1
|
Millar JE, Reddy K, Bos LDJ. Future Directions in Therapies for Acute Respiratory Distress Syndrome. Clin Chest Med 2024; 45:943-951. [PMID: 39443010 DOI: 10.1016/j.ccm.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is caused by a complex interplay among hyperinflammation, endothelial dysfunction, and alveolar epithelial injury. Targeted treatments toward the underlying pathways have been unsuccessful in unselected patient populations. The first reliable biological subphenotypes reflective of these biological disease states have been identified in the past decade. Subphenotype targeted intervention studies are needed to advance the pharmacologic treatment of ARDS.
Collapse
Affiliation(s)
- Jonathan E Millar
- Baillie-Gifford Pandemic Science Hub, Centre for Inflammation Research, Institute for Repair and Regeneration, University of Edinburgh, The Roslin Institute, Easter Bush Campus, Midlothian, Edinburgh EH25 9RG, UK; Department of Critical Care, Queen Elizabeth University Hospital, Glasgow, UK
| | - Kiran Reddy
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, University Road, Belfast BT7 1NN, UK
| | - Lieuwe D J Bos
- Intensive Care Department, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands.
| |
Collapse
|
2
|
Jiang R, Lou L, Shi W, Chen Y, Fu Z, Liu S, Sok T, Li Z, Zhang X, Yang J. Statins in Mitigating Anticancer Treatment-Related Cardiovascular Disease. Int J Mol Sci 2024; 25:10177. [PMID: 39337662 PMCID: PMC11432657 DOI: 10.3390/ijms251810177] [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: 08/20/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Certain anticancer therapies inevitably increase the risk of cardiovascular events, now the second leading cause of death among cancer patients. This underscores the critical need for developing effective drugs or regimens for cardiovascular protection. Statins possess properties such as antioxidative stress, anti-inflammatory effects, antifibrotic activity, endothelial protection, and immune modulation. These pathological processes are central to the cardiotoxicity associated with anticancer treatment. There is prospective clinical evidence confirming the protective role of statins in chemotherapy-induced cardiotoxicity. Numerous preclinical studies have demonstrated that statins can ameliorate heart and endothelial damage caused by radiotherapy, although clinical studies are scarce. In the animal models of trastuzumab-induced cardiomyopathy, statins provide protection through anti-inflammatory, antioxidant, and antifibrotic mechanisms. In animal and cell models, statins can mitigate inflammation, endothelial damage, and cardiac injury induced by immune checkpoint inhibitors. Chimeric antigen receptor (CAR)-T cell therapy-induced cardiotoxicity and immune effector cell-associated neurotoxicity syndrome are associated with uncontrolled inflammation and immune activation. Due to their anti-inflammatory and immunomodulatory effects, statins have been used to manage CAR-T cell therapy-induced immune effector cell-associated neurotoxicity syndrome in a clinical trial. However, direct evidence proving that statins can mitigate CAR-T cell therapy-induced cardiotoxicity is still lacking. This review summarizes the possible mechanisms of anticancer therapy-induced cardiotoxicity and the potential mechanisms by which statins may reduce related cardiac damage. We also discuss the current status of research on the protective effect of statins in anticancer treatment-related cardiovascular disease and provide directions for future research. Additionally, we propose further studies on using statins for the prevention of cardiovascular disease in anticancer treatment.
Collapse
Affiliation(s)
- Rong Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lian Lou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wen Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuxiao Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhaoming Fu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shuo Liu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Thida Sok
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhihang Li
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
3
|
Zhang X, Geng H, Shan C, Cui X, Zhang X, Ashokkumar M, Cui J, Zhang P. Assembly of Emulsion-Based Cascade Vehicles for Combination Oxygen-Chemotherapy in Diabetic Wound Healing. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:19766-19774. [PMID: 39235374 DOI: 10.1021/acs.langmuir.4c02549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
High blood glucose and insufficient angiogenesis in diabetic wounds prevent healing, often leading to amputation or death. To address this, a multifunctional emulsion loaded with simvastatin and stabilized by enzymes was synthesized using ultrasound-assisted emulsification. This emulsion promotes angiogenesis and reduces blood glucose levels. Glucose oxidase and catalase at the emulsion interface catalyze a glucose cascading response, lowering the glucose concentration at the diabetic wound site and improving the wound microenvironment. Simvastatin in the emulsion further promotes angiogenesis. The emulsion significantly accelerated wound healing in diabetic rats, offering a promising approach to diabetic wound management.
Collapse
Affiliation(s)
- Xunhui Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Huimin Geng
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Caiyun Shan
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Xiaomiao Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Xiaohui Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | | | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| | - Peiyu Zhang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong 250100, People's Republic of China
| |
Collapse
|
4
|
Kusumoto J, Muraki Y, Sakakibara A, Furudoi S, Akashi M. Effect of Statins on Patients With Osteoradionecrosis of the Jaw. J Oral Maxillofac Surg 2024:S0278-2391(24)00687-6. [PMID: 39208868 DOI: 10.1016/j.joms.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Osteoradionecrosis of the jaw (ORN) is a late complication of radiation therapy. Radiation-induced fibrosis is the most likely mechanism for developing ORN, and statins are effective against radiation-induced fibrosis. However, no reports have indicated the direct effectiveness of statins in treating ORN. PURPOSE This study aimed to measure the association between statin exposure and ORN disease resolution. STUDY DESIGN, SETTING, SAMPLE This retrospective cohort study included patients with ORN diagnosed between January 2008 and December 2020 at the Hospital's Department of Oral and Maxillofacial Surgery. Patients who were immunocompromised or followed up for < 6 months were excluded. PREDICTOR VARIABLE The predictor variable was statin exposure, which was defined as the use of statins for dyslipidemia. MAIN OUTCOME VARIABLE The main outcome variable was ORN disease progression status (good prognosis). Patients who showed full recovery and improvement were included in the good prognosis group, and those who showed invariance and deterioration were included in the poor prognosis group. COVARIATES We analyzed the clinicodemographic including the age of onset, sex, history of smoking, alcohol consumption, history of chemotherapy, tumor site, association with dental treatment, location (maxilla or mandible), the time to ORN onset from radiation therapy, and stage of ORN, and treatment characteristics including hyperbaric oxygen therapy, long-term macrolide administration, and sequestrectomy. ANALYSES We analyzed the association between statin exposure or covariates and time to ORN improvement using bivariate and multivariate Cox regression. The significance level was set at P = .05. RESULTS We analyzed 102 patients, and the improvement rate was 32.4%. The favorable prognostic factors were statin exposure (adjusted hazard ratio [HR], 3.71; 95% confidence interval [CI], 1.62 to 8.50; P = .002), onset in the maxilla (HR, 2.15; 95% CI, 1.02 to 4.55; P = .045), and stage 1 of ORN (HR, 2.65; 95% CI, 1.20 to 5.83; P = .016). CONCLUSION AND RELEVANCE In this study, statin exposure, onset in the maxilla, and stage 1 of Lyons's classification were favorable prognostic factors for ORN. Although this study's findings were insufficient to recommend statin use for ORN, statins may be a novel and effective treatment for ORN.
Collapse
Affiliation(s)
- Junya Kusumoto
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Yumi Muraki
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akiko Sakakibara
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan; Department Head, Department of Oral and Maxillofacial Surgery, Mitsubishi Kobe Hospital, Kobe, Japan
| | - Shungo Furudoi
- Department Head, Department of Oral Surgery, Konan Medical Center, Kobe, Japan
| | - Masaya Akashi
- Assistant Professor, Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan; Professor, Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
5
|
Kounatidis D, Tentolouris N, Vallianou NG, Mourouzis I, Karampela I, Stratigou T, Rebelos E, Kouveletsou M, Stamatopoulos V, Tsaroucha E, Dalamaga M. The Pleiotropic Effects of Lipid-Modifying Interventions: Exploring Traditional and Emerging Hypolipidemic Therapies. Metabolites 2024; 14:388. [PMID: 39057711 PMCID: PMC11278853 DOI: 10.3390/metabo14070388] [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: 06/18/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerotic cardiovascular disease poses a significant global health issue, with dyslipidemia standing out as a major risk factor. In recent decades, lipid-lowering therapies have evolved significantly, with statins emerging as the cornerstone treatment. These interventions play a crucial role in both primary and secondary prevention by effectively reducing cardiovascular risk through lipid profile enhancements. Beyond their primary lipid-lowering effects, extensive research indicates that these therapies exhibit pleiotropic actions, offering additional health benefits. These include anti-inflammatory properties, improvements in vascular health and glucose metabolism, and potential implications in cancer management. While statins and ezetimibe have been extensively studied, newer lipid-lowering agents also demonstrate similar pleiotropic effects, even in the absence of direct cardiovascular benefits. This narrative review explores the diverse pleiotropic properties of lipid-modifying therapies, emphasizing their non-lipid effects that contribute to reducing cardiovascular burden and exploring emerging benefits for non-cardiovascular conditions. Mechanistic insights into these actions are discussed alongside their potential therapeutic implications.
Collapse
Affiliation(s)
- Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Irene Karampela
- Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Theodora Stratigou
- Department of Endocrinology and Metabolism, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Eleni Rebelos
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | - Marina Kouveletsou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.K.); (N.T.); (E.R.); (M.K.)
| | | | - Eleni Tsaroucha
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
6
|
Kim YS, Jeong HG, Chang JY, Kim JY, Kim BJ, Bae HJ, Han MK. Effect of Statin Therapy on Cardiovascular Outcome in Stroke Patients with Low Baseline Low-Density Lipoprotein Cholesterol. Ann Neurol 2024; 95:876-885. [PMID: 38400785 DOI: 10.1002/ana.26895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/26/2024]
Abstract
OBJECTIVES To investigate whether post-stroke statin therapy reduces subsequent major vascular events in statin-naïve patients with pretreatment low-density lipoprotein cholesterol (LDL-C) below the recommended target (≤70 mg/dL for atherosclerotic stroke and ≤100 mg/dL for non-atherosclerotic stroke) at stroke onset. METHODS Patients from an ongoing stroke registry who had an ischemic stroke between 2011 and 2020 were screened. Statin naïve patients with baseline LDL-C below the target were assessed. The effect of post-stroke statin therapy on major vascular events (composite of recurrent stroke, myocardial infarction, and death) was investigated using weighted Cox regression analyses using stabilized inverse probability treatment weighting. RESULTS The baseline LDL-C level of the 1,858 patients (mean age 67.9 ± 15.3 years, 61.4% men, 13.2% atherosclerotic stroke) included in the study was 75.7 ± 17.0 mg/dL. Statins were prescribed to 1,256 (67.7%) patients (low-to-moderate intensity, 23.5%; high intensity, 44.1%). Post-stroke statin therapy was associated with a lower risk of major vascular events during 1-year follow-up (weighted hazard ratio 0.55, 95% confidence interval 0.42-0.71). In a subgroup of patients who were at very high risk of atherosclerotic cardiovascular disease with LDL-C <55 mg/dL or patients who were not at very high risk of atherosclerotic cardiovascular disease with LDL-C <70 mg/dL, post-stroke statin therapy was also associated with a reduction in major vascular events (weighted hazard ratio 0.45, 95% confidence interval 0.29-0.70). The intensity of the most beneficial statin varied by subtype of stroke. INTERPRETATION Statin therapy may improve vascular outcomes after ischemic stroke, even in cases of LDL-C below the target without pre-stroke lipid-lowering therapy. ANN NEUROL 2024;95:876-885.
Collapse
Affiliation(s)
- Yong Soo Kim
- Department of Neurology, Nowon Eulji Medical Center, Seoul, Korea
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Han-Gil Jeong
- Division of Neurocritical Care, Department of Neurosurgery and Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jun Young Chang
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jun Yup Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Beom Joon Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hee-Joon Bae
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Moon-Ku Han
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
7
|
Mehraj V, Chen J, Routy JP. Effects of statins beyond lipid-lowering agents in ART-treated HIV infection. Front Immunol 2024; 15:1339338. [PMID: 38655259 PMCID: PMC11035727 DOI: 10.3389/fimmu.2024.1339338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Antiretroviral therapies (ART) have reduced human immunodeficiency virus (HIV) infection-associated morbidity and mortality improving the life of people with HIV (PWH). However, ART lead to residual HIV production, which in conjunction with microbial translocation and immune dysfunction contributes to chronic inflammation and immune activation. PWH on ART remain at an increased risk for cardiovascular diseases (CVDs) including myocardial infarction and stroke; which in part is explained by chronic inflammation and immune activation. Lifestyle factors and certain ART are associated with dyslipidemia characterized by an increase of low-density lipoprotein (LDL), which further contributes in the increased risk for CVDs. Lipid-lowering agents like statins are emerging as immune modulators in decreasing inflammation in a variety of conditions including HIV. The international randomized clinical trial REPRIEVE has shed light on the reduction of CVDs with statin therapy among PWH. Such reports indicate a more than expected benefit of statins beyond their lipid-lowering effects. Bempedoic acid, a first-in-class non-statin LDL-lowering drug with immune modulatory effects, may further aid PWH in combination with statins. Herein, we critically reviewed studies aimed at lipid-lowering and immune-modulating roles of statins that may benefit aging PWH.
Collapse
Affiliation(s)
- Vikram Mehraj
- Research Centre McGill University Health Centre, Montreal, QC, Canada
| | - Jun Chen
- Department of Infectious Diseases and Immunology, Shanghai Public Health Clinical Center, Shanghai, China
| | - Jean-Pierre Routy
- Research Centre McGill University Health Centre, Montreal, QC, Canada
- Chronic Viral Illness Service and Division of Hematology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
8
|
Pordel S, McCloskey AP, Almahmeed W, Sahebkar A. The protective effects of statins in traumatic brain injury. Pharmacol Rep 2024; 76:235-250. [PMID: 38448729 DOI: 10.1007/s43440-024-00582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Traumatic brain injury (TBI), often referred to as the "silent epidemic", is the most common cause of mortality and morbidity worldwide among all trauma-related injuries. It is associated with considerable personal, medical, and economic consequences. Although remarkable advances in therapeutic approaches have been made, current treatments and clinical management for TBI recovery still remain to be improved. One of the factors that may contribute to this gap is that existing therapies target only a single event or pathology. However, brain injury after TBI involves various pathological mechanisms, including inflammation, oxidative stress, blood-brain barrier (BBB) disruption, ionic disturbance, excitotoxicity, mitochondrial dysfunction, neuronal necrosis, and apoptosis. Statins have several beneficial pleiotropic effects (anti-excitotoxicity, anti-inflammatory, anti-oxidant, anti-thrombotic, immunomodulatory activity, endothelial and vasoactive properties) in addition to promoting angiogenesis, neurogenesis, and synaptogenesis in TBI. Supposedly, using agents such as statins that target numerous and diverse pathological mechanisms, may be more effective than a single-target approach in TBI management. The current review was undertaken to investigate and summarize the protective mechanisms of statins against TBI. The limitations of conducted studies and directions for future research on this potential therapeutic application of statins are also discussed.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Mohammadi F, Harofteh FZ, Sahebnasagh A, Ghaneei N, Ardakani MEZ, Saghafi F. Efficacy and safety of topical rosuvastatin & melatonin vs. placebo in patients with mild to moderate plaque psoriasis: A preliminary randomized double-blinded clinical trial. Skin Res Technol 2024; 30:e13689. [PMID: 38563131 PMCID: PMC10985546 DOI: 10.1111/srt.13689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 01/16/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Considering the pathogenesis of psoriasis and also the anti-oxidant, immunomodulatory, and anti-inflammatory properties of rosuvastatin and melatonin, the current clinical trial aimed to evaluate the efficacy of topical rosuvastatin and melatonin in patients with mild to moderate psoriasis. METHODS The current randomized placebo-controlled clinical trial was conducted using a 3-arm parallel group included 77 adult patients (≥18 years old) with mild to moderate plaque psoriasis. Patients were randomized into a 1:1:1 ratio to one of three groups to receive one of the three interventions: melatonin cream, 5.0% (w/w), rosuvastatin cream, 5.0% (w/w), or placebo cream with a similar transparent appearance twice a day for 12 weeks. The primary outcome was severity of the disease using Psoriasis Area Severity Index (PASI). The secondary outcomes included the Dermatological Sum Score (DSS) to assess the erythema, scaling, and plaque elevation and the Dermatology Life Quality Index (DLQI). Photographs of the lesions were also taken at the baseline and at different periodic intervals thereafter. RESULTS Among 77 randomized patients, 52 (mean (SD) age, 40.67 (10.85) years; 22 (42.30%) men) completed the study. A significant reduction of 45% (mean (SD) of 2.67 (0.98) to 1.74 (1.12)) and 70% (mean (SD) of 2.67 (0.98) to 1.31 (1.13)) in PASI score, and 46% (mean (SD) of 2.91(1.85) to 1.57 (1.11)) and 77% (mean (SD) of 2.91 (1.85) to 0.87 (0.67)) in DSS score on days 30 and 60 with rosuvastatin cream, 5% w/w (P < 0.001) compared with baseline was observed, respectively. Also a significant decrease of 35% (mean (SD) of 2.67 (0.98) to 1.74 (1.12)) and 51% (mean (SD) of 2.67 (0.98) to 1.31 (1.13)) in PASI score, and 40% (mean (SD) of 5.00 (1.58) to 3.00 (1.76))and 61% (mean (SD) of 5.00 (1.58) to 1.92 (1.71)) in DSS score on days 30 and 60 with melatonin cream, 5% w/w (P < 0.001) compared with baseline were observed, respectively. In each of the melatonin or rosuvastatin groups, DLQI improved significantly on days 30 (P < 0.0001) and 60 (P < 0.001) while the changes in the control group were not significant. CONCLUSION The results of this clinical trial demonstrated that topical melatonin and rosuvastatin diminished the severity of mild to moderate plaque psoriasis with a satisfactory safety profile. Future clinical trials should assess both the long-term efficacy and safety of melatonin and rosuvastatin creams in larger study populations.
Collapse
Affiliation(s)
- Farhad Mohammadi
- Department of PharmaceuticsSchool of PharmacyShahid Sadoughi University of Medical Sciences and Health ServicesYazdIran
| | - Fatemeh Zare Harofteh
- Pharmaceutical Sciences Research CenterSchool of PharmacyStudent Research CommitteeShahid Sadoughi University of Medical Sciences and Health ServicesYazdIran
| | - Adeleh Sahebnasagh
- Department of Internal MedicineClinical Research CenterFaculty of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Narges Ghaneei
- Department of DermatologySchool of MedicineShahid Sadoughi University of Medical Sciences and Health ServicesYazdIran
| | | | - Fatemeh Saghafi
- Department of Clinical PharmacySchool of PharmacyShahid Sadoughi University of Medical Sciences and Health ServicesYazdIran
| |
Collapse
|
10
|
Chen WH, Chen CH, Hsu MC, Chang RW, Wang CH, Lee TS. Advances in the molecular mechanisms of statins in regulating endothelial nitric oxide bioavailability: Interlocking biology between eNOS activity and L-arginine metabolism. Biomed Pharmacother 2024; 171:116192. [PMID: 38262153 DOI: 10.1016/j.biopha.2024.116192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
Statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A, are widely used to treat hypercholesterolemia. In addition, statins have been suggested to reduce the risk of cardiovascular events owing to their pleiotropic effects on the vascular system, including vasodilation, anti-inflammation, anti-coagulation, anti-oxidation, and inhibition of vascular smooth muscle cell proliferation. The major beneficial effect of statins in maintaining vascular homeostasis is the induction of nitric oxide (NO) bioavailability by activating endothelial NO synthase (eNOS) in endothelial cells. The mechanisms underlying the increased NO bioavailability and eNOS activation by statins have been well-established in various fields, including transcriptional and post-transcriptional regulation, kinase-dependent phosphorylation and protein-protein interactions. However, the mechanism by which statins affect the metabolism of L-arginine, a precursor of NO biosynthesis, has rarely been discussed. Autophagy, which is crucial for energy homeostasis, regulates endothelial functions, including NO production and angiogenesis, and is a potential therapeutic target for cardiovascular diseases. In this review, in addition to summarizing the molecular mechanisms underlying increased NO bioavailability and eNOS activation by statins, we also discuss the effects of statins on the metabolism of L-arginine.
Collapse
Affiliation(s)
- Wen-Hua Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Man-Chen Hsu
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ru-Wen Chang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chih-Hsien Wang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
11
|
Zahedipour F, Hosseini SA, Reiner Ž, Tedeschi-Reiner E, Jamialahmadi T, Sahebkar A. Therapeutic Effects of Statins: Promising Drug for Topical and Transdermal Administration. Curr Med Chem 2024; 31:3149-3166. [PMID: 37157198 DOI: 10.2174/0929867330666230508141434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 05/10/2023]
Abstract
Statins are HMG-CoA reductase inhibitors and decrease plasma low-density lipoprotein cholesterol (LDL-C) levels. They are well tolerated, and because of their LDL-C-lowering effect, they are utilized to decrease the risk of atherosclerosis and cardiovascular disease. However, statins have pleiotropic effects, including immunomodulatory, anti-inflammatory, antioxidant, and anticancer. Currently, oral administration is the only Food and Drug Administration (FDA)-approved route of administration for statins. However, other administration routes have demonstrated promising results in different pre-clinical and clinical studies. For instance, statins also seem beneficial in dermatitis, psoriasis, vitiligo, hirsutism, uremic pruritus, and graft-versus-host disease. Topically applied statins have been studied to treat seborrhea, acne, rhinophyma, and rosacea. They also have beneficial effects in contact dermatitis and wound healing in animal studies, (HIV) infection, osseointegration, porokeratosis, and some ophthalmologic diseases. Topical and transdermal application of statins is a non-invasive drug administration method that has shown significant results in bypassing the first-pass metabolism in the liver, thereby reducing possible adverse effects. This study reviews the multifaceted molecular and cellular impacts of statins, their topical and transdermal application, novel delivery systems, such as nanosystems for topical and transdermal administration and the challenges concerning this approach.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyede Atefe Hosseini
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Željko Reiner
- University Hospital Center Zagreb, Department of Internal Medicine, Zagreb, Croatia
- Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | | | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- 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
|
12
|
Zhou X, Wu X, Wang R, Han L, Li H, Zhao W. Mechanisms of 3-Hydroxyl 3-Methylglutaryl CoA Reductase in Alzheimer's Disease. Int J Mol Sci 2023; 25:170. [PMID: 38203341 PMCID: PMC10778631 DOI: 10.3390/ijms25010170] [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: 11/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide and has a high incidence in the elderly. Unfortunately, there is no effective therapy for AD owing to its complicated pathogenesis. However, the development of lipid-lowering anti-inflammatory drugs has heralded a new era in the treatment of Alzheimer's disease. Several studies in recent years have shown that lipid metabolic dysregulation and neuroinflammation are associated with the pathogenesis of AD. 3-Hydroxyl 3-methylglutaryl CoA reductase (HMGCR) is a rate-limiting enzyme in cholesterol synthesis that plays a key role in cholesterol metabolism. HMGCR inhibitors, known as statins, have changed from being solely lipid-lowering agents to neuroprotective compounds because of their effects on lipid levels and inflammation. In this review, we first summarize the main regulatory mechanism of HMGCR affecting cholesterol biosynthesis. We also discuss the pathogenesis of AD induced by HMGCR, including disordered lipid metabolism, oxidative stress, inflammation, microglial proliferation, and amyloid-β (Aβ) deposition. Subsequently, we explain the possibility of HMGCR as a potential target for AD treatment. Statins-based AD treatment is an ascent field and currently quite controversial; therefore, we also elaborate on the current application prospects and limitations of statins in AD treatment.
Collapse
Affiliation(s)
- Xun Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Xiaolang Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Rui Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Lu Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| | - Huilin Li
- Department of Endocrinology, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China;
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; (X.Z.); (X.W.); (R.W.); (L.H.)
| |
Collapse
|
13
|
Piao X, Ma L, Xu Q, Zhang X, Jin C. Noncoding RNAs: Versatile regulators of endothelial dysfunction. Life Sci 2023; 334:122246. [PMID: 37931743 DOI: 10.1016/j.lfs.2023.122246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Noncoding RNAs have recently emerged as versatile regulators of endothelial dysfunction in atherosclerosis, a chronic inflammatory disease characterized by the formation of plaques within the arterial walls. Through their ability to modulate gene expression, noncoding RNAs, including microRNAs, long noncoding RNAs, and circular RNAs, play crucial roles in various cellular processes involved in endothelial dysfunction (ECD), such as inflammation, pyroptosis, migration, proliferation, apoptosis, oxidative stress, and angiogenesis. This review provides an overview of the current understanding of the regulatory roles of noncoding RNAs in endothelial dysfunction during atherosclerosis. It highlights the specific noncoding RNAs that have been implicated in the pathogenesis of ECD, their target genes, and the mechanisms by which they contribute to ECD. Furthermore, we have reviewed the current therapeutics in atherosclerosis and explore their interaction with noncoding RNAs. Understanding the intricate regulatory network of noncoding RNAs in ECD may open up new opportunities for the development of novel therapeutic strategies to combat ECD.
Collapse
Affiliation(s)
- Xiong Piao
- Cardiovascular Surgery, Yanbian University Hospital, Yanji 133000, China.
| | - Lie Ma
- Cardiovascular Surgery, Yanbian University Hospital, Yanji 133000, China
| | - Qinqi Xu
- Cardiovascular Surgery, Yanbian University Hospital, Yanji 133000, China
| | - Xiaomin Zhang
- Cardiovascular Surgery, Yanbian University Hospital, Yanji 133000, China
| | - Chengzhu Jin
- Cardiovascular Surgery, Yanbian University Hospital, Yanji 133000, China
| |
Collapse
|
14
|
Kiani M, Moraffah F, Khonsari F, Kharazian B, Dinarvand R, Shokrgozar MA, Atyabi F. Co-delivery of simvastatin and microRNA-21 through liposome could accelerates the wound healing process. BIOMATERIALS ADVANCES 2023; 154:213658. [PMID: 37866233 DOI: 10.1016/j.bioadv.2023.213658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/10/2023] [Accepted: 10/07/2023] [Indexed: 10/24/2023]
Abstract
The gene delivery approach, mainly microRNAs (miRNA) as key wound healing mediators, has recently received extensive attention. MicroRNA-21 (miR-21) has strongly impacted wound healing by affecting the inflammation and proliferation phases. Previous studies have also demonstrated the beneficial effect of simvastatin on wound healing. Therefore, we designed a dual-drug/gene delivery system using PEGylated liposomes that could simultaneously attain the co-encapsulation and co-delivery of miRNA and simvastatin (SIM) to explore the combined effect of this dual-drug delivery system on wound healing. The PEG-liposomes for simvastatin and miR-21 plasmid (miR-21-P/SIM/Liposomes) were prepared using the thin-film hydration method. The liposomes showed 85 % entrapment efficiency for SIM in the lipid bilayer and high physical entrapment of miR-21-P in the inner cavity. In vitro studies demonstrated no cytotoxicity for the carrier on normal human dermal fibroblast cells (NHDF) and 97 % cellular uptake over 2 h incubation. The scratch test revealed excellent cell proliferation and migration after treatment with miR-21-P/SIM/Liposomes. For the in vivo experiments, a full-thickness cutaneous wound model was used. The wound closure on day 8 was higher for Liposomal formulation containing miR-21-P promoting faster re-epithelialization. On day 12, all treated groups showed complete wound closure. However, following histological analysis, the miR-21-P/SIM/Liposomes revealed the best tissue regeneration, similar to normal functional skin, by reduced inflammation and increased re-epithelialization, collagen deposition and angiogenesis. In conclusion, the designed miR-21-P/SIM/Liposomes could significantly accelerate the process of wound healing, which provides a new strategy for the management of chronic wounds.
Collapse
Affiliation(s)
- Melika Kiani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Moraffah
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Khonsari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahar Kharazian
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; School of Pharmacy, De Mont Fort University, Leicester, UK
| | | | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Ishinoda Y, Masaki N, Hitomi Y, Taruoka A, Kawai A, Iwashita M, Yumita Y, Kagami K, Yasuda R, Ido Y, Toya T, Ikegami Y, Namba T, Nagatomo Y, Miyazaki K, Takase B, Adachi T. A Low Arginine/Ornithine Ratio is Associated with Long-Term Cardiovascular Mortality. J Atheroscler Thromb 2023; 30:1364-1375. [PMID: 36775332 PMCID: PMC10564648 DOI: 10.5551/jat.63779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/25/2022] [Indexed: 02/13/2023] Open
Abstract
AIMS The long-term prognostic value of the bioavailability of L-arginine, an important source of nitric oxide for the maintenance of vascular endothelial function, has not been investigated fully. We therefore investigated the relationship between amino acid profile and long-term prognosis in patients with a history of standby coronary angiography. METHODS We measured the serum concentrations of L-arginine, L-citrulline, and L-ornithine by high-speed liquid chromatography. We examined the relationship between the L-arginine/L-ornithine ratio and the incidence of all-cause death, cardiovascular death, and major adverse cardiovascular events (MACEs) in 262 patients (202 men and 60 women, age 65±13 years) who underwent coronary angiography over a period of ≤ 10 years. RESULTS During the observation period of 5.5±3.2 years, 31 (12%) patients died, including 20 (8%) of cardiovascular death, while 32 (12%) had MACEs. Cox regression analysis revealed that L-arginine/L-ornithine ratio was associated with an increased risk for all-cause death (unadjusted hazard ratio, 95% confidence interval) (0.940, 0.888-0.995) and cardiovascular death (0.895, 0.821-0.965) (p<0.05 for all). In a model adjusted for age, sex, hypertension, hyperlipidemia, diabetes, current smoking, renal function, and log10-transformed brain natriuretic peptide level, cardiovascular death (0.911, 0.839-0.990, p=0.028) retained an association with a low L-arginine/ L-ornithine ratio. When the patients were grouped according to an L-arginine/L-ornithine ratio of 1.16, the lower L-arginine/L-ornithine ratio group had significantly higher incidence of all-cause death, cardiovascular death, and MACEs. CONCLUSION A low L-arginine/L-ornithine ratio may be associated with increased 10-year cardiac mortality.
Collapse
Affiliation(s)
- Yuki Ishinoda
- Department of Endocrinology, National Defense Medical College, Saitama, Japan
| | - Nobuyuki Masaki
- Department o f Intensive Care Medicine, National Defense Medical College, Saitama, Japan
| | - Yasuhiro Hitomi
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Akira Taruoka
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Akane Kawai
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Midori Iwashita
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Yusuke Yumita
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Kazuki Kagami
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Risako Yasuda
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Yasuo Ido
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Takumi Toya
- Department o f Intensive Care Medicine, National Defense Medical College, Saitama, Japan
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Yukinori Ikegami
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Takayuki Namba
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Yuji Nagatomo
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| | - Koji Miyazaki
- Department of Comprehensive Internal Medicine, Tokai University Hachioji Hospital, Tokyo, Japan
| | - Bonpei Takase
- Department o f Intensive Care Medicine, National Defense Medical College, Saitama, Japan
| | - Takeshi Adachi
- Department of Cardiology, National Defense Medical College, Saitama, Japan
| |
Collapse
|
16
|
Li S, Zhang Y, Yang Y, Chen S, Yang Z, Kuang C, Zhong Y, Liu F. The impact of statin use before intensive care unit admission on patients with acute kidney injury after cardiac surgery. Front Pharmacol 2023; 14:1259828. [PMID: 37781714 PMCID: PMC10537929 DOI: 10.3389/fphar.2023.1259828] [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: 07/16/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Background: Cardiac surgery-associated acute kidney injury (CSA-AKI) is a common and serious complication after cardiac surgery. The influence of statin use before surgery on the renal outcome of patients undergoing cardiac surgery is controversial. The purpose of this study was to evaluate the effect of statins on postoperative renal outcomes in patients undergoing cardiac surgery. Methods: We included CSA-AKI patients in the Medical Information Mart for Intensive Care (MIMIC)-IV database and were divided into statin group and non-statin group according to whether they used statins before entering intensive care units (ICU). The main outcomes were hospitalization and 30-day mortality, and the secondary outcomes were 60-day mortality and 90-day mortality. We used propensity score matching (PSM) to adjust for confounding factors. The 95% confidence interval (CI) and risk ratio (RO) were calculated by the COX proportional regression model. At the same time, stratified analysis was used to explore whether the relationship between the statins use before intensive care units and mortality was different in each subgroup and whether the relationship between different doses of Atorvastatin and mortality was different. Result: We identified 675 pre-ICU statin users and 2095 non-statin users. In the COX proportional regression model, pre-ICU statin use was associated with decreased in-hospital (HR = 0.407, 95%confidence interval 0.278-0.595, p < 0.001) and 30-day mortality (HR = 0.407, 95%CI 0.279-0.595, p < 0.001). The survival rate of patients who took statins before entering ICU was significantly higher than that of those who did not use statins at 30 days, 60 days and 90 days. There is a significant interaction between patients with aged>65 years (HR = 0.373, 95%CI 0.240-0.581, p < 0.001), Acute kidney injury grade I (HR = 0.244, 95%CI 0.118-0.428, p < 0.001), and without post-myocardial infarction syndrome (HR = 0.344, 95%CI 0.218-0.542, p < 0.001). The mortality in hospital and 60 days of CSA-AKI patients treated with ≥80 mg Atorvastatin before operation was significantly reduced (p < 0.05). Conclusion: The pre-ICU statin use was significantly associated with decreased risk in hospital and 30-day mortality. The preoperative use of ≥80 mg Atorvastatin may improve the prognosis of CSA-AKI.
Collapse
Affiliation(s)
- Shishi Li
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Youlin Zhang
- Department of the Second Clinical, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Yan Yang
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Sining Chen
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhiqian Yang
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Chaoying Kuang
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Yuzhen Zhong
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Fanna Liu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
17
|
Malektaj H, Nour S, Imani R, Siadati MH. Angiogenesis induction as a key step in cardiac tissue Regeneration: From angiogenic agents to biomaterials. Int J Pharm 2023; 643:123233. [PMID: 37460050 DOI: 10.1016/j.ijpharm.2023.123233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. After myocardial infarction, the vascular supply of the heart is damaged or blocked, leading to the formation of scar tissue, followed by several cardiac dysfunctions or even death. In this regard, induction of angiogenesis is considered as a vital process for supplying nutrients and oxygen to the cells in cardiac tissue engineering. The current review aims to summarize different approaches of angiogenesis induction for effective cardiac tissue repair. Accordingly, a comprehensive classification of induction of pro-angiogenic signaling pathways through using engineered biomaterials, drugs, angiogenic factors, as well as combinatorial approaches is introduced as a potential platform for cardiac regeneration application. The angiogenic induction for cardiac repair can enhance patient treatment outcomes and generate economic prospects for the biomedical industry. The development and commercialization of angiogenesis methods often involves collaboration between academic institutions, research organizations, and biomedical companies.
Collapse
Affiliation(s)
- Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg 9220, Denmark
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, VIC 3010, Australia; Department of Chemical Engineering, The University of Melbourne, VIC 3010, Australia
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad H Siadati
- Materials Science and Engineering Faculty, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
18
|
Kabra S, Thosar NR, Malviya NS. Exploring the Synergistic Effect of Simvastatin in Oral Health Applications: A Literature Review. Cureus 2023; 15:e44411. [PMID: 37791218 PMCID: PMC10543113 DOI: 10.7759/cureus.44411] [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: 03/17/2023] [Accepted: 08/30/2023] [Indexed: 10/05/2023] Open
Abstract
Statins are the first line of treatment for hyperlipidaemia. Along with lowering lipids, it also lowers mortality and cardiovascular risk. Statins play a major role in maintaining the homeostasis of the oral cavity via a number of different mechanisms. It includes regeneration of dentin and pulp by differentiation and increased development of mineralized tissue via the bone morphogenetic proteins (BMP)-2 Pathway. It shows effective bone health by leading to osteogenic differentiation mesenchymal stem cells, by facilitating epithelization process in wound healing, anti-inflammatory, antioxidant, antimicrobial, antiviral, and fungicidal properties. To the finest of the information we have, there have been very few comprehensive studies that have investigated the effects of statin drugs on various aspects of dental and oral health. As a result, the main objective of this review was to examine the effect of statins on oral health applications. According to the findings of our extensive review, statins have noteworthy and promising effects on several aspects of oral health, including dental pulp cells, chronic periodontitis, alveolar bone loss, orthodontic tooth movement, and so on. Nevertheless, it is concluded that local or even systemic administration of simvastatin should be regarded as an innovative, easily accessible, and safe therapeutic agent that has a significant impact on enhancing the oral health.
Collapse
Affiliation(s)
- Sakshi Kabra
- Pediatric and Preventive Dentistry, Sharard Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research (Deemed to be University), Wardha, IND
| | - Nilima R Thosar
- Pediatric and Preventive Dentistry, Sharard Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research (Deemed to be University), Wardha, IND
| | - Nishi S Malviya
- Pediatric and Preventive Dentistry, Sharard Pawar Dental College and Hospital, Datta Meghe Institute of Higher Education and Research (Deemed to be University), Wardha, IND
| |
Collapse
|
19
|
Pham DL, Niemi A, Blank R, Lomenzo G, Tham J, Ko ML, Ko GYP. Peptide Lv Promotes Trafficking and Membrane Insertion of K Ca3.1 through the MEK1-ERK and PI3K-Akt Signaling Pathways. Cells 2023; 12:1651. [PMID: 37371121 PMCID: PMC10296961 DOI: 10.3390/cells12121651] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Peptide Lv is a small endogenous secretory peptide that is proangiogenic through hyperpolarizing vascular endothelial cells (ECs) by enhancing the current densities of KCa3.1 channels. However, it is unclear how peptide Lv enhances these currents. One way to enhance the current densities of ion channels is to promote its trafficking and insertion into the plasma membrane. We hypothesized that peptide Lv-elicited KCa3.1 augmentation occurs through activating the mitogen-activated protein kinase kinase 1 (MEK1)-extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt) signaling pathways, which are known to mediate ion channel trafficking and membrane insertion in neurons. To test this hypothesis, we employed patch-clamp electrophysiological recordings and cell-surface biotinylation assays on ECs treated with peptide Lv and pharmaceutical inhibitors of ERK and Akt. Blocking ERK or Akt activation diminished peptide Lv-elicited EC hyperpolarization and increase in KCa3.1 current densities. Blocking PI3K or Akt activation decreased the level of plasma membrane-bound, but not the total amount of KCa3.1 protein in ECs. Therefore, the peptide Lv-elicited EC hyperpolarization and KCa3.1 augmentation occurred in part through channel trafficking and insertion mediated by MEK1-ERK and PI3K-Akt activation. These results demonstrate the molecular mechanisms of how peptide Lv promotes EC-mediated angiogenesis.
Collapse
Affiliation(s)
- Dylan L. Pham
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
| | - Autumn Niemi
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
| | - Ria Blank
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
| | - Gabriella Lomenzo
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
| | - Jenivi Tham
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
| | - Michael L. Ko
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
- Department of Biology, Division of Natural and Physical Sciences, Blinn College, Bryan, TX 77802, USA
| | - Gladys Y.-P. Ko
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA; (D.L.P.); (A.N.); (R.B.); (G.L.); (J.T.); (M.L.K.)
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
20
|
German CA, Liao JK. Understanding the molecular mechanisms of statin pleiotropic effects. Arch Toxicol 2023; 97:1529-1545. [PMID: 37084080 PMCID: PMC10119541 DOI: 10.1007/s00204-023-03492-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 04/22/2023]
Abstract
Statins represent the cornerstone of pharmacotherapy for the prevention of atherosclerotic cardiovascular disease. These medications not only reduce low-density lipoprotein cholesterol (LDL-C) via inhibition of 3-hydroxy-3-methylglutarate attached to CoA reductase, the key rate-limiting step in the cholesterol biosynthetic pathway, but also upregulate expression of the low-density lipoprotein receptor, improving serum clearance. Given LDL-C is a causal risk factor for the development of atherosclerosis, these complementary mechanisms largely explain why statin therapy leads to reductions in major adverse cardiovascular events. However, decades of basic and clinical research have suggested that statins may exert other effects independent of LDL-C lowering, termed pleiotropic effects, which have become a topic of debate among the scientific community. While some literature suggests statins may improve plaque stability, reduce inflammation and thrombosis, decrease oxidative stress, and improve endothelial function and vascular tone, other studies have suggested potential harmful pleiotropic effects related to increased risk of muscle-related side effects, diabetes, hemorrhagic stroke, and cognitive decline. Furthermore, the introduction of newer, non-statin LDL-C lowering therapies, including ezetimibe, proprotein convertase subtilisin/Kexin Type 9, and bempedoic acid, have challenged the statin pleiotropy theory. This review aims to provide a historical background on the development of statins, explore the mechanistic underpinnings of statin pleiotropy, review the available literature, and provide up to date examples that suggest statins may exert effects outside of LDL-C lowering and the cardiovascular system.
Collapse
Affiliation(s)
- Charles A German
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA.
| | - James K Liao
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
21
|
Higashi Y. Endothelial Function in Dyslipidemia: Roles of LDL-Cholesterol, HDL-Cholesterol and Triglycerides. Cells 2023; 12:1293. [PMID: 37174693 PMCID: PMC10177132 DOI: 10.3390/cells12091293] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Dyslipidemia is associated with endothelial dysfunction. Endothelial dysfunction is the initial step for atherosclerosis, resulting in cardiovascular complications. It is clinically important to break the process of endothelial dysfunction to cardiovascular complications in patients with dyslipidemia. Lipid-lowering therapy enables the improvement of endothelial function in patients with dyslipidemia. It is likely that the relationships of components of a lipid profile such as low-density lipoprotein cholesterol, high-density lipoprotein cholesterol and triglycerides with endothelial function are not simple. In this review, we focus on the roles of components of a lipid profile in endothelial function.
Collapse
Affiliation(s)
- Yukihito Higashi
- Department of Regenerative Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 743-8551, Japan; ; Tel.: +81-82-257-5831
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8553, Japan
| |
Collapse
|
22
|
Li R, Zhang J, Shi J, Yue J, Cui Y, Ye Q, Wu G, Zhang Z, Guo Y, Fu D. An intelligent phase transformation system based on lyotropic liquid crystals for sequential biomolecule delivery to enhance bone regeneration. J Mater Chem B 2023; 11:2946-2957. [PMID: 36916173 DOI: 10.1039/d2tb02725a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Endogenous repair of critical bone defects is typically hampered by inadequate vascularization in the early stages and insufficient bone regeneration later on. Therefore, drug delivery systems with the ability to couple angiogenesis and osteogenesis in a spatiotemporal manner are highly desirable for vascularized bone formation. Herein, we devoted to develop a liquid crystal formulation system (LCFS) attaining a controlled temporal release of angiogenic and osteoinductive bioactive molecules that could orchestrate the coupling of angiogenesis and osteogenesis in an optimal way. It has been demonstrated that the release kinetics of biomolecules depend on the hydrophobicity of the loaded molecules, making the delivery profile programmable and controllable. The hydrophilic deferoxamine (DFO) could be released rapidly within 5 days to activate angiogenic signaling, while the lipophilic simvastatin (SIM) showed a slow and sustained release for continuous osteogenic induction. Apart from its good biocompatibility with mesenchymal stem cells derived from rat bone marrow (rBMSCs), the DFO/SIM loaded LCFS could stimulate the formation of a vascular morphology in human umbilical vein endothelial cells (HUVECs) and the osteogenic differentiation of rBMSCs in vitro. The in vivo rat femoral defect models have witnessed the prominent angiogenic and osteogenic effects induced by the sequential presentation of DFO and SIM. This study suggests that the sequential release of DFO and SIM from the LCFS results in enhanced bone formation, offering a facile and viable treatment option for bone defects by mimicking the physiological process of bone regeneration.
Collapse
Affiliation(s)
- Rui Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P. R. China
| | - Jiao Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P. R. China
| | - Jingyu Shi
- Department of Pharmacy, Liyuan Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P. R. China.
| | - Jiang Yue
- Department of Endocrinology and Metabolism, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201114, P. R. China
| | - Yongzhi Cui
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei 430066, P. R. China
| | - Gang Wu
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, The Netherlands
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P. R. China
| | - Yuanyuan Guo
- Department of Pharmacy, Liyuan Hospital, Tongji Medical School, Huazhong University of Science and Technology, Wuhan, Hubei 430077, P. R. China.
| | - Dehao Fu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, P. R. China.
| |
Collapse
|
23
|
Ahmadi Y, Fard JK, Ghafoor D, Eid AH, Sahebkar A. Paradoxical effects of statins on endothelial and cancer cells: the impact of concentrations. Cancer Cell Int 2023; 23:43. [PMID: 36899388 PMCID: PMC9999585 DOI: 10.1186/s12935-023-02890-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
In addition to their lipid-lowering functions, statins elicit additional pleiotropic effects on apoptosis, angiogenesis, inflammation, senescence, and oxidative stress. Many of these effects have been reported in cancerous and noncancerous cells like endothelial cells (ECs), endothelial progenitor cells (EPCs) and human umbilical vein cells (HUVCs). Not surprisingly, statins' effects appear to vary largely depending on the cell context, especially as pertains to modulation of cell cycle, senescence, and apoptotic processes. Perhaps the most critical reason for this discordance is the bias in selecting the applied doses in various cells. While lower (nanomolar) concentrations of statins impose anti-senescence, and antiapoptotic effects, higher concentrations (micromolar) appear to precipitate opposite effects. Indeed, most studies performed in cancer cells utilized high concentrations, where statin-induced cytotoxic and cytostatic effects were noted. Some studies report that even at low concentrations, statins induce senescence or cytostatic impacts but not cytotoxic effects. However, the literature appears to be relatively consistent that in cancer cells, statins, in both low or higher concentrations, induce apoptosis or cell cycle arrest, anti-proliferative effects, and cause senescence. However, statins' effects on ECs depend on the concentrations; at micromolar concentrations statins cause cell senescence and apoptosis, while at nonomolar concentrations statins act reversely.
Collapse
Affiliation(s)
- Yasin Ahmadi
- College of Science, Department of Medical Laboratory Sciences, Komar University of Science and Technology, 46001, Sulaymania, Iraq.
| | - Javad Khalili Fard
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dlzar Ghafoor
- College of Science, Department of Medical Laboratory Sciences, Komar University of Science and Technology, 46001, Sulaymania, Iraq
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran. .,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
24
|
Chen X, Yu W, Zhang J, Fan X, Liu X, Liu Q, Pan S, Dixon RAF, Li P, Yu P, Shi A. Therapeutic angiogenesis and tissue revascularization in ischemic vascular disease. J Biol Eng 2023; 17:13. [PMID: 36797776 PMCID: PMC9936669 DOI: 10.1186/s13036-023-00330-2] [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: 11/08/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Ischemic vascular disease is a major healthcare problem. The keys to treatment lie in vascular regeneration and restoration of perfusion. However, current treatments cannot satisfy the need for vascular regeneration to restore blood circulation. As biomedical research has evolved rapidly, a variety of potential alternative therapeutics has been explored widely, such as growth factor-based therapy, cell-based therapy, and material-based therapy including nanomedicine and biomaterials. This review will comprehensively describe the main pathogenesis of vascular injury in ischemic vascular disease, the therapeutic function of the above three treatment strategies, the corresponding potential challenges, and future research directions.
Collapse
Affiliation(s)
- Xinyue Chen
- grid.412455.30000 0004 1756 5980The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Wenlu Yu
- grid.260463.50000 0001 2182 8825School of Ophthalmology and Optometry of Nanchang University, Nanchang, 330006 China
| | - Jing Zhang
- grid.412455.30000 0004 1756 5980Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Xiao Fan
- grid.412455.30000 0004 1756 5980Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi China
| | - Xiao Liu
- grid.412536.70000 0004 1791 7851Department of Cardiovascular Medicine, The Second Affiliated Hospital of Sun Yat Sen University, Guangzhou, 51000 Guangdong China
| | - Qi Liu
- grid.416470.00000 0004 4656 4290Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX USA
| | - Su Pan
- grid.416470.00000 0004 4656 4290Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX USA
| | - Richard A. F. Dixon
- grid.416470.00000 0004 4656 4290Wafic Said Molecular Cardiology Research Laboratory, The Texas Heart Institute, Houston, TX USA
| | - Pengyang Li
- grid.224260.00000 0004 0458 8737Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA USA
| | - Peng Yu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China. .,Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, China.
| | - Ao Shi
- School of Medicine, St. George University of London, London, UK. .,School of Medicine, University of Nicosia, Nicosia, Cyprus.
| |
Collapse
|
25
|
Neuroprotective effects of pravastatin in cerebral venous infarction in a rat model. IBRO Neurosci Rep 2023; 14:202-209. [PMID: 36852215 PMCID: PMC9958423 DOI: 10.1016/j.ibneur.2023.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Objectives Pravastatin sodium is reported to have multiple beneficial effects in cerebral atherosclerosis and neuronal injury; however, the preventive effects on cerebral venous ischemia are still unknown. Herein, we aimed to examine the neuroprotective effects of transoral prior administration of pravastatin sodium against cerebral cortical venous ischemia with suppression of apoptosis. Methods Thirty 8-week-old male Wistar rats were divided equally into two study groups (n = 15 vs. n = 15); the pravastatin group was fed 1% pravastatin sodium with their usual diet for 2 weeks, while the control group only received the usual diet. Two-vein occlusion (2VO) model was applied for this study, and two adjacent cortical veins in each animal were permanently occluded photochemically with rose bengal dye. During photo-thrombosis, regional changes of the cerebral blood flow (CBF) in area of the venous ischemia were recorded. At 48-h after 2VO, animals were euthanized using perfusion fixation, and we histologically measured ratios of infarcted area to contralateral hemisphere, and counted Bax- and Bcl-2-positive cells in the penumbra to investigate the implications for apoptosis. Results The ratio of infarcted area was significantly decreased in the pravastatin group compared to the control group (P < 0.01). The number of Bax-positive cells also decreased significantly in the pravastatin group (P < 0.01). In contrast, immunolabeling for Bcl-2 was essentially negative in all areas in both groups. There were also no significant differences in regional CBF changes after 2VO between the two groups (P = 0.13). Conclusions Pre-emptive administration of pravastatin sodium mixed in the food has neuroprotective effects against cerebral cortical venous ischemia with suppression of apoptosis associated with inhibition of Bax expression but has little influence on regional CBF.
Collapse
Key Words
- 2VO, two-vein occlusion
- Akt, protein kinase B
- BBB, blood-brain-barrier
- CAI, cerebral arterial ischemia
- CBF, cerebral blood flow
- CVI, cerebral venous ischemia
- Cerebral blood flow
- Cerebral cortex
- Cerebral ischemia
- HMG-CoA, 3-hydroxy 3-methylglutaryl coenzyme A
- HSP, heat shock protein
- IL-6, Interleukin-6
- JNK, Jun-NH2-terminal kinase
- LDL, low-density lipoprotein
- LDU, Laser Doppler-unit
- MAPK, mitogen-activated protein kinase
- MCAO, middle cerebral artery occlusion
- Neuronal apoptosis
- PI3K, phosphatidylinositol 3-OH kinase
- Pravastatin sodium
- TNFα, Tumor Necrosis Factor‐α
- Vein
Collapse
|
26
|
Association between atherogenic risk-modulating proteins and endothelium-dependent flow-mediated dilation in coronary artery disease patients. Eur J Appl Physiol 2023; 123:367-380. [PMID: 36305972 PMCID: PMC9894982 DOI: 10.1007/s00421-022-05040-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 09/04/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE Endothelial dysfunction is an early and integral event in the development of atherosclerosis and coronary artery disease (CAD). Reduced NO bioavailability, oxidative stress, vasoconstriction, inflammation and senescence are all implicated in endothelial dysfunction. However, there are limited data examining associations between these pathways and direct in vivo bioassay measures of endothelial function in CAD patients. This study aimed to examine the relationships between in vivo measures of vascular function and the expression of atherogenic risk-modulating proteins in endothelial cells (ECs) isolated from the radial artery of CAD patients. METHODS Fifty-six patients with established CAD underwent trans-radial catheterization. Prior to catheterization, radial artery vascular function was assessed using a) flow-mediated dilation (FMD), and b) exercise-induced dilation in response to handgrip (HE%). Freshly isolated ECs were obtained from the radial artery during catheterization and protein content of eNOS, NAD(P)H oxidase subunit NOX2, NFκB, ET-1 and the senescence markers p53, p21 and p16 were evaluated alongside nitrotyrosine abundance and eNOS Ser1177 phosphorylation. RESULTS FMD was positively associated with eNOS Ser1177 phosphorylation (r = 0.290, P = 0.037), and protein content of p21 (r = 0.307, P = 0.027) and p16 (r = 0.426, P = 0.002). No associations were found between FMD and markers of oxidative stress, vasoconstriction or inflammation. In contrast to FMD, HE% was not associated with any of the EC proteins. CONCLUSION These data revealed a difference in the regulation of endothelium-dependent vasodilation measured in vivo between patients with CAD compared to previously reported data in subjects without a clinical diagnosis, suggesting that eNOS Ser1177 phosphorylation may be the key to maintain vasodilation in CAD patients.
Collapse
|
27
|
Combined Therapy with Simvastatin- and Coenzyme-Q10-Loaded Nanoparticles Upregulates the Akt-eNOS Pathway in Experimental Metabolic Syndrome. Int J Mol Sci 2022; 24:ijms24010276. [PMID: 36613727 PMCID: PMC9820291 DOI: 10.3390/ijms24010276] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
In addition to their LDL-cholesterol-lowering effect, statins have pleiotropic beneficial effects on the cardiovascular system. However, long-term treatment with statins may be associated with serious side effects. With the aim to make statin therapy more effective, we studied the effects of simvastatin- and coenzyme-Q10-loaded polymeric nanoparticles on the lipid profile and nitric oxide (NO)/reactive oxygen species (ROS) balance in the heart and aorta of adult male obese Zucker rats. The rats were divided into an untreated group, a group treated with empty nanoparticles, and groups treated with simvastatin-, coenzyme Q10 (CoQ10)-, or a combination of simvastatin- and CoQ10-loaded nanoparticles (SIMV+CoQ10). After 6 weeks, the lipid profile in the plasma and the concentration of conjugated dienes in the liver were determined. Nitric oxide synthase (NOS) activity, Akt, endothelial NOS (eNOS), phosphorylated eNOS (p-eNOS), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, and nuclear factor kappaB (NF-kappaB) protein expressions were measured in the heart and aorta. All simvastatin, CoQ10, and SIMV+CoQ10 treatments decreased plasma LDL levels, but only the combined SIMV+CoQ10 treatment increased NOS activity and the expression of Akt, eNOS, and p-eNOS in both the heart and the aorta. Interestingly, NADPH oxidase in the heart and NF-kappaB protein expression in the aorta were decreased by all treatments, including nanoparticles alone. In conclusion, only combined therapy with SIMV- and CoQ10-loaded nanoparticles increased NOS activity and upregulated the Akt-eNOS pathway in obese Zucker rats, which may represent a promising tool for the treatment of cardiometabolic diseases.
Collapse
|
28
|
Shcheblykin DV, Bolgov AA, Pokrovskii MV, Stepenko JV, Tsuverkalova JM, Shcheblykina OV, Golubinskaya PA, Korokina LV. Endothelial dysfunction: developmental mechanisms and therapeutic strategies. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.80376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: Every year the importance of the normal functioning of the endothelial layer of the vascular wall in maintaining the health of the body becomes more and more obvious.
The physiological role of the endothelium: The endothelium is a metabolically active organ actively involved in the regulation of hemostasis, modulation of inflammation, maintenance of hemovascular homeostasis, regulation of angiogenesis, vascular tone, and permeability.
Risk factors for the development of endothelial dysfunction: Currently, insufficient bioavailability of nitric oxide is considered the most significant risk factor for endothelial dysfunction.
Mechanisms of development of endothelial dysfunction: The genesis of endothelial dysfunction is a multifactorial process. Among various complex mechanisms, this review examines oxidative stress, inflammation, hyperglycemia, vitamin D deficiency, dyslipidemia, excess visceral fat, hyperhomocysteinemia, hyperuricemia, as well as primary genetic defect of endotheliocytes, as the most common causes in the population underlying the development of endothelial dysfunction.
Markers of endothelial dysfunction in various diseases: This article discusses the main biomarkers of endothelial dysfunction currently used, as well as promising biomarkers in the future for laboratory diagnosis of this pathology.
Therapeutic strategies: Therapeutic approaches to the endothelium in order to prevent or reduce a degree of damage to the vascular wall are briefly described.
Conclusion: Endothelial dysfunction is a typical pathological process involved in the pathogenesis of many diseases. Thus, pharmacological agents with endothelioprotective properties can provide more therapeutic benefits than a drug without such an effect.
Collapse
|
29
|
Motoji Y, Fukazawa R, Matsui R, Abe Y, Uehara I, Watanabe M, Hashimoto Y, Miyagi Y, Nagi-Miura N, Tanaka N, Ishii Y. Statins Show Anti-Atherosclerotic Effects by Improving Endothelial Cell Function in a Kawasaki Disease-like Vasculitis Mouse Model. Int J Mol Sci 2022; 23:ijms232416108. [PMID: 36555746 PMCID: PMC9780952 DOI: 10.3390/ijms232416108] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Kawasaki disease (KD) is an acute inflammatory syndrome of unknown etiology that is complicated by cardiovascular sequelae. Chronic inflammation (vasculitis) due to KD might cause vascular cellular senescence and vascular endothelial cell damage, and is a potential cause of atherosclerosis in young adults. This study examined the effect of KD and HMG-CoA inhibitors (statins) on vascular cellular senescence and vascular endothelial cells. Candida albicans water-soluble fraction (CAWS) was administered intraperitoneally to 5-week-old male apolipoprotein E-deficient (ApoE-) mice to induce KD-like vasculitis. The mice were then divided into three groups: control, CAWS, and CAWS+statin groups. Ten weeks after injection, the mice were sacrificed and whole aortic tissue specimens were collected. Endothelial nitric oxide synthase (eNOS) expression in the ascending aortic intima epithelium was evaluated using immunostaining. In addition, eNOS expression and levels of cellular senescence markers were measured in RNA and proteins extracted from whole aortic tissue. KD-like vasculitis impaired vascular endothelial cells that produce eNOS, which maintains vascular homeostasis, and promoted macrophage infiltration into the tissue. Statins also restored vascular endothelial cell function by promoting eNOS expression. Statins may be used to prevent secondary cardiovascular events during the chronic phase of KD.
Collapse
Affiliation(s)
- Yusuke Motoji
- Department of Cardiovascular Surgery, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Ryuji Fukazawa
- Department of Pediatrics, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
- Correspondence: ; Tel.: +81-3-3822-2131
| | - Ryosuke Matsui
- Department of Pediatrics, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yoshinori Abe
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Ikuno Uehara
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Makoto Watanabe
- Department of Pediatrics, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yoshiaki Hashimoto
- Department of Pediatrics, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yasuo Miyagi
- Department of Cardiovascular Surgery, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Noriko Nagi-Miura
- Laboratory for Immunopharmacology of Microbial Products, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan
| | - Nobuyuki Tanaka
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| | - Yosuke Ishii
- Department of Cardiovascular Surgery, Nippon Medical School Hospital, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8603, Japan
| |
Collapse
|
30
|
Gastric Cancer Cell-Derived Exosomal GRP78 Enhances Angiogenesis upon Stimulation of Vascular Endothelial Cells. Curr Issues Mol Biol 2022; 44:6145-6157. [PMID: 36547080 PMCID: PMC9776843 DOI: 10.3390/cimb44120419] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Exosomes containing glucose-regulated protein 78 (GRP78) are involved in cancer malignancy. GRP78 is thought to promote the tumor microenvironment, leading to angiogenesis. No direct evidence for this role has been reported, however, mainly because of difficulties in accurately measuring the GRP78 concentration in the exosomes. Recently, exosomal GRP78 concentrations were successfully measured using an ultrasensitive ELISA. In the present study, GRP78 concentrations in exosomes collected from gastric cancer AGS cells with overexpression of GRP78 (OE), knockdown of GRP78 (KD), or mock GRP78 (mock) were quantified. These three types of exosomes were then incubated with vascular endothelial cells to examine their effects on endothelial cell angiogenesis. Based on the results of a tube formation assay, GRP78-OE exosomes accelerated angiogenesis compared with GRP78-KD or GRP78-mock exosomes. To investigate the mechanisms underlying this effect, we examined the Ser473 phosphorylation state ratio of AKT, which is involved in the angiogenesis process, and found that AKT phosphorylation was increased by GRP78-OE exosome application to the endothelial cells. An MTT assay showed that GRP78-OE exosome treatment increased the proliferation rate of endothelial cells, and a wound healing assay showed that this treatment increased the migration capacity of the endothelial cells. These findings demonstrated that GRP78-containing exosomes promote the tumor microenvironment and induce angiogenesis.
Collapse
|
31
|
[STE029 Overcomes EGFR-TKI Resistance in Human Lung Adenocarcinoma]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:771-781. [PMID: 36419390 PMCID: PMC9720680 DOI: 10.3779/j.issn.1009-3419.2022.102.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Acquired and primary resistance to epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) is still the bottleneck of clinical treatment of advanced non-small cell lung cancer (NSCLC). STE029 is a novel anticancer drug which consists of 3-hydroxy-3-methylglutarylcoenzyme A reductase (HMGCR) inhibitor and novel cancer cell membrane targeting molecular. This study aimed to investigate the reversal mechanism of EGFR-TKI resistance by STE029 in lung adenocarcinoma. METHODS CCK8 test was used to test the cell viability and survival rate of EGFR mutated PC9 cell (Gefitinib sensitive), PC9/BB4 cell (acquired Gefitinib resistant), and EGFR wild type A549 cell after treatment of STE029, Gefitinib or combination of both. EdU test was applied to detect changes in cell cycle and Hoechst 33258 was applied to detect apoptosis rate in overcoming the EGFR-TKI resistance. The activity of EGFR/PI3K/Akt, cell cycle and apoptosis signal pathways were examined. In vivo, nude mice were exposed to STE029, Gefitinib and STE029+Gefitinib for 5 wk. And the the tumor volume was measured and tumor weight was obtained on the last day. RESULTS (1) PC9 cells was highly sensitive to Gefitinib, while PC9/BB4 and A549 cell showed significant resistance to Gefitinib treatment; (2) STE029+Gefitinib treatment could significantly decrease the 50% inhibitory concentrarion (IC₅₀) of Gefitinib in PC9, PC9/BB4 and A549 cells (P<0.05, respectively); (3) In PC9 and PC9/BB4 cells, STE029+Gefitinib can block cell cycle and inhibit cell proliferation (P<0.001), while there was no significant difference in apoptosis rate among three drug intervention groups (P>0.05); However, apoptosis rate was increased in STE029+Gefitinib group in A549 cell (P<0.01), while no significance detected in cell proliferation (P>0.05). (4) In PC9 and PC9/BB4 cells, the combination of STE029 and Gefitinib could downregulate p-EGFR, p-Akt, p-Cyclin D1 and Cyclin D1 (P<0.001), and upregulate the expression of GSK-3β (P<0.001), and the expression of cleaved caspase-8, caspase-8 cleaved caspase-9, caspase-9 showed no difference among groups (P>0.05). In A549 cells, the combination of STE029 and Gefitinib could downregulate p-Akt (P<0.001) and upregulate cleaved caspase-8 and cleaved caspase-9 (P<0.001); (5)In vivo, the combination of STE029 and Gefitinib effectively inhibited tumor development and progression compared to STE029 alone or Gefitinib alone, with significant difference (P<0.05) in PC9 and PC9/BB4 xenografted tumor. CONCLUSIONS STE029 could sensitize Gefitinib by inhibiting EGFR/PI3K/Akt pathway, blocking the tumor cell cycle and proliferation and inducing apoptosis through caspase-8 and caspase-9 dependent pathway. STE029 deserves further investigations in overcoming EGFR-TKI resistance in lung cancer.
Collapse
|
32
|
Zahedipour F, Butler AE, Eid AH, Sahebkar A. Pleiotropic properties of statins via angiogenesis modulation in cardiovascular disease. Drug Discov Today 2022; 27:103325. [PMID: 35872297 DOI: 10.1016/j.drudis.2022.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Accepted: 07/19/2022] [Indexed: 12/15/2022]
Abstract
Inhibition of hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase by statins is affected by inhibiting the active site of the enzyme in a competitive manner. Statins reduce plasma cholesterol by inhibiting its de novo synthesis. In addition, statins impart 'pleiotropic' activities that do not directly relate to their ability to decrease cholesterol. The proangiogenic and antiangiogenic characteristics of statins are among these pleiotropic effects. These angiogenic-modifying properties could offer new therapeutic applications. Statins stimulate or suppress angiogenesis in a biphasic manner. Whereas low doses of statin stimulate angiogenesis, high doses reduce protein prenylation and limit cell development and angiogenesis. In this review, we discuss how statins impact angiogenesis, with a particular focus on angiogenesis in stroke and cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, Adliya, Bahrain
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Australia, Perth, WA, Australia; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
33
|
Statins and angiogenesis in non-cardiovascular diseases. Drug Discov Today 2022; 27:103320. [PMID: 35850434 DOI: 10.1016/j.drudis.2022.07.005] [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: 04/26/2022] [Revised: 06/06/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022]
Abstract
Statins inhibit HMG-CoA reductase by competitively inhibiting the active site of the enzyme, thus preventing cholesterol synthesis and reducing the risk of developing cardiovascular disease. Many pleiotropic effects of statins have been demonstrated that can be either related or unrelated to their cholesterol-lowering ability. Among these effects are their proangiogenic and antiangiogenic properties that could offer new therapeutic applications. In this regard, pro- and anti-angiogenic properties of statins have been shown to be dose dependent. Statins also appear to have a variety of non-cardiovascular angiogenic effects in many diseases, some examples being ocular disease, brain disease, cancer, preeclampsia, diabetes and bone disease, which are discussed in this review using reports from in vitro and in vivo investigations.
Collapse
|
34
|
Izoe Y, Nagao M, Sato K, Sakai A, Ando K, Kanai M, Yamamoto A, Sakai S, Chida K. Dynamic coronary CT Angiography-Estimated coronary flow in Non-Obstructive, Plaque-free coronary Arteries: Association with dyslipidemia and diabetes. IJC HEART & VASCULATURE 2022; 42:101098. [PMID: 36032266 PMCID: PMC9399286 DOI: 10.1016/j.ijcha.2022.101098] [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: 05/21/2022] [Revised: 07/09/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022]
Abstract
Rationale and Objectives In this study, we implemented dynamic coronary CT angiography (CCTA) in order to estimate the coronary flow rate in morphologically normal coronary arteries as well as to identify factors affecting the coronary flow rate. Materials and Methods We retrospectively enrolled 95 consecutively presenting patients without stenosis or plaque in their major coronary arteries on CCTA conducted with a 320-detector scanner (mean age, 57 years; 43 % men). Time-attenuation curves of the distal sites of the major coronary arteries and the aortic root were extracted from dynamic CCTA data. Coronary flow rate, an indicator of coronary blood flow, was calculated via a convolution-integration method integrating the two curves. Patients with dyslipidemia were divided according to the presence or absence of familial hypercholesterolemia (FH) as well as according to the receipt of statin therapy. Results We found that the coronary flow rate was statistically significantly lower in statin-naïve patients with dyslipidemia (n = 27, 0.56 ± 0.10) than in patients without dyslipidemia (n = 32, 0.64 ± 0.10, p = 0.0013). In FH (n = 26), the coronary flow rate was statistically significantly lower in statin-naïve patients (n = 7, 0.65 ± 0.08) than in those taking statins (n = 19, 0.72 ± 0.10, p = 0.0221). Coronary flow rate likewise exhibited a statistically significant negative correlation with hemoglobin A1c (Pearson r, −0.437; p = 0.0003), but no correlation with other coronary risk factors. The coronary flow rate was statistically significantly lower in patients with diabetes (n = 14, 0.55 ± 0.10) than in those without diabetes (n = 81, 0.61 ± 0.11, p = 0.0461). Conclusion We found a reduction in coronary flow rate in patients with statin-naive dyslipidemia and diabetes, even within morphologically normal coronary arteries.
Collapse
Affiliation(s)
- Yukako Izoe
- Graduate School of Medicine, Health Sciences, Division of Radiological Examination and Technology Tohoku University, Sendai City, Japan
| | - Michinobu Nagao
- Department of Diagnostic Imaging & Nuclear Medicine, Tokyo Women’s Medical University, Tokyo, Japan
- Corresponding author.
| | - Kayoko Sato
- Department of Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Akiko Sakai
- Department of Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kiyoe Ando
- Department of Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Miwa Kanai
- Department of Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Astushi Yamamoto
- Department of Diagnostic Imaging & Nuclear Medicine, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Cardiology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Shuji Sakai
- Department of Diagnostic Imaging & Nuclear Medicine, Tokyo Women’s Medical University, Tokyo, Japan
| | - Koichi Chida
- Graduate School of Medicine, Health Sciences, Division of Radiological Examination and Technology Tohoku University, Sendai City, Japan
| |
Collapse
|
35
|
Degrush E, Shazeeb MS, Drachman D, Vardar Z, Lindsay C, Gounis MJ, Henninger N. Cumulative effect of simvastatin, L-arginine, and tetrahydrobiopterin on cerebral blood flow and cognitive function in Alzheimer's disease. Alzheimers Res Ther 2022; 14:134. [PMID: 36115980 PMCID: PMC9482313 DOI: 10.1186/s13195-022-01076-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVES Vascular disease is a known risk factor for Alzheimer's disease (AD). Endothelial dysfunction has been linked to reduced cerebral blood flow. Endothelial nitric oxide synthase pathway (eNOS) upregulation is known to support endothelial health. This single-center, proof-of-concept study tested whether the use of three medications known to augment the eNOS pathway activity improves cognition and cerebral blood flow (CBF). METHODS Subjects with mild AD or mild cognitive impairment (MCI) were sequentially treated with the HMG-CoA reductase synthesis inhibitor simvastatin (weeks 0-16), L-arginine (weeks 4-16), and tetrahydrobiopterin (weeks 8-16). The primary outcome of interest was the change in CBF as measured by MRI from baseline to week 16. Secondary outcomes included standard assessments of cognition. RESULTS A total of 11 subjects were deemed eligible and enrolled. One subject withdrew from the study after enrollment, leaving 10 subjects for data analysis. There was a significant increase in CBF from baseline to week 8 by ~13% in the limbic and ~15% in the cerebral cortex. Secondary outcomes indicated a modest but significant increase in the MMSE from baseline (24.2±3.2) to week 16 (26.0±2.7). Exploratory analysis indicated that subjects with cognitive improvement (reduction of the ADAS-cog 13) had a significant increase in their respective limbic and cortical CBF. CONCLUSIONS Treatment of mild AD/MCI subjects with medications shown to augment the eNOS pathway was well tolerated and associated with modestly increased cerebral blood flow and cognitive improvement. TRIAL REGISTRATION This study is registered in https://www. CLINICALTRIALS gov ; registration identifier: NCT01439555; date of registration submitted to registry: 09/23/2011; date of first subject enrollment: 11/2011.
Collapse
Affiliation(s)
- Elizabeth Degrush
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA.
- Department of Psychiatry, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA.
| | - Mohammed Salman Shazeeb
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - David Drachman
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA
| | - Zeynep Vardar
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Clifford Lindsay
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Matthew J Gounis
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- New England Center for Stroke Research, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Nils Henninger
- Department of Neurology, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA
- Department of Psychiatry, University of Massachusetts Chan Medical School, 55 Lake Ave, North, Worcester, MA, 01655, USA
| |
Collapse
|
36
|
Kang YR, Kim JT, Lee JS, Kim BJ, Kang K, Lee SJ, Kim JG, Cha JK, Kim DH, Park TH, Lee KB, Lee J, Hong KS, Cho YJ, Park HK, Lee BC, Yu KH, Oh MS, Kim DE, Ryu WS, Choi JC, Kwon JH, Kim WJ, Shin DI, Sohn SI, Hong JH, Park MS, Choi KH, Cho KH, Park JM, Lee SH, Lee J, Bae HJ. Differential influences of LDL cholesterol on functional outcomes after intravenous thrombolysis according to prestroke statin use. Sci Rep 2022; 12:15478. [PMID: 36104394 PMCID: PMC9474509 DOI: 10.1038/s41598-022-19852-8] [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: 12/08/2021] [Accepted: 09/06/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractThis study aimed to elucidate whether low-density lipoprotein cholesterol (LDL-C) levels differentially affect functional outcomes after intravenous thrombolysis (IVT) depending on prestroke statin use. Patients with acute ischemic stroke treated with IVT were categorized into low, intermediate, and high LDL-C groups based on LDL-C levels at admission (< 100/100–130/ > 130 mg/dl, respectively). Multivariable logistic regression analyses were performed to explore the relationships between LDL-C and clinical outcomes (good outcomes at 3 months, modified Rankin Scale scores 0–2). The interaction between LDL-C levels and prestroke statin use regarding functional outcomes was investigated. Among the 4711 patients (age, 67 ± 12 years; males, 62.1%) who met the eligibility criteria, compared with the high LDL-C group, the low and intermediate LDL-C groups were not associated with good outcomes at 3 months according to the multivariable analysis. A potential interaction between the LDL-C group and prestroke statin use on good outcomes at 3 months was observed (Pinteraction = 0.07). Among patients with prestroke statin use, low (aOR 1.84 [1.04–3.26]) and intermediate (aOR 2.31 [1.20–4.47]) LDL-C groups were independently associated with a greater likelihood of having a 3-month good outcome. Our study showed that LDL-C was not associated with a 3-month good outcome, but prestroke statin use could modify the influence of LDL-C levels on functional outcomes after IVT.
Collapse
|
37
|
de Paula LJC, Uchida AH, Rezende PC, Soares P, Scudeler TL. Protective or Inhibitory Effect of Pharmacological Therapy on Cardiac Ischemic Preconditioning: A Literature Review. Curr Vasc Pharmacol 2022; 20:409-428. [PMID: 35986546 DOI: 10.2174/1570161120666220819163025] [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: 03/21/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 01/25/2023]
Abstract
Ischemic preconditioning (IP) is an innate phenomenon, triggered by brief, non-lethal cycles of ischemia/reperfusion applied to a tissue or organ that confers tolerance to a subsequent more prolonged ischemic event. Once started, it can reduce the severity of myocardial ischemia associated with some clinical situations, such as percutaneous coronary intervention (PCI) and intermittent aortic clamping during coronary artery bypass graft surgery (CABG). Although the mechanisms underlying IP have not been completely elucidated, several studies have shown that this phenomenon involves the participation of cell triggers, intracellular signaling pathways, and end-effectors. Understanding this mechanism enables the development of preconditioning mimetic agents. It is known that a range of medications that activate the signaling cascades at different cellular levels can interfere with both the stimulation and the blockade of IP. Investigations of signaling pathways underlying ischemic conditioning have identified a number of therapeutic targets for pharmacological manipulation. This review aims to present and discuss the effects of several medications on myocardial IP.
Collapse
Affiliation(s)
| | | | - Paulo Cury Rezende
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Paulo Soares
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Thiago Luis Scudeler
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
HMG-CoA Reductase Inhibitor Statins Activate the Transcriptional Activity of p53 by Regulating the Expression of TAZ. Pharmaceuticals (Basel) 2022; 15:ph15081015. [PMID: 36015162 PMCID: PMC9412369 DOI: 10.3390/ph15081015] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 12/18/2022] Open
Abstract
Transcriptional coactivator with PDZ-binding motif (TAZ) is a downstream transcriptional regulator of the Hippo pathway that controls cell growth and differentiation. The aberrant activation of TAZ correlates with a poor prognosis in human cancers, such as breast and colon cancers. We previously demonstrated that TAZ inhibited the tumor suppressor functions of p53 and enhanced cell proliferation. Statins, which are used to treat dyslipidemia, have been reported to suppress the activity of TAZ and exert anti-tumor effects. In the present study, we focused on the regulation of p53 functions by TAZ and investigated whether statins modulate these functions via TAZ. The results obtained suggest that statins, such as simvastatin and fluvastatin, activated the transcriptional function of p53 by suppressing TAZ protein expression. Furthermore, co-treatment with simvastatin and anti-tumor agents that cooperatively activate p53 suppressed cancer cell survival. These results indicate a useful mechanism by which statins enhance the effects of anti-tumor agents through the activation of p53 and may represent a novel approach to cancer therapy.
Collapse
|
39
|
Seidi F, Zhong Y, Xiao H, Jin Y, Crespy D. Degradable polyprodrugs: design and therapeutic efficiency. Chem Soc Rev 2022; 51:6652-6703. [PMID: 35796314 DOI: 10.1039/d2cs00099g] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Prodrugs are developed to increase the therapeutic properties of drugs and reduce their side effects. Polyprodrugs emerged as highly efficient prodrugs produced by the polymerization of one or several drug monomers. Polyprodrugs can be gradually degraded to release therapeutic agents. The complete degradation of polyprodrugs is an important factor to guarantee the successful disposal of the drug delivery system from the body. The degradation of polyprodrugs and release rate of the drugs can be controlled by the type of covalent bonds linking the monomer drug units in the polymer structure. Therefore, various types of polyprodrugs have been developed based on polyesters, polyanhydrides, polycarbonates, polyurethanes, polyamides, polyketals, polymetallodrugs, polyphosphazenes, and polyimines. Furthermore, the presence of stimuli-responsive groups, such as redox-responsive linkages (disulfide, boronate ester, metal-complex, and oxalate), pH-responsive linkages (ester, imine, hydrazone, acetal, orthoester, P-O and P-N), light-responsive (metal-complex, o-nitrophenyl groups) and enzyme-responsive linkages (ester, peptides) allow for a selective degradation of the polymer backbone in targeted tumors. We envision that new strategies providing a more efficient synergistic therapy will be developed by combining polyprodrugs with gene delivery segments and targeting moieties.
Collapse
Affiliation(s)
- Farzad Seidi
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China. .,Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| | - Yajie Zhong
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China.
| | - Huining Xiao
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| | - Yongcan Jin
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China.
| | - Daniel Crespy
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| |
Collapse
|
40
|
Damay VA, Setiawan S, Lesmana R, Akbar MR, Lukito AA. Effects of Moderate Intensity Aerobic Exercise to FSTL-1 Regulation in Atherosclerosis: A Systematic Review. Int J Angiol 2022; 32:1-10. [PMID: 36727145 PMCID: PMC9886452 DOI: 10.1055/s-0042-1750184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Moderate intensity exercise is considered as a primary step to prevent coronary artery diseases (CADs) by stimulated FSTL-1 secretion as a novel myokines to improve endothelial cell function, prevent arterial stiffness, or vascular inflammation. This review aims to provide the current evident role of FSTL-1 as a novel myokine secreted during exercise to prevent atherosclerosis progression. A systematic review using databases from (PubMed), ScienceDirect, and The Cochrane Library, was conducted up to October 2021 to identify all the eligible experimental and observational studies that assess how moderate intensity exercises stimulate FSTL-1 secretion to prevent atherosclerosis. Results were described through narrative synthesis of the evidence. From 84 retrieved references, 15 studies met the inclusion criteria and were included in this review. The overall results suggest that exercise or physical activity can stimulate myokines secretion, especially in FSTL-1. FSTL-1 is a myokine or adipokine that plays a potential role in preventing atherosclerosis by various mechanisms such as via improvement of endothelial functions, suppression of smooth muscle cells (SMCs) proliferation, and reduction of arterial thickening. FSTL-1 is a relatively new and less known myokine, but probably holds a key role in assessing how moderate intensity aerobic exercise prevents atherosclerosis progression by preventing endothelial dysfunction, arterial stiffness, or vascular inflammation.
Collapse
Affiliation(s)
- Vito Anggarino Damay
- Department of Cardiovascular Medicine, Universitas Pelita Harapan, Banten, Indonesia,Address for correspondence Vito A. Damay, MD, Cardiologist Department of Cardiovascular Medicine, Universitas Pelita HarapanBantenIndonesia
| | - Setiawan Setiawan
- Department of Biomedical Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Ronny Lesmana
- Department of Biomedical Sciences, Universitas Padjadjaran, Bandung, Indonesia
| | - Muhammad Rizki Akbar
- Department of Cardiology and Vascular, Padjadjaran University, Bandung, Indonesia
| | - Antonia Anna Lukito
- Department of Cardiovascular Medicine, Universitas Pelita Harapan, Banten, Indonesia
| |
Collapse
|
41
|
Pharmacological prevention of intimal hyperplasia: A state-of-the-art review. Pharmacol Ther 2022; 235:108157. [PMID: 35183591 DOI: 10.1016/j.pharmthera.2022.108157] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/04/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023]
Abstract
Intimal hyperplasia (IH) occurs in a considerable number of cases of blood vessel reconstruction by stenting or balloon angioplasty, venous bypass grafting, and arteriovenous dialysis accesses. It is triggered by endothelial injury during the vascular intervention and leads to vessel restenosis with life-threatening consequences for patients. To date, the drugs used for IH prevention in clinics-paclitaxel and rapalog drugs-have been focusing primarily on the vascular smooth muscle cell (VSMC) proliferation pathway of IH development. Limitations, such as endothelial toxicity and inappropriate drug administration timing, have spurred the search for new and efficient pharmacological approaches to control IH. In this state-of-the-art review, we present the pathways of IH development, focusing on the key events and actors involved in IH. Subsequently, we discuss different drugs and drug combinations interfering with these pathways based on their effect on peripheral circulation IH models in animal studies, or on clinical reports. The reports were obtained through an extensive search of peer-reviewed publications in Pubmed, Embase, and Google Scholar, with search equations composed based on five concepts around IH and their various combinations. To improve vascular intervention outcomes, rethinking of conventional therapeutic approaches to IH prevention is needed. Exploring local application of drugs and drug combinations acting on different pathophysiological pathways of IH development has the potential to provide effective and safe restenosis prevention.
Collapse
|
42
|
Han J, Luo L, Marcelina O, Kasim V, Wu S. Therapeutic angiogenesis-based strategy for peripheral artery disease. Theranostics 2022; 12:5015-5033. [PMID: 35836800 PMCID: PMC9274744 DOI: 10.7150/thno.74785] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/14/2022] [Indexed: 01/12/2023] Open
Abstract
Peripheral artery disease (PAD) poses a great challenge to society, with a growing prevalence in the upcoming years. Patients in the severe stages of PAD are prone to amputation and death, leading to poor quality of life and a great socioeconomic burden. Furthermore, PAD is one of the major complications of diabetic patients, who have higher risk to develop critical limb ischemia, the most severe manifestation of PAD, and thus have a poor prognosis. Hence, there is an urgent need to develop an effective therapeutic strategy to treat this disease. Therapeutic angiogenesis has raised concerns for more than two decades as a potential strategy for treating PAD, especially in patients without option for surgery-based therapies. Since the discovery of gene-based therapy for therapeutic angiogenesis, several approaches have been developed, including cell-, protein-, and small molecule drug-based therapeutic strategies, some of which have progressed into the clinical trial phase. Despite its promising potential, efforts are still needed to improve the efficacy of this strategy, reduce its cost, and promote its worldwide application. In this review, we highlight the current progress of therapeutic angiogenesis and the issues that need to be overcome prior to its clinical application.
Collapse
Affiliation(s)
- Jingxuan Han
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China
| | - Lailiu Luo
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China
| | - Olivia Marcelina
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China
| | - Vivi Kasim
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.,✉ Corresponding authors: Vivi Kasim, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65112672, Fax: +86-23-65111802, ; Shourong Wu, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65111632, Fax: +86-23-65111802,
| | - Shourong Wu
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing 400044, China.,The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.,✉ Corresponding authors: Vivi Kasim, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65112672, Fax: +86-23-65111802, ; Shourong Wu, College of Bioengineering, Chongqing University, Chongqing, China; Phone: +86-23-65111632, Fax: +86-23-65111802,
| |
Collapse
|
43
|
The role of protein kinases as key drivers of metabolic dysfunction-associated fatty liver disease progression: New insights and future directions. Life Sci 2022; 305:120732. [PMID: 35760093 DOI: 10.1016/j.lfs.2022.120732] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD), proposed in 2020 is a novel term for non-alcoholic fatty liver disease (NAFLD) which was coined for the first time in 1980. It is a leading cause of the most chronic liver disease and hepatic failure all over the world, and unfortunately, with no licensed drugs for treatment yet. The progress of the disease is driven by the triggered inflammatory process, oxidative stress, and insulin resistance in many pathways, starting with simple hepatic steatosis to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and liver cancer. Protein kinases (PKs), such as MAPK, ErbB, PKC, PI3K/Akt, and mTOR, govern most of the pathological pathways by acting on various downstream key points in MAFLD and regulating both hepatic gluco- lipo-neogenesis and inflammation. Therefore, modulating the function of those potential protein kinases that are effectively involved in MAFLD might be a promising therapeutic approach for tackling this disease. In the current review, we have discussed the key role of protein kinases in the pathogenesis of MAFLD and performed a protein-protein interaction (PPI) network among the main proteins of each kinase pathway with MAFLD-related proteins to predict the most likely targets of the PKs in MAFLD. Moreover, we have reported the experimental, pre-clinical, and clinical data for the most recent investigated molecules that are activating p38-MAPK and AMPK proteins and inhibiting the other PKs to improve MAFLD condition by regulating oxidation and inflammation signalling.
Collapse
|
44
|
Elseweidy MM, Ali SI, Shershir NI, Ali AEA, Hammad SK. Vitamin D3 intake as modulator for the early biomarkers of myocardial tissue injury in diabetic hyperlipidaemic rats. Arch Physiol Biochem 2022; 128:628-636. [PMID: 32046509 DOI: 10.1080/13813455.2020.1716015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CONTEXT Myocardial cell death occurs within hours following the onset of myocardial ischaemia and its chief cause is atherosclerosis. There is a link between vitamin D3 deficiency and many cardiovascular risk factors. OBJECTIVE This study compared the effect of vitamin D3 on early biomarkers of myocardial injury, to that of atorvastatin. METHODS Diabetic hyperlipidaemia was induced in Wistar rats, which were divided into 3 groups: diabetic hyperlipidaemic control, diabetic hyperlipidaemic rats treated with atorvastatin and diabetic hyperlipidaemic rats treated with vitamin D3. Blood glucose, glycated haemoglobin and lipid profile were evaluated. Markers of myocardial injury were examined including cardiac troponin, heart fatty acid binding protein (HFABP) and C-terminal pro-endothelin-1 (CT-pro-ET-1). RESULTS Vitamin D3 and atorvastatin intake improved lipid profile and glucose homeostasis, and reduced levels of predictive biomarkers of myocardial injury. CONCLUSION Vitamin D3 can be used in a suitable dose as a safe and protective candidate against myocardial injury.
Collapse
Affiliation(s)
- Mohamed M Elseweidy
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Sousou I Ali
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Noura I Shershir
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Abd Elmonem A Ali
- Pathology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Sally K Hammad
- Biochemistry Department, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
45
|
Handley EL, Callanan A. Modulation of Tissue Microenvironment Following Myocardial Infarction. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Ella Louise Handley
- Institute for Bioengineering School of Engineering University of Edinburgh Edinburgh EH9 3DW UK
| | - Anthony Callanan
- Institute for Bioengineering School of Engineering University of Edinburgh Edinburgh EH9 3DW UK
| |
Collapse
|
46
|
Matsumoto T, Yoshino S, Furuyama T, Morisaki K, Nakano K, Koga JI, Maehara Y, Komori K, Mori M, Egashira K. Pitavastatin-Incorporated Nanoparticles for Chronic Limb Threatening Ischemia: A Phase I/IIa Clinical Trial. J Atheroscler Thromb 2022; 29:731-746. [PMID: 33907060 PMCID: PMC9135659 DOI: 10.5551/jat.58941] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/28/2021] [Indexed: 12/02/2022] Open
Abstract
AIM To assess the results of a phase I/IIa open-label dose-escalation clinical trial of 5-day repeated intramuscular administration of pitavastatin-incorporated poly (lactic-co-glycolic acid) nanoparticles (NK-104-NP) in patients with chronic limb threatening ischemia (CLTI). METHODS NK-104-NP was formulated using an emulsion solvent diffusion method. NK-104-NP at four doses (nanoparticles containing 0.5, 1, 2, and 4 mg of pitavastatin calcium, n=4 patients per dose) was investigated in a dose-escalation manner and administered intramuscularly into the ischemic limbs of 16 patients with CLTI. The safety and therapeutic efficacy of treatment were investigated over a 26-week follow-up period. RESULTS No cardiovascular or other serious adverse events caused by NK-104-NP were detected during the follow-up period. Improvements in Fontaine and Rutherford classifications were noted in five patients (one, three, and one in the 1-, 2-, and 4-mg dose groups, respectively). Pharmacokinetic parameters including the maximum serum concentration and the area under the blood concentration-time curve increased with pitavastatin treatment in a dose-dependent manner. The area under the curve was slightly increased at day 5 compared with that at day 1 of treatment, although the difference was not statistically significant. CONCLUSIONS This is the first clinical trial of pitavastatin-incorporated nanoparticles in patients with CLTI. Intramuscular administration of NK-104-NP to the ischemic limbs of patients with CLTI was safe and well tolerated and resulted in improvements in limb function.
Collapse
Affiliation(s)
- Takuya Matsumoto
- Department of Vascular Surgery, National Hospital Organization Fukuoka-higashi Medical Center, Fukuoka, Japan
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinichiro Yoshino
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tadashi Furuyama
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Morisaki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kaku Nakano
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Disruptive Cardiovascular Innovation, Kyushu University, Fukuoka, Japan
| | - Jun-ichiro Koga
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Disruptive Cardiovascular Innovation, Kyushu University, Fukuoka, Japan
- Department of Cardiovascular Medicine, Kyusyu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kimihiro Komori
- Division of Vascular Surgery, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kensuke Egashira
- Department of Cardiovascular Research, Development, and Translational Medicine, Center for Disruptive Cardiovascular Innovation, Kyushu University, Fukuoka, Japan
- Department of Translational Medicine, Kyushu University Graduate School of Pharmaceutical Sciences, Fukuoka, Japan
| |
Collapse
|
47
|
HIF1A promotes miR-210/miR-424 transcription to modulate the angiogenesis in HUVECs and HDMECs via sFLT1 under hypoxic stress. Mol Cell Biochem 2022; 477:2107-2119. [PMID: 35488146 DOI: 10.1007/s11010-022-04428-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/30/2022] [Indexed: 01/08/2023]
Abstract
Angiogenesis is a critical process during human skin wound healing. However, hypoxia might lead to the dysfunction of the cellular interplay of endothelial cells and subcutaneous fibroblasts, resulting in the deregulation of angiogenesis. HIF1A is a key regulatory of the recovery of intracellular homeostasis under hypoxia. In the present study, the detailed role and mechanism of HIF1A in the angiogenesis under hypoxia were investigated. Via bioinformatic analyses on microarray profiles (GSE1041 and GSE17944), solube fms-related tyrosine kinase 1 (sFLT1, also known as sVEGFR1) and miR-210/miR-424 might be involved in HIF1A function on the angiogenesis under hypoxia in human umbilical vascular endothelium cells (HUVECs) and human dermal microvascular endothelial cells (HDMECs). In the present study, we identified sFLT1 as a downregulated gene in response to hypoxia and HIF1A overexpression in HUVECs and HDMECs. sFLT1 overexpression inhibited the capacity of migration and angiogenesis and significantly reversed the inducible effects of HIF1A on the migration and angiogenesis in both cell lines. miR-210 and miR-424 were upregulated by hypoxia and targeted sFLT1 3'-UTR to negatively modulate its expression. HIF1A modulated sFLT1 expression, VEGF signaling, and the migration and angiogenesis in HUVECs and HDMECs via miR-210/miR-424. Regarding the molecular mechanism, HIF1A bound the promoter region of miR-210 and miR-424 to activate their transcription, while miR-210/miR-424 bound sFLT1 3'-UTR to suppress its expression. In summary, HIF1A/miR-210/miR-424/sFLT1 axis modulates the angiogenesis in HUVECs and HDMECs upon hypoxic condition via VEGF signaling.
Collapse
|
48
|
le Noble F, Kupatt C. Interdependence of Angiogenesis and Arteriogenesis in Development and Disease. Int J Mol Sci 2022; 23:ijms23073879. [PMID: 35409246 PMCID: PMC8999596 DOI: 10.3390/ijms23073879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 02/04/2023] Open
Abstract
The structure of arterial networks is optimized to allow efficient flow delivery to metabolically active tissues. Optimization of flow delivery is a continuous process involving synchronization of the structure and function of the microcirculation with the upstream arterial network. Risk factors for ischemic cardiovascular diseases, such as diabetes mellitus and hyperlipidemia, adversely affect endothelial function, induce capillary regression, and disrupt the micro- to macrocirculation cross-talk. We provide evidence showing that this loss of synchronization reduces arterial collateral network recruitment upon arterial stenosis, and the long-term clinical outcome of current revascularization strategies in these patient cohorts. We describe mechanisms and signals contributing to synchronized growth of micro- and macrocirculation in development and upon ischemic challenges in the adult organism and identify potential therapeutic targets. We conclude that a long-term successful revascularization strategy should aim at both removing obstructions in the proximal part of the arterial tree and restoring “bottom-up” vascular communication.
Collapse
Affiliation(s)
- Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131 Karlsruhe, Germany
- Institute for Biological and Chemical Systems—Biological Information Processing, Karlsruhe Institute of Technology (KIT), P.O. Box 3640, 76021 Karlsruhe, Germany
- Institute of Experimental Cardiology, Heidelberg Germany and German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, University of Heidelberg, 69117 Heidelberg, Germany
- Correspondence: (F.l.N.); (C.K.)
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
- DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, 80802 Munich, Germany
- Correspondence: (F.l.N.); (C.K.)
| |
Collapse
|
49
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
50
|
Crosstalk between Statins and Cancer Prevention and Therapy: An Update. Pharmaceuticals (Basel) 2021; 14:ph14121220. [PMID: 34959621 PMCID: PMC8704600 DOI: 10.3390/ph14121220] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
The importance of statins in cancer has been discussed in many studies. They are known for their anticancer properties against solid tumors of the liver or lung, as well as diffuse cancers, such as multiple myeloma or leukemia. Currently, the most commonly used statins are simvastatin, rosuvastatin and atorvastatin. The anti-tumor activity of statins is largely related to their ability to induce apoptosis by targeting cancer cells with high selectivity. Statins are also involved in the regulation of the histone acetylation level, the disturbance of which can lead to abnormal activity of genes involved in the regulation of proliferation, differentiation and apoptosis. As a result, tumor growth and its invasion may be promoted, which is associated with a poor prognosis. High levels of histone deacetylases are observed in many cancers; therefore, one of the therapeutic strategies is to use their inhibitors. Combining statins with histone deacetylase inhibitors can induce a synergistic anticancer effect.
Collapse
|