, thrombotic processes drive the catastrophic organ failure and mortality in these patients. In addition to a well-established cytokine storm associated with the disease, perturbations in platelets, endothelial cells, and the coagulation system are key in triggering systemic coagulopathy, involving both the macro- and microvasculatures of different organs. Of the several mechanisms that might contribute to dysregulation of these cells following SARS-CoV-2 infection, the current review focuses on the role of activated Janus kinase (JAK) signaling in augmenting thrombotic processes and organ dysfunction. The review concludes with presenting the current understanding and emerging controversies concerning the potential therapeutic applications of JAK inhibitors for ameliorating the inflammation-thrombosis phenotype in COVID-19 patients.
Collapse
|
23
|
Aydın E, Kant A, Yilmaz G. Evaluation of the cardio-ankle vascular index in COVID-19 patients. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2021; 68:73-76. [PMID: 34909966 DOI: 10.1590/1806-9282.20210781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 09/17/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aimed to investigate the relationship and prognostic significance of cardio-ankle vascular index, which is a measure of arterial stiffness that can lead to endothelial dysfunction and poor cardiovascular issues in COVID-19 patients, with COVID-19. METHODS The study included 115 patients, of which 65 patients in the case group with Real time reversetranscription-polymerasechainreaction test positive and diagnosed for COVID-19 and 50 volunteers in the control group. Patients with COVID-19 were classified as moderate/severe or mild COVID-19 in the subgroup analysis based on the severity of the disease. We investigated the relationship between cardio-ankle vascular index and COVID-19 by using the VaSera VS-1000 device to automatically measure each patient's cardio-ankle vascular index and ankle-brachial pressure index. RESULTS The mean age of participants included in the study was 65.7±10.7 years. Patients and volunteers were statistically similar in terms of age, gender, comorbidities, Charlson comorbidity index scores, and body mass index values (p>0.05). The right-cardio-ankle vascular index value was 9.6±2.4 in the case group and 8.5±1.1 in the control group (p=0.004). The left-cardio-ankle vascular index value was 9.4±2.7 in the case group and 8.5±1.2 in the control group (p=0.01). The right-cardio-ankle vascular index value was 10.8±3.4 in the moderate/severe disease group and 8.8±0.9 in the mild disease group (p=0.008). The left-cardio-ankle vascular index value was 10.7±3.6 in the moderate/severe disease group and 8.5±1.5 in the mild disease group (p<0.001). The right-cardio-ankle vascular index and left-cardio-ankle vascular index values were found to be significantly higher in COVID-19 patients in our study. When receiver operating characteristic analysis was performed to distinguish moderate/severe COVID-19 patients from mild patients, right-cardio-ankle vascular index was area under the curve 0.757 (0.630-0.884), and left-cardio-ankle vascular index was area under the curve 0.782 (0.661-0.902). CONCLUSION The right-cardio-ankle vascular index and left-cardio-ankle vascular index values increased in COVID-19 patients in our study, and this was thought to be prognostically significant.
Collapse
Affiliation(s)
- Ercan Aydın
- Trabzon Kanuni Education and Research Hospital, Department of Cardiology - Trabzon, Turkey
| | - Aydın Kant
- Vakfıkebir State Hospital, Department of Chest Diseases - Trabzon, Turkey
| | - Gürdal Yilmaz
- Karadeniz Technical University, Faculty of Medicine, Department of Infectious Diseases and Clinical Microbiology - Trabzon, Turkey
| |
Collapse
|
24
|
Yilmaz B, Somay E, Selek U, Topkan E. Pretreatment Systemic Immune-Inflammation Index Predict Needs for Teeth Extractions for Locally Advanced Head and Neck Cancer Patients Undergoing Concurrent Chemoradiotherapy. Ther Clin Risk Manag 2021; 17:1113-1121. [PMID: 34703240 PMCID: PMC8536877 DOI: 10.2147/tcrm.s334556] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/10/2021] [Indexed: 12/13/2022] Open
Abstract
Background To evaluate the utility of pretreatment systemic immune-inflammation index (SII) in predicting the teeth caries and need for tooth extraction after concurrent chemoradiotherapy (C-CRT) for locally advanced squamous-cell head and neck cancer (LA-SCHNC) patients. Methods The records of LA-SCHNC patients who underwent formal dental evaluations at pre- and post-C-CRT periods were retrospectively analyzed. The pretreatment SII values were calculated using the platelet, neutrophil, and lymphocyte measures acquired on the first day of C-CRT: SII=Platelets×neutrophils/lymphocytes. Receiver operating characteristic (ROC) curve analysis was employed to identify the ideal pre-C-CRT SII cutoff that may predict the teeth caries and the need for tooth extraction after the C-CRT. The primary endpoint was the link between the pre-C-CRT SII and the need for tooth extraction during the follow-up period. Results A sum of 126 patients were included. Median follow-up was 4.9 years (range: 2.7–7.8). Nasopharyngeal and laryngeal cancers comprised the majority (75.4%) study cohort. Post-treatment teeth extractions were reported in 62.7% patients. The optimal cutoff was 558 [Area under the curve (AUC): %76.8 sensitivity: 72.3%; and specificity: 70.9%] that grouped the patients into two subgroups with significantly different post-C-CRT tooth extraction rates: Group 1: SII≤558 (n = 70) and SII>558 (n = 56), respectively. Correlation analysis revealed a significant relationship between the pretreatment SII and the tooth extraction rates after the C-CRT (rs:0.89: P = 0.001). The comparative analysis displayed that the teeth extractions rates were significantly higher in the SII>558 group (77.1% versus 51.4% for SII≤558; Hazard ratio: 1.68; P = 0.001). Further analyses showed that the pre-C-CRT SII>558 was the unique factor associated with meaningfully higher necessities for post-C-CRT teeth extractions. Conclusion The present outcomes intimated that high pretreatment SII levels were linked to significantly increased post-treatment teeth extractions in LA-SCHNC patients undergoing definitive C-CRT.
Collapse
Affiliation(s)
- Busra Yilmaz
- Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Baskent University, Ankara, Turkey
| | - Efsun Somay
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Baskent University, Ankara, Turkey
| | - Ugur Selek
- Department of Radiation Oncology, School of Medicine, Koc University, Istanbul, Turkey.,Department of Radiation Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Erkan Topkan
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana, Turkey
| |
Collapse
|
25
|
COVID-19 and Acute Coronary Syndromes: From Pathophysiology to Clinical Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4936571. [PMID: 34484561 PMCID: PMC8410438 DOI: 10.1155/2021/4936571] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023]
Abstract
Acute coronary syndromes (ACS) are frequently reported in patients with coronavirus disease 2019 (COVID-19) and may impact patient clinical course and mortality. Although the underlying pathogenesis remains unclear, several potential mechanisms have been hypothesized, including oxygen supply/demand imbalance, direct viral cellular damage, systemic inflammatory response with cytokine-mediated injury, microvascular thrombosis, and endothelial dysfunction. The severe hypoxic state, combined with other conditions frequently reported in COVID-19, namely sepsis, tachyarrhythmias, anemia, hypotension, and shock, can induce a myocardial damage due to the mismatch between oxygen supply and demand and results in type 2 myocardial infarction (MI). In addition, COVID-19 promotes atherosclerotic plaque instability and thrombus formation and may precipitate type 1 MI. Patients with severe disease often show decrease in platelets count, higher levels of d-dimer, ultralarge von Willebrand factor multimers, tissue factor, and prolongation of prothrombin time, which reflects a prothrombotic state. An endothelial dysfunction has been described as a consequence of the direct viral effects and of the hyperinflammatory environment. The expression of tissue factor, von Willebrand factor, thromboxane, and plasminogen activator inhibitor-1 promotes the prothrombotic status. In addition, endothelial cells generate superoxide anions, with enhanced local oxidative stress, and endothelin-1, which affects the vasodilator/vasoconstrictor balance and platelet aggregation. The optimal management of COVID-19 patients is a challenge both for logistic and clinical reasons. A deeper understanding of ACS pathophysiology may yield novel research insights and therapeutic perspectives in higher cardiovascular risk subjects with COVID-19.
Collapse
|
26
|
Kim HJ, Lee JH, Jang SY, Bae MH, Yang DH, Park HS, Cho Y, Jeong MH, Park JS, Kim HS, Hur SH, Seong IW, Cho MC, Chae SC. Prognostic value of novel neutrophil-to-hemoglobin and lymphocyte score in patients with acute myocardial infarction. EUR J INFLAMM 2021. [DOI: 10.1177/20587392211039095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We developed and assessed whether a novel neutrophil-to-hemoglobin and lymphocyte (NHL) score would improve the ability to predict clinical outcome compared with neutrophil-to-lymphocyte ratio (NLR) and systemic immune-inflammation index (SII) in acute myocardial infarction (AMI). We examined 13,072 AMI patients from the Korean AMI Registry–National Institute of Health database. NHL score was calculated as follows: NHL score (U) = N/(Hb × L), where N, Hb, and L are baseline blood neutrophil, hemoglobin, and lymphocyte count. The primary outcome was the occurrence of major adverse cerebrocardiovascular events (MACCEs) at 2 years. The NLR, SII, and NHL score were independent predictors of 2-year MACCEs. The area under the curve of the NHL score (0.637) for predicting 2-year MACCEs was significantly higher compared with those of SII (0.589) and NLR (0.607). The NHL score significantly improved the reclassification and integrated discrimination compared with NLR ( p < 0.0001) and SII ( p < 0.0001). A high NHL score (≥ 0.35 U) was an independent predictor of 2-year MACCEs (adjusted hazard ratio, 1.41; 95% confidence interval, 1.29–1.55; p < 0.001). The NHL score could be a novel model for predicting long-term MACCEs in patients with AMI.
Collapse
Affiliation(s)
- Hyeon Jeong Kim
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jang Hoon Lee
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Se Yong Jang
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Myung Hwan Bae
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Heon Yang
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hun Sik Park
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Yongkeun Cho
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Myung H Jeong
- Department of Internal Medicine, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Jong-Seon Park
- Department of Internal Medicine, Yeungnam University Hospital, Daegu, Republic of Korea
| | - Hyo-Soo Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung-Ho Hur
- Department of Internal Medicine, Keimyung University Dongsan Medical Center, Daegu, Republic of Korea
| | - In-Whan Seong
- Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Myeong-Chan Cho
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Shung Chull Chae
- Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
27
|
Matsuura T, Soeki T, Fukuda D, Uematsu E, Tobiume T, Hara T, Kusunose K, Ise T, Yamaguchi K, Yagi S, Yamada H, Wakatsuki T, Sata M. Activated Factor X Signaling Pathway via Protease-Activated Receptor 2 Is a Novel Therapeutic Target for Preventing Atrial Fibrillation. Circ J 2021; 85:1383-1391. [PMID: 33746155 DOI: 10.1253/circj.cj-20-1006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Activated factor X (FXa), which contributes to chronic inflammation via protease-activated receptor 2 (PAR2), might play an important role in atrial fibrillation (AF) arrhythmogenesis. This study aimed to assess whether PAR2 signaling contributes to AF arrhythmogenesis and whether rivaroxaban ameliorates atrial inflammation and prevents AF. METHODS AND RESULTS In Study 1, PAR2 deficient (PAR2-/-) and wild-type mice were infused with angiotensin II (Ang II) or a vehicle via an osmotic minipump for 2 weeks. In Study 2, spontaneously hypertensive rats (SHRs) were treated with rivaroxaban, warfarin, or vehicle for 2 weeks after 8 h of right atrial rapid pacing. The AF inducibility and atrial remodeling in both studies were examined. Ang II-treated PAR2-/- mice had a lower incidence of AF and less mRNA expression of collagen1 and collagen3 in the atrium compared to wild-type mice treated with Ang II. Rivaroxaban significantly reduced AF inducibility compared with warfarin or vehicle. In SHRs treated with a vehicle, rapid atrial pacing promoted gene expression of inflammatory and fibrosis-related biomarkers in the atrium. Rivaroxaban, but not warfarin, significantly reduced expression levels of these genes. CONCLUSIONS The FXa-PAR2 signaling pathway might contribute to AF arrhythmogenesis associated with atrial inflammation. A direct FXa inhibitor, rivaroxaban, could prevent atrial inflammation and reduce AF inducibility, probably by inhibiting the pro-inflammatory activation.
Collapse
Affiliation(s)
- Tomomi Matsuura
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences
| | - Daiju Fukuda
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Etsuko Uematsu
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takeshi Tobiume
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Tomoya Hara
- Department of Cardiovascular Medicine, Shikoku Medical Center for Children and Adults
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Takayuki Ise
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Koji Yamaguchi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Hirotsugu Yamada
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences
| | - Tetsuzo Wakatsuki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| |
Collapse
|
28
|
Shen Z, Li H. Long non-coding RNA GAS5 knockdown facilitates proliferation and impedes apoptosis by regulating miR-128-3p/FBLN2 axis in ox-LDL-induced THP-1 cells. Clin Hemorheol Microcirc 2021; 77:153-164. [PMID: 33074219 DOI: 10.3233/ch-200897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are found to involve in modulating the development of atherosclerosis (AS). But the molecular mechanism of lncRNA growth-arrest specific transcript 5 (GAS5) in AS is not fully understood. METHODS QRT-PCR was performed to measure the abundances of GAS5, miR-128-3p and fibulin 2 (FBLN2). Oxidized low-density lipoprotein (ox-LDL)-treated THP-1 cells were employed as cell models of AS. The cell proliferation and apoptosis were analyzed using CCK-8 and Flow cytometry assays, respectively. Levels of all protein were examined by western blot. The interaction among GAS5, miR-128-3p and FBLN2 was confirmed via dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. RESULTS GAS5 was elevated and miR-128-3p was decreased in the serum of patients with AS and ox-LDL-stimulated THP-1 cells. Ox-LDL stimulation inhibited proliferation and induced apoptosis of THP-1 cells. Meanwhile, GAS5 directly targeted miR-128-3p and inversely modulated its expression. Importantly, GAS5 depletion facilitated cell proliferation and impaired apoptosis in ox-LDL-induced THP-1 cells. Additionally, GAS5 augmented FBLN2 expression through sponging miR-128-3p, and miR-128-3p facilitated proliferation and retarded apoptosis of ox-LDL-induced THP-1 cells by targeting FBLN2. CONCLUSION GAS5 knockdown promoted the growth of ox-LDL-induced THP-1 cells through down-modulating FBLN2 and increasing miR-128-3p, suggesting the potential value of GAS5 for treatment of AS.
Collapse
Affiliation(s)
- Zijian Shen
- Deparment of Vasculocardiology, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow, Jiangsu, China
| | - Haigang Li
- Deparment of Vasculocardiology, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow, Jiangsu, China
| |
Collapse
|
29
|
Migita S, Kitano D, Li Y, Koyama Y, Shimodai-Yamada S, Onishi A, Fuchimoto D, Suzuki S, Nakamura Y, Matsuyama TA, Hirota S, Sakuma M, Tsujimoto M, Hirayama A, Okumura Y, Hao H. Pathological findings after third- and second-generation everolimus-eluting stent implantations in coronary arteries from autopsy cases and an atherosclerotic porcine model. Sci Rep 2021; 11:6281. [PMID: 33737695 PMCID: PMC7973802 DOI: 10.1038/s41598-021-85740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/23/2021] [Indexed: 01/10/2023] Open
Abstract
Pathological changes after third-generation drug-eluting stent implantation remain unclear. We compared the tissue responses of coronary arteries after the implantation of third-generation abluminal biodegradable-polymer everolimus-eluting stent (3rd EES) and second-generation durable-polymer EES (2nd EES) using autopsy specimens and an atherosclerotic porcine model. We compared the histology of stented coronary arteries obtained by autopsy performed 1–10 months after 3rd EES (n (number of cases) = 4, stent-implanted period of 3–7 months) and 2nd EES (n (number of cases) = 9, stent-implanted period of 1–10 months) implantations. The ratio of covered stent struts was higher with 3rd EESs than with 2nd EESs (3rd; 0.824 ± 0.032 vs. 2nd; 0.736 ± 0.022, p = 0.035). Low-density lipoprotein receptor knockout minipigs were stented with 3rd or 2nd EES in the coronary arteries and the stented regions were investigated. The fibrin deposition around the 2nd EES was more prominent. Additionally, higher density of smooth muscle cells was confirmed after the 3rd EES implantation. Pathological examination after the 3rd EES demonstrated a combination of less fibrin deposition and more rapid acquisition of well-developed neointima as compared to the 2nd EES at autopsy and the atherosclerotic porcine model.
Collapse
Affiliation(s)
- Suguru Migita
- Division of Human Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan.,Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Daisuke Kitano
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yuxin Li
- Division of Cell Regeneration and Transplantation, Department of Functional Morphology, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan.
| | - Yutaka Koyama
- Division of Human Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan.,Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Sayaka Shimodai-Yamada
- Division of Human Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Akira Onishi
- Department of Animal Science and Resources, College of Bioresource Sciences, Nihon University, Kanagawa, Japan
| | - Daiichiro Fuchimoto
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Ibaraki, Japan
| | - Shunichi Suzuki
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization (NARO), Ibaraki, Japan
| | - Yoshiyuki Nakamura
- Swine and Poultry Research, Saitama Prefectural Agricultural Technology Research Center, Saitama, Japan
| | - Taka-Aki Matsuyama
- Department of Legal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masashi Sakuma
- Department of Cardiovascular Medicine, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | | | | | - Yasuo Okumura
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroyuki Hao
- Division of Human Pathology, Department of Pathology and Microbiology, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo, 173-8610, Japan.
| |
Collapse
|
30
|
Hong LZ, Xue Q, Shao H. Inflammatory Markers Related to Innate and Adaptive Immunity in Atherosclerosis: Implications for Disease Prediction and Prospective Therapeutics. J Inflamm Res 2021; 14:379-392. [PMID: 33628042 PMCID: PMC7897977 DOI: 10.2147/jir.s294809] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/21/2021] [Indexed: 12/20/2022] Open
Abstract
Several lines of evidence have linked a dysregulated inflammatory setting to the pathogenesis of atherosclerosis, which is a form of chronic vascular inflammation. Various inflammatory biomarkers have been associated with inflammation and are recognized as potential tools to monitor the progression of atherosclerosis. A well-studied inflammatory marker in the context of cardiovascular diseases is C-reactive protein (CRP) or, more accurately, highly sensitive-CRP (hs-CRP), which has been established as an inflammatory biomarker for atherosclerotic events. In addition, a growing body of investigations has attempted to disclose the potential of inflammatory cytokines, enzymes, and genetic polymorphisms related to innate and adaptive immunity as biomarkers for predicting the development of atherosclerosis. In this review article, we clarify both traditional and novel inflammatory biomarkers related to components of the innate and adaptive immune system that may mirror the progression or phases of atherosclerotic inflammation/lesions. Furthermore, the contribution of the inflammatory biomarkers in developing potential therapeutics against atherosclerotic treatment will be discussed.
Collapse
Affiliation(s)
- Ling-Zhi Hong
- Emergency Department, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, 311700, Zhejiang Province, People’s Republic of China
| | - Qi Xue
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, People’s Republic of China
| | - Hong Shao
- Department of Cardiology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, People’s Republic of China
| |
Collapse
|
31
|
Qiu H, Tu Q, Gao P, Li X, Maitz MF, Xiong K, Huang N, Yang Z. Phenolic-amine chemistry mediated synergistic modification with polyphenols and thrombin inhibitor for combating the thrombosis and inflammation of cardiovascular stents. Biomaterials 2020; 269:120626. [PMID: 33418199 DOI: 10.1016/j.biomaterials.2020.120626] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
Antithrombogenicity, anti-inflammation, and rapid re-endothelialization are central requirements for the long-term success of cardiovascular stents. In this work, a plant-inspired phenolic-amine chemistry strategy was developed to combine the biological functions of a plant polyphenol, tannic acid (TA), and the thrombin inhibitor bivalirudin (BVLD) for tailoring the desired multiple surface functionalities of cardiovascular stents. To realize the synergistic modification of TA and BVLD on a stent surface, an amine-bearing coating of plasma polymerized allylamine was firstly prepared on the stent surface, followed by the sequential conjugation of TA and BVLD in alkaline solution based on phenolic-amine chemistry (i.e., Michael addition reaction). TA and BVLD were successfully immobilized onto the stent surface with considerable amounts of 330 ± 12 and 930 ± 80 ng/cm2, respectively. The abundant phenolic hydroxyl groups of TA imparted the stent with ability to suppress inflammation. Meanwhile, BVLD provided an antithrombogenic and endothelial-friendly microenvironment. As a result, the combined functions of the TA and BVLD facilitate the rapid stent re-endothelialization for reduced intimal hyperplasia in vivo, and may be a promising strategy to address the clinical complications associated with restenosis and late stent thrombosis.
Collapse
Affiliation(s)
- Hua Qiu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qiufen Tu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Peng Gao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Xiangyang Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Manfred F Maitz
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Max Bergmann Center of Biomaterials, Leibniz Institute of Polymer Research Dresden, Dresden, 01069, Germany
| | - Kaiqin Xiong
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Nan Huang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhilu Yang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
32
|
Kelham M, Choudry FA, Hamshere S, Beirne AM, Rathod KS, Baumbach A, Ahluwalia A, Mathur A, Jones DA. Therapeutic Implications of COVID-19 for the Interventional Cardiologist. J Cardiovasc Pharmacol Ther 2020; 26:203-216. [PMID: 33331160 DOI: 10.1177/1074248420982736] [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] [Indexed: 12/23/2022]
Abstract
Although COVID-19 is viewed primarily as a respiratory disease, cardiovascular risk factors and disease are prevalent among infected patients and are associated with worse outcomes. In addition, among multiple extra-pulmonary manifestations, there has been an increasing recognition of specific cardiovascular complications of COVID-19. Despite this, in the initial stages of the pandemic there was evidence of a reduction in patients presenting to acute cardiovascular services. In this masterclass review, with the aid of 2 exemplar cases, we will focus on the important therapeutic implications of COVID-19 for interventional cardiologists. We summarize the existing evidence base regarding the varied cardiovascular presentations seen in COVID-19 positive patients and the prognostic importance and potential mechanisms of acute myocardial injury in this setting. Importantly, through the use of a systematic review of the literature, we focus our discussion on the observed higher rates of coronary thrombus burden in patients with COVID-19 and acute coronary syndromes.
Collapse
Affiliation(s)
- Matthew Kelham
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Fizzah A Choudry
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Stephen Hamshere
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom
| | - Anne-Marie Beirne
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Krishnaraj S Rathod
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Andreas Baumbach
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Amrita Ahluwalia
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Anthony Mathur
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| | - Daniel A Jones
- Department of Cardiology, 560754Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom.,Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, 4617Queen Mary University of London, London, United Kingdom
| |
Collapse
|
33
|
Casolo G, Del Meglio J, Tessa C. Epidemiology and pathophysiologic insights of coronary atherosclerosis relevant for contemporary non-invasive imaging. Cardiovasc Diagn Ther 2020; 10:1906-1917. [PMID: 33381434 PMCID: PMC7758762 DOI: 10.21037/cdt-20-157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/17/2020] [Indexed: 12/20/2022]
Abstract
In the past few years significant changes have taken place in the diagnostic and therapeutic approach to patients with coronary artery disease (CAD) and/or ischemic heart disease (IHD). New discoveries about the development and progression of coronary atherosclerosis have changed the clinical landscape. At the same time a marked decrease in cardiovascular (CV) mortality and CAD incidence have been observed in many Countries but particularly in the most industrialized ones. This fall has been also observed in the incidence of stroke, sudden death, myocardial ischemia, myocardial infarction (MI), and prevalence of CAD. As a consequence, an increasing number of patients with chest pain exhibits non-significant stenosis at both invasive and non-invasive coronary angiography and the rate of coronary vessels revascularizations has greatly reduced. Coronary atherosclerosis and its characteristics have shown to be both diagnostic and therapeutic targets beyond obstructive CAD. The decreased prevalence of CAD in the general population has modified the pre-test probability (PTP) of disease. In this landscape the conventional stress imaging tests appear to have limited accuracy making the diagnosis of obstructive CAD very challenging. These diagnostic tests have been introduced and tested in a population with a much higher probability of disease and therefore the contemporary accuracy of these old tests appear much lower than in the past. In addition, in the past few years the relevance of the traditional ischemia guided coronary intervention strategy has been questioned. Given the low CV events granted by an optimal medical therapy in CAD the major attention has been directed on detecting coronary atherosclerosis. The earlier the better. At the same time, a growing number of data from clinical studies have shown a significant prognostic role for non-obstructive CAD and coronary atherosclerosis. All these facts have shifted the clinicians' attention from the functional evaluation of the coronary circulation to the anatomic burden of disease.
Collapse
Affiliation(s)
- Giancarlo Casolo
- Cardiology Department, Versilia Hospital, Lido di Camaiore, Italy
| | | | - Carlo Tessa
- Radiology Department, Versilia Hospital, Lido di Camaiore, Italy
| |
Collapse
|
34
|
Weaver JC, Ullah I, Qi M, Giannakopoulos B, Rye KA, Kockx M, Kritharides L, Krilis SA. Free Thiol β2-GPI (β-2-Glycoprotein-I) Provides a Link Between Inflammation and Oxidative Stress in Atherosclerotic Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2020; 40:2794-2804. [DOI: 10.1161/atvbaha.120.315156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective:
Atherosclerotic coronary artery disease is well recognised as an inflammatory disorder that is also influenced by oxidative stress. β2-GPI (β-2-glycoprotein-I) is a circulating plasma protein that undergoes post-translational modification and exists in free thiol as well as oxidized forms. The aim of this study was to assess the association between these 2 post-translational redox forms of β2-GPI and atherosclerotic coronary artery disease.
Approach and Results:
Stable patients presenting for elective coronary angiography or CT coronary angiography were prospectively recruited. A separate group of patients after reperfused ST-segment–elevation myocardial infarction formed an acute coronary syndrome subgroup. All patients had collection of fasting serum and plasma for quantification of total and free thiol β2-GPI. Coronary artery disease extent was quantified by the Syntax and Gensini scores. A total of 552 patients with stable disease and 44 with acute coronary syndrome were recruited. While total β2-GPI was not associated with stable coronary artery disease, a higher free thiol β2-GPI was associated with its presence and extent. This finding remained significant after correcting for confounding variables, and free thiol β2-GPI was a better predictor of stable coronary artery disease than hs-CRP (high-sensitivity C-reactive protein). Paradoxically, there were lower levels of free thiol β2-GPI after ST-segment–elevation myocardial infarction.
Conclusions:
Free thiol β2-GPI is a predictor of coronary artery disease presence and extent in stable patients. Free thiol β2-GPI was a better predictor than high-sensitivity C-reactive protein.
Collapse
Affiliation(s)
- James C. Weaver
- University of NSW, Sydney, Australia (J.C.W., I.U., M.Q., B.G., K.A.R., S.A.K.)
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, Sydney, Australia (J.C.W.)
| | - Inaam Ullah
- University of NSW, Sydney, Australia (J.C.W., I.U., M.Q., B.G., K.A.R., S.A.K.)
- Department of Cardiology, St George Hospital, Kogarah, Sydney, Australia (I.U.)
| | - Miao Qi
- University of NSW, Sydney, Australia (J.C.W., I.U., M.Q., B.G., K.A.R., S.A.K.)
- Department of Infectious Disease, Immunology and Sexual Health, St George Hospital, Kogarah, Sydney, Australia (M.Q., B.G., S.A.K.)
| | - Bill Giannakopoulos
- University of NSW, Sydney, Australia (J.C.W., I.U., M.Q., B.G., K.A.R., S.A.K.)
- Department of Infectious Disease, Immunology and Sexual Health, St George Hospital, Kogarah, Sydney, Australia (M.Q., B.G., S.A.K.)
- Department of Rheumatology, St George Hospital, Kogarah, Sydney, Australia (B.G.)
| | - Kerry Anne Rye
- University of NSW, Sydney, Australia (J.C.W., I.U., M.Q., B.G., K.A.R., S.A.K.)
| | - Maaike Kockx
- ANZAC Research Institute, University of Sydney, Concord, Australia (M.K., L.K.)
| | - Leonard Kritharides
- ANZAC Research Institute, University of Sydney, Concord, Australia (M.K., L.K.)
- Department of Cardiology, Concord Hospital, Sydney, Australia (L.K.)
| | - Steven A. Krilis
- University of NSW, Sydney, Australia (J.C.W., I.U., M.Q., B.G., K.A.R., S.A.K.)
- Department of Infectious Disease, Immunology and Sexual Health, St George Hospital, Kogarah, Sydney, Australia (M.Q., B.G., S.A.K.)
| |
Collapse
|
35
|
Zhang Q, Si D, Zhang Z, Wang C, Zheng H, Li S, Huang S, Zhang W. Value of the platelet-to-lymphocyte ratio in the prediction of left ventricular thrombus in anterior ST-elevation myocardial infarction with left ventricular dysfunction. BMC Cardiovasc Disord 2020; 20:428. [PMID: 32993501 PMCID: PMC7526106 DOI: 10.1186/s12872-020-01712-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/22/2020] [Indexed: 12/23/2022] Open
Abstract
Background The predictors of left ventricular thrombus (LVT) formation are not well defined in the contemporary era, especially in those patients at high risk. We aimed to evaluate whether the platelet/lymphocyte ratio (PLR) is valuable in the determination of LVT formation in patients with anterior ST-elevation myocardial infarction (STEMI) and left ventricular (LV) dysfunction. Methods The LVT group (n = 46) was identified from anterior STEMI patients with LV dysfunction who were treated with primary percutaneous coronary intervention (PCI) from January 2017 to December 2019 at the China-Japan Union Hospital of Jilin University. The no-LVT group (n = 92) were also selected from the same batch of patients and were age- and sex-matched to the patients with LVT. The PLR was determined at admission and was calculated as the ratio of the platelet count to the lymphocyte count using the complete blood count. The presence of LVT was determined by echocardiography. Results The PLR were significantly higher in patients with LVT than in no-LVT group (p = 0.001). In a receiver operator characteristic curve (ROC) analysis, using a cut-off value of 118.07 (AUC 0.673, 95% CI: 0.574–0.771, P = 0.001), the PLR could independently predict the occurrence of LVT. Multivariate analysis showed that an increased PLR (OR = 1.011, 95% CI: 1.004–1.018, P = 0.002), the presence of a left ventricular aneurysm (OR = 46.350, 95% CI: 5.659–379.615, P < 0.001) and increased DTBT (OR = 1.005, 95% CI: 1.001–1.009, P = 0.012) were independent predictors of LVT formation. Conclusions In acute anterior STEMI patients with LV dysfunction, an increased PLR and DTBT and the presence of an LV aneurysm were independent predictors of LVT formation. A larger prospective study is warranted to evaluate this result. Trial registration This study was registered (May 4, 2019) on Chinese Clinical Trial Registry (ChiCTR-DDD-17011214).
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China
| | - Daoyuan Si
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China
| | - Zhongfan Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China
| | - Chengbing Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Haikuo Zheng
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China
| | - Shouping Li
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China
| | - Shijian Huang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China
| | - Wenqi Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Xiantai Street NO.126, Changchun, Jilin, China.
| |
Collapse
|
36
|
Libby P, Lüscher T. COVID-19 is, in the end, an endothelial disease. Eur Heart J 2020; 41:3038-3044. [PMID: 32882706 PMCID: PMC7470753 DOI: 10.1093/eurheartj/ehaa623] [Citation(s) in RCA: 635] [Impact Index Per Article: 158.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/07/2020] [Accepted: 07/17/2020] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelium provides the crucial interface between the blood compartment and tissues, and displays a series of remarkable properties that normally maintain homeostasis. This tightly regulated palette of functions includes control of haemostasis, fibrinolysis, vasomotion, inflammation, oxidative stress, vascular permeability, and structure. While these functions participate in the moment-to-moment regulation of the circulation and coordinate many host defence mechanisms, they can also contribute to disease when their usually homeostatic and defensive functions over-reach and turn against the host. SARS-CoV-2, the aetiological agent of COVID-19, causes the current pandemic. It produces protean manifestations ranging from head to toe, wreaking seemingly indiscriminate havoc on multiple organ systems including the lungs, heart, brain, kidney, and vasculature. This essay explores the hypothesis that COVID-19, particularly in the later complicated stages, represents an endothelial disease. Cytokines, protein pro-inflammatory mediators, serve as key danger signals that shift endothelial functions from the homeostatic into the defensive mode. The endgame of COVID-19 usually involves a cytokine storm, a phlogistic phenomenon fed by well-understood positive feedback loops that govern cytokine production and overwhelm counter-regulatory mechanisms. The concept of COVID-19 as an endothelial disease provides a unifying pathophysiological picture of this raging infection, and also provides a framework for a rational treatment strategy at a time when we possess an indeed modest evidence base to guide our therapeutic attempts to confront this novel pandemic.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Lüscher
- Heart Division, Royal Brompton & Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
37
|
Chang C, Pan Y, Du H, Wang X, Li X. Serum amyloid A1 can be a novel biomarker for evaluating the presence and severity of acute coronary syndrome. Clin Biochem 2020; 85:27-32. [PMID: 32805223 DOI: 10.1016/j.clinbiochem.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Serum amyloid A (SAA) is an acute phase protein and a novel inflammatory biomarker of cardiovascular diseases. Of the four subtypes, SAA1 is the most representative biomarker. In this study, we aimed to assess the value of SAA1 as a novel biomarker for evaluating the presence and severity of acute coronary syndrome (ACS) in Chinese patients. METHODS AND RESULTS A total of 140 ACS patients and 88 non-ACS patients (including 36 stable coronary artery disease (SCAD) patients and 52 healthy controls) who underwent coronary angiography were enrolled. The SAA1 level was significantly higher in ACS patients compared with the SCAD and healthy control subgroups (P < 0.001, respectively), and was significantly higher in the high SYNTAX Score II (SS II) group compared with the medium SS II group and low SS II group (P < 0.001, respectively) in ACS patients. The cutoff level of SAA1 for indicating the presence of ACS was 324.65 ng/mL (sensitivity of 77.9%, specificity of 60.2% and an area under the curve of 0.717). The increased SAA1 levels were positively associated with the presence (OR = 1.013, P < 0.001) and severity (OR = 1.023, P < 0.001) of ACS. Furthermore, there was a positive correlation between SAA1 levels and SS II (r = 0.467, P < 0.001). CONCLUSIONS Our results suggest that elevated SAA1 levels may be a novel biomarker for evaluating the presence of ACS and the severity of CAD in ACS patients. Measuring SAA1 levels makes it possible to evaluate the presence of ACS and severity of CAD in ACS patients.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Hongjiao Du
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiao Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China
| | - Xiaodong Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang 110004, People's Republic of China.
| |
Collapse
|
38
|
Marchini JF, Manica A, Crestani P, Dutzmann J, Folco EJ, Weber H, Libby P, Croce K. Oxidized Low-Density Lipoprotein Induces Macrophage Production of Prothrombotic Microparticles. J Am Heart Assoc 2020; 9:e015878. [PMID: 32750308 PMCID: PMC7792235 DOI: 10.1161/jaha.120.015878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Activated vascular cells produce submicron prothrombotic and proinflammatory microparticle vesicles. Atherosclerotic plaques contain high levels of microparticles. Plasma microparticle levels increase during acute coronary syndromes and the thrombotic consequences of plaque rupture likely involve macrophage-derived microparticles (MΦMPs). The activation pathways that promote MΦMP production remain poorly defined. This study tested the hypothesis that signals implicated in atherogenesis also stimulate MΦMP production. Methods and Results We stimulated human primary MΦs with proinflammatory cytokines and atherogenic lipids, and measured MΦMP production by flow cytometry. Oxidized low-density lipoprotein (oxLDL; 25 µg/mL) induced MΦMP production in a concentration-dependent manner (293% increase; P<0.001), and these oxLDL MΦMP stimulatory effects were mediated by CD36. OxLDL stimulation increased MΦMP tissue factor content by 78% (P<0.05), and oxLDL-induced MΦMP production correlated with activation of caspase 3/7 signaling pathways. Salvionolic acid B, a CD36 inhibitor and a CD36 inhibitor antibody reduced oxLDL-induced MΦMP by 67% and 60%, respectively. Caspase 3/7 inhibition reduced MΦMP release by 52% (P<0.01) and caspase 3/7 activation increased MΦMP production by 208% (P<0.01). Mevastatin pretreatment (10 µM) decreased oxLDL-induced caspase 3/7 activation and attenuated oxLDL-stimulated MΦMP production and tissue factor content by 60% (P<0.01) and 43% (P<0.05), respectively. Conclusions OxLDL induces the production of prothrombotic microparticles in macrophages. This process depends on caspases 3 and 7 and CD36 and is inhibited by mevastatin pretreatment. These findings link atherogenic signaling pathways, inflammation, and plaque thrombogenicity and identify a novel potential mechanism for antithrombotic effects of statins independent of LDL lowering.
Collapse
Affiliation(s)
- Julio F Marchini
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA.,Departamento de Clínica Médica Instituto Central do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo SP Brazil
| | - Andre Manica
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA.,Instituto de Cardiologia/Fundação Universitária de Cardiologia Porto Alegre RS Brazil
| | - Paulo Crestani
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Jochen Dutzmann
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Eduardo J Folco
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Heinz Weber
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Peter Libby
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| | - Kevin Croce
- Cardiovascular Division Department of Medicine Brigham and Women's HospitalHarvard Medical School Boston MA
| |
Collapse
|
39
|
Platelet-to-lymphocyte ratio is a predictor of long-term mortality in patients with acute coronary syndrome. ADVANCES IN INTERVENTIONAL CARDIOLOGY 2020; 16:170-176. [PMID: 32636901 PMCID: PMC7333190 DOI: 10.5114/aic.2020.95859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/27/2020] [Indexed: 01/23/2023] Open
Abstract
Introduction Atherosclerosis is a chronic inflammatory process and inflammation is an important component of acute coronary syndrome (ACS). Platelet-to-lymphocyte ratio (PLR) is a useful parameter showing the degree of the inflammatory response. Aim To explore the association between PLR and long-term mortality in patients with ACS. Material and methods A total of 538 patients who had a diagnosis of ACS between January 2012 and August 2013 were followed up to 60 months. On admission, blood sampling to calculate PLR and detailed clinical data were obtained. Results In total, 538 patients with a mean age of 61.5 ±13.1 years (69% male) were enrolled in the study. Median follow-up was 79 months (IQR: 74–83 months). Patients were divided into 3 tertiles based on PLR levels. Five-year mortality of the patients was significantly higher among patients in the upper PLR tertile when compared with the lower and middle PLR tertile groups (55 (30.7%) vs. 27 (15.0%) and 34 (19.0%); p < 0.001, p = 0.010 respectively). In the Cox regression analysis, a high level of PLR was an independent predictor of 5-year mortality (OR = 1.005, 95% CI: 1.001–1.008, p = 0.004). Kaplan-Meier analysis according to the long-term mortality-free survival revealed the higher occurrence of mortality in the third PLR tertile group compared to the first (p < 0.001) and second tertiles (p = 0.009). Conclusions PLR, which is an easily calculated and universally available marker, may be useful in long-term risk classification of patients presenting with ACS.
Collapse
|
40
|
Maduzia D, Ceranowicz P, Cieszkowski J, Gałązka K, Kuśnierz-Cabala B, Warzecha Z. Pretreatment with Warfarin Attenuates the Development of Ischemia/Reperfusion-Induced Acute Pancreatitis in Rats. Molecules 2020; 25:E2493. [PMID: 32471279 PMCID: PMC7321200 DOI: 10.3390/molecules25112493] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022] Open
Abstract
In acute pancreatitis (AP), pancreatic damage leads to local vascular injury, manifesting as endothelial damage and activation, increased vascular permeability, leukocyte rolling, sticking and transmigration to pancreatic tissue as well as activation of coagulation. Previous studies have shown that pretreatment with heparin or acenocoumarol inhibits the development of AP. The aim of the present study was to check the impact of pretreatment with warfarin, an oral vitamin K antagonist, on the development of ischemia/reperfusion-induced AP in rats. AP was induced by pancreatic ischemia followed by reperfusion of the gland. Warfarin (90, 180 or 270 µg/kg/dose) or vehicle were administered intragastrically once a day for 7 days before induction of AP. The effect of warfarin on the severity of AP was assessed 6 h after pancreatic reperfusion. The assessment included histological, functional, and biochemical analyses. Pretreatment with warfarin given at a dose of 90 or 180 µg/kg/dose increased the international normalized ratio and reduced morphological signs of pancreatic damage such as pancreatic edema, vacuolization of acinar cells, necrosis and the number of hemorrhages. These effects were accompanied by an improvement of pancreatic blood flow and a decrease in serum level amylase, lipase, pro-inflammatory interleukin-1β and plasma level of D-dimer. In contrast, pretreatment with warfarin given at a dose of 270 µg/kg/dose led to an increase in severity of pancreatic damage and biochemical indicators of AP. In addition, this dose of warfarin resulted in deaths in some animals. Pretreatment with low doses of warfarin inhibits the development of AP induced by pancreatic ischemia followed by reperfusion.
Collapse
Affiliation(s)
- Dawid Maduzia
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.M.); (J.C.); (Z.W.)
- Department of Anatomy, Faculty of Medicine, Jagiellonian University Medical College, 31-034 Cracow, Poland
| | - Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.M.); (J.C.); (Z.W.)
| | - Jakub Cieszkowski
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.M.); (J.C.); (Z.W.)
| | - Krystyna Gałązka
- Department of Pathology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland;
| | - Beata Kuśnierz-Cabala
- Department of Diagnostics, Chair of Clinical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Cracow, Poland;
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 31-531 Cracow, Poland; (D.M.); (J.C.); (Z.W.)
| |
Collapse
|
41
|
Wu H, Wang Y, Zhang Y, Xu F, Chen J, Duan L, Zhang T, Wang J, Zhang F. Breaking the vicious loop between inflammation, oxidative stress and coagulation, a novel anti-thrombus insight of nattokinase by inhibiting LPS-induced inflammation and oxidative stress. Redox Biol 2020; 32:101500. [PMID: 32193146 PMCID: PMC7078552 DOI: 10.1016/j.redox.2020.101500] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/01/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022] Open
Abstract
Thrombosis is a principle cause of cardiovascular disease, the leading cause of morbidity and mortality worldwide; however, the conventional anti-thrombotic approach often leads to bleeding complications despite extensive clinical management and monitoring. In view of the intense crosstalk between inflammation and coagulation, plus the contributing role of ROS to both inflammation and coagulation, it is highly desirable to develop safer anti-thrombotic agent with preserved anti-inflammatory and anti-oxidative stress activities. Nattokinase (NK) possesses many beneficial effects on cardiovascular system due to its strong thrombolytic and anticoagulant activities. Herein, we demonstrated that NK not only effectively prevented xylene-induced ear oedema in mice, but also remarkably protected against LPS-induced acute kidney injury in mice through restraining inflammation and oxidative stress, a central player in the initiation and progression of inflammation. Fascinatingly, in line with our in vivo data, NK elicited prominent anti-inflammatory activity in RAW264.7 macrophages via suppressing the LPS-induced TLR4 and NOX2 activation, thereby repressing the corresponding ROS production, MAPKs activation, and NF-κB translocation from the cytoplasm to the nucleus, where it mediates the expression of pro-inflammatory mediators, such as TNF-α, IL-6, NO, and PAI-1 in activated macrophage cells. In particular, consistent with the macrophage studies, NK markedly inhibited serum PAI-1 levels induced by LPS, thereby blocking the deposition of fibrin in the glomeruli of endotoxin-treated animals. In summary, we extended the anti-thrombus mechanism of NK by demonstrating the anti-inflammatory and anti-oxidative stress effects of NK in ameliorating LPS-activated macrophage signaling and protecting against LPS-stimulated AKI as well as glomeruler thrombus in mice, opening a comprehensive anti-thrombus strategy by breaking the vicious cycle between inflammation, oxidative stress and thrombosis. NK protects against LPS-induced AKI via inhibiting inflammation and oxidative stress. NK inhibits LPS-induced TRL4 and NOX2 activation in macrophages. NK inhibits inflammation and oxidative stress both in vitro and in vivo. NK inhibits LPS-induced PAI-I levels, thereby blocking glomerular thrombus in mice. NK may break the vicious loop between inflammation, oxidative stress and coagulation.
Collapse
Affiliation(s)
- Hao Wu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Ying Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yupeng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Feng Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Jiepeng Chen
- Sungen Biotech Co., Ltd, Shantou, 515000, PR China
| | - Lili Duan
- Sungen Biotech Co., Ltd, Shantou, 515000, PR China
| | - Tingting Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Fengjiao Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
42
|
Ye Y, Wang Y, Yang Y, Tao L. Aloperine suppresses LPS-induced macrophage activation through inhibiting the TLR4/NF-κB pathway. Inflamm Res 2020; 69:375-383. [PMID: 32144444 DOI: 10.1007/s00011-019-01313-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/13/2019] [Accepted: 12/26/2019] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE The currently available anti-inflammatory drugs often cause diverse side effects with long-term use. Exploring anti-inflammatory drugs with better efficacy and lower toxicity presents an ongoing challenge. Aloperine is an alkaloid extracted from the leaves and seeds of Sophora alopecuroides L. However, the anti-inflammatory effects of Aloperine have not been fully elucidated. This study aimed to investigate whether Aloperine suppresses lipopolysaccharide (LPS)-induced inflammatory responses in RAW264.7 macrophages. METHODS RAW264.7 macrophages were stimulated with LPS (1 μg/mL) in the presence or absence of Aloperine (50 and 100 μM). mRNA expression was measured by real-time PCR, and protein expression was assessed by western blot analysis. The secretion of pro-inflammatory cytokines was measured by ELISA. The levels of nitric oxide (NO) and reactive oxygen species (ROS) were measured by staining. The transcriptional activity of NF-κB was assayed by a luciferase activity assay. RESULTS The results proved that Aloperine inhibited the expression of LPS-induced pro-inflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-17A (IL-17A)] in macrophages. Treatment with Aloperine inhibited NO production through suppressing inducible nitric oxide synthase (iNOS) expression and the secretion of prostaglandin E2 (PGE2) by inhibiting cyclooxygenase 2 (COX-2) expression. Aloperine prevented LPS-induced oxidative stress by reducing the generation of ROS. Furthermore, aloperine significantly reduced Toll-like receptor 4 (TLR4) and myeloid differentiation factor (Myd-88) levels and prevented the nuclear translocation of nuclear factor-κB (NF-κB) in LPS-treated macrophages. CONCLUSION Taken together, our findings show that Aloperine could suppress LPS-induced macrophage activation by inhibiting the TLR4/Myd-88/NF-κB pathway.
Collapse
Affiliation(s)
- Yinyin Ye
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, # 92 Zheshan Road, Wuhu, 241002, Anhui Province, People's Republic of China.
| | - Yuwei Wang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, # 92 Zheshan Road, Wuhu, 241002, Anhui Province, People's Republic of China
| | - Yanlang Yang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, # 92 Zheshan Road, Wuhu, 241002, Anhui Province, People's Republic of China
| | - Liangfei Tao
- Medical Emergency Center, #158 Yueming Road, Baoshan District, Shanghai, 201900, People's Republic of China.
| |
Collapse
|
43
|
Alamgeer, Asif H, Sandhu MZA, Aziz M, Irfan HM, Moreno KGT, Junior AG. Ameliorative Effects and Cellular Aspects of Phytoconstituents in Atherosclerosis. Curr Pharm Des 2020; 26:2574-2582. [PMID: 32056518 DOI: 10.2174/1381612826666200214161139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 01/17/2020] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a cardiovascular disease that involves vessels through the development of fatty streaks and plaques. Plant-based compounds can help treat or prevent atherosclerosis by affecting various factors that are involved in the disease. The present review discusses our current knowledge of the major cellular and molecular mechanisms of phytotherapeutics for the treatment of atherosclerosis. Numerous studies have evaluated the antiatherosclerotic activity of phytoconstituents to provide preliminary evidence of efficacy, but only a few studies have delineated the underlying molecular mechanisms. Plant-derived phytotherapeutics primarily targets abnormal levels of lipoproteins, endothelial dysfunction, smooth muscle cell migration, foam cell development, and atheromatous plaque formation. Nonetheless, the principal mechanisms that are responsible for their therapeutic actions remain unclear. Further pharmacological studies are needed to elucidate the underlying molecular mechanisms of the antiatherosclerotic response to these phytoconstituents.
Collapse
Affiliation(s)
- Alamgeer
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Hira Asif
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan,Department of Pharmacy, University of Lahore, Gujrat Campus, Gujrat, Pakistan
| | - Muhammad Z A Sandhu
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Madiha Aziz
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Hafiz M Irfan
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Karyne G T Moreno
- Laboratory of Cardiovascular Research and Integrative Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Arquimedes Gasparotto Junior
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, MS, Brazil
| |
Collapse
|
44
|
Abstract
Platelets - blood cells continuously produced from megakaryocytes mainly in the bone marrow - are implicated not only in haemostasis and arterial thrombosis, but also in other physiological and pathophysiological processes. This Review describes current evidence for the heterogeneity in platelet structure, age, and activation properties, with consequences for a diversity of platelet functions. Signalling processes of platelet populations involved in thrombus formation with ongoing coagulation are well understood. Genetic approaches have provided information on multiple genes related to normal haemostasis, such as those encoding receptors and signalling or secretory proteins, that determine platelet count and/or responsiveness. As highly responsive and secretory cells, platelets can alter the environment through the release of growth factors, chemokines, coagulant factors, RNA species, and extracellular vesicles. Conversely, platelets will also adapt to their environment. In disease states, platelets can be positively primed to reach a pre-activated condition. At the inflamed vessel wall, platelets interact with leukocytes and the coagulation system, interactions mediating thromboinflammation. With current antiplatelet therapies invariably causing bleeding as an undesired adverse effect, novel therapies can be more beneficial if directed against specific platelet responses, populations, interactions, or priming conditions. On the basis of these novel concepts and processes, we discuss several initiatives to target platelets therapeutically.
Collapse
|
45
|
A Novel Index for Prompt Prediction of Severity in Patients with Unstable Angina Pectoris. Emerg Med Int 2020; 2020:7651610. [PMID: 32377438 PMCID: PMC7199558 DOI: 10.1155/2020/7651610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/26/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022] Open
Abstract
Objectives Rapid risk stratification by emergency department (ED) physicians to evaluate patients with chest pain for predicting the short-term occurrence of major adverse cardiac event (MACE) is crucial. The aim of this study was to investigate the predictive value of platelet-lymphocyte ratio (PLR) levels and compare with the modified heart score (m-HS) and stress testing to predict the severity of high-risk patients with unstable angina pectoris (UAP) in the ED. Methods This study is prospective which included 316 patients with UAP and 316 control healthy subjects. The study took place from 01 April 2016, until 01 April 2017, in Medipol University. Result The mean PLR levels in the UAP group were higher than those in the control group (p < 0.001). The mean PLR of the m-HS ≥4 group was higher than that in the m-HS ≤3 group (p < 0.001). The mean levels of PLR in the subgroups based on the stress testing positivity were higher than those in the stress testing negative subgroup (p < 0.001). PLR levels were positively correlated with the m-HS, stress testing, and treatment decision in this study (r = 0.559; p < 0.001; r = 0.582; p < 0.001; r = 0.789; p < 0.001, respectively). Conclusion A positive correlation was determined with an increase in m-HS, treatment decision, and positive exercise testing as the PLR levels increased, indicating the severity of high risk of UAP in the ED.
Collapse
|
46
|
Rahadian A, Fukuda D, Salim HM, Yagi S, Kusunose K, Yamada H, Soeki T, Shimabukuro M, Sata M. Thrombin inhibition by dabigatran attenuates endothelial dysfunction in diabetic mice. Vascul Pharmacol 2019; 124:106632. [PMID: 31759113 DOI: 10.1016/j.vph.2019.106632] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 09/19/2019] [Accepted: 11/19/2019] [Indexed: 11/16/2022]
Abstract
Diabetic patients have coagulation abnormalities, in which thrombin plays a key role. Whereas accumulating evidence suggests that it also contributes to the development of vascular dysfunction through the activation of protease-activated receptors (PARs). Here we investigated whether the blockade of thrombin attenuates endothelial dysfunction in diabetic mice. Induction of diabetes by streptozotocin (STZ) increased the expression of PAR1, PAR3, and PAR4 in the aorta. STZ-induced diabetic mice showed impairment of endothelial function, while the administration of dabigatran etexilate, a direct thrombin inhibitor, significantly attenuated endothelial dysfunction in diabetic mice with no alteration of metabolic parameters including blood glucose level. Dabigatran did not affect endothelium-independent vasodilation. Dabigatran decreased the expression of inflammatory molecules (e.g., MCP-1 and ICAM-1) in the aorta of diabetic mice. Thrombin increased the expression of these inflammatory molecules and the phosphorylation of IκBα, and decreased the phosphorylation of eNOSSer1177 in human umbilical endothelial cells (HUVEC). Thrombin significantly impaired the endothelium-dependent vascular response of aortic rings obtained from wild-type mice. Inhibition of NF-κB attenuated thrombin-induced inflammatory molecule expression in HUVEC and ameliorated thrombin-induced endothelial dysfunction in aortic rings. Dabigatran attenuated the development of diabetes-induced endothelial dysfunction. Thrombin signaling may serve as a potential therapeutic target in diabetic condition.
Collapse
Affiliation(s)
- Arief Rahadian
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Daiju Fukuda
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan.
| | - Hotimah Masdan Salim
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Hirotsugu Yamada
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| | - Michio Shimabukuro
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan; Department of Diabetes, Endocrinology and Metabolism School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima 770-8503, Japan
| |
Collapse
|
47
|
Bi C, Li PL, Liao Y, Rao HY, Li PB, Yi J, Wang WY, Su WW. Pharmacodynamic effects of Dan-hong injection in rats with blood stasis syndrome. Biomed Pharmacother 2019; 118:109187. [DOI: 10.1016/j.biopha.2019.109187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/14/2019] [Accepted: 06/28/2019] [Indexed: 01/23/2023] Open
|
48
|
Hara T, Phuong PT, Fukuda D, Yamaguchi K, Murata C, Nishimoto S, Yagi S, Kusunose K, Yamada H, Soeki T, Wakatsuki T, Imoto I, Shimabukuro M, Sata M. Protease-Activated Receptor-2 Plays a Critical Role in Vascular Inflammation and Atherosclerosis in Apolipoprotein E-Deficient Mice. Circulation 2019; 138:1706-1719. [PMID: 29700120 DOI: 10.1161/circulationaha.118.033544] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The coagulation system is closely linked with vascular inflammation, although the underlying mechanisms are still obscure. Recent studies show that protease-activated receptor (PAR)-2, a major receptor of activated factor X, is expressed in both vascular cells and leukocytes, suggesting that PAR-2 may contribute to the pathogenesis of inflammatory diseases. Here we investigated the role of PAR-2 in vascular inflammation and atherogenesis. METHODS We generated apolipoprotein E-deficient ( ApoE-/-) mice lacking systemic PAR-2 expression ( PAR-2-/- ApoE-/-). ApoE-/- mice, which lack or express PAR-2 only in bone marrow (BM) cells, were also generated by BM transplantation. Atherosclerotic lesions were investigated after 20 weeks on a Western-type diet by histological analyses, quantitative reverse transcription polymerase chain reaction, and Western blotting. In vitro experiments using BM-derived macrophages were performed to confirm the proinflammatory roles of PAR-2. The association between plasma activated factor X level and the severity of coronary atherosclerosis was also examined in humans who underwent coronary intervention. RESULTS PAR-2-/- ApoE-/- mice showed reduced atherosclerotic lesions in the aortic arch ( P<0.05) along with features of stabilized atherosclerotic plaques, such as less lipid deposition ( P<0.05), collagen loss ( P<0.01), macrophage accumulation ( P<0.05), and inflammatory molecule expression ( P<0.05) compared with ApoE-/- mice. Systemic PAR2 deletion in ApoE-/-mice significantly decreased the expression of inflammatory molecules in the aorta. The results of BM transplantation experiments demonstrated that PAR-2 in hematopoietic cells contributed to atherogenesis in ApoE-/- mice. PAR-2 deletion did not alter metabolic parameters. In vitro experiments demonstrated that activated factor X or a specific peptide agonist of PAR-2 significantly increased the expression of inflammatory molecules and lipid uptake in BM-derived macrophages from wild-type mice compared with those from PAR-2-deficient mice. Activation of nuclear factor-κB signaling was involved in PAR-2-associated vascular inflammation and macrophage activation. In humans who underwent coronary intervention, plasma activated factor X level independently correlated with the severity of coronary atherosclerosis as determined by Gensini score ( P<0.05) and plaque volume ( P<0.01). CONCLUSIONS PAR-2 signaling activates macrophages and promotes vascular inflammation, increasing atherosclerosis in ApoE-/- mice. This signaling pathway may also participate in atherogenesis in humans.
Collapse
Affiliation(s)
- Tomoya Hara
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Pham Tran Phuong
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Daiju Fukuda
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan.,Cardio-Diabetes Medicine (D.F., M.Shimabukuro), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Koji Yamaguchi
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Chie Murata
- Human Genetics (C.M., I.I.), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Sachiko Nishimoto
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | | | - Kenya Kusunose
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Hirotsugu Yamada
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Takeshi Soeki
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Tetsuzo Wakatsuki
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Issei Imoto
- Human Genetics (C.M., I.I.), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Michio Shimabukuro
- Cardio-Diabetes Medicine (D.F., M.Shimabukuro), Tokushima University Graduate School of Biomedical Sciences, Japan
| | - Masataka Sata
- Departments of Cardiovascular Medicine (T.H., P.T.P., D.F., K.Y., S.N., S.Y., K.K., H.Y., T.S., T.W., M.Sata), Tokushima University Graduate School of Biomedical Sciences, Japan
| |
Collapse
|
49
|
Libby P, Hansson GK. From Focal Lipid Storage to Systemic Inflammation: JACC Review Topic of the Week. J Am Coll Cardiol 2019; 74:1594-1607. [PMID: 31537270 PMCID: PMC6910128 DOI: 10.1016/j.jacc.2019.07.061] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 06/12/2019] [Accepted: 07/07/2019] [Indexed: 12/24/2022]
Abstract
Concepts of atherogenesis have evolved considerably with time. Early animal experiments showed that a cholesterol-rich diet could induce fatty lesion formation in arteries. The elucidation of lipoprotein metabolism ultimately led to demonstrating the clinical benefits of lipid lowering. The view of atheromata as bland accumulations of smooth muscle cells that elaborated an extracellular matrix that could entrap lipids then expanded to embrace inflammation as providing pathways that could link risk factors to atherogenesis. The characterization of leukocyte adhesion molecules and their control by proinflammatory cytokines, the ability of chemokines to recruit leukocytes, and the identification of inflammatory cell subtypes in lesions spurred the unraveling of innate and adaptive immune pathways that contribute to atherosclerosis and its thrombotic complications. Such pathophysiologic insights have led to the identification of biomarkers that can define categories of risk and direct therapies and to the development of new treatments.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Göran K Hansson
- Department of Medicine, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Acharya AB, Shetty IP, Jain S, Padakannaya I, Acharya S, Shettar L, Thakur S. Neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in chronic periodontitis before and after nonsurgical therapy. J Indian Soc Periodontol 2019; 23:419-423. [PMID: 31543614 PMCID: PMC6737853 DOI: 10.4103/jisp.jisp_622_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Various biomarkers have been evaluated for understanding the systemic inflammatory response (SIR) to periodontitis. Hematological markers have been reported to be useful biomarkers in a variety of diseases, including periodontal diseases. The role of neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in periodontitis and their possible role in the SIR are not extensively documented. Therefore, this study assessed NLR and PLR in chronic periodontitis (CP) patients before and after periodontal treatment, which to the best of knowledge has not been reported in the literature. Materials and Methods: Sixty participants were grouped as systemically and periodontally healthy (H) (n = 30) and with CP (n = 30). Plaque index, gingival index, probing pocket depth, clinical attachment loss, leukocyte counts, platelet (PLT) counts, NLR, and PLR were estimated at baseline and also after treatment in the CP group. NLR was calculated as total neutrophil count/absolute lymphocyte count, and PLR was calculated as total PLT count/absolute lymphocyte count. The data were statistically analyzed. Results: Periodontal parameters differed significantly between groups H and CP at baseline and posttreatment. A pair-wise comparison of NLR and PLR between CP patients at baseline and posttreatment was significant. Correlation analyses were not remarkable. Receiver operating characteristics analyses provided significant NLR and PLR predictive cutoff values to differentiate between CP patients at baseline and posttreatment. Conclusion: NLR and PLR may serve as potential biomarkers of the SIR to CP to bridge the association between periodontal and systemic conditions.
Collapse
Affiliation(s)
- Anirudh Balakrishna Acharya
- Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, SDM University, Dharwad, Karnataka, India
| | | | - Shrinidhi Jain
- Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, SDM University, Dharwad, Karnataka, India
| | | | - Swetha Acharya
- Department of Oral Pathology and Microbiology, S.D.M. College of Dental Sciences and Hospital, SDM University, Dharwad, Karnataka, India
| | - Leena Shettar
- Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, SDM University, Dharwad, Karnataka, India
| | - Srinath Thakur
- Department of Periodontics, S.D.M. College of Dental Sciences and Hospital, SDM University, Dharwad, Karnataka, India
| |
Collapse
|