1
|
Beura SK, Panigrahi AR, Yadav P, Kulkarni PP, Lakhanpal V, Singh B, Singh SK. Role of Thrombosis in Neurodegenerative Diseases: An Intricate Mechanism of Neurovascular Complications. Mol Neurobiol 2024:10.1007/s12035-024-04589-4. [PMID: 39482419 DOI: 10.1007/s12035-024-04589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | | | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Bhupinder Singh
- Department of Cardiology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
2
|
Martí-Carvajal AJ, Gemmato-Valecillos MA, Monge Martín D, Dayer M, Alegría-Barrero E, De Sanctis JB, Parise Vasco JM, Riera Lizardo RJ, Nicola S, Martí-Amarista CE, Correa-Pérez A. Interleukin-receptor antagonist and tumour necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Cochrane Database Syst Rev 2024; 9:CD014741. [PMID: 39297531 PMCID: PMC11411914 DOI: 10.1002/14651858.cd014741.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
BACKGROUND Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.
Collapse
Key Words
- humans
- angina, unstable
- angina, unstable/mortality
- angina, unstable/prevention & control
- antibodies, monoclonal, humanized
- antibodies, monoclonal, humanized/administration & dosage
- antibodies, monoclonal, humanized/adverse effects
- atherosclerosis
- atherosclerosis/mortality
- atherosclerosis/prevention & control
- bias
- cause of death
- myocardial infarction
- myocardial infarction/mortality
- myocardial infarction/prevention & control
- primary prevention
- primary prevention/methods
- randomized controlled trials as topic
- receptors, interleukin-1
- receptors, interleukin-1/antagonists & inhibitors
- secondary prevention
- secondary prevention/methods
- tumor necrosis factor-alpha
- tumor necrosis factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Arturo J Martí-Carvajal
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
- Facultad de Medicina (Centro Cochrane Madrid), Universidad Francisco de Vitoria, Madrid, Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia , Venezuela
| | - Mario A Gemmato-Valecillos
- Icahn School of Medicine at Mount Sinai/ NYCHH Elmhurst Hospital Center, 79-01 Broadway, Elmhurst, New York 11373, USA
| | | | - Mark Dayer
- Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland
- Faculty of Health, University of Plymouth, Plymouth, UK
| | | | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University, Faculty of Medicine and Dentistry, Olomouc, Czech Republic
| | - Juan Marcos Parise Vasco
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | - Ricardo J Riera Lizardo
- Cátedra Rectoral de Medicina Basada en la Evidencia, Universidad de Carabobo, Valencia, Venezuela
| | - Susana Nicola
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, Ecuador
| | | | - Andrea Correa-Pérez
- Faculty of Medicine, Universidad Francisco de Vitoria, Madrid, Spain
- Hospital Pharmacy and Medical Devices Department, Hospital Central de la Defensa "Gómez Ulla" CSVE, Madrid, Spain
| |
Collapse
|
3
|
Feng S, Wang J, Yin C, Li H, Wang T, Liu J, Liang Y, Liu J, Han D. The association between lower extremity function and cardiovascular diseases risk in older Chinese adults: Longitudinal evidence from a nationwide cohort. Arch Gerontol Geriatr 2024; 124:105463. [PMID: 38723574 DOI: 10.1016/j.archger.2024.105463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 06/17/2024]
Abstract
BACKGROUND Older adults in China are at a high risk of cardiovascular diseases (CVD), and impaired lower extremity function (LEF) is commonly observed in this demographic. This study aimed at assessing the association between LEF and CVD, thus providing valuable insights for clinical practice and public health policies. METHODS A sample of 4,636 individuals was included from the China Health and Retirement Longitudinal Study (CHARLS) dataset. Logistic regression and cox proportional hazard regression model was utilized to study the association between LEF and CVD incidence. Cross-lagged panel models were utilized to investigate the potential causal association between LEF and CVD over time. RESULTS Poor LEF was significantly associated with a higher risk of CVD in the total population [OR (95 % CI): 1.62 (1.27-2.05), P < 0.001]. Individuals with poor LEF demonstrated an increased risk of developing CVD [HR (95 % CI): 1.11 (1.02-1.23), P < 0.05], particularly stroke, compared to those with good LEF. And those with poor LEF had higher risks for heart disease [1.21 (1.00-1.45), P < 0.05] and stroke [1.98 (1.47-2.67), P < 0.001]. CONCLUSION The results suggest the potential usefulness of the Short Physical Performance Battery (SPPB) for classifying stroke risk in older Chinese adults, which also suggested that preventing and/or improving LEF may be beneficial for reducing stroke incidence and promoting healthy aging for older adults.
Collapse
Affiliation(s)
- Shixing Feng
- Dongfang Hospital Beijing University of Chinese Medicine, Beijing 100000, China; School of Life and Science, Beijing University of Chinese Medicine, Beijing, China; Centre France Chine de la Médecine Chinoise, Selles sur Cher, France
| | - Junqi Wang
- Dongzhimen Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Chaohui Yin
- School of Resources and Environment, Henan Agricultural University, Zhengzhou 450046, China
| | - Hangyu Li
- School of Life and Science, Beijing University of Chinese Medicine, Beijing, China
| | - Tianyi Wang
- School of Management, Beijing University of Chinese Medicine, Beijing, China
| | - Jialin Liu
- Dongfang Hospital Beijing University of Chinese Medicine, Beijing 100000, China
| | - Yafeng Liang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jinmin Liu
- Dongfang Hospital Beijing University of Chinese Medicine, Beijing 100000, China.
| | - Dongran Han
- School of Life and Science, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
4
|
Zhang WJ, Chen RQ, Tang X, Li PB, Wang J, Wu HK, Xu N, Zou MF, Luo SR, Ouyang ZQ, Chen ZK, Liao XX, Wu H. Naoxintong capsule for treating cardiovascular and cerebrovascular diseases: from bench to bedside. Front Pharmacol 2024; 15:1402763. [PMID: 38994201 PMCID: PMC11236728 DOI: 10.3389/fphar.2024.1402763] [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: 03/18/2024] [Accepted: 06/06/2024] [Indexed: 07/13/2024] Open
Abstract
Naoxintong Capsule (NXT), a renowned traditional Chinese medicine (TCM) formulation, has been broadly applied in China for more than 30 years. Over decades, accumulating evidences have proven satisfactory efficacy and safety of NXT in treating cardiovascular and cerebrovascular diseases (CCVD). Studies have been conducted unceasingly, while this growing latest knowledge of NXT has not yet been interpreted properly and summarized comprehensively. Hence, we systematically review the advancements in NXT research, from its chemical constituents, quality control, pharmacokinetics, to its profound pharmacological activities as well as its clinical applications in CCVD. Moreover, we further propose specific challenges for its future perspectives: 1) to precisely clarify bioactivities of single compound in complicated mixtures; 2) to evaluate the pharmacokinetic behaviors of NXT feature components in clinical studies, especially drug-drug interactions in CCVD patients; 3) to explore and validate its multi-target mechanisms by integrating multi-omics technologies; 4) to re-evaluate the safety and efficacy of NXT by carrying out large-scale, multicenter randomized controlled trials. In brief, this review aims to straighten out a paradigm for TCM modernization, which help to contribute NXT as a piece of Chinese Wisdom into the advanced intervention strategy for CCVD therapy.
Collapse
Affiliation(s)
- Wei-jian Zhang
- Department of Neurosurgery, First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Rui-qi Chen
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-market Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xuan Tang
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-market Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pei-bo Li
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-market Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jian Wang
- Department of Neurosurgery, Foshan Sanshui District People’s Hospital, Foshan, Guangdong, China
| | - Hai-ke Wu
- Department of Neurology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Ning Xu
- Second People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Ming-fei Zou
- Second People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Sen-rong Luo
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zi-qi Ouyang
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhi-kai Chen
- Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xu-xing Liao
- Department of Neurosurgery, First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-market Traditional Chinese Medicine, Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Alsieni M, Esmat A, Bazuhair MA, Altayb HN. Fragment-based drug design of novel inhibitors targeting lipoprotein (a) kringle domain KIV-10-mediated cardiovascular disease. J Bioenerg Biomembr 2024; 56:247-259. [PMID: 38483739 DOI: 10.1007/s10863-024-10013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/11/2024] [Indexed: 05/24/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death globally, attributed to a complex etiology involving metabolic, genetic, and protein-related factors. Lipoprotein(a) (Lp(a)), identified as a genetic risk factor, exhibits elevated levels linked to an increased risk of cardiovascular diseases. The lipoprotein(a) kringle domains have recently been identified as a potential target for the treatment of CVDs, in this study we utilized a fragment-based drug design approach to design a novel, potent, and safe inhibitor for lipoprotein(a) kringle domain. With the use of fragment library (61,600 fragments) screening, combined with analyses such as MM/GBSA, molecular dynamics simulation (MD), and principal component analysis, we successfully identified molecules effective against the kringle domains of Lipoprotein(a). The hybridization process (Breed) of the best fragments generated a novel 249 hybrid molecules, among them 77 exhibiting superior binding affinity (≤ -7 kcal/mol) compared to control AZ-02 (-6.9 kcal/mol), Importantly, the top ten molecules displayed high similarity to the control AZ-02. Among the top ten molecules, BR1 exhibited the best docking energy (-11.85 kcal/mol ), and higher stability within the protein LBS site, demonstrating the capability to counteract the pathophysiological effects of lipoprotein(a) [Lp(a)]. Additionally, principal component analysis (PCA) highlighted a similar trend of motion during the binding of BR1 and the control compound (AZ-02), limiting protein mobility and reducing conformational space. Moreover, ADMET analysis indicated favorable drug-like properties, with BR1 showing minimal violations of Lipinski's rules. Overall, the identified compounds hold promise as potential therapeutics, addressing a critical need in cardiovascular medicine. Further preclinical and clinical evaluations are needed to validate their efficacy and safety, potentially ushering in a new era of targeted therapies for CVDs.
Collapse
Affiliation(s)
- Mohammed Alsieni
- Department of Clinical Pharmacology Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Ahmed Esmat
- Department of Clinical Pharmacology Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Mohammed A Bazuhair
- Department of Clinical Pharmacology Faculty of Medicine, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Centre of Research Excellence for Drug Research and Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hisham N Altayb
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, 23589, Saudi Arabia.
| |
Collapse
|
6
|
Wang A, Yue K, Yan X, Zhong W, Zhang G, Wang L, Zhang H, Zhang X. Inhibition of platelet adhesion to exposed subendothelial collagen by steric hindrance with blocking peptide nanoparticles. Colloids Surf B Biointerfaces 2024; 237:113866. [PMID: 38520952 DOI: 10.1016/j.colsurfb.2024.113866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
The inhibition of platelet adhesion to collagen in exposed vessels represents an innovative approach to the treatment of atherosclerosis and thrombosis. This study aimed to engineer peptide-based nanoparticles that prevent platelet binding to subendothelial collagen by engaging with collagen with high affinity. We examined the interactions between integrin α2/ glycoprotein VI/ von Willebrand factor A3 domain and collagen, as well as between the synthesized peptide nanoparticles and collagen, utilizing molecular dynamics simulations and empirical assays. Our findings indicated that the bond between von Willebrand factor and collagen was more robust. Specifically, the sequences SITTIDV, VDVMQRE, and YLTSEMH in von Willebrand factor were identified as essential for its attachment to collagen. Based on these sequences, three peptide nanoparticles were synthesized (BPa: Capric-GNNQQNYK-SITTIDV, BPb: Capric-GNNQQNYK-VDVMQRE, BPc: Capric-GNNQQNYK-YLTSEMH), each displaying significant affinity towards collagen. Of these, the BPa nanoparticles exhibited the most potent interaction with collagen, leading to a 75% reduction in platelet adhesion.
Collapse
Affiliation(s)
- Anqi Wang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China.
| | - Xiaotong Yan
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Lei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
| | - Hua Zhang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xinxin Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province 528399, China
| |
Collapse
|
7
|
Schepp M, Freuer D, Peters A, Heier M, Teupser D, Meisinger C, Linseisen J. Is the Habitual Dietary Intake of Foods of Plant or Animal Origin Associated with Circulating Hemostatic Factors?-Results of the Population-Based KORA-Fit Study. Nutrients 2024; 16:432. [PMID: 38337715 PMCID: PMC10857183 DOI: 10.3390/nu16030432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Blood coagulation is a complex physiological process critical for maintaining hemostasis, and disruptions in this system can lead to various health complications. Since the effects of specific food groups on a series of circulating coagulation parameters in the population are not well established, this study examines such associations in the population-based KORA-Fit study. A total of 595 subjects (263 men and 332 women) born between 1945 and 1964 and living in the study region of Augsburg were included in the study. Habitual food intake was estimated based on a combination of repeated 24-h food lists (24HFLs) and a food frequency questionnaire (FFQ). Antithrombin III, D-dimers, factor VIII, fibrinogen, protein C, protein S, aPTT, Quick value and INR were measured in citrate plasma. Multivariable linear regression models were applied to investigate associations between the consumption of specific foods of plant or animal origin and hemostatic factors. We found that the consumption of plant-based food groups, including green leafy vegetables (rich in vitamin K1), were hardly associated with coagulation parameters. Surprisingly, a high consumption of dairy products and especially butter were associated with higher D-dimer concentrations. These findings need further evaluation in prospective studies.
Collapse
Affiliation(s)
- Michael Schepp
- Epidemiology, University Hospital Augsburg, University of Augsburg, 86156 Augsburg, Germany; (D.F.); (C.M.); (J.L.)
| | - Dennis Freuer
- Epidemiology, University Hospital Augsburg, University of Augsburg, 86156 Augsburg, Germany; (D.F.); (C.M.); (J.L.)
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; (A.P.); (M.H.)
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Margit Heier
- Institute of Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany; (A.P.); (M.H.)
- KORA Study Centre, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, Medical Faculty, Ludwig-Maximilians-Universität München, 81377 Munich, Germany;
| | - Christine Meisinger
- Epidemiology, University Hospital Augsburg, University of Augsburg, 86156 Augsburg, Germany; (D.F.); (C.M.); (J.L.)
| | - Jakob Linseisen
- Epidemiology, University Hospital Augsburg, University of Augsburg, 86156 Augsburg, Germany; (D.F.); (C.M.); (J.L.)
| |
Collapse
|
8
|
Ferdous J, Rahman ME, Sraboni FS, Dutta AK, Rahman MS, Ali MR, Sikdar B, Khan A, Hasan MF. Assessment of the hypoglycemic and anti-hemostasis effects of Paederia foetida (L.) in controlling diabetes and thrombophilia combining in vivo and computational analysis. Comput Biol Chem 2023; 107:107954. [PMID: 37738820 DOI: 10.1016/j.compbiolchem.2023.107954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/24/2023]
Abstract
Paederia foetida is valued for its folk medicinal properties. This research aimed to assess the acute toxicity, hypoglycemic and anti-hemostasis properties of the methanolic extract of P. foetida leaves (PFLE). Acute toxicity of PFLE was performed on a mice model. Hypoglycemic and anti-hemostasis properties of PFLE were investigated on normal and streptozotocin-induced mice models. Deep learning, molecular docking, density functional theory, and molecular simulation techniques were employed to understand the underlying mechanisms through in silico study. Oral administration of PFLE at a dosage of 300 µg/kg body weight (BW) showed no signs of toxicity. Treatment with PFLE (300 µg/kg/BW) for 14 days resulted in a hypoglycemic condition and a 30.47% increase in body weight. Additionally, PFLE mixed with blood exhibited a 44.6% anti-hemostasis effect. Deep learning predicted the inhibitory concentration (pIC50, nM) of Cleomiscosins against SGLT2 and FXa to be 7.478 and 6.017, respectively. Molecular docking analysis revealed strong binding interactions of Cleomiscosins with crucial residues of the target proteins, exhibiting binding energies of -8.2 kcal/mol and -7.1 kcal/mol, respectively. ADME/Tox predictions indicated favorable pharmacokinetic properties of Cleomiscosins, and DFT calculations of frontier molecular orbitals analyzed the stability and reactivity of these compounds. Molecular simulation dynamics, principal component analysis and MM-PBSA calculation demonstrated the stable, compact, and rigid nature of the protein-ligand complexes. The methanolic PFLE exhibited significant hypoglycemic and anti-hemostasis properties. Cleomiscosin may have inhibitory properties for the development of novel drugs to manage diabetes and thrombophilia in the near future.
Collapse
Affiliation(s)
- Jannatul Ferdous
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Md Ekhtiar Rahman
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Farzana Sayed Sraboni
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Amit Kumar Dutta
- Department of Microbiology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Md Siddikur Rahman
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Md Roushan Ali
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Biswanath Sikdar
- Department of Microbiology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| | - Alam Khan
- Department of Pharmacy, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md Faruk Hasan
- Department of Microbiology, University of Rajshahi, Rajshahi 6205, Bangladesh.
| |
Collapse
|
9
|
Ye X, Wang S, Liu X, Wu Q, Lv Y, Lv Q, Li J, Li L, Yang Y. Effects of PCSK9 inhibitors on coronary microcirculation, inflammation and cardiac function in patients with CHD after PCI: a protocol for systematic review and meta-analysis. BMJ Open 2023; 13:e074067. [PMID: 37723117 PMCID: PMC10510950 DOI: 10.1136/bmjopen-2023-074067] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/14/2023] [Indexed: 09/20/2023] Open
Abstract
INTRODUCTION Coronary heart disease (CHD) is one of the common cardiovascular diseases that seriously jeopardise human health, and endothelial inflammation and dyslipidaemia are the initiating links leading to its occurrence. Percutaneous coronary intervention (PCI) is one of the most effective surgical treatments for CHD with narrowed or blocked blood vessels, which can quickly unblock the blocked vessels and restore coronary blood supply. However, most patients may experience coronary microcirculation disorders (CMDs) and decreased cardiac function after PCI treatment, which directly affects the efficacy of PCI and the prognosis of patients. Preprotein converting enzyme subtilisin/Kexin 9 (PCSK9) inhibitors are novel pleiotropy lipid-lowering drug with dual anti-inflammation and lipid-lowering effects, and represent a new clinical pathway for rapid correction of dyslipidaemia. Therefore, we designed this protocol to systematically evaluate the effects of PCSK9 inhibitors on coronary microcirculation and cardiac function in patients with CHD after PCI, and to provide high-quality evidence-based evidence for the clinical application of PCSK9 inhibitors. METHODS AND ANALYSIS This protocol is reported strictly in accordance with the 2020 Preferred Reporting Items for Systematic Reviews and Meta-analyses Protocols Guidelines. We will search PubMed, EMBASE, Web of Science and three Chinese databases (CNKI, Wanfang and VIP database) according to preset search strategies, without language and publication data restrictions. We will work with manual retrieval to screen references that have been included in the literature. Google Scholar will be used to search for grey literature. The final included literature must meet the established inclusion criteria. Titles, abstracts and full text will be extracted independently by two reviewers, and disagreements will be resolved through discussion or the involvement of a third reviewer. Extracted data will be analysed using Review Manager V.5.3. The Cochrane Risk of Bias Tool will be used to evaluate the risk of bias. Publication bias will be assessed by funnel plots. Heterogeneity will be assessed by I2 test and subgroup analyses will be used to further investigate potential sources of heterogeneity. The quality of the literature will be assessed by GRADE score. This protocol will start in January 2026 and end in December 2030. ETHICS AND DISSEMINATION This study is a systematic review of published literature data and no special ethical approval was required. PROSPERO REGISTRATION NUMBER CRD42022346189.
Collapse
Affiliation(s)
- Xuejiao Ye
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shihan Wang
- Department of Cardiology, China Academy of Traditional Chinese Medicine Guang'anmen Hospital, Beijing, China
| | | | - Qian Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Lv
- Shanghai Qianhe Technology Co LTD, Shanghai, China
| | - Qianyu Lv
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junjia Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lanlan Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingtian Yang
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
10
|
Hu N, Liu J, Cui W, Wang J, Wang Z, Chen X. Extracutaneous/osteoarticular manifestations in patients with SAPHO syndrome. Int J Rheum Dis 2023; 26:1649-1652. [PMID: 37664960 DOI: 10.1111/1756-185x.14779] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/24/2023] [Accepted: 05/28/2023] [Indexed: 09/05/2023]
Affiliation(s)
- Nandong Hu
- Department of Radiology, The Affiliated Hospital of Nanjing, University of Chinese Medicine, Nanjing, China
| | - Jingjing Liu
- Department of Radiology, The Affiliated Hospital of Nanjing, University of Chinese Medicine, Nanjing, China
- The First Clinical Institute, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjing Cui
- Department of Radiology, The Affiliated Hospital of Nanjing, University of Chinese Medicine, Nanjing, China
| | - Jianhua Wang
- Department of Radiology, The Affiliated Hospital of Nanjing, University of Chinese Medicine, Nanjing, China
| | - Zhongqiu Wang
- Department of Radiology, The Affiliated Hospital of Nanjing, University of Chinese Medicine, Nanjing, China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing, University of Chinese Medicine, Nanjing, China
| |
Collapse
|
11
|
Gigante B, Levy JH, van Gorp E, Bartoloni A, Bochaton-Piallat ML, Bäck M, Ten Cate H, Christersson C, Ferreiro JL, Geisler T, Lutgens E, Schulman S, Storey RF, Thachil J, Vilahur G, Liaw PC, Rocca B. Management of patients on antithrombotic therapy with severe infections: a joint clinical consensus statement of the ESC Working Group on Thrombosis, the ESC Working Group on Atherosclerosis and Vascular Biology, and the International Society on Thrombosis and Haemostasis. Eur Heart J 2023; 44:3040-3058. [PMID: 37439553 DOI: 10.1093/eurheartj/ehad388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 05/01/2023] [Accepted: 05/29/2023] [Indexed: 07/14/2023] Open
Abstract
Patients with severe infections and a pre-existing indication for antithrombotic therapy, i.e. antiplatelet agents, anticoagulant drugs, or their combinations, require integrated clinical counselling among coagulation, infectious disease, and cardiology specialists, due to sepsis-induced coagulopathy that frequently occurs. Bacterial and viral pathogens constitute an increasing threat to global public health, especially for patients with ongoing antithrombotic treatment who have a high risk of thrombotic recurrences and high susceptibility to severe infections with increased morbidity and mortality. Similarly, sepsis survivors are at increased risk for major vascular events. Coagulopathy, which often complicates severe infections, is associated with a high mortality and obligates clinicians to adjust antithrombotic drug type and dosing to avoid bleeding while preventing thrombotic complications. This clinical consensus statement reviews the best available evidence to provide expert opinion and statements on the management of patients hospitalized for severe bacterial or viral infections with a pre-existing indication for antithrombotic therapy (single or combined), in whom sepsis-induced coagulopathy is often observed. Balancing the risk of thrombosis and bleeding in these patients and preventing infections with vaccines, if available, are crucial to prevent events or improve outcomes and prognosis.
Collapse
Affiliation(s)
- Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Solnavägen 30. 17164 and Department of Cardiology, Danderyds Hospital, Entrévägen 2, 182 88, Stockholm, Sweden
| | - Jerrold H Levy
- Departments of Anesthesiology, Critical Care, and Surgery (Cardiothoracic), Duke University School of Medicine, Durham, United States; 2301 Erwin Road, Durham, NC 27710, USA
| | - Eric van Gorp
- Department of Viroscience, Erasmus MC, Rotterdam, PO box 2040 [Room Ee1726], 3000 CA Rotterdam, The Netherlands
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, Infectious Diseases Unit, University of Florence, Largo Brambilla 3, 50100 Florence, Italy
| | - Marie-Luce Bochaton-Piallat
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet 1, CH-1211 Genève, Switzerland
| | - Magnus Bäck
- Department of Translational Cardiology, Karolinska Institutet and Karolinska University Hospital, CMM L8:01, 171 76 Stockholm, Sweden
- INSERM U1116, University of Lorraine, Nancy University Hospital, 2 rue Jean Lamour, 54505 Vandoeuvre les Nancy Cedex, France
| | - Hugo Ten Cate
- Department of Internal medicine, Thrombosis Expertise Center, Maastricht University Medical Center and CARIM school for cardiovascular diseases, Universiteitsingel 50, PO Box 616, 6200 MD Maastricht, The Netherlands
- Center for Thrombosis and Haemostasis, Gutenberg University Medical Center, Langenbeckstr. 1, Bldg. 403, 55131 Mainz, Germany
| | - Christina Christersson
- Department of Medical Sciences, Cardiology, Uppsala University, Akademiska Sjukhuset, 75185, Uppsala, Sweden
| | - José Luis Ferreiro
- Department of Cardiology and Bio-Heart Cardiovascular Diseases Research Group; Bellvitge University Hospital - Bellvitge Biomedical Research Institute (IDIBELL); CIBERCV; L'Hospitalet de Llobregat, Hospital Duran i Reynals - Edifici Terapèutic - 2a planta Gran Via de l'Hospitalet, 199, 08908 Hospitalet de Llobregat Barcelona -Spain
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany
| | - Esther Lutgens
- Cardiovascular Medicine, Experimental CardioVascular Immunology Laboratory, Mayo Clinic, 200 First St SW, 55905, Rochester, MN, USA
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, München, Germany & German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstrasse 9, 80336, Munich, Germany
| | - Sam Schulman
- Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada HHS - General Hospital 237, Barton Street East, Hamilton, ON, L8L 2X2, Canada
- Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Bol'shaya Pirogovskaya Ulitsa, 2, стр. 4, Moscow 119435, Russia
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Pegasus House, 463a Glossop Road, Sheffield, S10 2QD, UK
| | - Jecko Thachil
- Department of Haematology, Manchester University Hospitals, Oxford road, Manchester, M13 9WL, UK
| | - Gemma Vilahur
- Institut de Recerca Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, CIBERCV, Avda. Sant Antoni Maria Claret 167, 08025 Barcelona, Spain
| | - Patricia C Liaw
- Department of Medicine, Thrombosis & Atherosclerosis Research Institute (TaARI), McMaster University, 237 Barton Street East Hamilton, Ontario L8L 2X2, Canada
| | - Bianca Rocca
- Department of Safety and Bioethics, Section on Pharmacology, Catholic University School of Medicine, Largo F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
12
|
Zhao Z, Lian H, Liu Y, Sun L, Zhang Y. Application of systemic inflammation indices and lipid metabolism-related factors in coronary artery disease. Coron Artery Dis 2023; 34:306-313. [PMID: 37102240 PMCID: PMC10309097 DOI: 10.1097/mca.0000000000001239] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/28/2023] [Indexed: 04/28/2023]
Abstract
OBJECTIVE We aimed to investigate the relationship between coronary artery disease (CAD) and systemic inflammation indices and lipid metabolism-related factors and subsequently, discuss the clinical application of these factors in CAD. METHODS We enrolled 284 consecutive inpatients with suspected CAD and divided them into a CAD group and a non-CAD group according to coronary angiography results. Serum levels of angiopoietin-like protein 3 (ANGPTL3), angiopoietin-like protein 4 (ANGPTL4), fatty acid-binding protein 4 (FABP4), and tumor necrosis factor-α (TNF-α) levels were assessed using the ELISA and the systemic inflammation indices were calculated. Multivariate logistic regression was used to assess the risk factors of CAD. The receiver operating characteristic curve was used to determine the cutoff and diagnostic values. RESULTS The neutrophil-to-high density lipoprotein cholesterol ratio (5.04 vs. 3.47), neutrophil-to-lymphocyte ratio (3.25 vs. 2.45), monocyte-to-high density lipoprotein cholesterol ratio (MHR) (0.46 vs. 0.36), monocyte-to-lymphocyte ratio (0.31 vs. 0.26), systemic immune-inflammation index (SII) (696.00 vs. 544.82), serum TNF-α (398.15 ng/l vs. 350.65 ng/l), FABP4 (1644.00 ng/l vs. 1553.00 ng/l), ANGPTL3 (57.60 ng/ml vs. 52.85 ng/ml), and ANGPTL4 (37.35 ng/ml vs. 35.20 ng/ml) values showed a significant difference between the CAD and non-CAD groups ( P < 0.05). After adjusting for confounding factors, the following values were obtained: ANGPTL3 > 67.53 ng/ml [odds ratio (OR) = 8.108, 95% confidence interval (CI) (1.022-65.620)]; ANGPTL4 > 29.95 ng/ml [OR = 5.599, 95% CI (1.809-17.334)]; MHR > 0.47 [OR = 4.872, 95% CI (1.715-13.835)]; SII > 589.12 [OR = 5.131, 95% CI (1.995-13.200)]. These factors were found to be independently associated with CAD ( P < 0.05). Diabetes combined with MHR > 0.47, SII > 589.12, TNF-α >285.60 ng/l, ANGPTL3 > 67.53 ng/ml, and ANGPTL4 > 29.95 ng/l had the highest diagnostic value for CAD [area under the curve: 0.921, 95% CI, (0.881-0.960), Sensitivity: 88.9%, Specificity: 82.2%, P < 0.001]. CONCLUSION MHR > 0.47, SII > 589.12, TNF-α >285.60 ng/l, ANGPTL3 > 67.53 ng/ml, and ANGPTL4 > 29.95 ng/l were identified as independent CAD risk factors and have valuable clinical implications in the diagnosis and treatment of CAD.
Collapse
Affiliation(s)
- Zhuoyan Zhao
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Huan Lian
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yixiang Liu
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Ying Zhang
- Department of Cardiology, The Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
13
|
Dimitroglou Y, Aggeli C, Theofilis P, Tsioufis P, Oikonomou E, Chasikidis C, Tsioufis K, Tousoulis D. Novel Anti-Inflammatory Therapies in Coronary Artery Disease and Acute Coronary Syndromes. Life (Basel) 2023; 13:1669. [PMID: 37629526 PMCID: PMC10455741 DOI: 10.3390/life13081669] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/27/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Evidence suggests that inflammation plays an important role in atherosclerosis and the consequent clinical presentation, including stable coronary artery disease (CAD) and acute coronary syndromes (ACS). The most essential elements are cytokines, proteins with hormone-like properties that are produced by the immune cells, endothelial cells, platelets, fibroblasts, and some stromal cells. Interleukins (IL-1β and IL-6), chemokines, interferon-γ (IFN-γ), and tumor necrosis factor-alpha (TNF-α) are the cytokines commonly associated with endothelial dysfunction, vascular inflammation, and atherosclerosis. These molecules can be targeted by commonly used therapeutic substances or selective molecules that exert targeted anti-inflammatory actions. The most significant anti-inflammatory therapies are aspirin, statins, colchicine, IL-1β inhibitors, and IL-6 inhibitors, along with novel therapies such as TNF-α inhibitors and IL-1 receptor antagonists. Aspirin and statins are well-established therapies for atherosclerosis and CAD and their pleiotropic and anti-inflammatory actions contribute to their efficacy and favorable profile. Colchicine may also be considered in high-risk patients if recurrent ACS episodes occur when on optimal medical therapy according to the most recent guidelines. Recent randomized studies have also shown that therapies specifically targeting inflammatory interleukins and inflammation can reduce the risk for cardiovascular events, but these therapies are yet to be fully implemented in clinical practice. Preclinical research is also intense, targeting various inflammatory mediators that are believed to be implicated in CAD, namely repeated transfers of the soluble mutant of IFN-γ receptors, NLRP3 inflammasome inhibitors, IL-10 delivery by nanocarriers, chemokine modulatory treatments, and reacting oxygen species (ROS) targeting nanoparticles. Such approaches, although intriguing and promising, ought to be tested in clinical settings before safe conclusions can be drawn. Although the link between inflammation and atherosclerosis is significant, further studies are needed in order to elucidate this association and improve outcomes in patients with CAD.
Collapse
Affiliation(s)
- Yannis Dimitroglou
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Constantina Aggeli
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Theofilis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Panagiotis Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece;
| | - Christos Chasikidis
- Department of Cardiology, General Hospital of Corinth, 20100 Corinth, Greece;
| | - Konstantinos Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece; (Y.D.); (C.A.); (P.T.); (K.T.); (D.T.)
| |
Collapse
|
14
|
Theofilis P, Oikonomou E, Chasikidis C, Tsioufis K, Tousoulis D. Pathophysiology of Acute Coronary Syndromes-Diagnostic and Treatment Considerations. Life (Basel) 2023; 13:1543. [PMID: 37511918 PMCID: PMC10381786 DOI: 10.3390/life13071543] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Coronary artery disease and acute coronary syndromes are accountable for significant morbidity and mortality, despite the preventive measures and technological advancements in their management. Thus, it is mandatory to further explore the pathophysiology in order to provide tailored and more effective therapies, since acute coronary syndrome pathogenesis is more varied than previously assumed. It consists of plaque rupture, plaque erosion, and calcified nodules. The advancement of vascular imaging tools has been critical in this regard, redefining the epidemiology of each mechanism. When it comes to acute coronary syndrome management, the presence of ruptured plaques almost always necessitates emergent reperfusion, whereas the presence of plaque erosions may indicate the possibility of conservative management with potent antiplatelet and anti-atherosclerotic medications. Calcified nodules, on the other hand, are an uncommon phenomenon that has largely gone unexplored in terms of the best management plan. Future studies should further establish the importance of detecting the underlying mechanism and the role of various treatment plans in each of these distinct entities.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- First Department of Cardiology, "Hippokration" General Hospital, University of Athens Medical School, 115 27 Athens, Greece
| | - Evangelos Oikonomou
- Third Department of Cardiology, Thoracic Diseases General Hospital "Sotiria", University of Athens Medical School, 115 27 Athens, Greece
| | - Christos Chasikidis
- Department of Cardiology, General Hospital of Corinth, 201 00 Corinth, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, "Hippokration" General Hospital, University of Athens Medical School, 115 27 Athens, Greece
| | - Dimitris Tousoulis
- First Department of Cardiology, "Hippokration" General Hospital, University of Athens Medical School, 115 27 Athens, Greece
| |
Collapse
|
15
|
Yalameha B, Reza Nejabati H. Urinary Exosomal Metabolites: Overlooked Clue for Predicting Cardiovascular Risk. Clin Chim Acta 2023:117445. [PMID: 37315726 DOI: 10.1016/j.cca.2023.117445] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
Over the last decade, increasing research has focused on urinary exosomes (UEs) in biological fluids and their relationship with physiological and pathological processes. UEs are membranous vesicles with a size of 40-100 nm, containing a number of bioactive molecules such as proteins, lipids, mRNAs, and miRNAs. These vesicles are an inexpensive non-invasive source that can be used in clinical settings to differentiate healthy patients from diseased patients, thereby serving as potential biomarkers for the early identification of disease. Recent studies have reported the isolation of small molecules called exosomal metabolites from individuals' urine with different diseases. These metabolites could utilize for a variety of purposes, such as the discovery of biomarkers, investigation of mechanisms related to disease development, and importantly prediction of cardiovascular diseases (CVDs) risk factors, including thrombosis, inflammation, oxidative stress, hyperlipidemia as well as homocysteine. It has been indicated that alteration in urinary metabolites of N1-methylnicotinamide, 4-aminohippuric acid, and citric acid can be valuable in predicting cardiovascular risk factors, providing a novel approach to evaluating the pathological status of CVDs. Since the UEs metabolome has been clearly and precisely so far unexplored in CVDs, the present study has specifically addressed the role of the mentioned metabolites in the prediction of CVDs risk factors.
Collapse
Affiliation(s)
- Banafsheh Yalameha
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Cui L, Liu Y, Hu Y, Dong J, Deng Q, Jiao B, Sun Y, Wu Y, Liu T, Wang W, Li C. Shexiang Tongxin Dropping Pill alleviates M1 macrophage polarization-induced inflammation and endothelial dysfunction to reduce coronary microvascular dysfunction via the dectin-1/Syk/IRF5 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023:116742. [PMID: 37290736 DOI: 10.1016/j.jep.2023.116742] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shexiang Tongxin Dropping Pill (STDP), a traditional Chinese medicine compound, is fragrant, invigorates the qi, unblocks pulses, activates the blood circulation, removes blood stasis, and relieves pain. It is used clinically to treat coronary heart disease and angina pectoris. Coronary microvascular dysfunction (CMD) is associated with increased morbidity and mortality from cardiovascular events. Endothelial dysfunction and inflammation have been verified as its underlying causes. STDP can ameliorate CMD, but the mechanism has not been fully elucidated. AIM OF THE STUDY To explore the effects of STDP on M1 macrophage polarization-induced inflammation and endothelial dysfunction as an inhibitor of CMD, and to determine its mechanisms of action. MATERIALS AND METHODS The CMD rat model was established by left anterior descending artery (LAD) ligation. The efficacy of STDP against CMD was evaluated by echocardiography, optical microangiography, Evans blue staining, and histological examination. The OGD/R-induced endothelial injury model, the endothelial injury-induced sterile inflammation model, the Dectin-1 overexpression model, and the Dectin-1-overexpressing RAW264.7 macrophage supernatant-stimulated HUVEC-induced secondary injury of endothelial function model were established to confirm the efficacy of STDP against M1 macrophage polarization-induced inflammation and endothelial dysfunction. RESULTS STDP blunted the deterioration of cardiac function and ameliorated CMD by reducing inflammatory cell infiltration and endothelial dysfunction in CMD rats. Endothelial injury and Dectin-1 overexpression induced M1 macrophage polarization and inflammation. Mechanically, STDP hindered M1 macrophage polarization and inflammation by inhibiting the Dectin-1/Syk/IRF5 pathway both in vivo and in vitro. STDP alleviated endothelial dysfunction induced by Dectin-1 overexpression in macrophages. CONCLUSION STDP can alleviate M1 macrophage polarization-induced inflammation and endothelial dysfunction against CMD via the Dectin-1/Syk/IRF5 pathway. Dectin-1-associated M1 macrophage polarization might be developed as a novel target for ameliorating CMD.
Collapse
Affiliation(s)
- Lingwen Cui
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yizhou Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yueyao Hu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jianteng Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiong Deng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Boyang Jiao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ying Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yan Wu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Tianhua Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Ministry of Education, Beijing, 100029, China; Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Ministry of Education, Beijing, 100029, China.
| |
Collapse
|
17
|
Polzin A, Dannenberg L, Benkhoff M, Barcik M, Helten C, Mourikis P, Ahlbrecht S, Wildeis L, Ziese J, Zikeli D, Metzen D, Hu H, Baensch L, Schröder NH, Keul P, Weske S, Wollnitzke P, Duse D, Saffak S, Cramer M, Bönner F, Müller T, Gräler MH, Zeus T, Kelm M, Levkau B. Revealing concealed cardioprotection by platelet Mfsd2b-released S1P in human and murine myocardial infarction. Nat Commun 2023; 14:2404. [PMID: 37100836 PMCID: PMC10133218 DOI: 10.1038/s41467-023-38069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
Antiplatelet medication is standard of care in acute myocardial infarction (AMI). However, it may have obscured beneficial properties of the activated platelet secretome. We identify platelets as major source of a sphingosine-1-phosphate (S1P) burst during AMI, and find its magnitude to favorably associate with cardiovascular mortality and infarct size in STEMI patients over 12 months. Experimentally, administration of supernatant from activated platelets reduces infarct size in murine AMI, which is blunted in platelets deficient for S1P export (Mfsd2b) or production (Sphk1) and in mice deficient for cardiomyocyte S1P receptor 1 (S1P1). Our study reveals an exploitable therapeutic window in antiplatelet therapy in AMI as the GPIIb/IIIa antagonist tirofiban preserves S1P release and cardioprotection, whereas the P2Y12 antagonist cangrelor does not. Here, we report that platelet-mediated intrinsic cardioprotection is an exciting therapeutic paradigm reaching beyond AMI, the benefits of which may need to be considered in all antiplatelet therapies.
Collapse
Affiliation(s)
- Amin Polzin
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lisa Dannenberg
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marcel Benkhoff
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maike Barcik
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Carolin Helten
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Mourikis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Samantha Ahlbrecht
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Laura Wildeis
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Justus Ziese
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dorothee Zikeli
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Metzen
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Hao Hu
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Leonard Baensch
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Nathalie H Schröder
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Petra Keul
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sarah Weske
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philipp Wollnitzke
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dragos Duse
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Süreyya Saffak
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Mareike Cramer
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Florian Bönner
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Tina Müller
- Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care, University Hospital Jena, Jena, Germany
| | - Tobias Zeus
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Medical Faculty of the Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital, Düsseldorf, Germany
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
18
|
Chen G, Jiang N, Zheng J, Hu H, Yang H, Lin A, Hu B, Liu H. Structural characterization and anti-inflammatory activity of polysaccharides from Astragalus membranaceus. Int J Biol Macromol 2023; 241:124386. [PMID: 37054858 DOI: 10.1016/j.ijbiomac.2023.124386] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/18/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023]
Abstract
In this study, two homogeneous polysaccharides (APS-A1 and APS-B1) were isolated from Astragalus membranaceus by DEAE-52 cellulose and Sephadex G-100 column chromatography. Their chemical structures were characterized by molecular weight distribution, monosaccharide composition, infrared spectrum, methylation analysis, and NMR. The results revealed that APS-A1 (2.62 × 106 Da) was a 1,4-α-D-Glcp backbone with a 1,4,6-α-D-Glcp branch every ten residues. APS-B1 (4.95 × 106 Da) was a heteropolysaccharide composed of glucose, galactose, and arabinose (75.24:17.27:19.35). Its backbone consisted of 1,4-α-D-Glcp, 1,4,6-α-D-Glcp, 1,5-α-L-Araf and the sidechains composed of 1,6-α-D-Galp and T-α/β-Glcp. Bioactivity assays showed that APS-A1 and APS-B1 had potential anti-inflammatory activity. They could inhibit the production of inflammatory factors (TNF-α, IL-6, and MCP-1) in LPS-stimulated RAW264.7 macrophages via NF-κB and MAPK (ERK, JNK) pathways. These results suggested that the two polysaccharides could be potential anti-inflammatory supplements.
Collapse
Affiliation(s)
- Guangming Chen
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Nan Jiang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, PR China; Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, PR China
| | - Junping Zheng
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China
| | - Aizhen Lin
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, PR China; Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, PR China
| | - Baifei Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, PR China.
| |
Collapse
|
19
|
Zhou Y, Chen Y, Yin G, Xie Q. Calciphylaxis and its co-occurrence with connective tissue diseases. Int Wound J 2023; 20:1316-1327. [PMID: 36274216 PMCID: PMC10031236 DOI: 10.1111/iwj.13972] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 03/23/2023] Open
Abstract
Calciphylaxis, also known as calcific uremic arteriopathy, is a rare calcification syndrome that presents as ischemic skin necrosis and severe pain. It has a high mortality rate and is characterised by calcification of the small and medium arteries and micro-thrombosis. Calciphylaxis mainly occurs in patients with end-stage renal disease. In recent years, there have been an increasing number of cases of calciphylaxis associated with connective tissue diseases. Given the absence of clear diagnostic criteria for calciphylaxis thus far, an early diagnosis is crucial for designing an effective multidisciplinary treatment plan. In this article, we review the research progress on calciphylaxis and describe its characteristics in the context of connective tissue diseases.
Collapse
Affiliation(s)
- Yueyuan Zhou
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuehong Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Geng Yin
- Department of General Practice, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qibing Xie
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Zhang H, Jing L, Zhai C, Xiang Q, Tian H, Hu H. Intestinal Flora Metabolite Trimethylamine Oxide Is Inextricably Linked to Coronary Heart Disease. J Cardiovasc Pharmacol 2023; 81:175-182. [PMID: 36607700 PMCID: PMC9988214 DOI: 10.1097/fjc.0000000000001387] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/01/2022] [Indexed: 01/07/2023]
Abstract
ABSTRACT Atherosclerotic coronary heart disease is a common cardiovascular disease with high morbidity and mortality. In recent years, the incidence of coronary heart disease has gradually become younger, and biomarkers for predicting coronary heart disease have demonstrated valuable clinical prospects. Several studies have established an association between coronary heart disease and intestinal flora metabolites, including trimethylamine oxide (TMAO), which has attracted widespread attention from researchers. Investigations have also shown that plasma levels of TMAO and its precursors can predict cardiovascular risk in humans; however, TMAO's mechanism of action in causing coronary heart disease is not fully understood. This review examines TMAO's generation, the mechanism through which it causes coronary heart disease, and the approaches used to treat TMAO-caused coronary heart disease to possible avenues for future research on coronary heart disease and find new concepts for the treatment of the condition.
Collapse
Affiliation(s)
- Honghong Zhang
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University; and
| | - Lele Jing
- Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing
| | - Changlin Zhai
- Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing
| | - Qiannan Xiang
- Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing
| | - Hongen Tian
- Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing
| | - Huilin Hu
- Affiliated Hospital of Jiaxing University: First Hospital of Jiaxing
| |
Collapse
|
21
|
Diabetes Mellitus and Heart Failure: Epidemiology, Pathophysiologic Mechanisms, and the Role of SGLT2 Inhibitors. Life (Basel) 2023; 13:life13020497. [PMID: 36836854 PMCID: PMC9968235 DOI: 10.3390/life13020497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Diabetes mellitus (DM) and heart failure (HF) are frequently encountered afflictions that are linked by a common pathophysiologic background. According to landmark studies, those conditions frequently coexist, and this interaction represents a poor prognostic indicator. Based on mechanistic studies, HF can be propagated by multiple pathophysiologic pathways, such as inflammation, oxidative stress, endothelial dysfunction, fibrosis, cardiac autonomic neuropathy, and alterations in substrate utilization. In this regard, DM may augment myocardial inflammation, fibrosis, autonomic dysfunction, and lipotoxicity. As the interaction between DM and HF appears critical, the new cornerstone in DM and HF treatment, sodium-glucose cotransporter-2 inhibitors (SGLT2i), may be able to revert the pathophysiology of those conditions and lead to beneficial HF outcomes. In this review, we aim to highlight the deleterious pathophysiologic interaction between DM and HF, as well as demonstrate the beneficial role of SGLT2i in this field.
Collapse
|
22
|
Roka-Moiia Y, Ammann K, Miller-Gutierrez S, Sheriff J, Bluestein D, Italiano JE, Flaumenhaft RC, Slepian MJ. Shear-Mediated Platelet Microparticles Demonstrate Phenotypic Heterogeneity as to Morphology, Receptor Distribution, and Hemostatic Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527675. [PMID: 36798322 PMCID: PMC9934663 DOI: 10.1101/2023.02.08.527675] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Objective Implantable cardiovascular therapeutic devices (CTD) including stents, percutaneous heart valves and ventricular assist devices, while lifesaving, impart supraphysiologic shear stress to platelets resulting in thrombotic and bleeding device-related coagulopathy. We previously demonstrated that shear-mediated platelet dysfunction is associated with downregulation of platelet GPIb-IX-V and αIIbβ3 receptors via generation of platelet-derived microparticles (PDMPs). Here, we test the hypothesis that shear-generated PDMPs manifest phenotypical heterogeneity of their morphology and surface expression of platelet receptors, and modulate platelet hemostatic function. Approach and Results Human gel-filtered platelets were exposed to continuous shear stress and sonication. Alterations of platelet morphology were visualized using transmission electron microscopy. Surface expression of platelet receptors and PDMP generation were quantified by flow cytometry. Thrombin generation was quantified spectrophotometrically, and platelet aggregation in plasma was measured by optical aggregometry. We demonstrate that platelet exposure to shear stress promotes notable alterations in platelet morphology and ejection of several distinctive types of PDMPs. Shear-mediated microvesiculation is associated with the differential remodeling of platelet receptors with PDMPs expressing significantly higher levels of both adhesion (α IIb β 3 , GPIX, PECAM-1, P-selectin, and PSGL-1) and agonist-evoked receptors (P 2 Y 12 & PAR1). Shear-mediated PDMPs have a bidirectional effect on platelet hemostatic function, promoting thrombin generation and inhibiting platelet aggregation induced by collagen and ADP. Conclusions Shear-generated PDMPs demonstrate phenotypic heterogeneity as to morphologic features and defined patterns of surface receptor alteration, and impose a bidirectional effect on platelet hemostatic function. PDMP heterogeneity suggests that a range of mechanisms are operative in the microvesiculation process, contributing to CTD coagulopathy and posing opportunities for therapeutic manipulation.
Collapse
|
23
|
Gong Y, Liu H, Ke S, Zhuo L, Wang H. Latest advances in biomimetic nanomaterials for diagnosis and treatment of cardiovascular disease. Front Cardiovasc Med 2023; 9:1037741. [PMID: 36684578 PMCID: PMC9846151 DOI: 10.3389/fcvm.2022.1037741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/09/2022] [Indexed: 01/05/2023] Open
Abstract
Cardiovascular disease remains one of the leading causes of death in China, with increasingly serious negative effects on people and society. Despite significant advances in preventing and treating cardiovascular diseases, such as atrial fibrillation/flutter and heart failure over the last few years, much more remains to be done. Therefore, developing innovative methods for identifying and managing cardiovascular disorders is critical. Nanomaterials provide multiple benefits in biomedicine, primarily better catalytic activity, drug loading, targeting, and imaging. Biomimetic materials and nanoparticles are specially combined to synthesize biomimetic nanoparticles that successfully reduce the nanoparticles' toxicity and immunogenicity while enhancing histocompatibility. Additionally, the biological targeting capability of nanoparticles facilitates the diagnosis and therapy of cardiovascular disease. Nowadays, nanomedicine still faces numerous challenges, which necessitates creating nanoparticles that are highly selective, toxic-free, and better clinically applicable. This study reviews the scientific accomplishments in this field over the past few years covering the classification, applications, and prospects of noble metal biomimetic nanozymes and biomimetic nanocarriers.
Collapse
Affiliation(s)
- Yuxuan Gong
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China
| | - Huaying Liu
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China
| | - Shen Ke
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China
| | - Li Zhuo
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China,Li Zhuo,
| | - Haibin Wang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing, China,*Correspondence: Haibin Wang,
| |
Collapse
|
24
|
Metabolic Dysfunction-Associated Fatty Liver Disease in Newly Diagnosed, Treatment-Naive Hypertensive Patients and Its Association with Cardiorenal Risk Markers. High Blood Press Cardiovasc Prev 2023; 30:63-72. [PMID: 36626077 DOI: 10.1007/s40292-023-00558-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Patients with arterial hypertension frequently present with comorbidities that are associated with increased cardiorenal risk, such as metabolic dysfunction-associated fatty liver disease (MAFLD). AIMS Our study aimed to assess the prevalence and the association of MAFLD with cardiorenal risk markers in newly diagnosed, treatment-naïve hypertensive patients. METHODS We recruited 281 individuals with new-onset hypertension who were not prescribed any medication. Medical history, clinical examination findings, and laboratory test results were recorded. Liver steatosis was assessed through fatty liver index (FLI) calculation. Patients with FLI ≥ 60 together with one main metabolic abnormality (type 2 diabetes mellitus or overweight/obesity) or at least two metabolic risk abnormalities (increased waist circumference, blood pressure, plasma triglycerides, presence of prediabetes or insulin resistance, decreased plasma high-density lipoprotein) fulfilled the diagnostic criteria for MAFLD. RESULTS The prevalence of MAFLD in our study population was 28.7%. Individuals with MAFLD were more frequently male and had increased body mass index. Systolic, diastolic, and pulse pressure values were significantly higher in this group of patients. Moreover, lipid, renal, glucose, and inflammatory markers were considerably deranged in patients with MAFLD. After multivariate regression analysis, uric acid, ferritin, and apoE emerged as independent predictors of MAFLD. Area under receiver operating characteristics curve revealed that uric acid had the greatest diagnostic accuracy, with the ideal cutoff being ≥ 5.2 mg/dl (sensitivity: 77.6%, specificity: 76.3%). CONCLUSION MAFLD represents a common comorbidity in hypertensive patients and is associated with markers of cardiorenal risk. Uric acid may be indicative of MAFLD in particular.
Collapse
|
25
|
Liu Y, Qu J, Xu J, Gu A, Deng D, Jia X, Wang B. Trimethylamine-N-oxide: a potential biomarker and therapeutic target in ischemic stroke. Front Neurol 2023; 14:1156879. [PMID: 37153682 PMCID: PMC10160411 DOI: 10.3389/fneur.2023.1156879] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
Ischemic stroke is by far the most common cerebrovascular disease and a major burden to the global economy and public health. Trimethylamine-N-oxide (TMAO), a small molecule compound produced by the metabolism of intestinal microorganisms, is reportedly associated with the risk of stroke, as well as the severity and prognosis of stroke; however, this conclusion remains contentious. This article reviews the production of TMAO, TMAO's relationship with different etiological types of ischemic stroke, and the possibility of reducing TMAO levels to improve the prognosis of ischemic stroke.
Collapse
Affiliation(s)
- Yuan Liu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, China
| | - Juan Qu
- Department of Neurology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| | - Junjie Xu
- Department of Neurology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| | - Aiming Gu
- Department of Neurology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| | - Dezhi Deng
- Department of Neurology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| | - Xiaodan Jia
- Department of Neurology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
| | - Baoxiang Wang
- Department of Neurology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, China
- *Correspondence: Baoxiang Wang,
| |
Collapse
|
26
|
Tsioufis P, Theofilis P, Tsioufis K, Tousoulis D. The Impact of Cytokines in Coronary Atherosclerotic Plaque: Current Therapeutic Approaches. Int J Mol Sci 2022; 23:ijms232415937. [PMID: 36555579 PMCID: PMC9788180 DOI: 10.3390/ijms232415937] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary atherosclerosis is a chronic pathological process that involves inflammation together with endothelial dysfunction and lipoprotein dysregulation. Experimental studies during the past decades have established the role of inflammatory cytokines in coronary artery disease, namely interleukins (ILs), tumor necrosis factor (TNF)-α, interferon-γ, and chemokines. Moreover, their value as biomarkers in disease development and progression further enhance the validity of this interaction. Recently, cytokine-targeted treatment approaches have emerged as potential tools in the management of atherosclerotic disease. IL-1β, based on the results of the CANTOS trial, remains the most validated option in reducing the residual cardiovascular risk. Along the same line, colchicine was also proven efficacious in preventing major adverse cardiovascular events in large clinical trials of patients with acute and chronic coronary syndrome. Other commercially available agents targeting IL-6 (tocilizumab), TNF-α (etanercept, adalimumab, infliximab), or IL-1 receptor antagonist (anakinra) have mostly been assessed in the setting of other inflammatory diseases and further testing in atherosclerosis is required. In the future, potential targeting of the NLRP3 inflammasome, anti-inflammatory IL-10, or atherogenic chemokines could represent appealing options, provided that patient safety is proven to be of no concern.
Collapse
|
27
|
Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis K, Tousoulis D. The Anti-Inflammatory Effect of Novel Antidiabetic Agents. Life (Basel) 2022; 12:1829. [PMID: 36362984 PMCID: PMC9696750 DOI: 10.3390/life12111829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/28/2022] [Accepted: 11/05/2022] [Indexed: 08/10/2023] Open
Abstract
The incidence of type 2 diabetes (T2DM) has been increasing worldwide and remains one of the leading causes of atherosclerotic disease. Several antidiabetic agents have been introduced in trying to regulate glucose control levels with different mechanisms of action. These agents, and sodium-glucose cotransporter-2 inhibitors in particular, have been endorsed by contemporary guidelines in patients with or without T2DM. Their widespread usage during the last three decades has raised awareness in the scientific community concerning their pleiotropic mechanisms of action, including their putative anti-inflammatory effect. In this review, we delve into the anti-inflammatory role and mechanism of the existing antidiabetic agents in the cardiovascular system and their potential use in other chronic sterile inflammatory conditions.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Cardiology Department, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Marios Sagris
- 3rd Cardiology Department, Thoracic Diseases Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
- 3rd Cardiology Department, Thoracic Diseases Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
| | - Alexios S. Antonopoulos
- 1st Cardiology Department, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
- 3rd Cardiology Department, Thoracic Diseases Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
| | - Kostas Tsioufis
- 1st Cardiology Department, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Department, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| |
Collapse
|
28
|
Theofilis P, Vordoni A, Kalaitzidis RG. Metabolic Dysfunction-Associated Fatty Liver Disease in the National Health and Nutrition Examination Survey 2017-2020: Epidemiology, Clinical Correlates, and the Role of Diagnostic Scores. Metabolites 2022; 12:1070. [PMID: 36355156 PMCID: PMC9697527 DOI: 10.3390/metabo12111070] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 08/10/2023] Open
Abstract
The recent establishment of metabolic dysfunction-associated fatty liver disease (MAFLD) has led to a reevaluation of its epidemiology, diagnosis, and clinical implications. In this study, we aimed to evaluate MAFLD's epidemiology and its association with other pathologic states and biomarkers, as well as to assess the prevalence of the different fibrosis stages in the MAFLD population, together with the importance of diagnostic scores in the preliminary determination of significant fibrosis. After analyzing the National Health and Nutrition Examination Survey (NHANES) 2017-2020, we found a high prevalence of MAFLD, at 58.6% of the studied population. MAFLD was accompanied by numerous comorbidities, which were increasingly common in individuals with higher grades of liver fibrosis. Fatty liver index emerged as a reliable indicator of MAFLD, as well as significant fibrosis. The estimation of fatty liver index could be a reasonable addition to the evaluation of patients with metabolic risk factors and could lead a diagnosis in the absence of liver elastography or biopsy. Further studies are needed to enhance our knowledge regarding its prognosis, as well as the role of novel therapies in its prevention or regression.
Collapse
|
29
|
Farraj AK, Martin BL, Schladweiler MC, Miller CN, Smoot J, Williams W, Fisher A, Oshiro W, Tennant A, Martin WK, Henriquez AR, Grindstaff R, Gavett SH, Gilmour MI, Kodavanti UP, Hazari MS, Dye JA. Mild allergic airways responses to an environmental mixture increase cardiovascular risk in rats. Toxicol Sci 2022; 191:106-122. [PMID: 36269214 PMCID: PMC9887678 DOI: 10.1093/toxsci/kfac112] [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: 02/03/2023] Open
Abstract
Recent epidemiological findings link asthma to adverse cardiovascular responses. Yet, the precise cardiovascular impacts of asthma have been challenging to disentangle from the potential cardiovascular effects caused by asthma medication. The purpose of this study was to determine the impacts of allergic airways disease alone on cardiovascular function in an experimental model. Female Wistar rats were intranasally sensitized and then challenged once per week for 5 weeks with saline vehicle or a mixture of environmental allergens (ragweed, house dust mite, and Aspergillus fumigatus). Ventilatory and cardiovascular function, measured using double-chamber plethysmography and implantable blood pressure (BP) telemetry and cardiovascular ultrasound, respectively, were assessed before sensitization and after single and final allergen challenge. Responses to a single 0.5 ppm ozone exposure and to the cardiac arrhythmogenic agent aconitine were also assessed after final challenge. A single allergen challenge in sensitized rats increased tidal volume and specific airways resistance in response to provocation with methacholine and increased bronchoalveolar lavage fluid (BALF) eosinophils, neutrophils, lymphocytes, cytokines interleukin (IL)-4, IL-5, IL-10, IL-1β, tumor necrosis factor-α, and keratinocyte chemoattract-growth-related oncogene characteristic of allergic airways responses. Lung responses after final allergen challenge in sensitized rats were diminished, although ozone exposure increased BALF IL-6, IL-13, IL-1 β, and interferon-γ and modified ventilatory responses only in the allergen group. Final allergen challenge also increased systolic and mean arterial BP, stroke volume, cardiac output, end-diastolic volume, sensitivity to aconitine-induced cardiac arrhythmia, and cardiac gene expression with lesser effects after a single challenge. These findings demonstrate that allergic airways responses may increase cardiovascular risk in part by altering BP and myocardial function and by causing cardiac electrical instability.
Collapse
Affiliation(s)
- Aimen K Farraj
- To whom correspondence should be addressed at US Environmental Protection Agency, 109 T.W. Alexander Drive, Mail Code: B105-02, Research Triangle Park, NC 27709, USA. E-mail:
| | - Brandi L Martin
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Mette C Schladweiler
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Colette N Miller
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Jacob Smoot
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Wanda Williams
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Anna Fisher
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Wendy Oshiro
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Alan Tennant
- Biomolecular and Computational Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - W Kyle Martin
- Curriculum of Toxicology and Environmental Medicine, UNC Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Andres R Henriquez
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Rachel Grindstaff
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37830, USA
| | - Stephen H Gavett
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - M Ian Gilmour
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Urmila P Kodavanti
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Mehdi S Hazari
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Janice A Dye
- Public Health and Integrated Toxicology Division, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| |
Collapse
|
30
|
Theofilis P, Vordoni A, Kalaitzidis RG. Interplay between metabolic dysfunction-associated fatty liver disease and chronic kidney disease: Epidemiology, pathophysiologic mechanisms, and treatment considerations. World J Gastroenterol 2022; 28:5691-5706. [PMID: 36338895 PMCID: PMC9627426 DOI: 10.3748/wjg.v28.i39.5691] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/06/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
The recently proposed nomenclature change from non-alcoholic fatty liver disease to metabolic dysfunction-associated fatty liver disease (MAFLD) has resulted in the reappraisal of epidemiological trends and associations with other chronic diseases. In this context, MAFLD appears to be tightly linked to incident chronic kidney disease (CKD). This association may be attributed to multiple shared risk factors including type 2 diabetes mellitus, arterial hypertension, obesity, dyslipidemia, and insulin resistance. Moreover, similarities in their molecular pathophysiologic mechanisms can be detected, since inflammation, oxidative stress, fibrosis, and gut dysbiosis are highly prevalent in these pathologic states. At the same time, lines of evidence suggest a genetic predisposition to MAFLD due to gene polymorphisms, such as the PNPLA3 rs738409 G allele polymorphism, which may also propagate renal dysfunction. Concerning their management, available treatment considerations for obesity (bariatric surgery) and novel antidiabetic agents (glucagon-like peptide 1 receptor agonists, sodium-glucose co-transporter 2 inhibitors) appear beneficial in preclinical and clinical studies of MAFLD and CKD modeling. Moreover, alternative approaches such as melatonin supplementation, farnesoid X receptor agonists, and gut microbiota modulation may represent attractive options in the future. With a look to the future, additional adequately sized studies are required, focusing on preventing renal complications in patients with MAFLD and the appropriate management of individuals with concomitant MAFLD and CKD.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- Center for Nephrology “G. Papadakis,” General Hospital of Nikaia-Piraeus “Agios Panteleimon,” Nikaia 18454, Greece
| | - Aikaterini Vordoni
- Center for Nephrology “G. Papadakis,” General Hospital of Nikaia-Piraeus “Agios Panteleimon,” Nikaia 18454, Greece
| | - Rigas G Kalaitzidis
- Center for Nephrology “G. Papadakis,” General Hospital of Nikaia-Piraeus “Agios Panteleimon,” Nikaia 18454, Greece
| |
Collapse
|
31
|
Richter P, Cardoneanu A, Rezus C, Burlui AM, Rezus E. Non-Traditional Pro-Inflammatory and Pro-Atherosclerotic Risk Factors Related to Systemic Lupus Erythematosus. Int J Mol Sci 2022; 23:ijms232012604. [PMID: 36293458 PMCID: PMC9604037 DOI: 10.3390/ijms232012604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/16/2022] [Accepted: 10/17/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases (CVD) are one of the leading causes of high mortality in patients with systemic lupus erythematosus (SLE). The Framingham risk score and other traditional risk factors do not fully reflect the CVD risk in SLE patients. Therefore, in order to stratify these high-risk patients, additional biomarkers for subclinical CVD are needed. The mechanisms of atherogenesis in SLE are still being investigated. During the past decades, many reports recognized that inflammation plays a crucial role in the development of atherosclerosis. The aim of this report is to present novel proinflammatory and pro-atherosclerotic risk factors that are closely related to SLE inflammation and which determine an increased risk for the occurrence of early cardiovascular events.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Anca Cardoneanu
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence: (A.C.); (C.R.); Tel.: +40232301615 (A.C. & C.R.)
| | - Ciprian Rezus
- Department of Internal Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- “Sfantul Spiridon” Emergency Hospital, 700111 Iasi, Romania
- Correspondence: (A.C.); (C.R.); Tel.: +40232301615 (A.C. & C.R.)
| | - Alexandra Maria Burlui
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
32
|
Zhang J, Xu H, Yao M, Jia H, Cong H. The abnormal level and prognostic potency of multiple inflammatory cytokines in PCI-treated STEMI patients. J Clin Lab Anal 2022; 36:e24730. [PMID: 36245413 DOI: 10.1002/jcla.24730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Inflammatory cytokines modulate atherogenesis and plaque rupture to involve in ST-segment elevation myocardial infarction (STEMI) progression. The present study determined eight inflammatory cytokine levels in 212 percutaneous coronary intervention (PCI)-treated STEMI patients, aiming to comprehensively investigate their potency in estimating major adverse cardiac event (MACE) risk. METHODS Serum tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, IL-8, IL-10, IL-17A, vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) of 212 PCI-treated STEMI patients and 30 angina pectoris patients were determined using enzyme-linked immunosorbent assay. RESULTS TNF-α (52.5 (43.9-62.6) pg/ml versus 46.4 (39.0-59.1) pg/ml, p = 0.031), IL-8 (61.6 (49.6-81.7) pg/ml versus 46.7 (32.5-63.1) pg/ml, p = 0.001), IL-17A (57.4 (45.7-77.3) pg/ml versus 43.2 (34.2-64.6) pg/ml, p = 0.001), and VCAM-1 (593.6 (503.4-811.4) ng/ml versus 493.8 (390.3-653.7) ng/ml, p = 0.004) levels were elevated in STEMI patients compared to angina pectoris patients, while IL-1β (p = 0.069), IL-6 (p = 0.110), IL-10 (p = 0.052), and ICAM-1 (p = 0.069) were of no difference. Moreover, both IL-17A high (vs. low) (p = 0.026) and VCAM-1 high (vs. low) (p = 0.012) were linked with increased cumulative MACE rate. The multivariable Cox's analysis exhibited that IL-17A high (vs. low) (p = 0.034) and VCAM-1 high (vs. low) (p = 0.014) were independently associated with increased cumulative MACE risk. Additionally, age, diabetes mellitus, C-reactive protein, multivessel disease, stent length, and stent type were also independent factors for cumulative MACE risk. CONCLUSION IL-17A and VCAM-1 high level independently correlate with elevated MACE risk in STEMI patients, implying its potency in identifying patients with poor prognoses.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Cardiology, Clinical School of Thoracic, Tianjin Medical University, Tianjin, China.,Department of Cardiology, Affiliated Hospital of Hebei University, Baoding, China
| | - Huichuan Xu
- Department of Cardiology, Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Mingyan Yao
- Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Hongdan Jia
- Department of Cardiology, Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Hongliang Cong
- Department of Cardiology, Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| |
Collapse
|
33
|
Posadas-Sánchez R, Vargas-Alarcón G, Pérez-Méndez Ó, Pérez-Hernández N, Rodríguez-Pérez JM. Increased Carotid Intima-Media Thickness in Asymptomatic Individuals Is Associated with the PCSK9 (rs2149041) Gene Polymorphism in the Mexican Mestizo Population: Results of the GEA Cohort. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101531. [PMID: 36294964 PMCID: PMC9604912 DOI: 10.3390/life12101531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
The increase in carotid intima-media thickness (CIMT) and coronary artery calcification (CAC) are features of subclinical atherosclerosis that might be determined by the genetic background of patients. Among the multiple risk factors, the proprotein convertase subtilisin kexin type 9 (PCSK9) has a great impact on atheroma development. Then, we focused on the potential association of the PCSK9 gene polymorphism (rs2149041) with the risk of an increased CIMT. We included 881 unrelated, asymptomatic individuals (732 normal CIMT and 149 increased CIMT) who lacked coronary calcification (CAC score = 0). Under the recessive inheritance model and adjusted by several cardiovascular risk factors, the rs2149041 polymorphism, determined by TaqMan genotyping assay, was associated with a high risk of increased CIMT (OR = 2.10, 95% IC = 1.26–3.47, P recessive = 0.004). Our results suggest that the rs2149041 polymorphism could be a risk marker for increased CIMT in asymptomatic individuals without coronary artery disease determined by the absence of a CAC score.
Collapse
Affiliation(s)
- Rosalinda Posadas-Sánchez
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Óscar Pérez-Méndez
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
| | - Nonanzit Pérez-Hernández
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
- Correspondence: (N.P.-H.); (J.M.R.-P.); Tel.: +52-55-55732911 (ext. 26301) (N.P.-H. & J.M.R.-P.)
| | - José Manuel Rodríguez-Pérez
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México 14080, Mexico
- Correspondence: (N.P.-H.); (J.M.R.-P.); Tel.: +52-55-55732911 (ext. 26301) (N.P.-H. & J.M.R.-P.)
| |
Collapse
|
34
|
Granulocyte Colony-Stimulating Factor Ameliorates Endothelial Activation and Thrombotic Diathesis Biomarkers in a Murine Model of Hind Limb Ischemia. Biomedicines 2022; 10:biomedicines10092303. [PMID: 36140404 PMCID: PMC9496113 DOI: 10.3390/biomedicines10092303] [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: 07/30/2022] [Revised: 08/29/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Novel therapies in peripheral arterial disease, such as granulocyte colony-stimulating factor (GCSF) administration, might result in anti-atherosclerotic effects. In this study, we used 10-week-old male ApoE−/− mice, which were fed an atherosclerosis-inducing diet for four weeks. At the end of the four weeks, hind limb ischemia was induced through left femoral artery ligation, the atherosclerosis-inducing diet was discontinued, and a normal diet was initiated. Mice were then randomized into a control group (intramuscular 0.4 mL normal saline 0.9% for 7 days) and a group in which GCSF was administrated intramuscularly in the left hind limb for 7 days (100 mg/kg). In the GCSF group, but not in the control group, we observed significant reductions in the soluble adhesion molecules (vascular cell adhesion molecule-1 (sVCAM-1) and intercellular adhesion molecule-1 (sICAM-1)), sE-Selectin, and plasminogen activator inhibitor (PAI)-1 when they were measured through ELISA on the 1st and the 28th days after hind limb ischemia induction. Therefore, GCSF administration in an atherosclerotic mouse model of hind limb ischemia led to decreases in the biomarkers associated with endothelial activation and thrombosis. These findings warrant further validation in future preclinical studies.
Collapse
|
35
|
Jia F, Fei SF, Tong DB, Zhang S, Li JJ. Do Patients with Myocardial Infarction with Non-Obstructive Coronary Arteries Have Similar Prognosis Compared to Ones with MI-CAD? Angiology 2022; 74:407-416. [PMID: 35993693 DOI: 10.1177/00033197221121191] [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: 11/16/2022]
Abstract
Acute myocardial infarction (AMI) is a leading cause of morbidity and mortality worldwide and it is primarily caused by acute plaque disruption and coronary occlusion. Recent studies suggest that myocardial infarction with non-obstructive coronary arteries (MINOCA) also occurs but the underlying mechanisms have not been fully understood until recently. The evidence also suggests that the clinical outcomes of patients presenting with MINOCA are similar to AMI patients with obstructive coronary artery disease (MI-CAD), including all-cause mortality and major adverse cardiovascular events. The present narrative review considers the risk factors, pathological changes, and outcomes associated with MINOCA and compares them with MI-CAD.
Collapse
Affiliation(s)
- Fang Jia
- Department of Cardiology, 117850The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Si-Fan Fei
- Department of Cardiology, 117850The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - De-Bing Tong
- Department of Cardiology, 117850The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Sheng Zhang
- Department of Cardiology, 117850The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jian-Jun Li
- Cardio-Metabolic Center, 569172Fu Wai Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
36
|
Theofilis P, Sagris M, Oikonomou E, Antonopoulos AS, Siasos G, Tsioufis K, Tousoulis D. The impact of SGLT2 inhibitors on inflammation: A systematic review and meta-analysis of studies in rodents. Int Immunopharmacol 2022; 111:109080. [PMID: 35908505 DOI: 10.1016/j.intimp.2022.109080] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Inhibition of sodium-glucose cotransporter-2 (SGLT2) has received remarkable attention due to the beneficial effects observed in diabetes mellitus, heart failure, and kidney disease. Several mechanisms have been proposed for these pleiotropic effects, including anti-inflammatory ones. Our systematic review and meta-analysis aimed to assess the effect of SGLT2 inhibition on inflammatory markers in experimental models. METHODS A literature search was conducted to detect studies examining the effect of SGLT2 inhibitors on inflammatory markers [interleukin-6 (IL-6), C reactive protein (CRP), tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1)]. Consequently, a meta-analysis of the included studies was performed, assessing the differences in the levels of the inflammatory markers between the treatment groups as its primary outcome. Moreover, risk of bias, sensitivity analysis and publication bias were evaluated. RESULTS The systematic literature review yielded 30 studies whose meta-analysis suggested that treatment with an SGLT2 inhibitor resulted in decreases of IL-6 [standardized mean difference (SMD): -1.56, 95% CI -2.06 to -1.05), CRP (SMD: -2.17, 95% CI -2.80 to -1.53), TNF-α (SMD: -1.75, 95% CI -2.14 to -1.37), and MCP-1 (SMD: -2.04, 95% CI -2.91 to -1.17). The effect on CRP and TNF-α was of lesser magnitude in cases of empagliflozin use. Moderate-to-substantial heterogeneity and possible publication bias were noted. The findings remained largely unaffected after the sensitivity analyses, the exclusion of outlying studies, and trim-and-fill analyses. CONCLUSION The present meta-analysis suggests that SGLT2 inhibition results in reduction of inflammatory markers in animal models, further validating the suggested anti-inflammatory mechanism of action.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Marios Sagris
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece; 3rd Cardiology Department, "Sotiria" Regional Hospital for Chest Diseases, University of Athens Medical School, 11527 Athens, Greece
| | - Alexios S Antonopoulos
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece; 3rd Cardiology Department, "Sotiria" Regional Hospital for Chest Diseases, University of Athens Medical School, 11527 Athens, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, 11527 Athens, Greece.
| |
Collapse
|
37
|
Theofilis P, Sagris M, Antonopoulos AS, Oikonomou E, Tsioufis K, Tousoulis D. Non-Invasive Modalities in the Assessment of Vulnerable Coronary Atherosclerotic Plaques. Tomography 2022; 8:1742-1758. [PMID: 35894012 PMCID: PMC9326642 DOI: 10.3390/tomography8040147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/04/2022] [Accepted: 07/04/2022] [Indexed: 12/26/2022] Open
Abstract
Coronary atherosclerosis is a complex, multistep process that may lead to critical complications upon progression, revolving around plaque disruption through either rupture or erosion. Several high-risk features are associated with plaque vulnerability and may add incremental prognostic information. Although invasive imaging modalities such as optical coherence tomography or intravascular ultrasound are considered to be the gold standard in the assessment of vulnerable coronary atherosclerotic plaques (VCAPs), contemporary evidence suggests a potential role for non-invasive methods in this context. Biomarkers associated with deleterious pathophysiologic pathways, including inflammation and extracellular matrix degradation, have been correlated with VCAP characteristics and adverse prognosis. However, coronary computed tomography (CT) angiography has been the most extensively investigated technique, significantly correlating with invasive method-derived VCAP features. The estimation of perivascular fat attenuation as well as radiomic-based approaches represent additional concepts that may add incremental information. Cardiac magnetic resonance imaging (MRI) has also been evaluated in clinical studies, with promising results through the various image sequences that have been tested. As far as nuclear cardiology is concerned, the implementation of positron emission tomography in the VCAP assessment currently faces several limitations with the myocardial uptake of the radiotracer in cases of fluorodeoxyglucose use, as well as with motion correction. Moreover, the search for the ideal radiotracer and the most adequate combination (CT or MRI) is still ongoing. With a look to the future, the possible combination of imaging and circulating inflammatory and extracellular matrix degradation biomarkers in diagnostic and prognostic algorithms may represent the essential next step for the assessment of high-risk individuals.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
- Correspondence:
| | - Marios Sagris
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Alexios S. Antonopoulos
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Evangelos Oikonomou
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
- 3rd Cardiology Department, Thoracic Diseases Hospital “Sotiria”, University of Athens Medical School, 11527 Athens, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| | - Dimitris Tousoulis
- 1st Cardiology Department, “Hippokration” General Hospital, Medical School, University of Athens, 11527 Athens, Greece; (M.S.); (A.S.A.); (E.O.); (K.T.); (D.T.)
| |
Collapse
|
38
|
Martí-Carvajal AJ, De Sanctis JB, Hidalgo R, Martí-Amarista CE, Alegría E, Correa-Pérez A, Monge Martín D, Riera Lizardo RJ. Colchicine for the primary prevention of cardiovascular events. Hippokratia 2022. [DOI: 10.1002/14651858.cd015003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Arturo J Martí-Carvajal
- Cochrane Ecuador. Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC). Facultad de Ciencias de la Salud Eugenio Espejo ; Universidad UTE; Quito Ecuador
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
- Cátedra Rectoral de Médicina Basada en la Evidencia; Universidad de Carabobo; Valencia Venezuela
| | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine; Palacky University Olomouc, Faculty of Medicine and Dentistry; Czech Republic Czech Republic
| | - Ricardo Hidalgo
- Cochrane Ecuador. Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC). Facultad de Ciencias de la Salud Eugenio Espejo; Universidad Tecnológica Equinoccial; Quito Ecuador
| | | | - Eduardo Alegría
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
| | - Andrea Correa-Pérez
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
- Clinical Biostatistics Unit; Hospital Universitario Ramón y Cajal (IRYCIS); Madrid Spain
| | | | | |
Collapse
|
39
|
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
|
40
|
Li F, Ye P, Hao Y, Du J, Zhang H, Wang Z, Wang X, Zeng H, Ma Y, Lin J. A PCSK9 inhibitor induces a transient decrease in the neutrophil-lymphocyte ratio and monocyte-lymphocyte ratio in homozygous familial hypercholesterolemia patients. ATHEROSCLEROSIS PLUS 2022; 49:12-19. [PMID: 36644203 PMCID: PMC9833253 DOI: 10.1016/j.athplu.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/24/2022] [Accepted: 05/09/2022] [Indexed: 01/18/2023]
Abstract
Background and aims Extremely elevated levels of low-density lipoprotein-cholesterol (LDL-C) contribute to long-term chronic systemic inflammation in homozygous familial hypercholesterolemia (HoFH) patients. The aims of this study is to describe the inflammatory profile of HoFH patients and explore the effect of a PCSK9 inhibitor (PCSK9i) on a series of inflammatory biomarkers, including the neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), monocyte-HDL ratio (MHR), monocyte-lymphocyte ratio (MLR) and mean platelet volume-lymphocyte ratio (MPVLR). Methods In this prospective cohort study, 25 definitive HoFH patients on high-intensity statins plus ezetimibe were administered subcutaneous injections of 420 mg PCSK9i every 4 weeks (Q4W). The biochemical parameters and inflammatory profile were analyzed on the day before PCSK9i therapy and 3 months and 6 months after PCSK9i therapy. Results HoFH on the maximum tolerated statin dose plus ezetimibe displayed elevated lipid and disturbed blood biomarker profiles. After 3 months of add-on PCSK9i therapy, a significant reduction in LDL-C was observed. Moreover, the percentage and count of neutrophils, monocyte counts, MPV, and two inflammatory biomarkers, the NLR and MLR, were reduced. However, at 6 months of PCSK9i treatment, the NLR and MLR returned to pre-PCSK9i treatment levels. Conclusions PCSK9i induces a transient decrease in the NLR and MLR in HoFH patients in a lipid-lowering independent manner.
Collapse
Affiliation(s)
- Fangyuan Li
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Peking University Ditan Teaching Hospital, Beijing, China
| | - Pucong Ye
- Department of Critical Care Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Yu Hao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Juan Du
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hang Zhang
- Department of Atherosclerosis, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Zengtao Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xumin Wang
- College of Life Sciences, Yantai University, Yantai, China
| | - Hui Zeng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Peking University Ditan Teaching Hospital, Beijing, China,Corresponding author.. Beijing Ditan Hospital, Jingshundongjie 8, Beijing, 100015, China.
| | - Yaluan Ma
- The Institute of Basic Medical Theory of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China,Corresponding author.
| | - Jie Lin
- Department of Atherosclerosis, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China,Corresponding author.. Department of Atherosclerosis, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
41
|
Zheng Q, Chen J, Yuan Y, Zhang X, Li L, Zhai Y, Gong X, Li B. Structural characterization, antioxidant, and anti-inflammatory activity of polysaccharides from Plumula Nelumbinis. Int J Biol Macromol 2022; 212:111-122. [PMID: 35594937 DOI: 10.1016/j.ijbiomac.2022.05.097] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/18/2022]
Abstract
A polysaccharide from Plumula Nelumbinis (PNP), was isolated and purified. PNP had a molecular weight of 450 kDa and consisted five monosaccharides, including rhamnose, galacturonic acid, xylose, galactose, and arabinose. The methylation and nuclear magnetic resonance (NMR) analysis revealed that the main glycosidic linkage types of PNP were →5)-α-L-Araf-(1→, →3)-β-D-Galp-(1→, β-D-Xylp-(→1, →3,4)-β-D-Rhap-(1→, →4)-β-D-GalpA-(1→. In the range of 25-1200 μg/mL, PNP had no cytotoxicity to RAW264.7 cells. PNP could protect RAW264.7 cell from oxidative damage by reducing the production of ROS and MDA and the secretion of LDH, enhancing the activity of SOD, CAT, and GSH-Px, and increasing the content of GSH. Anti-inflammatory activity experiments showed that PNP inhibited the expression of NO, TNF-α, INF-γ, IL-1β, and IL-6. PNP could inhibit the activation of MAPK/NF-κB cell pathways. PNP could be used as a potential natural antioxidant and anti-inflammatory substance in functional foods and pharmaceuticals.
Collapse
Affiliation(s)
- Qingsong Zheng
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China
| | - Juncheng Chen
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China
| | - Yi Yuan
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China
| | - Xia Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China
| | - Lin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China; School of Chemical Engineering and Energy Technology, Dongguan University of Technology, College Road 1, Dongguan, 523808, China
| | - Yongzhen Zhai
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China
| | - Xiao Gong
- Agricultural Product Processing Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524001, China.
| | - Bing Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Ministry of Education Engineering Research Center of Starch & Protein Processing, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
42
|
Garcia C, Compagnon B, Poëtte M, Gratacap MP, Lapébie FX, Voisin S, Minville V, Payrastre B, Vardon-Bounes F, Ribes A. Platelet Versus Megakaryocyte: Who Is the Real Bandleader of Thromboinflammation in Sepsis? Cells 2022; 11:1507. [PMID: 35563812 PMCID: PMC9104300 DOI: 10.3390/cells11091507] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets are mainly known for their key role in hemostasis and thrombosis. However, studies over the last two decades have shown their strong implication in mechanisms associated with inflammation, thrombosis, and the immune system in various neoplastic, inflammatory, autoimmune, and infectious diseases. During sepsis, platelets amplify the recruitment and activation of innate immune cells at the site of infection and contribute to the elimination of pathogens. In certain conditions, these mechanisms can lead to thromboinflammation resulting in severe organ dysfunction. Here, we discuss the interactions of platelets with leukocytes, neutrophil extracellular traps (NETs), and endothelial cells during sepsis. The intrinsic properties of platelets that generate an inflammatory signal through the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome are discussed. As an example of immunothrombosis, the implication of platelets in vaccine-induced immune thrombotic thrombocytopenia is documented. Finally, we discuss the role of megakaryocytes (MKs) in thromboinflammation and their adaptive responses.
Collapse
Affiliation(s)
- Cédric Garcia
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - Baptiste Compagnon
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Michaël Poëtte
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Marie-Pierre Gratacap
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - François-Xavier Lapébie
- Service de Médecine Vasculaire, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Sophie Voisin
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
| | - Vincent Minville
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Bernard Payrastre
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - Fanny Vardon-Bounes
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Agnès Ribes
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| |
Collapse
|
43
|
Mak A, Chan JKY. Endothelial function and endothelial progenitor cells in systemic lupus erythematosus. Nat Rev Rheumatol 2022; 18:286-300. [PMID: 35393604 DOI: 10.1038/s41584-022-00770-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/13/2022]
Abstract
The observations that traditional cardiovascular disease (CVD) risk factors fail to fully account for the excessive cardiovascular mortality in patients with systemic lupus erythematosus (SLE) compared with the general population have prompted in-depth investigations of non-traditional, SLE-related risk factors that contribute to cardiovascular complications in patients with SLE. Of the various perturbations of vascular physiology, endothelial dysfunction, which is believed to occur in the earliest step of atherosclerosis, has been extensively investigated for its contribution to CVD risk in SLE. Endothelial progenitor cells (EPCs), which play a crucial part in vascular repair, neovascularization and maintenance of endothelial function, are quantitatively and functionally reduced in patients with SLE. Yet, the lack of a unified definition of EPCs, standardization of the quantity and functional assessment of EPCs as well as endothelial function measurement pose challenges to the translation of endothelial function measurements and EPC levels into prognostic markers for CVD in patients with SLE. This Review discusses factors that contribute to CVD in SLE, with particular focus on how endothelial function and EPCs are evaluated currently, and how EPCs are quantitatively and functionally altered in patients with SLE. Potential strategies for the use of endothelial function measurements and EPC quantification as prognostic markers of CVD in patients with SLE, and the limitations of their prognostication potential, are also discussed.
Collapse
Affiliation(s)
- Anselm Mak
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Division of Rheumatology, University Medicine Cluster, National University Health System, Singapore, Singapore.
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Academic Clinical Programme in Obstetrics and Gynaecology, Duke-NUS Medical School, Singapore, Singapore.,Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
44
|
Yue M, Hu M, Fu F, Ruan H, Wu C. Emerging Roles of Platelets in Allergic Asthma. Front Immunol 2022; 13:846055. [PMID: 35432313 PMCID: PMC9010873 DOI: 10.3389/fimmu.2022.846055] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a complex chronic inflammatory disease of the airways, driven by Th2 immune responses and characterized by eosinophilic pulmonary inflammation, airway hyperresponsiveness, excessive mucus production, and airway remodeling. Overwhelming evidence from studies in animal models and allergic asthmatic patients suggests that platelets are aberrantly activated and recruited to the lungs. It has been established that platelets can interact with other immune cells and secrete various biochemical mediators to promote allergic sensitization and airway inflammatory response, and platelet deficiency may alleviate the pathological features and symptoms of allergic asthma. However, the comprehensive roles of platelets in allergic asthma have not been fully clarified, leaving attempts to treat allergic asthma with antiplatelet agents questionable. In this review, we summarize the role of platelet activation and pulmonary accumulation in allergic asthma; emphasis is placed on the different interactions between platelets with crucial immune cell types and the contribution of platelet-derived mediators in this context. Furthermore, clinical antiplatelet approaches to treat allergic asthma are discussed. This review provides a clearer understanding of the roles of platelets in the pathogenesis of allergic asthma and could be informative in the development of novel strategies for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Ming Yue
- Department of Physiology, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengjiao Hu
- Department of Immunology and Microbiology, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangda Fu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongfeng Ruan
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Hongfeng Ruan,
| | - Chengliang Wu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
45
|
Schönknecht YB, Crommen S, Stoffel-Wagner B, Coenen M, Fimmers R, Stehle P, Egert S. Influence of a proinflammatory state on postprandial outcomes in elderly subjects with a risk phenotype for cardiometabolic diseases. Eur J Nutr 2022; 61:3077-3083. [PMID: 35352134 PMCID: PMC9363402 DOI: 10.1007/s00394-022-02870-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 03/08/2022] [Indexed: 12/04/2022]
Abstract
Purpose Low-grade inflammation in obesity is associated with insulin resistance and other metabolic disturbances. In response to high-energy meal intake, blood concentrations of inflammatory markers, glucose and insulin rise. The aim of this study was to examine whether a basal inflammatory state influences postprandial responses. Methods A randomized crossover trial was performed in 60 participants with a cardiometabolic risk phenotype (age 70 ± 5 years; BMI 30.9 ± 3.1 kg/m2). Each participant consumed three different iso-energetic meals (4300 kJ): a Western diet-like high-fat meal (WDHF), a Western diet-like high-carbohydrate meal (WDHC) and a Mediterranean diet-like meal (MED). Blood samples were collected when fasted and hourly for 5 h postprandially and analyzed for glucose, insulin, interleukin-1β (IL-1β), interleukin-6 (IL-6) and endothelial adhesion molecules. Based on fasting serum C-reactive protein (CRP) concentrations, participants were assigned to a high inflammation (CRP ≥ 2.0 mg/L; n = 30) or low inflammation (CRP < 2.0 mg/L; n = 30) group, and postprandial outcomes were compared. Results Plasma IL-6, glucose and serum insulin increased after all meals, while IL-1β and endothelial adhesion molecules were unchanged. The high inflammation group had higher fasting and postprandial IL-6 concentrations than the low inflammation group, although the IL-6 response slope was similar between groups. In response to the WDHC meal, participants in the high inflammation group experienced a higher glycaemic response than those in the low inflammation group. Conclusion A basal proinflammatory state results in higher absolute fasting and postprandial IL-6 concentrations, but the increase in IL-6 relative to basal levels is not different between high and low inflammation groups. Elevated glycaemic response in the high inflammation group may be due to inflammation-induced short-term insulin resistance. The trial was registered at http://www.germanctr.de and http://www.drks.de under identifier DRKS00009861 (registration date, January 22, 2016).
Collapse
Affiliation(s)
- Yannik Bernd Schönknecht
- Department of Nutrition and Food Science, Nutritional Physiology, University of Bonn, Nussallee 9, 53115, Bonn, Germany
| | - Silke Crommen
- Department of Nutrition and Food Science, Nutritional Physiology, University of Bonn, Nussallee 9, 53115, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Martin Coenen
- Clinical Study Core Unit, Study Center Bonn, Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Rolf Fimmers
- Institute of Medical Biometry, Informatics and Epidemiology, University Hospital Bonn, Bonn, Germany
| | - Peter Stehle
- Department of Nutrition and Food Science, Nutritional Physiology, University of Bonn, Nussallee 9, 53115, Bonn, Germany
| | - Sarah Egert
- Department of Nutrition and Food Science, Nutritional Physiology, University of Bonn, Nussallee 9, 53115, Bonn, Germany.
| |
Collapse
|
46
|
Factors Associated with Platelet Activation-Recent Pharmaceutical Approaches. Int J Mol Sci 2022; 23:ijms23063301. [PMID: 35328719 PMCID: PMC8955963 DOI: 10.3390/ijms23063301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Platelets are at the forefront of human health and disease following the advances in their research presented in past decades. Platelet activation, their most crucial function, although beneficial in the case of vascular injury, may represent the initial step for thrombotic complications characterizing various pathologic states, primarily atherosclerotic cardiovascular diseases. In this review, we initially summarize the structural and functional characteristics of platelets. Next, we focus on the process of platelet activation and its associated factors, indicating the potential molecular mechanisms involving inflammation, endothelial dysfunction, and miRs. Finally, an overview of the available antiplatelet agents is being portrayed, together with agents possessing off-set platelet-inhibitory actions, while an extensive presentation of drugs under investigation is being given.
Collapse
|
47
|
Zeng S, Yi R, Tan F, Sun P, Cheng Q, Zhao X. Lactobacillus plantarum HFY05 Attenuates Carrageenan-Induced Thrombosis in Mice by Regulating NF-κB Pathway-Associated Inflammatory Responses. Front Nutr 2022; 9:813899. [PMID: 35308280 PMCID: PMC8931398 DOI: 10.3389/fnut.2022.813899] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/31/2022] [Indexed: 01/02/2023] Open
Abstract
In this study, a carrageenan-induced thrombus model was established in mice to observe the ability of Lactobacillus plantarum KFY05 (LP-KFY05) to inhibit thrombosis through an NF-κB-associated pathway. Biochemical analysis, microscopical observations, quantitative polymerase chain reactions (qPCR) and western blot analysis were used to examine relevant serum and tissue indexes, and the composition of intestinal microorganisms was determined by examining the abundance of microorganisms in feces. The results showed that LP-KFY05 could markedly reduce the degree of black tail in thrombotic mice; increase the activated partial thromboplastin time (APTT); and decrease the thrombin time (TT), fibrinogen (FIB) level, and prothrombin time (PT). LP-KFY05 could also reduce tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) levels in sera and renal tissues of thrombotic mice. Hematoxylin and eosin staining showed that LP-KFY05 could alleviate renal tissue lesions and tail vein thrombosis. qPCR results showed that LP-KFY05 could down-regulate nuclear factor kappa-B (NF-κB) p65, IL-6, TNF-α, and interferon γ (IFN-γ) mRNA expression in renal tissues, as well as NF-κB p65, intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin mRNA expression in tail vein vascular tissues of thrombotic mice. Western blot analysis showed that LP-KFY05 also down-regulated NF-κB protein expression in renal and tail vein vascular tissues of thrombotic mice. Lastly, LP-KFY05 increased the abundances of Bacteroidetes, Lactobacillus, and Bifidobacterium, as well as decreased the abundance of Firmicutes. These results show that LP-KFY05 can reduce inflammation and inhibit thrombosis in thrombotic mice, and the effects of high concentrations of LP-KFY05 were most pronounced, which were similar to the effects of dipyridamole.
Collapse
Affiliation(s)
- Shi Zeng
- Department of Neurosurgery, People's Hospital of Chongqing Banan District, Chongqing, China
| | - Ruokun Yi
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Fang Tan
- Department of Public Health, Our Lady of Fatima University, Valenzuela, Philippines
| | - Peng Sun
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| | - Qiang Cheng
- Department of Neurosurgery, People's Hospital of Chongqing Banan District, Chongqing, China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing Engineering Research Center of Functional Food, Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing, China
| |
Collapse
|
48
|
Ahmad F, Kannan M, Ansari AW. Role of SARS-CoV-2 -induced cytokines and growth factors in coagulopathy and thromboembolism. Cytokine Growth Factor Rev 2022; 63:58-68. [PMID: 34750061 PMCID: PMC8541834 DOI: 10.1016/j.cytogfr.2021.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/08/2023]
Abstract
Severe COVID-19 patients frequently present thrombotic complications which commonly lead to multiorgan failure and increase the risk of death. Severe SARS-CoV-2 infection induces the cytokine storm and is often associated with coagulation dysfunction. D-dimer, a hallmark of venous thromboembolism (VTE), is observed at a higher level in the majority of hospitalized COVID-19 patients. The precise molecular mechanism of the disproportionate effect of SARS-CoV-2 infection on the coagulation system is largely undefined. SARS-CoV-2 -induced endotheliopathy and, induction of cytokines and growth factors (GFs) most likely play important roles in platelet activation, coagulopathy, and VTE. Generally, viral infections lead to systemic inflammation and induction of numerous cytokines and GFs and many of them are reported to be associated with increased VTE. Most importantly, platelets play key thromboinflammatory roles linking coagulation to immune mediators in a variety of infections including response to viral infection. Since the pathomechanism of coagulopathy and VTE in COVID-19 is largely undefined, herein we highlight the association of dysregulated inflammatory cytokines and GFs with thrombotic complications and coagulopathy in COVID-19.
Collapse
Affiliation(s)
- Firdos Ahmad
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE.
| | - Meganathan Kannan
- Blood and Vascular Biology Research Lab, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610005, India
| | - Abdul W Ansari
- Dermatology Institute, Translational Research Institute, Academic Health Systems, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| |
Collapse
|
49
|
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology. Cells 2022; 11:cells11020213. [PMID: 35053329 PMCID: PMC8773869 DOI: 10.3390/cells11020213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The manifold actions of the pro-inflammatory and regenerative chemokine CXCL12/SDF-1α are executed through the canonical GProteinCoupledReceptor CXCR4, and the non-canonical ACKR3/CXCR7. Platelets express CXCR4, ACKR3/CXCR7, and are a vital source of CXCL12/SDF-1α themselves. In recent years, a regulatory impact of the CXCL12-CXCR4-CXCR7 axis on platelet biogenesis, i.e., megakaryopoiesis, thrombotic and thrombo-inflammatory actions have been revealed through experimental and clinical studies. Platelet surface expression of ACKR3/CXCR7 is significantly enhanced following myocardial infarction (MI) in acute coronary syndrome (ACS) patients, and is also associated with improved functional recovery and prognosis. The therapeutic implications of ACKR3/CXCR7 in myocardial regeneration and improved recovery following an ischemic episode, are well documented. Cardiomyocytes, cardiac-fibroblasts, endothelial lining of the blood vessels perfusing the heart, besides infiltrating platelets and monocytes, all express ACKR3/CXCR7. This review recapitulates ligand induced differential trafficking of platelet CXCR4-ACKR3/CXCR7 affecting their surface availability, and in regulating thrombo-inflammatory platelet functions and survival through CXCR4 or ACKR3/CXCR7. It emphasizes the pro-thrombotic influence of CXCL12/SDF-1α exerted through CXCR4, as opposed to the anti-thrombotic impact of ACKR3/CXCR7. Offering an innovative translational perspective, this review also discusses the advantages and challenges of utilizing ACKR3/CXCR7 as a potential anti-thrombotic strategy in platelet-associated cardiovascular disorders, particularly in coronary artery disease (CAD) patients post-MI.
Collapse
|
50
|
Saiki P, Yoshihara M, Kawano Y, Miyazaki H, Miyazaki K. Anti-Inflammatory Effects of Heliangin from Jerusalem Artichoke (Helianthus tuberosus) Leaves Might Prevent Atherosclerosis. Biomolecules 2022; 12:biom12010091. [PMID: 35053238 PMCID: PMC8774036 DOI: 10.3390/biom12010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is considered the major cause of cardiovascular and cerebrovascular diseases, which are the leading causes of death worldwide. Excessive nitric oxide production and inflammation result in dysfunctional vascular endothelial cells, which are critically involved in the initiation and progression of atherosclerosis. The present study aimed to identify a bioactive compound from Jerusalem artichoke leaves with anti-inflammatory activity that might prevent atherosclerosis. We isolated bioactive heliangin that inhibited NO production in LPS-induced macrophage-like RAW 264.7 cells. Heliangin suppressed ICAM-1, VCAM-1, E-selectin, and MCP-1 expression, as well as NF-κB and IκBα phosphorylation, in vascular endothelial cells stimulated with TNF-α. These results suggested that heliangin suppresses inflammation by inhibiting excessive NO production in macrophages and the expression of the factors leading to the development of atherosclerosis via the NF-κB signaling pathway in vascular endothelial cells. Therefore, heliangin in Jerusalem artichoke leaves could function in the prevention of atherosclerosis that is associated with heart attacks and strokes.
Collapse
Affiliation(s)
- Papawee Saiki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
- Correspondence: ; Tel.: +81-29-861-4304
| | - Mizuki Yoshihara
- Graduate School of Life and Environment Sciences, University of Tsukuba, Tsukuba 305-8577, Japan; (M.Y.); (H.M.)
| | - Yasuhiro Kawano
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
| | - Hitoshi Miyazaki
- Graduate School of Life and Environment Sciences, University of Tsukuba, Tsukuba 305-8577, Japan; (M.Y.); (H.M.)
| | - Koyomi Miyazaki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
| |
Collapse
|