1
|
Huang X, Hu L, Li J, Xie X, Tao S, Xue T, Zhang W. The association of nutritional and inflammatory status with cardiovascular and all-cause mortality risk among US patients with metabolic syndrome. Sci Rep 2025; 15:9589. [PMID: 40113950 PMCID: PMC11926066 DOI: 10.1038/s41598-025-94061-7] [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: 08/16/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
To investigate the relationship between nutritional and inflammatory status and all-cause and cardiovascular mortality in the U.S. population with cardiometabolic syndrome (CMS) and to identify the optimal nutrition-inflammation index for assessing long-term prognosis. Health data from the 1999-2010 National Health and Nutrition Examination Survey (NHANES) were used. Kaplan-Meier analysis explored associations between nutrition-inflammation indices and mortality in the CMS population. Significant indices were selected for ROC curve analysis, and the most effective index was analyzed using COX regression models. Restricted cubic splines (RCS) analyzed nonlinear associations, and a recursive algorithm determined inflection points. Subgroup and sensitivity analyses assessed the stability of the model. The study included data from 5,969 participants (2,900 males, 3,069 females), with 1,753 all-cause deaths and 607 cardiovascular deaths. Kaplan-Meier analysis indicated that good nutrition and low inflammation were linked to better long-term outcomes. The Advanced Lung Cancer Inflammation Index (ALI) was the most effective assessing index. COX regression showed a negative association between ALI and both mortality types. RCS analysis revealed a U-shaped relationship for all-cause mortality and an L-shaped curve for cardiovascular mortality, with an inflection point at 106.24. Subgroup analysis and sensitivity analysis confirmed the robustness of ALI in assessing the mortality risk in the CMS population. ALI is an ideal indicator for evaluating the relationship between nutritional and inflammatory status and both all-cause and cardiovascular mortality in the CMS population. Maintaining an appropriate ALI level can help reduce the risk of all-cause and cardiovascular mortality in CMS patients.
Collapse
Affiliation(s)
- Xuanchun Huang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Lanshuo Hu
- Xiyuan Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Jun Li
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China.
| | - Xiaoling Xie
- Zhangzhou Hospital of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Zhangzhou, Fujian, China
| | - Shiyi Tao
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Tiantian Xue
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China.
| | - Wenjie Zhang
- Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Shabani E, Hasanzadi A, Allela OQB, Kareem RA, Abed RE, Al-Nuaimi AMA, Athab ZH, Khodarahmi S. The role of lipids and lipids lowering drugs in human papillomavirus (HPV) and HPV-associated cancers. Infect Agent Cancer 2025; 20:4. [PMID: 39876011 PMCID: PMC11773819 DOI: 10.1186/s13027-025-00635-5] [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: 11/27/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025] Open
Abstract
Both women and men are now confronted with the grave threat of cancers caused by the human papillomavirus (HPV). It is estimated that 80% of women may encounter HPV over their lives. In the preponderance of cases involving anal, head and neck, oral, oropharyngeal, penile, vaginal, vulvar, and cervical malignancies, high-risk HPV (HR-HPV) is the causative agent. In 2019, HPV is believed to have been the cause of 620,000 new cases of cancer in women and 70,000 new cases of cancer in men worldwide. The bulk of the 530,000 cervical cancer cases (~ 270,000 fatalities) caused by HPV infection (86% of cases, 88% of deaths) happen in poor nations each year. Lipid metabolism is crucial in HPV infection and cancer development related to HPV. One of the most noticeable metabolic abnormalities in cancer is lipid metabolism reprogramming, in which cancer cells dysregulate lipid metabolism to obtain sufficient energy, building blocks for cell membranes, and signaling molecules necessary for invasion, metastasis, proliferation, and survival. Moreover, HPV proteins' stimulation of lipid production in infected cells will probably have a significant effect on oncogenesis. In addition, lipids are critical in producing cellular energy, the epithelial-mesenchymal transition (EMT) process, and therapy resistance of HPV-related cancers (HRCs). Therefore, lipids are essential in HPV infection and HRC development and may also be an important target for new approaches associated with treatments during HPV infection or cancer development. This review study looked at the role of lipids and lipid-lowering drugs in HPV and related cancers.
Collapse
Affiliation(s)
- Ehsan Shabani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Aida Hasanzadi
- Department of Obstetric and Gynecology, University of Gilan, Rasht, Iran
| | | | | | - Riyad E Abed
- College of Health and Medical Technology, National University of Science and Technology, Nasiriyah, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Shiva Khodarahmi
- Shahid Beheshti University of Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
3
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
4
|
Drăgan A, Drăgan AŞ, Ştiru O. The Predictive Value of Perioperative Inflammatory Indexes in Major Arterial Surgical Revascularization from Leriche Syndrome. J Clin Med 2024; 13:6338. [PMID: 39518478 PMCID: PMC11546754 DOI: 10.3390/jcm13216338] [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: 09/05/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Objectives: The role of inflammation in the pathophysiology of atherosclerosis is extensive. Our study aims to assess the predictive role of inflammatory indexes regarding in-hospital mortality in major vascular surgery of Leriche syndrome as a convenient, low-cost, and noninvasive prognostic marker to optimize the patient's perioperative course. Methods: Our retrospective single-center study enrolled consecutive patients diagnosed with aortoiliac occlusive disease, Leriche syndrome, who underwent elective major vascular surgery between 2017 and 2023 in a tertiary cardiovascular center. Preoperative, postoperative, and day-one after-surgery data, including systemic immune-inflammation index (SII), systemic inflammation response index (SIRI), aggregate index of systemic inflammation (AISI), neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio, and monocyte-lymphocyte ratio, were studied to the endpoint, in-hospital death. We also tested the delta values of the indexes to the endpoint. The indexes were compared to the Revised Cardiac Risk Index (RCRI) and Vascular Surgery Group Cardiac Risk Index (VSG-CRI) for outcome prediction. Results: The tested inflammatory indexes significantly increased from the preoperative to postoperative and, further, to the day-one settings. Preoperative AISI (p = 0.040) emerged as the only independent risk factor regarding in-hospital death occurrence in Leriche patients who underwent major revascularization surgery. While RCRI did not significantly predict the endpoint (AUC = 0.698, p = 0.057), VSG-CRI (AUC = 0.864, p = 0.001) presented the best result in ROC analysis. Postoperative NLR (AUC = 0.758, p = 0.006) was next, followed by NLR postoperative-preoperative (_Preop-_Postop) delta value (AUC = 0.725, p = 0.004), postoperative SIRI (AUC = 0.716, p = 0.016), SIRI_Preop-_Postop delta value (AUC = 0.712, p = 0.016), postoperative SII (AUC = 0.692, p = 0.032), and SII_Preop-_Postop delta value (AUC = 0.631, p = 0.030). Conclusions: Inflammatory indexes are valuable tools for assessing perioperative risk in major vascular surgery, enhancing the value of the already validated risk scores.
Collapse
Affiliation(s)
- Anca Drăgan
- Department of Cardiovascular Anaesthesiology and Intensive Care, “Prof. Dr. C.C. Iliescu” Emergency Institute for Cardiovascular Diseases, 258 Fundeni Road, 022328 Bucharest, Romania
| | - Adrian Ştefan Drăgan
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (A.Ş.D.); (O.Ş.)
| | - Ovidiu Ştiru
- Faculty of General Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania; (A.Ş.D.); (O.Ş.)
- Department of Cardiovascular Surgery, “Prof. Dr. C.C. Iliescu” Emergency Institute for Cardiovascular Diseases, 258 Fundeni Road, 022328 Bucharest, Romania
| |
Collapse
|
5
|
Barbieri L, Tumminello G, Fichtner I, Corsini A, Santos RD, Carugo S, Ruscica M. PCSK9 and Coronary Artery Plaque-New Opportunity or Red Herring? Curr Atheroscler Rep 2024; 26:589-602. [PMID: 39150672 PMCID: PMC11393034 DOI: 10.1007/s11883-024-01230-6] [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] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE OF REVIEW Although the clinical benefit of reducing low-density lipoprotein cholesterol (LDLc) in patients with coronary artery disease (CAD) is well-established, the impact on plaque composition and stability is less clear. Our narrative review aimed to assess the clinical effects of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors on coronary plaque characteristics specifically focusing from atheroma progression to regression and stabilization. RECENT FINDINGS The combination of statin therapy and PCSK9 inhibitors (evolocumab and alirocumab) promotes plaque stability in patients following an acute coronary syndrome. The GLAGOV study highlighted the relationship between achieved LDLc levels and changes in percentage atheroma volume. Similarly, the PACMAN-AMI study concluded that the qualitative and quantitative changes in coronary plaque were associated with the levels of LDLc. Assessing the severity of coronary artery stenosis and the extent of atherosclerotic burden by means of imaging techniques (e.g., IVUS, OCT and near-infrared spectroscopic) have significantly advanced our understanding of the benefits from promoting plaque regression and achieving to features of plaque stabilization through increasingly intensive lipid-lowering strategies.
Collapse
Affiliation(s)
- Lucia Barbieri
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriele Tumminello
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy
| | - Raul D Santos
- Heart Institute (InCor), Lipid Clinic, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pharmacological and Biomolecular Sciences, Rodolfo Paoletti", Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
6
|
Lamia TH, Shah-Riar P, Khanam M, Khair F, Sadat A, Tania MK, Haque SM, Saaki SS, Ferdausi A, Naurin SA, Tabassum M, Rahie RET, Hasan R. Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors as Adjunct Therapy to Statins: A New Frontier in Cardiovascular Risk Reduction. Cureus 2024; 16:e71365. [PMID: 39539858 PMCID: PMC11558015 DOI: 10.7759/cureus.71365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2024] [Indexed: 11/16/2024] Open
Abstract
Lowering low-density lipoprotein cholesterol (LDL-C) plasma levels is crucial for the prevention of primary and secondary cardiovascular diseases (CVDs). Many patients struggle to obtain goal LDL-C levels, despite the availability of several lipid-lowering medications, because of limited efficaciousness and unfavorable side effects. Proprotein convertase subtilisin/kexin type 9 (PCSK9) targeting has drawn interest recently as a novel approach to further lower cardiovascular (CV) risk. The number of receptors accessible to remove LDL-C from the bloodstream is reduced when PCSK9 attaches to LDL-C receptors and directs them toward lysosomal destruction. LDL receptor activity is increased by PCSK9 inhibition, which attracts therapeutic intervention. Despite concurrent statin therapy, phase 3 clinical trials have demonstrated encouraging outcomes with monoclonal antibodies against PCSK9, such as evolocumab and alirocumab, resulting in significant reductions in LDL-C levels. This study intends to investigate recent advancements in the field to evaluate PCSK9 inhibitors' safety, effectiveness, and potential for preventing CVD. The investigation will also review potential future paths and wider effects of using PCSK9 inhibitors in therapeutic settings.
Collapse
Affiliation(s)
| | - Prince Shah-Riar
- Internal Medicine, Doctors Hospital at Renaissance (DHR) Health, Edinburg, USA
| | - Mousumi Khanam
- Internal Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | - Farzana Khair
- Internal Medicine, Bangladesh Medical College, Dhaka, BGD
| | - Anahita Sadat
- Internal Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | - Maksuda Khan Tania
- Internal Medicine, Ibrahim Medical College and Birdem General Hospital, Dhaka, BGD
| | - Siddiqi M Haque
- Internal Medicine, University of California, Riverside School of Medicine, Riverside, USA
| | - Shaila S Saaki
- Internal Medicine, Dhaka Medical College and Hospital, Dhaka, BGD
| | | | | | - Maliha Tabassum
- Internal Medicine, Holy Family Red Crescent Medical College, Dhaka, BGD
| | | | - Rashedul Hasan
- Internal Medicine, Desert Valley Hospital, Victorville, USA
| |
Collapse
|
7
|
Pang S, Li X. Early use of SGLT2 inhibitors reduces the progression of diabetic kidney disease: a retrospective cohort study. Am J Transl Res 2024; 16:4967-4978. [PMID: 39398587 PMCID: PMC11470299 DOI: 10.62347/arya8831] [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: 05/29/2024] [Accepted: 07/29/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To evaluate the potential of sodium-glucose cotransporter 2 (SGLT2) inhibitors in preventing the progression of diabetic kidney disease and to provide guidance for clinical practice to improve renal health management strategies for diabetic patients. METHODS A retrospective analysis was conducted on 178 patients with diabetic kidney disease admitted to Baoji High Tech Hospital from March 2023 to March 2024. Of these, 88 patients who received early treatment with the SGLT2 inhibitor dapagliflozin were included in the early SGLT2-i group, while 90 patients receiving later treatment with SGLT2 inhibitor dapagliflozin were included in the late SGLT2-i group. Clinical data, overall effectiveness, adverse reactions, blood glucose, renal function, lipid levels, and inflammatory markers were compared between the two groups. RESULTS Prior to treatment, there were no differences in blood glucose indicators between the two groups (all P > 0.05). Following treatment, both groups showed reductions in 2-hour postprandial blood glucose (2hPG), fasting plasma glucose (FPG), and glycosylated hemoglobin (HbA1c), with the early SGLT2-i group demonstrating significantly lower values compared to the late SGLT2-i group (all P < 0.05). Similarly, there were no differences in renal function indicators between the two groups before treatment (all P > 0.05). However, following treatment, the early SGLT2-i group showed more noticeable improvements compared to the late SGLT2-i group (P < 0.05). Inflammatory markers and lipid levels followed similar patterns. The overall effectiveness of the early SGLT2-i group was higher than that of the late SGLT2-i group (92.05% vs. 78.89%, P < 0.05), while the incidence of adverse reactions did not differ statistically between the two groups (6.82% vs. 10.00%, P > 0.05). CONCLUSION Early use of SGLT2 inhibitors in diabetic kidney disease patients effectively controls blood glucose and lipid levels, improves renal function, reduces inflammatory responses, and exhibits a low incidence of adverse reactions. This demonstrates high safety and an important role in delaying disease progression. Therefore, it is worth considering clinical promotion and use for this patient population.
Collapse
Affiliation(s)
- Shaowei Pang
- Nephrology Department, Baoji High Tech Hospital Baoji 721000, Shaanxi, China
| | - Xiaoli Li
- Nephrology Department, Baoji High Tech Hospital Baoji 721000, Shaanxi, China
| |
Collapse
|
8
|
Yang Y, Zhang F, Jiang Z, Du Z, Liu S, Zhang M, Jin Y, Qin Y, Yang X, Wang C, Gao H. Microplastics are associated with elevated atherosclerotic risk and increased vascular complexity in acute coronary syndrome patients. Part Fibre Toxicol 2024; 21:34. [PMID: 39164741 PMCID: PMC11337598 DOI: 10.1186/s12989-024-00596-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Microplastics, widely present in the environment, are implicated in disease pathogenesis through oxidative stress and immune modulation. Prevailing research, primarily based on animal and cell studies, falls short in elucidating microplastics' impact on human cardiovascular health. This cross-sectional study detected blood microplastic concentrations in patients presenting with chest pain using pyrolysis-gas chromatography/mass spectrometry and evaluating inflammatory and immune markers through flow cytometry, to explore the potential effects of microplastic on acute coronary syndrome. RESULTS The study included 101 participants, comprising 19 controls and 82 acute coronary syndrome cases. Notably, acute coronary syndrome patients exhibited elevated microplastic concentrations, with those suffering from acute myocardial infarction presenting higher loads compared to those with unstable angina. Furthermore, patients at intermediate to high risk of coronary artery disease displayed significantly higher microplastic accumulations than their low-risk counterparts. A significant relationship was observed between increased microplastic levels and enhanced IL-6 and IL-12p70 contents, alongside elevated B lymphocyte and natural killer cell counts. CONCLUSION These results suggest an association between microplastics and both vascular pathology complexity and immunoinflammatory response in acute coronary syndrome, underscoring the critical need for targeted research to delineate the mechanisms of this association. HIGHLIGHTS 1 Blood microplastic levels escalate from angiographic patency, to angina patients, peaking in myocardial infarction patients. 2 Microplastics in acute coronary syndrome patients are predominantly PE, followed by PVC, PS, and PP. 3 Microplastics may induce immune cell-associated inflammatory responses in acute coronary syndrome patients.
Collapse
Affiliation(s)
- Yunxiao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Feng Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhili Jiang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Zhiyong Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Sheng Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Ming Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yanyan Jin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Yanwen Qin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xiubin Yang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Chenggang Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| | - Hai Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China.
| |
Collapse
|
9
|
Santangelo G, Gherbesi E, Donisi L, Faggiano A, Bergamaschi L, Pizzi C, Carugo S, Ruscica M, Faggiano P. Imaging approaches in risk stratification of patients with coronary artery disease: a narrative review. Arch Med Sci 2024; 21:16-31. [PMID: 40190322 PMCID: PMC11969509 DOI: 10.5114/aoms/188808] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 04/09/2025] Open
Abstract
Coronary artery disease (CAD) is the most common cause of mortality among adults worldwide. In the prognostic risk stratification of these patients, crucial determinants are lumen stenosis, total volume and composition of the plaque. Considering that most of the myocardial infarctions are due to non-obstructive plaques or are associated with high-risk features, plaque composition can serve as an independent predictor of cardiac outcomes. Conversely, although there is a close relationship between ischemia and CAD severity, the assessment of the degree of ischemia, as a surrogate marker of the coronary plaque burden, remains a controversial issue. Thus, aim of this narrative review is to discuss the usefulness of the imaging methodologies to differentiate the ischemia vs the plaque burden in clinical practice. New diagnostic tools to evaluate the extent of the atheromatous coronary artery could help in tailoring a personalized therapeutic approach.
Collapse
Affiliation(s)
- Gloria Santangelo
- Department of Cardio-Thoracic-Vascular Diseases, IRCCS Foundation Ca’ Granda Hospital Maggiore Polyclinic, Milan, Italy
| | - Elisa Gherbesi
- Department of Cardio-Thoracic-Vascular Diseases, IRCCS Foundation Ca’ Granda Hospital Maggiore Polyclinic, Milan, Italy
| | - Luca Donisi
- Department of Cardio-Thoracic-Vascular Diseases, IRCCS Foundation Ca’ Granda Hospital Maggiore Polyclinic, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Andrea Faggiano
- Department of Cardio-Thoracic-Vascular Diseases, IRCCS Foundation Ca’ Granda Hospital Maggiore Polyclinic, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Luca Bergamaschi
- Cardiology Unit, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences – DIMEC – Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Carmine Pizzi
- Cardiology Unit, IRCCS University Hospital of Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences – DIMEC – Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, IRCCS Foundation Ca’ Granda Hospital Maggiore Polyclinic, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Massimiliano Ruscica
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, University of Milan, Milan, Italy
| | - Pompilio Faggiano
- Poliambulance Foundation, Department of Cardiothoracic, Brescia, Italy
| |
Collapse
|
10
|
Di Fiore V, Cappelli F, Del Punta L, De Biase N, Armenia S, Maremmani D, Lomonaco T, Biagini D, Lenzi A, Mazzola M, Tricò D, Masi S, Mengozzi A, Pugliese NR. Novel Techniques, Biomarkers and Molecular Targets to Address Cardiometabolic Diseases. J Clin Med 2024; 13:2883. [PMID: 38792427 PMCID: PMC11122330 DOI: 10.3390/jcm13102883] [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: 04/06/2024] [Revised: 05/01/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
Cardiometabolic diseases (CMDs) are interrelated and multifactorial conditions, including arterial hypertension, type 2 diabetes, heart failure, coronary artery disease, and stroke. Due to the burden of cardiovascular morbidity and mortality associated with CMDs' increasing prevalence, there is a critical need for novel diagnostic and therapeutic strategies in their management. In clinical practice, innovative methods such as epicardial adipose tissue evaluation, ventricular-arterial coupling, and exercise tolerance studies could help to elucidate the multifaceted mechanisms associated with CMDs. Similarly, epigenetic changes involving noncoding RNAs, chromatin modulation, and cellular senescence could represent both novel biomarkers and targets for CMDs. Despite the promising data available, significant challenges remain in translating basic research findings into clinical practice, highlighting the need for further investigation into the complex pathophysiology underlying CMDs.
Collapse
Affiliation(s)
- Valerio Di Fiore
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Federica Cappelli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Lavinia Del Punta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Nicolò De Biase
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Silvia Armenia
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Davide Maremmani
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy; (T.L.)
| | - Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy; (T.L.)
| | - Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy; (T.L.)
| | - Matteo Mazzola
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Domenico Tricò
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| | - Nicola Riccardo Pugliese
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 67, 56124 Pisa, Italy (F.C.)
| |
Collapse
|
11
|
Raschi E, Casula M, Cicero AFG, Corsini A, Borghi C, Catapano A. Beyond statins: New pharmacological targets to decrease LDL-cholesterol and cardiovascular events. Pharmacol Ther 2023; 250:108507. [PMID: 37567512 DOI: 10.1016/j.pharmthera.2023.108507] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/13/2023]
Abstract
The pharmacological treatment of dyslipidemia, a major modifiable risk factor for developing atherosclerotic cardiovascular disease (ASCVD), remains a debated and controversial issue, not only in terms of the most appropriate therapeutic range for lipid levels, but also with regard to the optimal strategy and sequence approach (stepwise vs upstream therapy). Current treatment guidelines for the management of dyslipidemia focus on the intensity of low-density lipoprotein cholesterol (LDL-C) reduction, stratified according to risk for developing ASCVD. Beyond statins and ezetimibe, different medications targeting LDL-C have been recently approved by regulatory agencies with potential innovative mechanisms of action, including proprotein convertase subtilisin/kexin type 9 modulators (monoclonal antibodies such as evolocumab and alirocumab; small interfering RNA molecules such as inclisiran), ATP-citrate lyase inhibitors (bempedoic acid), angiopoietin-like 3 inhibitors (evinacumab), and microsomal triglyceride transfer protein inhibitors (lomitapide). An understanding of their pharmacological aspects, benefit-risk profile, including impact on hard cardiovascular endpoints beyond LDL-C reduction, and potential advantages from the patient perspective (e.g., adherence) - the focus of this evidence-based review - is crucial for practitioners across medical specialties to minimize therapeutic inertia and support clinical practice.
Collapse
Affiliation(s)
- Emanuel Raschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Manuela Casula
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Arrigo F G Cicero
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy; IRCCS AOU S. Orsola-Malpighi, Bologna, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Claudio Borghi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy; IRCCS AOU S. Orsola-Malpighi, Bologna, Italy
| | | |
Collapse
|
12
|
Xiu J, Lin X, Chen Q, Yu P, Lu J, Yang Y, Chen W, Bao K, Wang J, Zhu J, Zhang X, Pan Y, Tu J, Chen K, Chen L. The aggregate index of systemic inflammation (AISI): a novel predictor for hypertension. Front Cardiovasc Med 2023; 10:1163900. [PMID: 37265570 PMCID: PMC10229810 DOI: 10.3389/fcvm.2023.1163900] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
Abstract
Objective Inflammation plays an important role in the pathophysiology of hypertension (HTN). Aggregate index of systemic inflammation (AISI), as a new inflammatory and prognostic marker has emerged recently. Our goal was to determine whether there was a relationship between HTN and AISI. Methods We analyzed patients with HTN from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2018. The primary end point was cardiovascular mortality. A total of 23,765 participants were divided into four groups according to the AISI quartile level. The association between AISI and cardiovascular mortality in patients with HTN was assessed by survival curves and Cox regression analyses based on NHANES recommended weights. Results High levels of AISI were significantly associated with cardiovascular mortality in patients with HTN. After full adjustment for confounders, there was no significant difference in the risk of cardiovascular mortality in Q2 and Q3 compared to Q1, while Q4 (HR: 1.91, 95% CI: 1.42-2.58; P < 0.001) had a higher risk of cardiovascular mortality compared to Q1. Results remained similar in subgroup analyses stratified by age (P for interaction = 0.568), gender (P for interaction = 0.059), and obesity (P for interaction = 0.289). Conclusions In adults with HTN, elevated AISI levels are significantly associated with an increased risk of cardiovascular mortality and may serve as an early warning parameter for poor prognosis.
Collapse
Affiliation(s)
- Jiaming Xiu
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Xueqin Lin
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Qiansheng Chen
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Pei Yu
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Jin Lu
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Yanfang Yang
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Weihua Chen
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Kunming Bao
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Junjie Wang
- Department of Cardiology, Fuzhou First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinlong Zhu
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Xiaoying Zhang
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Yuxiong Pan
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Jiabin Tu
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
| | - Kaihong Chen
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| | - Liling Chen
- Department of Cardiology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China
| |
Collapse
|
13
|
Boyalla V, Gallego-Colon E, Spartalis M. Immunity and inflammation in cardiovascular disorders. BMC Cardiovasc Disord 2023; 23:148. [PMID: 36959565 PMCID: PMC10035189 DOI: 10.1186/s12872-023-03185-z] [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: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023] Open
Abstract
Recent studies have developed our understanding of the role of the immune system and inflammation in Cardiovascular Disease (CVD), opening new avenues for risk stratification and therapeutic intervention. However, gaps in our knowledge remain. To address this issue, BMC Cardiovascular Disorders has launched a Collection on "Immunity and Inflammation in Cardiovascular Disorders".
Collapse
Affiliation(s)
| | | | - Michael Spartalis
- National and Kapodistrian University of Athens, 32 Tsakalof Ave, Athens, 10673, Greece.
| |
Collapse
|
14
|
Horinouchi Y, Murashima Y, Yamada Y, Yoshioka S, Fukushima K, Kure T, Sasaki N, Imanishi M, Fujino H, Tsuchiya K, Shinomiya K, Ikeda Y. Pemafibrate inhibited renal dysfunction and fibrosis in a mouse model of adenine-induced chronic kidney disease. Life Sci 2023; 321:121590. [PMID: 36940907 DOI: 10.1016/j.lfs.2023.121590] [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: 01/27/2023] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
AIMS Peroxisome proliferator-activated receptor-alpha (PPARα) levels are markedly lower in the kidneys of chronic kidney disease (CKD) patients. Fibrates (PPARα agonists) are therapeutic agents against hypertriglyceridemia and potentially against CKD. However, conventional fibrates are eliminated by renal excretion, limiting their use in patients with impaired renal function. Here, we aimed to evaluate the renal risks associated with conventional fibrates via clinical database analysis and investigate the renoprotective effects of pemafibrate, a novel selective PPARα modulator mainly excreted into the bile. MAIN METHODS The risks associated with conventional fibrates (fenofibrate, bezafibrate) to the kidneys were evaluated using the Food and Drug Administration Adverse Event Reporting System. Pemafibrate (1 or 0.3 mg/kg/day) was administered daily using an oral sonde. Its renoprotective effects were examined in unilateral ureteral obstruction (UUO)-induced renal fibrosis model mice (UUO mice) and adenine-induced CKD model mice (CKD mice). KEY FINDINGS The ratios of glomerular filtration rate decreased and blood creatinine increased were markedly higher after conventional fibrate use. Pemafibrate administration suppressed increased gene expressions of collagen-I, fibronectin, and interleukin 1 beta (IL-1β) in the kidneys of UUO mice. In CKD mice, it suppressed increased plasma creatinine and blood urea nitrogen levels and decreased red blood cell count, hemoglobin, and hematocrit levels, along with renal fibrosis. Moreover, it inhibited the upregulation of monocyte chemoattractant protein-1, IL-1β, tumor necrosis factor-alpha, and IL-6 in the kidneys of CKD mice. SIGNIFICANCE These results demonstrated the renoprotective effects of pemafibrate in CKD mice, confirming its potential as a therapeutic agent for renal disorders.
Collapse
Affiliation(s)
- Yuya Horinouchi
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Yuka Murashima
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Yuto Yamada
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Shun Yoshioka
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Keijo Fukushima
- Department of Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences & Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
| | - Takumi Kure
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Naofumi Sasaki
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Masaki Imanishi
- Department of Medical Pharmacology, Graduate School of Pharmaceutical Sciences & Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
| | - Hiromichi Fujino
- Department of Pharmacology for Life Sciences, Graduate School of Pharmaceutical Sciences & Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Graduate School of Pharmaceutical Sciences & Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan.
| | - Kazuaki Shinomiya
- Department of Pharmaceutical Care and Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan.
| | - Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| |
Collapse
|
15
|
Donniacuo M, De Angelis A, Telesca M, Bellocchio G, Riemma MA, Paolisso P, Scisciola L, Cianflone E, Torella D, Castaldo G, Capuano A, Urbanek K, Berrino L, Rossi F, Cappetta D. Atrial fibrillation: Epigenetic aspects and role of sodium-glucose cotransporter 2 inhibitors. Pharmacol Res 2023; 188:106591. [PMID: 36502999 DOI: 10.1016/j.phrs.2022.106591] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022]
Abstract
Atrial fibrillation (AF) is the most frequent arrhythmia and is associated with substantial morbidity and mortality. Pathophysiological aspects consist in the activation of pro-fibrotic signaling and Ca2+ handling abnormalities at atrial level. Structural and electrical remodeling creates a substrate for AF by triggering conduction abnormalities and cardiac arrhythmias. The care of AF patients focuses predominantly on anticoagulation, symptoms control and the management of risk factors and comorbidities. The goal of AF therapy points to restore sinus rhythm, re-establish atrioventricular synchrony and improve atrial contribution to the stroke volume. New layer of information to better comprehend AF pathophysiology, and identify targets for novel pharmacological interventions consists of the epigenetic phenomena including, among others, DNA methylation, histone modifications and noncoding RNAs. Moreover, the benefits of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in diabetic and non-diabetic patients at cardiovascular risk as well as emerging evidence on the ability of SGLT2i to modify epigenetic signature in cardiovascular diseases provide a solid background to investigate a possible role of this drug class in the onset and progression of AF. In this review, following a summary of pathophysiology and management, epigenetic mechanisms in AF and the potential of sodium-glucose SGLT2i in AF patients are discussed.
Collapse
Affiliation(s)
- M Donniacuo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - A De Angelis
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - M Telesca
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - G Bellocchio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - M A Riemma
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - P Paolisso
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium; Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via A. Pansini 5, 80131 Naples, Italy
| | - L Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - E Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - D Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - G Castaldo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Via A. Pansini 5, 80131 Naples, Italy; CEINGE-Advanced, Via G. Salvatore 486, 80131 Naples, Italy
| | - A Capuano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - K Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Via A. Pansini 5, 80131 Naples, Italy; CEINGE-Advanced, Via G. Salvatore 486, 80131 Naples, Italy.
| | - L Berrino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - F Rossi
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - D Cappetta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
16
|
Baumeister SE, Holtfreter B, Reckelkamm SL, Kocher T, Alayash Z, Ehmke B, Baurecht H, Nolde M. Genotype-driven NPC1L1 and PCSK9 inhibition and reduced risk of periodontitis. J Clin Periodontol 2023; 50:114-120. [PMID: 36054135 DOI: 10.1111/jcpe.13719] [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: 05/12/2022] [Revised: 08/02/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022]
Abstract
AIM Epidemiological and pre-clinical studies suggest a chemoprotective role of lipid-lowering agents in periodontitis. We tested the association of genetically proxied inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR), Niemann-Pick C1-Like 1 (NPC1L1) and proprotein convertase subtilisin/kexin type 9 (PCSK9) with periodontitis. MATERIALS AND METHODS Genetic variants in HMGCR, NCP1L1 and PCSK9 associated with low-density lipoprotein (LDL) cholesterol in a genome-wide association study (GWAS) meta-analysis (N = 188,578) were used to proxy therapeutic inhibition of HMGCR, NPC1L1 and PCSK9. For these genetic variants, associations with periodontitis were obtained from GWAS of 17,353 cases and 28,210 controls in the GeneLifestyle Interactions in Dental Endpoints consortium. Generalized weighted least squares analysis accounted for linkage disequilibrium of genotypes to derive pooled estimates. RESULTS While genetically proxied HMGCR inhibition equivalent to 1 mmol/L reduction in LDL was not associated with odds of periodontitis (odds ratio [OR] = 0.92 [95% confidence interval [CI]: 0.73; 1.16]; p = .4905; false discovery rate [FDR] = 0.4905), genetically proxied NPC1L1 (OR = 0.53 [95% CI: 0.35; 0.81]; p = .0038; FDR = 0.0077) and PCSK9 (OR = 0.84 [95% CI: 0.74; 0.95]; p = .0051; FDR = 0.0077) inhibition lowered the odds of periodontitis. CONCLUSIONS Genetically proxied inhibition of NCP1L1 and PCSK9 was associated with lower odds of periodontitis.
Collapse
Affiliation(s)
| | - Birte Holtfreter
- Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Lars Reckelkamm
- Institute of Health Services Research in Dentistry, University of Münster, Münster, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, University Medicine Greifswald, Greifswald, Germany
| | - Zoheir Alayash
- Institute of Health Services Research in Dentistry, University of Münster, Münster, Germany
| | - Benjamin Ehmke
- Clinic for Periodontology and Conservative Dentistry, University of Münster, Münster, Germany
| | - Hansjörg Baurecht
- Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Michael Nolde
- Institute of Health Services Research in Dentistry, University of Münster, Münster, Germany
| |
Collapse
|
17
|
Salunke AS, Nile SH, Kharat AS. A comparative study on fibrinolytic enzymes extracted from six Bacillus spp. isolated from fruit-vegetable waste biomass. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
18
|
Tummala R, Gupta M, Devanabanda AR, Bandyopadhyay D, Aronow WS, Ray KK, Mamas M, Ghosh RK. Bempedoic acid and its role in contemporary management of hyperlipidemia in atherosclerosis. Ann Med 2022; 54:1287-1296. [PMID: 35533049 PMCID: PMC9090378 DOI: 10.1080/07853890.2022.2059559] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Atherosclerotic heart disease is the leading cause of mortality and morbidity in the USA. Low density lipoprotein (LDL) has been the target for many hypolipidemic agents to modify atherosclerotic risk. Bempedoic acid is a novel hypolipidemic drug that inhibits the enzymatic activity of ATP citrate lyase in the cholesterol synthesis pathway. CLEAR Harmony, CLEAR Wisdom, CLEAR Tranquillity and CLEAR Serenity have shown safety and efficacy associated with long term administration of this drug. Studies have shown effectiveness in reducing LDL-C in both statin intolerant patients and in patients on maximally tolerated doses of statin. The fixed drug combination of bempedoic acid and ezetimibe in a recent phase III showed significant reduction in LDL compared with placebo, which might be a promising future for LDL reduction among statin intolerant patients. Bempedoic acid also reduced inflammatory markers like hs-CRP. Given these results, bempedoic acid alone and in combination with ezetimibe received the USA FDA approval for adults with heterozygous familial hypercholesterolaemia or established atherosclerotic cardiovascular disease. We present a comprehensive review exploring the underlying mechanism, pre-clinical studies, and clinical trials of bempedoic acid and discuss the potential future role of the drug in treating hyperlipidaemia.
Collapse
Affiliation(s)
| | - Manasvi Gupta
- Department of Internal Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - Arvind Reddy Devanabanda
- Department of Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Dhrubajyoti Bandyopadhyay
- Department of Cardiology, Westchester Medical Center and New York Medical College, New York, NY, USA
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center and New York Medical College, New York, NY, USA
| | - Kausik K Ray
- Imperial Centre for Cardiovascular Disease Prevention, London, UK
| | - Mamas Mamas
- Keele Cardiac Research Group, Institutes of Science and Technology in Medicine and Primary Care Keele University, Stoke-on-Trent, UK
| | - Raktim K Ghosh
- MedStar Heart and Vascular Institute, Union Memorial Hospital, Baltimore, MD, USA
| |
Collapse
|
19
|
Grześk G, Dorota B, Wołowiec Ł, Wołowiec A, Osiak J, Kozakiewicz M, Banach J. Safety of PCSK9 inhibitors. Biomed Pharmacother 2022; 156:113957. [DOI: 10.1016/j.biopha.2022.113957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
|
20
|
Wang Z, Li G, Li M, Hu L, Hao Z, Li Q, Sun C. Periostin contributes to the adventitial remodeling of atherosclerosis by activating adventitial fibroblasts. ATHEROSCLEROSIS PLUS 2022; 50:57-64. [PMID: 36643802 PMCID: PMC9833252 DOI: 10.1016/j.athplu.2022.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
Background and aims Adventitial remodeling is an important pathological process of atherosclerosis, but cues implicated in adventitial remodeling are far from fully understood. Periostin (POSTN), a matricellular protein, has been demonstrated to have multiple roles in cardiovascular diseases. The aim of the study was to explore the function of POSTN in adventitial remodeling during atherosclerosis. Methods An atherosclerosis model was constructed based on ApoE-/- mice fed a high-fat and high-cholesterol diet. The expression of POSTN in the adventitia of mouse atherosclerotic vascular specimens was detected by immunohistochemical staining. The roles of POSTN in regulating adventitial fibroblast activation were assessed by cell contractility and activation marker α-smooth muscle actin (α-SMA) expression evaluation in adventitial fibroblasts overexpressing POSTN. In addition, we performed quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting to examine the expression of the proinflammatory chemokines transforming growth factor-β1 (TGF-β1) and monocyte chemotactic protein 1 (MCP1), as well as some extracellular matrix (ECM)-related proteins, in POSTN-overexpressing adventitial fibroblasts. Finally, the integrin-related signaling pathway was detected upon POSTN overexpression in adventitial fibroblasts. Results POSTN was highly expressed in the adventitia of atherosclerotic aortae in the mouse atherosclerosis model and promoted the activation and contraction of adventitial fibroblasts. Meanwhile, POSTN also induced adventitial fibroblasts to express TGF-β1, monocyte chemotactic protein-1 (MCP1), and ECM-related proteins and activated the phosphorylation of focal adhesion kinase (FAK) and Src. Conclusions Our results revealed that POSTN is elevated in adventitia during atherosclerosis and contributes to the adventitial remodeling of atherosclerosis by activating adventitial fibroblasts.
Collapse
Key Words
- Adventitial fibroblasts
- Adventitial remodeling
- Atherosclerosis
- COL1A1, collagen Ⅰ
- COL3A1, collagen Ⅲ
- DMEM, Dulbecco's modified Eagle's medium
- ECM, extracellular matrix
- FAK, focal adhesion kinase
- FBS, fetal bovine serum
- MCP1, monocyte chemotactic protein-1
- MMPs, matrix metalloproteinases
- POSTN
- POSTN, periostin
- TGF-β1
- TGF-β1, transforming growth factor-β1
- qRT-PCR, quantitative real-time polymerase chain reaction
- α-SMA, α-smooth muscle actin
Collapse
Affiliation(s)
- Zhonghua Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China,Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Guoliang Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Mingpeng Li
- Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Lu Hu
- Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Zichen Hao
- Department of Cardiovascular Medicine, Affiliated to the First People's Hospital of Chenzhou of University of South China, Chenzhou No. 1 People's Hospital, The First Affiliated Hospital of Xiangnan University, Chenzhou, Hunan, China
| | - Qian Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chaofeng Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China,Corresponding author. Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
21
|
Awad K, Zaki MM, Mohammed M, Lewek J, Lavie CJ, Banach M. Effect of the Renin-Angiotensin System Inhibitors on Inflammatory Markers: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Mayo Clin Proc 2022; 97:1808-1823. [PMID: 36202494 DOI: 10.1016/j.mayocp.2022.06.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 05/01/2022] [Accepted: 06/30/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To synthesize more conclusive evidence on the anti-inflammatory effects of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs). METHODS PubMed, Scopus, and Embase were searched from inception until March 1, 2021. We included randomized controlled trials (RCTs) that assessed the effect of ACEIs or ARBs, compared with placebo, on any of the following markers: C-reactive protein (CRP), interleukin 6 (IL-6), or tumor necrosis factor α (TNF-α). Mean changes in the levels of these markers were pooled as a weighted mean difference (WMD) with a 95% CI. RESULTS Thirty-two RCTs (n=3489 patients) were included in the final analysis. Overall pooled analysis suggested that ACEIs significantly reduced plasma levels of CRP (WMD, -0.54 [95% CI, -0.88 to -0.21]; P=.002; I2=96%), IL-6 (WMD, -0.84 [95% CI, -1.03 to -0.64]; P<.001; I2=0%), and TNF-α (WMD, -12.75 [95% CI, -17.20 to -8.29]; P<.001; I2=99%). Moreover, ARBs showed a significant reduction only in IL-6 (WMD, -1.34 [95% CI, -2.65 to -0.04]; P=.04; I2=85%) and did not significantly affect CRP (P=.15) or TNF-α (P=.97) levels. The lowering effect of ACEIs on CRP levels remained significant with enalapril (P=.006) and perindopril (P=.01) as well as with a treatment duration of less than 24 weeks (WMD, -0.67 [95% CI, -1.07 to -0.27]; P=.001; I2=94%) and in patients with coronary artery disease (WMD, -0.75 [95% CI, -1.17 to -0.33]; P<.001; I2=96%). CONCLUSION Based on this meta-analysis, ACEIs showed a beneficial lowering effect on CRP, IL-6, and TNF-α, whereas ARBs were effective as a class in reduction of IL-6 only.
Collapse
Affiliation(s)
- Kamal Awad
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt; Zagazig University Hospitals, Zagazig, El-Sharkia, Egypt.
| | - Mahmoud Mohamed Zaki
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt; Zagazig University Hospitals, Zagazig, El-Sharkia, Egypt
| | - Maged Mohammed
- Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt; Zagazig University Hospitals, Zagazig, El-Sharkia, Egypt
| | - Joanna Lewek
- Department of Preventive Cardiology and Lipidology, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland; Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Carl J Lavie
- Department of Cardiovascular Diseases, John Ochsner Heart and Vascular Institute, Ochsner Clinical School-The University of Queensland School of Medicine, New Orleans, Louisiana
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland; Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The aim of creating an orally active non-statin cholesterol-lowering drug was achieved with bempedoic acid, a small linear molecule providing both a significant low-density lipoprotein cholesterol (LDL-C) reduction and an anti-inflammatory effect by decreasing high-sensitivity C-reactive protein. Bempedoic acid antagonizes ATP citrate-lyase, a cytosolic enzyme upstream of HMGCoA reductase which is the rate-limiting step of cholesterol biosynthesis. Bempedoic acid is a pro-drug converted to its active metabolite by very-long-chain acyl-CoA synthetase 1 which is present mostly in the liver and absent in skeletal muscles. This limits the risk of myalgia and myopathy. The remit of this review is to give clinical insights on the safety and efficacy of bempedoic acid and to understand for whom it should be prescribed. RECENT FINDINGS Bempedoic acid with a single daily dose (180 mg) reduces LDL-C by a mean 24.5% when given alone, by 18% when given on top of a major statin and by 38-40% when given in a fixed-dose combination with ezetimibe. Bempedoic acid does not lead to the risk of new-onset diabetes, and moderately improves the glycaemic profile. The extensive knowledge on bempedoic acid mechanism, metabolism and side effects has led to an improved understanding of the potential benefits of this agent and offers a possible alternative to cardiologists and clinical practitioners somewhat worn out today by the occurrence of the muscular side effects of statins.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy
| | - Stefano Carugo
- Department of Clinical Sciences and Community Health, Università Degli Studi Di Milano, Milan, Italy
- Fondazione Ospedale Maggiore IRCCS Policlinico Di Milano, Milan, Italy
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, 93-338, Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, 65-046, Zielona Gora, Poland
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università Degli Studi Di Milano, Milan, Italy
| |
Collapse
|
23
|
Ferri N, Ruscica M, Lupo MG, Vicenzi M, Sirtori CR, Corsini A. Pharmacological rationale for the very early treatment of acute coronary syndrome with monoclonal antibodies anti-PCSK9. Pharmacol Res 2022; 184:106439. [PMID: 36100012 DOI: 10.1016/j.phrs.2022.106439] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022]
Abstract
Immediate and aggressive lipid lowering therapies after acute coronary syndromes (ACS) and percutaneous coronary interventions (PCI) are supported by the ESC/EAS dyslipidemia guidelines, recommending the initiation of high-intensity statin therapy within the first 1-4 days of hospitalization. However, whether non statin lipid-lowering agents, added to statin treatment, could produce a further reduction in the risk of major adverse cardiovascular events (MACE) is still unknown. Thus, the efficacy of early treatment post-ACS with monoclonal antibodies (mAbs) anti PCSK9, evolocumab and alirocumab, is under investigation. The rationale to explore the rapid and aggressive pharmacological intervention with PCSK9 mAbs is supported by at least five confirmatory data in ACS: 1) circulating PCSK9 levels are raised during ACS 2) PCSK9 may stimulate platelet reactivity, this last being pivotal in the recurrence of ischemic events; 3) PCSK9 is associated with intraplaque inflammation, macrophage activation and endothelial dysfunction; 4) PCSK9 concentrations are associated with inflammation in the acute phase of ACS; and 5) statins raise PCSK9 levels promptly and, at times, dramatically. In this scenario, appropriate pharmacodynamic characteristics of anti PCSK9 therapies are a prerequisite for an effective response. Monoclonal antibodies act on circulating PCSK9 with a direct and rapid binding by blocking the interaction with the low-density lipoprotein receptor (LDLR). Evolocumab and alirocumab show a very rapid (within 4 h) and effective suppression of circulating unbound PCSK9 (- 95 % ÷ - 97 %). This inhibition results in a significant reduction of LDL-cholesterol (LDL-C) after 48 h (- 35 %) post injection with a full effect after 7-10 days (55-75 %). The complete and swift inhibitory action by evolocumab and alirocumab could have a potential clinical impact in ACS patients, also considering their potential inhibition of PCSK9 within the atherosclerotic plaque. Thus, administration of evolocumab or alirocumab is effective in lowering LDL-C levels in ACS, although the efficacy to prevent further cardiovascular (CV) events is still undetermined. The answer to this question will be provided by the ongoing clinical trials with evolocumab and alirocumab in ACS. In the present review we will discuss the pharmacological and biological rationale supporting the potential use of PCSK9 mAbs in ACS patients and the emerging evidence of evolocumab and alirocumab treatment in this clinical setting.
Collapse
Affiliation(s)
- Nicola Ferri
- Dipartimento di Medicina, Università degli Studi di Padova, Padua, Italy.
| | - Massimiliano Ruscica
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | | | - Marco Vicenzi
- Cardiovascular Disease Unit, Internal Medicine Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cesare R Sirtori
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
24
|
Jia X, Al Rifai M, Saeed A, Ballantyne CM, Virani SS. PCSK9 Inhibitors in the Management of Cardiovascular Risk: A Practical Guidance. Vasc Health Risk Manag 2022; 18:555-566. [PMID: 35898405 PMCID: PMC9309324 DOI: 10.2147/vhrm.s275739] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/13/2022] [Indexed: 01/19/2023] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors are potent medications in the toolkit for treatment of atherosclerotic cardiovascular disease. These agents have been well studied in clinical trials supporting their efficacy in dramatically reducing low-density lipoprotein cholesterol (LDL-C) and impact on cardiovascular outcomes. Since the approval of commercial use for PCSK9 inhibitors in 2015, we have also gained significant experience in the use of these therapeutics in the real-world setting. In this article, we review current guideline recommendations, clinical trial evidence on efficacy and safety as well as data on cost-effectiveness, prescription and adherence. We focus primarily on the monoclonal antibody class of PCSK9 inhibitors in this review while also touching on other types of therapeutics that are under development.
Collapse
Affiliation(s)
- Xiaoming Jia
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mahmoud Al Rifai
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Anum Saeed
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Salim S Virani
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| |
Collapse
|
25
|
Amioka N, Miyoshi T, Yonezawa T, Kondo M, Akagi S, Yoshida M, Saito Y, Nakamura K, Ito H. Pemafibrate Prevents Rupture of Angiotensin II-Induced Abdominal Aortic Aneurysms. Front Cardiovasc Med 2022; 9:904215. [PMID: 35845076 PMCID: PMC9280056 DOI: 10.3389/fcvm.2022.904215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/14/2022] [Indexed: 12/26/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA) is a life-threatening disease that lacks effective preventive therapies. This study aimed to evaluate the effect of pemafibrate, a selective peroxisome proliferator-activated receptor alpha (PPARα) agonist, on AAA formation and rupture. Methods Experimental AAA was induced by subcutaneous angiotensin II (AngII) infusion in ApoE - / - mice for 4 weeks. Pemafibrate (0.1 mg/kg/day) was administered orally. Dihydroethidium staining was used to evaluate the reactive oxygen species (ROS). Results The size of the AngII-induced AAA did not differ between pemafibrate- and vehicle-treated groups. However, a decreased mortality rate due to AAA rupture was observed in pemafibrate-treated mice. Pemafibrate ameliorated AngII-induced ROS and reduced the mRNA expression of interleukin-6 and tumor necrosis factor-α in the aortic wall. Gelatin zymography analysis demonstrated significant inhibition of matrix metalloproteinase-2 activity by pemafibrate. AngII-induced ROS production in human vascular smooth muscle cells was inhibited by pre-treatment with pemafibrate and was accompanied by an increase in catalase activity. Small interfering RNA-mediated knockdown of catalase or PPARα significantly attenuated the anti-oxidative effect of pemafibrate. Conclusion Pemafibrate prevented AAA rupture in a murine model, concomitant with reduced ROS, inflammation, and extracellular matrix degradation in the aortic wall. The protective effect against AAA rupture was partly mediated by the anti-oxidative effect of catalase induced by pemafibrate in the smooth muscle cells.
Collapse
Affiliation(s)
- Naofumi Amioka
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Miyoshi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tomoko Yonezawa
- Department of Molecular Biology and Biochemistry, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Megumi Kondo
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoshi Akagi
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masashi Yoshida
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroshi Ito
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
26
|
Kytikova OY, Novgorodtseva TP, Denisenko YK, Antonyuk MV, Gvozdenko TA, Atamas OV. Brain-Derived Neurotrophic Factor And Coronary Artery Disease. RUSSIAN OPEN MEDICAL JOURNAL 2022. [DOI: 10.15275/rusomj.2022.0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Coronary artery disease (CAD) is defined as myocardial damage developing as a result of its organic and functional changes, and leading to impaired blood flow through the coronary arteries. An important pathogenetic component of CAD is atherosclerosis. Currently, key aspects of the molecular relationship between inflammation and atherosclerosis are being actively studied, the immunometabolic theory of atherosclerosis is being discussed, along with an involvement of perivascular adipose tissue in the pathogenesis of this pathology, due to its ability to respond to atherogenic stimuli via developing inflammatory reactions. Evidence has been accumulated that in patients with CAD, both in their blood and perivascular adipose tissue, the level of neurotrophic factors (in particular, brain-derived neurotrophic factor, BDNF) changes, which may be a promising area of research from the standpoint of studying this factor as a therapeutic target for atherosclerosis in CAD. Neurotrophic growth factors control the functioning of both immune and nervous systems, and the balance of energy metabolism and innervation of adipose tissue. They affect vascular homeostasis, and are also involved in causing and stopping inflammation. Currently, there are data on the role of BDNF in the pathogenesis of cardiovascular, neurodegenerative and metabolic diseases, and on the effect of polyunsaturated fatty acids and eicosanoids on the level of BDNF and, accordingly, the development and progression of coronary artery atherosclerosis. Our review summarizes published data (2019-2021) on the pathophysiological and pathogenetic mechanisms of the relationship between BDNF and CAD (atherosclerosis).
Collapse
Affiliation(s)
- Oksana Yu. Kytikova
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Scientific Research Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia
| | - Tatyana P. Novgorodtseva
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Scientific Research Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia
| | - Yulia K. Denisenko
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Scientific Research Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia
| | - Marina V. Antonyuk
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Scientific Research Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia
| | - Tatyana A. Gvozdenko
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Scientific Research Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia
| | - Olga V. Atamas
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Scientific Research Institute of Medical Climatology and Rehabilitation Therapy, Vladivostok, Russia
| |
Collapse
|
27
|
Zhou L, Liu J, An Y, Wang Y, Wang G. Plasma Homocysteine Level Is Independently Associated With Conventional Atherogenic Lipid Profile and Remnant Cholesterol in Adults. Front Cardiovasc Med 2022; 9:898305. [PMID: 35770226 PMCID: PMC9234129 DOI: 10.3389/fcvm.2022.898305] [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: 03/17/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundHomocysteine (Hcy) is an independent risk factor for cardiovascular disease, while mechanisms are unclear. Despite inconsistent and limited, epidemiological and experimental studies indicated that hyperhomocysteinemia (HHcy) affected lipid metabolism. This study aims to investigate the association of plasma Hcy with traditional lipid profiles and remnant cholesterol (RC) in Chinese adults.MethodsIn total, 7,898 subjects aged 20–79 years who underwent a physical examination at Beijing Chao-Yang Hospital in Beijing were included in this study. Fasting plasma total cholesterol (TC), triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), apolipoprotein A1 (ApoA1), apolipoprotein B (ApoB), lipoprotein (a) [Lp(a)], Hcy, and other metabolic risk factors were measured by routine automated laboratory methods. RC was calculated as TC minus HDL-C and LDL-C. The linear regression model and logistic regression model were used to assess the relationship between Hcy and lipids after adjusting potential confounders.ResultsOf the subjects, the median level of plasma Hcy was 13.0 μmol/L and 32.3% had HHcy. Plasma Hcy was negatively associated with HDL-C, ApoA1, and Lp(a) and positively associated with TG levels after adjusting age, sex, body mass index, blood pressure, alanine transaminase, aspartate transaminase, creatinine, uric acid, and glucose. HHcy significantly increased the risk of low HDL-C [odds ratio (OR) 1.26; 95%CI (1.11–1.44); p < 0.001]. The net mediation effects of ApoA1 on the relationship between Hcy and HDL-C before and after adjusting confounders were 46.9 and 30.6%, respectively. More interestingly, the RC level was significantly elevated in subjects with HHcy after adjusting other influencing factors (p = 0.025). Hcy presented a positive correlation with RC levels after adjusting the above confounding factors (β = 0.073, p = 0.004), and the correlation was still significant even after controlling other lipids, including TG, LDL-C, HDL-C, ApoA1, ApoB, and Lp(a).ConclusionOur study showed that plasma Hcy was not only significantly associated with conventional atherogenic lipids but also independently correlated with RC levels beyond other lipids after controlling potential confounders. This finding proposes that identifying Hcy-related dyslipidemia risk, both traditional lipids and RC residual risk, is clinically relevant as we usher in a new era of targeting Hcy-lowering therapies to fight against dyslipidemia or even cardiovascular disease.
Collapse
Affiliation(s)
- Liyuan Zhou
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jia Liu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yu An
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ying Wang
- Medical Examination Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- *Correspondence: Ying Wang,
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Guang Wang,
| |
Collapse
|
28
|
Estimated cardiovascular benefits of bempedoic acid in patients with established cardiovascular disease. ATHEROSCLEROSIS PLUS 2022; 49:20-27. [PMID: 36644205 PMCID: PMC9833227 DOI: 10.1016/j.athplu.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/05/2022] [Accepted: 05/25/2022] [Indexed: 01/18/2023]
Abstract
Background and aims Cardiovascular outcomes trials have demonstrated that lowering low-density lipoprotein cholesterol (LDL-C) reduces the risk for future cardiovascular events. We assessed the potential cardiovascular benefits of bempedoic acid through a simulation study in patients with atherosclerotic cardiovascular disease (ASCVD) and elevated LDL-C. Methods The validated SMART prediction model was used to estimate the baseline 10-year risk of three-point major adverse cardiovascular events (cardiovascular death, non-fatal myocardial infarction, and non-fatal stroke) in patients with ASCVD who were enrolled in four Phase 3, randomized, placebo-controlled bempedoic acid studies. The predicted change in 10-year cardiovascular risk associated with bempedoic acid was estimated for each patient based on the Cholesterol Treatment Trialists' model. Data were analyzed in two cohorts: Cohort 1 included mostly patients treated with moderate-high intensity statins, and Cohort 2 included patients who were intolerant of more than low-intensity statin. Results A total of 2884 patients were included in Cohort 1 and 226 in Cohort 2. Weighted average baseline 10-year cardiovascular event risk was 26.1% and 31.6% for Cohorts 1 and 2, respectively. The least squares mean percent difference (95% confidence interval (CI) of the predicted absolute change in 10-year cardiovascular event risk with bempedoic acid was -3.3% (-3.7% to -2.9%) for patients in Cohort 1 and -6.0% (-7.7% to -4.3%) for patients in Cohort 2 compared with placebo (p < 0.0001 for both). Conclusions Among patients with ASCVD who could potentially benefit from additional LDL-C lowering, our simulation predicted a lower absolute cardiovascular event risk after initiating bempedoic acid as compared with placebo.
Collapse
|
29
|
Åkra S, Seljeflot I, Braathen B, Bratseth V, Hansen CH, Arnesen H, Tønnessen T, Solheim S. The NLRP3 inflammasome activation in subcutaneous, epicardial and pericardial adipose tissue in patients with coronary heart disease undergoing coronary by-pass surgery. ATHEROSCLEROSIS PLUS 2022; 48:47-54. [PMID: 36644557 PMCID: PMC9833236 DOI: 10.1016/j.athplu.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023]
Abstract
Background and aims Epicardial and pericardial adipose tissue (EAT and PAT) associate with atherosclerosis, however, discussed to have different inflammatory properties. We examined the NLRP3 inflammasome related pathway, playing a pivotal role in atherosclerosis, in EAT, PAT and subcutaneous AT (SAT), their relationship to cell types and anthropometric measures in patients undergoing coronary artery bypass grafting. Methods Biopsies from EAT, PAT and SAT were collected from 52 patients with coronary heart disease (CHD) (median body weight 85.0 kg) and 22 controls. RNA was extracted and expression of interleukin (IL)-1β, IL-18, NLRP3, Caspase-1, toll-like receptor 4 (TLR4), IL-6, IL-6 receptor and gp130 were analyzed by RT-PCR. Results Limited differences in any genes between CHD patients and controls. IL-18 and IL-6 were 4-fold higher expressed in EAT versus PAT (p < 0.01, both) and SAT (p < 0.001, both), whereas caspase-1, IL-6R and gp130 were higher expressed in SAT compared to the other compartments (all p = 0.06-<0.001). Significant correlations between SAT and PAT gene expressions (r = 0.358-0.579, all p ≤ 0.01). Especially NLRP3 and TLR4 associated with the expression of macrophages in all compartments (all p < 0.001). In EAT IL-18 correlated inversely with the expression of macrophages and T-cells. In SAT and PAT most of the mediators associated with body weight. Conclusions Higher expression of IL-18 and IL-6 was observed in EAT in our non-obese CHD patients, not related to inflammatory cells. The NLRP3 inflammasome activation in SAT that mirrored PAT, both related to anthropometrics, suggest that SAT samples, being easily available, to a certain degree, represent adipose tissue inflammation in general.
Collapse
Affiliation(s)
- Sissel Åkra
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway,Faculty of Medicine, University of Oslo, Oslo, Norway,Corresponding author. Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Ullevål, Pb 4954 Nydalen, N-0240, Oslo, Norway.
| | - Bjørn Braathen
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Vibeke Bratseth
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Charlotte Holst Hansen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Harald Arnesen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Theis Tønnessen
- Faculty of Medicine, University of Oslo, Oslo, Norway,Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Svein Solheim
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| |
Collapse
|
30
|
Bytyçi I, Bajraktari G, Penson PE, Henein MY, Banach M. Efficacy and safety of colchicine in patients with coronary artery disease: A systematic review and meta-analysis of randomized controlled trials. Br J Clin Pharmacol 2022; 88:1520-1528. [PMID: 34409634 DOI: 10.1111/bcp.15041] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/05/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023] Open
Abstract
AIMS Inflammation plays a central role in the pathogenesis and clinical manifestations of atherosclerosis. Randomized controlled trials have investigated the potential benefit of colchicine in reducing cardiovascular (CV) events in patients with coronary artery disease (CAD) but produced conflicting results. The aim of this meta-analysis was to evaluate the efficacy and safety of colchicine in patients with CAD. METHODS We systematically searched selected electronic databases from inception until 10 December 2020. Primary clinical endpoints were: major adverse cardiac events; all-cause mortality; CV mortality; recurrent myocardial infarction; stroke; hospitalization; and adverse medication effects. Secondary endpoints were short-term effect of colchicine on inflammatory markers. RESULTS Twelve randomized controlled trials with a total of 13 073 patients with CAD (colchicine n = 6351 and placebo n = 6722) were included in the meta-analysis. At mean follow-up of 22.5 months, the colchicine group had lower risk of major adverse cardiac events (6.20 vs. 8.87%; P < .001), recurrent myocardial infarction (3.41 vs. 4.41%; P = .005), stroke (0.40 vs. 0.90%; P = .002) and hospitalization due to CV events (0.90 vs. 2.87%; P = .02) compared to the control group. The 2 patient groups had similar risk for all-cause mortality (2.08 vs. 1.88%; P = .82) and CV mortality (0.71 vs. 1.01%; P = .38). Colchicine significantly reduced high-sensitivity C-reactive protein (-4.25, P = .001) compared to controls but did not significantly affect interleukin (IL)-β1 and IL-18 levels. CONCLUSION Colchicine reduced CV events and inflammatory markers, high-sensitivity C-reactive protein and IL-6, in patients with coronary disease compared to controls. Its impact on cardiovascular and all-cause mortality requires further investigation.
Collapse
Affiliation(s)
- Ibadete Bytyçi
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina, Kosovo
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
| | - Gani Bajraktari
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina, Kosovo
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
- Medical Faculty, University of Prishtina, Prishtina, Kosovo
| | - Peter E Penson
- Liverpool Centre for Cardiovascular Science, University of Liverpool & Liverpool Heart & Chest Hospital, Liverpool, UK
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Michael Y Henein
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
- Molecular & Clinical Sciences Research Institute, St George University London, UK
- Brunel University, Middlesex, UK
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | | | | |
Collapse
|
31
|
Macchi C, Greco MF, Favero C, Dioni L, Cantone L, Hoxha M, Vigna L, Solazzo G, Corsini A, Banach M, Pesatori AC, Bollati V, Ruscica M. Associations Among PCSK9 Levels, Atherosclerosis-Derived Extracellular Vesicles, and Their miRNA Content in Adults With Obesity. Front Cardiovasc Med 2022; 8:785250. [PMID: 35071356 PMCID: PMC8782054 DOI: 10.3389/fcvm.2021.785250] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/13/2021] [Indexed: 01/16/2023] Open
Abstract
Background: Extracellular vesicles (EV) concentration is generally increased in patients with cardiovascular diseases, although the protective role of EVs in atherosclerosis has been reported. Among the specific cargo of EVs, miRNAs contribute to different stages of atherosclerosis. Aim of the present report has been to investigate, in individuals with obesity, the interplay among EVs derived from cells relevant for the atherosclerotic process (i.e., platelets, endothelium, monocytes/macrophages, and neutrophils), their miRNA content and proprotein convertase subtilisin/kexin type 9 (PCSK9), one of the main regulators of low-density lipoprotein receptor (LDLR). Methods and Results: EVs have been isolated from 936 individuals with obesity (body mass index = 33.6 ± 5.6 Kg/m2) and a raised cardiovascular risk (e.g., LDL-C = 131.6 ± 36.4 mg/dL, HOMA-IR = 3.1, and roughly 50% on anti-hypertensive medications). PCSK9 levels were negatively associated with EV count in the range 150–400 nm and with those derived from macrophages (CD14+), endothelium (CD105+), and neutrophils (CD66+). The association between PCSK9 and platelet-derived EVs (CD61+) was modified by platelet counts. PCSK9 was significantly associated with five EV-derived miRNAs (hsa-miRNA−362−5p,−150,−1244,−520b-3p,−638). Toll-like receptor 4 and estrogen receptor 1 were targeted by all five miRNAs and LDLR by four. The effect on LDLR expression is mainly driven by hsa-miR-150. Considering the implication of EV in atherosclerosis onset and progression, our findings show a potential role of PCSK9 to regulate EV-derived miRNAs, especially those involved in inflammation and expression of low-density lipoprotein receptor (LDLR) receptor.
Collapse
Affiliation(s)
- Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria Francesca Greco
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Favero
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Dioni
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Laura Cantone
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mirjam Hoxha
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Luisella Vigna
- Occupational Medicine Unit, Fondazione Cà Granda, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Solazzo
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,Istituto di Ricovero e Cura a Carattere ScientificoI (RCCS) Multimedica, Milan, Italy
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland.,Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Angela C Pesatori
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Valentina Bollati
- Epidemiology, Epigenetics and Toxicology (EPIGET) Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
32
|
Shinge SAU, Zhang D, Din AU, Yu F, Nie Y. Emerging Piezo1 signaling in inflammation and atherosclerosis; a potential therapeutic target. Int J Biol Sci 2022; 18:923-941. [PMID: 35173527 PMCID: PMC8771847 DOI: 10.7150/ijbs.63819] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose of Review: Atherosclerosis is the principal cause of cardiovascular diseases (CVDs) which are the major cause of death worldwide. Mechanical force plays an essential role in cardiovascular health and disease. To bring the awareness of mechanosensitive Piezo1 role in atherosclerosis and its therapeutic potentials we review recent literature to highlight its involvement in various mechanisms of the disease. Recent Findings: Recent studies reported Piezo1 channel as a sensor, and transducer of various mechanical forces into biochemical signals, which affect various cellular activities such as proliferation, migration, apoptosis and vascular remodeling including immune/inflammatory mechanisms fundamental phenomenon in atherogenesis. Summary: Numerous evidences suggest Piezo1 as a player in different mechanisms of cell biology, including immune/inflammatory and other cellular mechanisms correlated with atherosclerosis. This review discusses mechanistic insight about this matter and highlights the drugability and therapeutic potentials consistent with emerging functions Piezo1 in various mechanisms of atherosclerosis. Based on the recent works, we suggest Piezo1 as potential therapeutic target and a valid candidate for future research. Therefore, a deeper exploration of Piezo1 biology and translation towards the clinic will be a novel strategy for treating atherosclerosis and other CVDs.
Collapse
Affiliation(s)
- Shafiu A. Umar Shinge
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
| | - Daifang Zhang
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Clinical Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - Ahmad Ud Din
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - FengXu Yu
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| | - YongMei Nie
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| |
Collapse
|
33
|
Ugovšek S, Zupan J, Rehberger Likozar A, Šebeštjen M. Influence of lipid-lowering drugs on inflammation: what is yet to be done? Arch Med Sci 2022; 18:855-869. [PMID: 35832698 PMCID: PMC9266870 DOI: 10.5114/aoms/133936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/04/2021] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that is associated with risk of cardiovascular events. The best-characterised and well-standardised clinical indicator of inflammation is C-reactive protein. Current evidence-based drug therapies for prevention and treatment of cardiovascular diseases are mainly focused on reduction of low-density lipoprotein cholesterol. However, these drugs do not provide sufficient protection against recurrent cardiovascular events. One of the possible mechanisms behind this recurrence might be the persistence of residual inflammation. For the most commonly used lipid-lowering drugs, the statins, their reduction of cardiovascular events goes beyond lowering of low-density lipoprotein cholesterol. Here, we review the effects of these lipid-lowering drugs on inflammation, considering statins, ezetimibe, fibrates, niacin, proprotein convertase subtilisin/kexin type 9 inhibitors, bempedoic acid, ethyl eicosapentaenoic acid and antisense oligonucleotides. We focus in particular on C-reactive protein, and discuss how the effects of the statins might be related to reduced rates of cardiovascular events.
Collapse
Affiliation(s)
- Sabina Ugovšek
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Janja Zupan
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Miran Šebeštjen
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Vascular Diseases, University Medical Centre, Ljubljana, Slovenia
- University Medical Centre Ljubljana, Department of Cardiology, Slovenia
| |
Collapse
|
34
|
Vicente-Valor J, García-González X, Ibáñez-García S, Durán-García ME, de Lorenzo-Pinto A, Rodríguez-González C, Méndez-Fernández I, Percovich-Hualpa JC, Herranz-Alonso A, Sanjurjo-Sáez M. PCSK9 inhibitors revisited: Effectiveness and safety of PCSK9 inhibitors in a real-life Spanish cohort. Biomed Pharmacother 2021; 146:112519. [PMID: 34968928 DOI: 10.1016/j.biopha.2021.112519] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) have emerged as a therapeutic option for patients with hypercholesterolemia who do not attain low-density lipoprotein cholesterol (LDL-C) goals and/or are intolerant to other lipid-lowering drugs. Our aim was to analyze the effectiveness and safety of PCSK9i in routine clinical practice and factors related to poor outcomes. MATERIALS AND METHODS We conducted an ambispective study in 115 patients who recieved alirocumab or evolocumab, in a tertiary level hospital. From February 2017 to April 2020, patients were recruited and followed up for a median of 20.4 months. The main outcomes were relative reduction in LDL-C, percentage of patients achieving the therapeutic goals established by 2016 ESC/EAS guidelines, incidence of major cardiovascular events (MACEs) and drug-related adverse events (ADRs). RESULTS The median LDL-C achieved was 57.0 mg/dL (relative reduction of 59.9% from baseline, p< 0.001). After adjusting for confounders, smaller LDL-C reductions were related to female sex, absence of concomitant lipid-lowering therapy and treatment with alirocumab. Overall, 84.6% of the patients achieved the therapeutic goals. During follow-up, 7 MACEs were detected. ADRs, generally considered mild, affected 38.1% of the participants (mainly mialgias and arthralgias) and triggered discontinuations in 8.7% of cases. CONCLUSIONS PCSK9i are effective and safe, although certain factors may influence their effectiveness. Interestingly, our results suggest that alirocumab and evolocumab may not be therapeutic equivalents, as initially suggested.
Collapse
Affiliation(s)
- Juan Vicente-Valor
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| | - Xandra García-González
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Sara Ibáñez-García
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - María Esther Durán-García
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ana de Lorenzo-Pinto
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Carmen Rodríguez-González
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Irene Méndez-Fernández
- Cardiology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Juan Carlos Percovich-Hualpa
- Endocrinology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ana Herranz-Alonso
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - María Sanjurjo-Sáez
- Pharmacy Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
35
|
Marques P, Domingo E, Rubio A, Martinez-Hervás S, Ascaso JF, Piqueras L, Real JT, Sanz MJ. Beneficial effects of PCSK9 inhibition with alirocumab in familial hypercholesterolemia involve modulation of new immune players. Biomed Pharmacother 2021; 145:112460. [PMID: 34864314 DOI: 10.1016/j.biopha.2021.112460] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 12/27/2022] Open
Abstract
Familial hypercholesterolemia (FH) is associated with low-grade systemic inflammation, a key driver of premature atherosclerosis. We investigated the effects of inhibiting proprotein convertase subtilisin/kexin type 9 (PCSK9) function on inflammatory state, endothelial dysfunction and cardiovascular outcomes in patients with FH. Fourteen patients with FH were evaluated before and 8 weeks after administration of a PCSK9 blocking monoclonal antibody (alirocumab, 150 mg/subcutaneous/14 days). In vivo and ex vivo analysis revealed that alirocumab blunted the attachment of leukocytes to TNFα-stimulated human umbilical arterial endothelial cells (HUAEC) and suppressed the activation of platelets and most leukocyte subsets, which was accompanied by the diminished expression of CX3CR1, CXCR6 and CCR2 on several leukocyte subpopulations. By contrast, T-regulatory cell activation was enhanced by alirocumab treatment, which also elevated anti-inflammatory IL-10 plasma levels and lowered circulating pro-inflammatory cytokines. Plasma levels of IFNγ positively correlated with levels of total and LDL-cholesterol, whereas circulating IL-10 levels negatively correlated with these key lipid parameters. In vitro analysis revealed that TNFα stimulation of HUAEC increased the expression of PCSK9, whereas endothelial PCSK9 silencing reduced TNFα-induced mononuclear cell adhesion mediated by Nox5 up-regulation and p38-MAPK/NFκB activation, concomitant with reduced SREBP2 expression. PCSK9 silencing also decreased endothelial CX3CL1 and CXCL16 expression and chemokine generation. In conclusion, PCSK9 inhibition impairs systemic inflammation and endothelial dysfunction by constraining leukocyte-endothelium interactions. PCSK9 blockade may constitute a new therapeutic approach to control the inflammatory state associated with FH, preventing further cardiovascular events in this cardiometabolic disorder.
Collapse
Affiliation(s)
- Patrice Marques
- Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; Institute of Health Research INCLIVA, Av. Menéndez Pelayo 4, 46010 Valencia, Spain
| | - Elena Domingo
- Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain
| | - Arantxa Rubio
- Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain
| | - Sergio Martinez-Hervás
- Department of Medicine, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; Institute of Health Research INCLIVA, Av. Menéndez Pelayo 4, 46010 Valencia, Spain; Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Juan F Ascaso
- Department of Medicine, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; Institute of Health Research INCLIVA, Av. Menéndez Pelayo 4, 46010 Valencia, Spain; Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Laura Piqueras
- Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; Institute of Health Research INCLIVA, Av. Menéndez Pelayo 4, 46010 Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - José T Real
- Department of Medicine, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; Institute of Health Research INCLIVA, Av. Menéndez Pelayo 4, 46010 Valencia, Spain; Endocrinology and Nutrition Service, University Clinic Hospital of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain.
| | - Maria-Jesus Sanz
- Department of Pharmacology, Faculty of Medicine and Odontology, University of Valencia, Av. Blasco Ibáñez 15, 46010 Valencia, Spain; Institute of Health Research INCLIVA, Av. Menéndez Pelayo 4, 46010 Valencia, Spain; CIBERDEM-Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, ISCIII, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain.
| |
Collapse
|
36
|
Lv X, Sun Y, Tan W, Liu Y, Wen N, Fu S, Yu L, Liu T, Qi X, Shu N, Du Y, Zhang W, Meng Y. NONMMUT140591.1 may serve as a ceRNA to regulate Gata5 in UT-B knockout-induced cardiac conduction block. Open Life Sci 2021; 16:1240-1251. [PMID: 34901457 PMCID: PMC8627919 DOI: 10.1515/biol-2021-0106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/22/2021] [Accepted: 07/30/2021] [Indexed: 01/16/2023] Open
Abstract
We intended to explore the potential molecular mechanisms underlying the cardiac conduction block inducted by urea transporter (UT)-B deletion at the transcriptome level. The heart tissues were harvested from UT-B null mice and age-matched wild-type mice for lncRNA sequencing analysis. Based on the sequencing data, the differentially expressed mRNAs (DEMs) and lncRNAs (DELs) between UT-B knockout and control groups were identified, followed by function analysis and mRNA-lncRNA co-expression analysis. The miRNAs were predicted, and then the competing endogenous RNA (ceRNA) network was constructed. UT-B deletion results in the aberrant expression of 588 lncRNAs and 194 mRNAs. These DEMs were significantly enriched in the inflammation-related pathway. A lncRNA-mRNA co-expression network and a ceRNA network were constructed on the basis of the DEMs and DELs. The complement 7 (C7)-NONMMUT137216.1 co-expression pair had the highest correlation coefficient in the co-expression network. NONMMUT140591.1 had the highest degree in the ceRNA network and was involved in the ceRNA of NONMMUT140591.1-mmu-miR-298-5p-Gata5 (GATA binding protein 5). UT-B deletion may promote cardiac conduction block via inflammatory process. The ceRNA NONMMUT140591.1-mmu-miR-298-5p-Gata5 may be a potential molecular mechanism of UT-B knockout-induced cardiac conduction block.
Collapse
Affiliation(s)
- Xuejiao Lv
- Department of Respiratory Medicine and Pathophysiology, Jilin University, No. 218, Ziqiang Road, Nanguan District, Changchun, 130041 Jilin, China
| | - Yuxin Sun
- Department of Otolaryngology, Jilin University, Changchun, Jilin, 130021, China
| | - Wenxi Tan
- Department of Respiratory Medicine and Pathophysiology, Jilin University, No. 218, Ziqiang Road, Nanguan District, Changchun, 130041 Jilin, China
| | - Yang Liu
- Department of Respiratory Medicine and Pathophysiology, Jilin University, No. 218, Ziqiang Road, Nanguan District, Changchun, 130041 Jilin, China
| | - Naiyan Wen
- Department of Nursing, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Shuang Fu
- Department of Pathology and Pathophysiology, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Lanying Yu
- Department of Pathology and Pathophysiology, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Tiantian Liu
- Department of Pathology and Pathophysiology, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Xiaocui Qi
- Department of Pathology and Pathophysiology, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Nanqi Shu
- Department of Pathology and Pathophysiology, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Yanwei Du
- Department of Pathology and Pathophysiology, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Wenfeng Zhang
- Department of Prescriptions, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Yan Meng
- Department of Respiratory Medicine and Pathophysiology, Jilin University, No. 218, Ziqiang Road, Nanguan District, Changchun, 130041 Jilin, China
| |
Collapse
|
37
|
Lupo MG, Arcidiacono D, Zaramella A, Fimiani F, Calabrò P, Cefalù AB, Averna M, D'Erasmo L, Arca M, De Martin S, Zambon A, Ferri N. Lomitapide does not alter PCSK9 and Lp(a) levels in homozygous familial hypercholesterolemia patients: Analysis on cytokines and lipid profile. ATHEROSCLEROSIS PLUS 2021; 43:7-9. [PMID: 36644506 PMCID: PMC9833247 DOI: 10.1016/j.athplu.2021.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 06/17/2023]
Abstract
Lomitapide, a drug for the treatment of homozygous familial hypercholesterolemia patients, reduced total and LDL cholesterol but no significant changes were observed on PCSK9 and Lp(a) plasma levels. Some changes of inflammatory mediators were also observed, including hsCRP, which may suggest an anti-inflammatory effect.
Collapse
Affiliation(s)
- Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Diletta Arcidiacono
- Digestive Endoscopy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Alice Zaramella
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italia
| | - Fabio Fimiani
- A.O.R.N. Sant’Anna e San Sebastiano, Caserta Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paolo Calabrò
- A.O.R.N. Sant’Anna e San Sebastiano, Caserta Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Angelo Baldassare Cefalù
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza, University of Rome, Rome, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza, University of Rome, Rome, Italy
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Alberto Zambon
- Department of Medicine, University of Padova, Padua, Italy
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| |
Collapse
|
38
|
Ruscica M, Sirtori CR, Corsini A, Watts GF, Sahebkar A. Lipoprotein(a): Knowns, unknowns and uncertainties. Pharmacol Res 2021; 173:105812. [PMID: 34450317 DOI: 10.1016/j.phrs.2021.105812] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023]
Abstract
Over the last 10 years, there have been advances on several aspects of lipoprotein(a) which are reviewed in the present article. Since the standard immunoassays for measuring lipoprotein(a) are not fully apo(a) isoform-insensitive, the application of an LC-MS/MS method for assaying molar concentrations of lipoprotein(a) has been advocated. Genome wide association, epidemiological, and clinical studies have established high lipoprotein(a) as a causal risk factor for atherosclerotic cardiovascular diseases (ASCVD). However, the relative importance of molar concentration, apo(a) isoform size or variants within the LPA gene is still controversial. Lipoprotein(a)-raising single nucleotide polymorphisms has not been shown to add on value in predicting ASCVD beyond lipoprotein(a) concentrations. Although hyperlipoproteinemia(a) represents an important confounder in the diagnosis of familial hypercholesterolemia (FH), it enhances the risk of ASCVD in these patients. Thus, identification of new cases of hyperlipoproteinemia(a) during cascade testing can increase the identification of high-risk individuals. However, it remains unclear whether FH itself increases lipoprotein(a). The ASCVD risk associated with lipoprotein(a) seems to follow a linear gradient across the distribution, regardless of racial subgroups and other risk factors. The inverse association with the risk of developing type 2 diabetes needs consideration as effective lipoprotein(a) lowering therapies are progressing towards the market. Considering that Mendelian randomization analyses have identified the degree of lipoprotein(a)-lowering that is required to achieve ASCVD benefit, the findings of the ongoing outcome trial with pelacarsen will clarify whether dramatically lowering lipoprotein(a) levels can reduce the risk of ASCVD.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy.
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy; IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, Australia; Lipid Disorders Clinic, Cardiometabolic Services, Department of Cardiology, Royal Perth Hospital, Australia
| | - Amirhossein Sahebkar
- School of Medicine, University of Western Australia, Perth, Australia; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
39
|
Shinge SAU, Zhang D, Achu Muluh T, Nie Y, Yu F. Mechanosensitive Piezo1 Channel Evoked-Mechanical Signals in Atherosclerosis. J Inflamm Res 2021; 14:3621-3636. [PMID: 34349540 PMCID: PMC8328000 DOI: 10.2147/jir.s319789] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/03/2021] [Indexed: 12/18/2022] Open
Abstract
Recently, more and more works have focused and used extensive resources on atherosclerosis research, which is one of the major causes of death globally. Alongside traditional risk factors, such as hyperlipidemia, smoking, hypertension, obesity, and diabetes, mechanical forces, including shear stress, pressure and stretches exerted on endothelial cells by flow, is proved to be crucial in atherosclerosis development. Studies have recognized the mechanosensitive Piezo1 channel as a special sensor and transducer of various mechanical forces into biochemical signals, and recent studies report its role in atherosclerosis through different mechanical forces in pressure, stretching and turbulent shear stress. Based on our expertise in this field and considering the recent advancement of atherosclerosis research, we will be focusing on the function of Piezo1 and its involvement in various cellular mechanisms and consequent involvement in the development of atherosclerosis in this review. Also, we will discuss various functions of Piezo1 involvement in atherosclerosis and come up with new mechanistic insight for future research. Based on the recent findings, we suggest Piezo1 as a valid candidate for novel therapeutic innovations, in which deep exploration and translating its findings into the clinic will be a new therapeutic strategy for cardiovascular diseases, particularly atherosclerosis.
Collapse
Affiliation(s)
- Shafiu A Umar Shinge
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Daifang Zhang
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Clinical Research Center, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Tobias Achu Muluh
- Oncology Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yongmei Nie
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Fengxu Yu
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
40
|
Ruscica M, Penson PE, Ferri N, Sirtori CR, Pirro M, Mancini GBJ, Sattar N, Toth PP, Sahebkar A, Lavie CJ, Wong ND, Banach M. Impact of nutraceuticals on markers of systemic inflammation: Potential relevance to cardiovascular diseases - A position paper from the International Lipid Expert Panel (ILEP). Prog Cardiovasc Dis 2021; 67:40-52. [PMID: 34186099 DOI: 10.1016/j.pcad.2021.06.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 02/05/2023]
Abstract
Inflammation is a marker of arterial disease stemming from cholesterol-dependent to -independent molecular mechanisms. In recent years, the role of inflammation in atherogenesis has been underpinned by pharmacological approaches targeting systemic inflammation that have led to a significant reduction in cardiovascular disease (CVD) risk. Although the use of nutraceuticals to prevent CVD has largely focused on lipid-lowering (e.g, red-yeast rice and omega-3 fatty acids), there is growing interest and need, especially now in the time of coronavirus pandemic, in the use of nutraceuticals to reduce inflammatory markers, and potentially the inflammatory CVD burden, however, there is still not enough evidence to confirm this. Indeed, diet is an important lifestyle determinant of health and can influence both systemic and vascular inflammation, to varying extents, according to the individual nutraceutical constituents. Thus, the aim of this Position Paper is to provide the first attempt at recommendations on the use of nutraceuticals with effective anti-inflammatory properties.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- Department of Pharmacology and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK; Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Padova, Italy
| | - Cesare R Sirtori
- Department of Pharmacology and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Matteo Pirro
- Internal Medicine Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - G B John Mancini
- Center for Cardiovascular Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Peter P Toth
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Carl J Lavie
- Department of Medicine, John Ochsner Medical Center, New Orleans, LA, USA
| | - Nathan D Wong
- Heart Disease Prevention Program, Division of Cardiology, University of California Irvine, Irvine, CA, USA
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz (MUL), Lodz, Poland; Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland.
| | | |
Collapse
|
41
|
Dongiovanni P, Paolini E, Corsini A, Sirtori CR, Ruscica M. Nonalcoholic fatty liver disease or metabolic dysfunction-associated fatty liver disease diagnoses and cardiovascular diseases: From epidemiology to drug approaches. Eur J Clin Invest 2021; 51:e13519. [PMID: 33583033 DOI: 10.1111/eci.13519] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND A consensus of experts has proposed to replace the term nonalcoholic fatty liver disease (NAFLD), whose global prevalence is 25%, with metabolic dysfunction-associated fatty liver disease (MAFLD), to describe more appropriately the liver disease related to metabolic derangements. MAFLD is closely intertwined with type 2 diabetes, obesity, dyslipidaemia, all linked to a rise in the risk of cardiovascular disease (CVDs). Since controversy still stands on whether or not NAFLD/MAFLD raises the odds of CVD, the present review aims to evaluate the impact of NAFLD/MAFLD aetiologies on CV health and the potential correction by dietary and drug approaches. RESULTS Epidemiological studies indicate that NAFLD raises risk of fatal or non-fatal CVD events. NAFLD patients have a higher prevalence of arterial plaques and stiffness, coronary calcification, and endothelial dysfunction. Although genetic and environmental factors strongly contribute to NAFLD pathogenesis, a Mendelian randomization analysis indicated that the PNPLA3 genetic variant leading to NAFLD may not be causally associated with CVD risk. Among other genetic variants related to NAFLD, TM6SF2 appears to be protective, whereas MBOAT7 may favour venous thromboembolism. CONCLUSIONS NAFLD is correlated to a higher CVD risk which may be ameliorated by dietary interventions. This is not surprising, since new criteria defining MAFLD include other metabolic risk abnormalities fuelling development of serious adverse extrahepatic outcomes, for example CVD. The present lack of a targeted pharmacological approach makes the identification of patients with liver disease at higher CVD risk (eg diabetes, hypertension, obesity or high levels of C-reactive protein) of major clinical interest.
Collapse
Affiliation(s)
- Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Paolini
- General Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.,Multimedica IRCCS, Sesto San Giovanni (MI), Milan, Italy
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
42
|
de Souza JG, Starobinas N, Ibañez OCM. Unknown/enigmatic functions of extracellular ASC. Immunology 2021; 163:377-388. [PMID: 34042182 DOI: 10.1111/imm.13375] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/23/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Apoptosis-associated speck-like protein containing a caspase recruit domain (ASC), encoded by PYCARD gene, is a 22 kDa small molecule, which aggregates into ASC specks during inflammasome activation. ASC protein is an adaptor protein present in several inflammasome complexes that performs several intra- and extracellular functions, in monomeric form or as ASC specks, during physiological and pathological processes related to inflammation and adaptive immunity. Extracellular ASC specks (eASC specks) released during cell death by pyroptosis can contribute as a danger signal to the propagation of inflammation via phagocytosis and activation of surrounding cells. ASC specks are found in the circulation of patients with chronic inflammatory diseases and have been considered as relevant blood biomarkers of inflammation. eASC amplifies the inflammatory signal, may induce the production of autoantibodies, transports molecules that bind to this complex, contributing to the generation of antibodies, and can induce the maturation of cytokines promoting the modelling of the adaptive immunity. Although several advances have been registered in the last 21 years, there are numerous unknown or enigmatic gaps in the understanding of the role of eASC specks in the organism. Here, we provide an overview about the ASC protein focusing on the probable roles of eASC specks in several diseases, up to the most recent studies concerning COVID-19.
Collapse
Affiliation(s)
- Jean Gabriel de Souza
- Laboratory of Immunogenetics, Butantan Institute, São Paulo, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil.,Immunology Catalyst, GlaxoSmithKline, Stevenag, UK
| | - Nancy Starobinas
- Laboratory of Immunogenetics, Butantan Institute, São Paulo, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| | - Olga Celia Martinez Ibañez
- Laboratory of Immunogenetics, Butantan Institute, São Paulo, Brazil.,CENTD, Centre of Excellence in New Target Discovery, Butantan Institute, São Paulo, Brazil
| |
Collapse
|
43
|
de Gaetano M, Tighe C, Gahan K, Zanetti A, Chen J, Newson J, Cacace A, Marai M, Gaffney A, Brennan E, Kantharidis P, Cooper ME, Leroy X, Perretti M, Gilroy D, Godson C, Guiry PJ. Asymmetric Synthesis and Biological Screening of Quinoxaline-Containing Synthetic Lipoxin A 4 Mimetics (QNX-sLXms). J Med Chem 2021; 64:9193-9216. [PMID: 34138563 PMCID: PMC8279484 DOI: 10.1021/acs.jmedchem.1c00403] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Failure to resolve
inflammation underlies many prevalent pathologies.
Recent insights have identified lipid mediators, typified by lipoxins
(LXs), as drivers of inflammation resolution, suggesting potential
therapeutic benefit. We report the asymmetric preparation of novel
quinoxaline-containing synthetic-LXA4-mimetics (QNX-sLXms).
Eight novel compounds were screened for their impact on inflammatory
responses. Structure–activity relationship (SAR) studies showed
that (R)-6 (also referred to as AT-02-CT)
was the most efficacious and potent anti-inflammatory compound of
those tested. (R)-6 significantly attenuated
lipopolysaccharide (LPS)- and tumor-necrosis-factor-α (TNF-α)-induced
NF-κB activity in monocytes and vascular smooth muscle cells.
The molecular target of (R)-6 was investigated.
(R)-6 activated the endogenous LX receptor
formyl peptide receptor 2 (ALX/FPR2). The anti-inflammatory properties
of (R)-6 were further investigated in vivo in murine models of acute inflammation. Consistent
with in vitro observations, (R)-6 attenuated inflammatory responses. These results support
the therapeutic potential of the lead QNX-sLXm (R)-6 in the context of novel inflammatory regulators.
Collapse
Affiliation(s)
- Monica de Gaetano
- School of Medicine, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Catherine Tighe
- Centre for Synthesis and Chemical Biology, School of Chemistry, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Kevin Gahan
- Centre for Synthesis and Chemical Biology, School of Chemistry, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Andrea Zanetti
- Centre for Synthesis and Chemical Biology, School of Chemistry, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Jianmin Chen
- William Harvey Research Institute, Queen Mary University London, London EC1M 6BQ, U.K
| | - Justine Newson
- Centre for Clinical Pharmacology, University College London, London WC1E 6JF, U.K
| | - Antonino Cacace
- School of Medicine, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Mariam Marai
- School of Medicine, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Andrew Gaffney
- School of Medicine, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Eoin Brennan
- School of Medicine, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Phillip Kantharidis
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Mark E Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Xavier Leroy
- Domain Therapeutics SA, 67400 Strasbourg, Illkirch, France
| | - Mauro Perretti
- William Harvey Research Institute, Queen Mary University London, London EC1M 6BQ, U.K
| | - Derek Gilroy
- Centre for Clinical Pharmacology, University College London, London WC1E 6JF, U.K
| | - Catherine Godson
- School of Medicine, Diabetes Complications Research Centre, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| | - Patrick J Guiry
- Centre for Synthesis and Chemical Biology, School of Chemistry, UCD Conway Institute, University College Dublin, Belfield, Dublin D04 N2E5, Ireland
| |
Collapse
|
44
|
Rogula S, Błażejowska E, Gąsecka A, Szarpak Ł, Jaguszewski MJ, Mazurek T, Filipiak KJ. Inclisiran-Silencing the Cholesterol, Speaking up the Prognosis. J Clin Med 2021; 10:2467. [PMID: 34199468 PMCID: PMC8199585 DOI: 10.3390/jcm10112467] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/15/2022] Open
Abstract
The reduction of circulating low-density lipoprotein-cholesterol (LDL-C) is a primary target in cardiovascular risk reduction due to its well-established benefits in terms of decreased mortality. Despite the use of statin therapy, 10%-20% of high- and very-high-risk patients do not reach their LDL-C targets. There is an urgent need for improved strategies to manage dyslipidemia, especially among patients with homozygous familial hypercholesterolemia, but also in patients with established cardiovascular disease who fail to achieve LDL goals despite combined statin, ezetimibe, and PCSK9 inhibitor (PCSK9i) therapy. Inclisiran is a disruptive, first-in-class small interfering RNA (siRNA)-based therapeutic developed for the treatment of hypercholesterolemia that inhibits proprotein convertase subtilisin-kexin type 9 (PCSK9) synthesis, thereby upregulating the number of LDL receptors on the hepatocytes, thus lowering the plasma LDL-C concentration. Inclisiran decreases the LDL-C levels by over 50% with one dose every 6 months, making it a simple and well-tolerated treatment strategy. In this review, we summarize the general information regarding (i) the role of LDL-C in atherosclerotic cardiovascular disease, (ii) data regarding the role of PCSK9 in cholesterol metabolism, (iii) pleiotropic effects of PCSK9, and (iv) the effects of PCSK9 silencing. In addition, we focus on inclisiran, in terms of its (i) mechanism of action, (ii) biological efficacy and safety, (iii) results from the ORION trials, (iv) benefits of its combination with statins, and (v) its potential future role in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Sylwester Rogula
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (S.R.); (E.B.); (T.M.); (K.J.F.)
| | - Ewelina Błażejowska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (S.R.); (E.B.); (T.M.); (K.J.F.)
| | - Aleksandra Gąsecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (S.R.); (E.B.); (T.M.); (K.J.F.)
| | - Łukasz Szarpak
- Maria Sklodowska-Curie Białystok Oncology Centre, Ogrodowa 12, 15-027 Białystok, Poland;
- Maria Sklodowska-Curie Medical Academy in Warsaw, Solidarności 12, 03-411 Warsaw, Poland
| | - Milosz J. Jaguszewski
- 1st Department of Cardiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland;
| | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (S.R.); (E.B.); (T.M.); (K.J.F.)
| | - Krzysztof J. Filipiak
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1a, 02-097 Warsaw, Poland; (S.R.); (E.B.); (T.M.); (K.J.F.)
| |
Collapse
|
45
|
Gencer B, Bonomi M, Adorni MP, Sirtori CR, Mach F, Ruscica M. Cardiovascular risk and testosterone - from subclinical atherosclerosis to lipoprotein function to heart failure. Rev Endocr Metab Disord 2021; 22:257-274. [PMID: 33616800 PMCID: PMC8087565 DOI: 10.1007/s11154-021-09628-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
The cardiovascular (CV) benefit and safety of treating low testosterone conditions is a matter of debate. Although testosterone deficiency has been linked to a rise in major adverse CV events, most of the studies on testosterone replacement therapy were not designed to assess CV risk and thus excluded men with advanced heart failure or recent history of myocardial infarction or stroke. Besides considering observational, interventional and prospective studies, this review article evaluates the impact of testosterone on atherosclerosis process, including lipoprotein functionality, progression of carotid intima media thickness, inflammation, coagulation and thromboembolism, quantification of plaque volume and vascular calcification. Until adequately powered studies evaluating testosterone effects in hypogonadal men at increased CV risk are available (TRAVERSE trial), clinicians should ponder the use of testosterone in men with atherosclerotic cardiovascular disease and discuss benefit and harms with the patients.
Collapse
Affiliation(s)
- Baris Gencer
- Cardiology Division, Geneva University Hospitals, Geneva, Switzerland.
| | - Marco Bonomi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
- Department of Endocrine and Metabolic Diseases & Lab. of Endocrine and Metabolic Research, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Maria Pia Adorni
- Department of Medicine and Surgery-Unit of Neurosciences, University of Parma, Parma, Italy
| | - Cesare R Sirtori
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - François Mach
- Cardiology Division, Geneva University Hospitals, Geneva, Switzerland
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
46
|
Banerjee S, Katiyar P, Kumar L, Kumar V, Saini SS, Krishnan V, Sircar D, Roy P. Black pepper prevents anemia of inflammation by inhibiting hepcidin over-expression through BMP6-SMAD1/ IL6-STAT3 signaling pathway. Free Radic Biol Med 2021; 168:189-202. [PMID: 33771600 DOI: 10.1016/j.freeradbiomed.2021.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 01/03/2023]
Abstract
Hepcidin, a circulatory hepatic peptide hormone, is associated with systemic iron homeostasis. Inflammation leads to an increase in hepcidin expression, which dysregulates body iron level. The related disorder, anemia of inflammation, is the second most prevalent anemia-related disorder worldwide. In the present study, we conducted in vitro and in vivo studies to evaluate the effect of black pepper (BP) and its major bioactive alkaloid, piperine, on anemia of inflammation. The initial in vitro study using human hepatocyte cell line, HepG2, confirmed that among different black pepper extracts: methanol (BPME), ethanol (BPEE) and aqueous (BPAE), BPME to be most effective in downregulating transcription of hepcidin gene. Further, BPME and piperine significantly downregulated hepcidin protein expression at 200 μg/ml and 100 μM concentrations, respectively. In the next phase, BPME and piperine were found to significantly attenuate BMP-6 and IL-6 induced hepcidin overexpression by downregulating the increased level of pSMAD1 and pSTAT3 proteins, respectively. For in vivo study, we first subcutaneously injected male BALB/c mice with oil of turpentine, thrice within a period of two weeks, in order to enhance the expression of hepcidin. After that, the intraperitoneal administration of BPME and piperine at 70 and 25 mg/kg body weight, respectively, on alternate days for a period of another two weeks resulted in downregulation of hepcidin overexpression in diseased mice, as confirmed by RT-PCR and immunoblot analysis. The histopathology of liver tissue confirmed increased iron bioavailability in BPME and piperine treated animals. The molecular docking-based interaction studies demonstrated the binding potential of piperine with SMAD1 and STAT3 proteins. The binding patterns supported the proposed inhibition of hepcidin activating proteins. All together, these findings suggest black pepper as a therapeutic candidate for the treatment of anemia of inflammation.
Collapse
Affiliation(s)
- Somesh Banerjee
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| | - Parul Katiyar
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| | - Lokesh Kumar
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| | - Vijay Kumar
- Laboratory of Structural Microbiology, Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India.
| | - Shashank Sagar Saini
- Plant Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| | - Vengadesan Krishnan
- Laboratory of Structural Microbiology, Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India.
| | - Debabrata Sircar
- Plant Molecular Biology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India.
| |
Collapse
|
47
|
Puato M, Zambon A, Nardin C, Faggin E, Pesavento R, Spinazzè A, Pauletto P, Rattazzi M. Lipid Profile and Vascular Remodelling in Young Dyslipidemic Subjects Treated with Nutraceuticals Derived from Red Yeast Rice. Cardiovasc Ther 2021; 2021:5546800. [PMID: 33976708 PMCID: PMC8087481 DOI: 10.1155/2021/5546800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND AIMS A relevant role is emerging for functional foods in cardiovascular prevention. The aim of this study was to assess the effect of a nutraceutical multitargeted approach on lipid profile and inflammatory markers along with vascular remodelling in a cohort of dyslipidemic subjects without history of cardiovascular (CV) disease. METHODS AND RESULTS We enrolled 25 subjects (mean age 48.2 years) with low to moderate CV risk profile and total cholesterol (TC) levels between 150 and 250 mg/dl. The patients were assigned to receive for one year a tablet/die of a nutraceutical combination containing red yeast rice (RYR) extract (Monacolin 3 mg/tablet) and coenzyme Q10 (30 mg/tablet). Treatment with the nutraceutical compounds led to a significant reduction of TC (from 227 to 201 mg/dl, p < 0.001), LDL-c (from 150 to 130 mg/dl, p = 0.001), triglycerides (from 121 to 109 mg/dl, p = 0.013), non-HDL-cholesterol (from 168 to 141 mg/dl, p < 0.001), hs-CRP (from 1.74 to 1.20 mg/l, p = 0.015), and osteoprotegerin (from 1488 to 1328 pg/ml, p = 0.045). Levels of HDL-c, Lp(a), glucose, liver enzyme, CPK, or creatinine did not change over time. An ultrasound study was performed to assess changes in mean carotid intima-media thickness (IMT) and maximum IMT (M-MAX) as well as modification in local carotid stiffness by means of determining the carotid compliance coefficient (CC) and distensibility coefficient (DC). At the end of the treatment, we observed small but significant reductions in both mean-IMT (from 0.62 to 0.57 mm, p = 0.022) and M-MAX (from 0.79 to 0.73 mm, p = 0.002), and an improvement in carotid elasticity (DC from 22.4 to 24.3 × 10-3/kPa, p = 0.006 and CC from 0.77 to 0.85 mm2/kPa, p = 0.019). CONCLUSIONS A long-term treatment with a combination of RYR and coenzyme Q10 showed lipid-lowering activity along with a reduction of inflammatory mediators and an improvement of vascular properties in young subjects with a low-to-moderate CV risk profile.
Collapse
Affiliation(s)
| | - Alberto Zambon
- Department of Medicine (DIMED), University of Padova, Italy
| | - Chiara Nardin
- Department of Medicine (DIMED), University of Padova, Italy
- Medicina Generale I^, Ca' Foncello Hospital, Treviso, Italy
| | | | | | - Alice Spinazzè
- Department of Medicine (DIMED), University of Padova, Italy
| | - Paolo Pauletto
- ORAS Rehabilitation Hospital, Motta di Livenza, Treviso, Italy
| | - Marcello Rattazzi
- Department of Medicine (DIMED), University of Padova, Italy
- Medicina Generale I^, Ca' Foncello Hospital, Treviso, Italy
| |
Collapse
|
48
|
Vlad CE, Foia LG, Popescu R, Popa I, Aanicai R, Reurean-Pintilei D, Toma V, Florea L, Kanbay M, Covic A. Molecular Genetic Approach and Evaluation of Cardiovascular Events in Patients with Clinical Familial Hypercholesterolemia Phenotype from Romania. J Clin Med 2021; 10:jcm10071399. [PMID: 33807407 PMCID: PMC8036385 DOI: 10.3390/jcm10071399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/13/2022] Open
Abstract
This study identifies the genetic background of familial hypercholesterolemia (FH) patients in Romania and evaluates the association between mutations and cardiovascular events. We performed a prospective observational study of 61 patients with a clinical diagnosis of FH selected based on Dutch Lipid Clinic Network (DLCN) and Simon Broome score between 2017 and 2020. Two techniques were used to identify mutations: multiplex ligation-dependent probe amplification (MLPA) and Sanger sequencing. The mutation rate was 37.7%, i.e., 23 patients with mutations were identified, of which 7 subjects had pathogenic mutations and 16 had polymorphisms. Moreover, 10 variants of the low-density lipoprotein receptor (LDLR) gene were identified in 22 patients, i.e., one variant of the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene in six patients, and one variant of the apolipoprotein B (APOB) gene in three patients. Of the LDLR gene variants, four were LDLR pathogenic mutations (c.81C > G, c.502G > A, c.1618G > A mutations in exon 2, exon 4, exon 11, and exon 13–15 duplication). The PCSK9 and APOB gene variants were benign mutations. The pathogenic LDLR mutations were significant predictors of the new cardiovascular events, and the time interval for new cardiovascular events occurrence was significantly decreased, compared to FH patients without mutations. In total, 12 variants were identified, with four pathogenic variants identified in the LDLR gene, whereas 62.3% of the study population displayed no pathological mutations.
Collapse
Affiliation(s)
- Cristiana-Elena Vlad
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
- Department of Nephrology-Internal Medicine, “Dr. C.I. Parhon” Clinical Hospital, Carol I Street, No 50, 700503 Iasi, Romania
| | - Liliana Georgeta Foia
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
- Department of Biochemistry, “Sf. Spiridon” Clinical County Hospital, Independentei Street, 700111 Iasi, Romania
- Correspondence: ; Tel.: +40-744704452
| | - Roxana Popescu
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
| | - Ioana Popa
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
| | - Ruxandra Aanicai
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
| | - Delia Reurean-Pintilei
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
| | - Vasilica Toma
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
| | - Laura Florea
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
- Department of Nephrology-Internal Medicine, “Dr. C.I. Parhon” Clinical Hospital, Carol I Street, No 50, 700503 Iasi, Romania
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, 34450 Istanbul, Turkey;
| | - Adrian Covic
- Faculty of Medicine, “Grigore T Popa” University of Medicine and Pharmacy, University Street, No 16, 700115 Iasi, Romania; (C.-E.V.); (R.P.); (I.P.); (R.A.); (D.R.-P.); (V.T.); (L.F.); (A.C.)
- Department of Nephrology-Internal Medicine, “Dr. C.I. Parhon” Clinical Hospital, Carol I Street, No 50, 700503 Iasi, Romania
| |
Collapse
|
49
|
Abstract
Purpose of review Based on the recent data of the DA VINCI study, it is clear that, besides utilization of statins, there is a need to increase non-statin lipid lowering approaches to reduce the cardiovascular burden in patients at highest risk. Recent findings For hypercholesterolemia, the small synthetic molecule bempedoic acid has the added benefit of selective liver activation, whereas inclisiran, a hepatic inhibitor of the PCSK9 synthesis, has comparable effects with PCSK9 monoclonal antibodies. For hypertriglyceridemia, cardiovascular benefit has been achieved by the use of icosapent ethyl, whereas results with pemafibrate, a selective agonist of PPAR-α, are eagerly awaited. In the era of RNA-based therapies, new options are offered to dramatically reduce levels of lipoprotein(a) (APO(a)LRX) and of triglycerides (ANGPTL3LRX and APOCIII-LRx). Summary Despite the demonstrated benefits of statins, a large number of patients still remain at significant risk because of inadequate LDL-C reduction or elevated blood triglyceride-rich lipoproteins or lipoprotein(a). The area of lipid modulating agents is still ripe with ideas and major novelties are to be awaited in the next few years.
Collapse
|
50
|
Zhou E, Li Z, Nakashima H, Choukoud A, Kooijman S, Berbée JFP, Rensen PCN, Wang Y. Beneficial effects of brown fat activation on top of PCSK9 inhibition with alirocumab on dyslipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice. Pharmacol Res 2021; 167:105524. [PMID: 33667684 DOI: 10.1016/j.phrs.2021.105524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/19/2021] [Accepted: 02/28/2021] [Indexed: 12/13/2022]
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibition, by increasing hepatic low density lipoprotein (LDL) receptor (LDLR) levels, has emerged as a strategy to reduce atherosclerosis by lowering circulating very low density lipoprotein (VLDL)-cholesterol. We hypothesized that the therapeutic effectiveness of PCSK9 inhibition can be increased by accelerating the generation of VLDL remnants, which typically have a high affinity for the LDLR. Therefore, we aimed to investigate whether accelerating lipolytic processing of VLDL by brown fat activation can further lower (V)LDL and reduce atherosclerosis on top of PCSK9 inhibition. APOE*3-Leiden.CETP mice were fed a Western-type diet and treated with the anti-PCSK9 antibody alirocumab or saline. After 2 weeks, both groups of mice were randomized to receive either the selective β3-adrenergic receptor (AR) agonist CL316,243 to activate brown fat or saline for 3 additional weeks to evaluate VLDL clearance or 12 additional weeks to analyze atherosclerosis development. β3-AR agonism and alirocumab combined decreased (V)LDL-cholesterol compared to alirocumab alone, which was explained by an accelerated plasma clearance of VLDL-cholesteryl esters that were mainly taken up by the liver. In addition, the combination promoted the transfer of VLDL-phospholipids to HDL to a higher extent than alirocumab alone, accompanied by higher plasma HDL-cholesterol levels and increased cholesterol efflux capacity. Consequently, combination treatment largely reduced atherosclerotic lesion area compared to vehicle. Together, β3-AR agonism enhances the lipoprotein-modulating effects of alirocumab to further improve dyslipidemia and non-significantly further attenuate atherosclerosis development. Our findings demonstrate that brown fat activation may enhance the therapeutic effects of PCSK9 inhibition in dyslipidemia.
Collapse
Affiliation(s)
- Enchen Zhou
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Zhuang Li
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Hiroyuki Nakashima
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ahlam Choukoud
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China; Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Yanan Wang
- Department of Endocrinology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China; Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|