1
|
X S, Aitken C, Mehta V, Tardajos-Ayllon B, Serbanovic-Canic J, Zhu J, Miao B, Tzima E, Evans P, Fang Y, Schwartz MA. Controversy in mechanotransduction - the role of endothelial cell-cell junctions in fluid shear stress sensing. J Cell Sci 2024; 137:jcs262348. [PMID: 39143856 PMCID: PMC11423816 DOI: 10.1242/jcs.262348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
Fluid shear stress (FSS) from blood flow, sensed by the vascular endothelial cells (ECs) that line all blood vessels, regulates vascular development during embryogenesis, controls adult vascular physiology and determines the location of atherosclerotic plaque formation. Although a number of papers have reported a crucial role for cell-cell adhesions or adhesion receptors in these processes, a recent publication has challenged this paradigm, presenting evidence that ECs can very rapidly align in fluid flow as single cells without cell-cell contacts. To address this controversy, four independent laboratories assessed EC alignment in fluid flow across a range of EC cell types. These studies demonstrate a strict requirement for cell-cell contact in shear stress sensing over timescales consistent with previous literature and inconsistent with the newly published data.
Collapse
Affiliation(s)
- Shaka X
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA
| | - Claire Aitken
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Vedanta Mehta
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Blanca Tardajos-Ayllon
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | | | - Jiayu Zhu
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Bernadette Miao
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Ellie Tzima
- Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Paul Evans
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Yun Fang
- Department of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL 60637, USA
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA
- Departments of Internal Medicine (Cardiovascular Medicine) and Cell Biology, Yale School of Medicine, and Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
2
|
Alradwan I, AL Fayez N, Alomary MN, Alshehri AA, Aodah AH, Almughem FA, Alsulami KA, Aldossary AM, Alawad AO, Tawfik YMK, Tawfik EA. Emerging Trends and Innovations in the Treatment and Diagnosis of Atherosclerosis and Cardiovascular Disease: A Comprehensive Review towards Healthier Aging. Pharmaceutics 2024; 16:1037. [PMID: 39204382 PMCID: PMC11360443 DOI: 10.3390/pharmaceutics16081037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs) are classed as diseases of aging, which are associated with an increased prevalence of atherosclerotic lesion formation caused by such diseases and is considered as one of the leading causes of death globally, representing a severe health crisis affecting the heart and blood vessels. Atherosclerosis is described as a chronic condition that can lead to myocardial infarction, ischemic cardiomyopathy, stroke, and peripheral arterial disease and to date, most pharmacological therapies mainly aim to control risk factors in patients with cardiovascular disease. Advances in transformative therapies and imaging diagnostics agents could shape the clinical applications of such approaches, including nanomedicine, biomaterials, immunotherapy, cell therapy, and gene therapy, which are emerging and likely to significantly impact CVD management in the coming decade. This review summarizes the current anti-atherosclerotic therapies' major milestones, strengths, and limitations. It provides an overview of the recent discoveries and emerging technologies in nanomedicine, cell therapy, and gene and immune therapeutics that can revolutionize CVD clinical practice by steering it toward precision medicine. CVD-related clinical trials and promising pre-clinical strategies that would significantly impact patients with CVD are discussed. Here, we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD medicine.
Collapse
Affiliation(s)
- Ibrahim Alradwan
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Nojoud AL Fayez
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Mohammad N. Alomary
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Abdullah A. Alshehri
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Alhassan H. Aodah
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Fahad A. Almughem
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Khulud A. Alsulami
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Ahmad M. Aldossary
- Wellness and Preventative Medicine Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Abdullah O. Alawad
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Yahya M. K. Tawfik
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Essam A. Tawfik
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| |
Collapse
|
3
|
Iqbal MP, Azam I, Yousuf FA, Kazmi K. Relationship of adhesion molecules (ICAM-1 and E-selectin) with ABO blood groups in patients hospitalized with acute myocardial infarction. Pak J Med Sci 2024; 40:1551-1555. [PMID: 39092065 PMCID: PMC11255794 DOI: 10.12669/pjms.40.7.8978] [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: 10/19/2023] [Revised: 03/21/2024] [Accepted: 04/19/2024] [Indexed: 08/04/2024] Open
Abstract
Objectives Adhesion molecules, sICAM-1 and sE-selectin appear to have a major role in the pathogenesis of coronary artery disease (CAD). The focus of this study was to investigate the relationship of sICAM-1 and sE-selectin with ABO blood groups in Pakistani patients hospitalized with acute myocardial infarction (AMI). Methods In a case-control study, 116 patients of acute myocardial infarction (AMI) and 116 healthy controls (age range for both: 30 years to 70 years; both males and females) were randomly selected from the Aga Khan University and National Institute of Cardiovascular Diseases, Karachi with informed consent. The blood samples were obtained and analyzed for ABO blood groups and serum levels of sICAM-1 and sE-selectin using kit methods. Statistical tests including independent sample t-test and Two-way ANOVA were used to study the association of these adhesion molecules with blood groups in AMI patients and healthy controls. Duration of the study was from July 2021 to June 30, 2023. Results Mean serum levels of sICAM-1 were significantly higher in AMI patients compared to healthy controls (342±159 mg/dl vs. 227±104 mg/dl; p-value<0.001). Similarly, serum levels of sE-selectin were also significantly higher in AMI patients compared to healthy controls (53.6±26.9 mg/dl vs. 40.7± mg/dl; p-value<0.001). Moreover, mean concentrations of sICAM-1 and sE-selectin for the interaction between subject type (cases and control) and blood groups were statistically significant (p-value = 0.007 and p-value = 0.035, respectively). Conclusion There is an association of adhesion molecules, sICAM-1 and sE-selectin with ABO blood groups in Pakistani patients hospitalized with AMI.
Collapse
Affiliation(s)
- Mohammad Perwaiz Iqbal
- Mohammad Perwaiz Iqbal, Department of Life Sciences, University of Management and Technology, Lahore, Pakistan. Pakistan Academy of Sciences, Islamabad, Pakistan. Department of Biological & Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Iqbal Azam
- Iqbal Azam, Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | - Farzana Abubaker Yousuf
- Farzana Abubaker Yousuf, Department of Biological & Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Khawar Kazmi
- Khawar Kazmi, Department of Preventive Cardiology, National Institute of Cardiovascular Diseases, Karachi, Pakistan
| |
Collapse
|
4
|
Peng D, Zhuge F, Wang M, Zhang B, Zhuang Z, Zhou R, Zhang Y, Li J, Yu Z, Shi J. Morus alba L. (Sangzhi) alkaloids mitigate atherosclerosis by regulating M1/M2 macrophage polarization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155526. [PMID: 38564921 DOI: 10.1016/j.phymed.2024.155526] [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: 09/25/2023] [Revised: 01/01/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Atherosclerosis (AS) is an important cause of cardiovascular disease, posing a substantial health risk. Recognized as a chronic inflammatory disorder, AS hinges on the pivotal involvement of macrophages in arterial inflammation, participating in its formation and progression. Sangzhi alkaloid (SZ-A) is a novel natural alkaloid extracted from the mulberry branches, has extensive pharmacological effects and stable pharmacokinetic characteristics. However, the effects and mechanisms of SZ-A on AS remain unclear. PURPOSE To explore the effect and underlying mechanisms of SZ-A on inflammation mediated by macrophages and its role in AS development. METHODS Atherosclerosis was induced in vivo in apolipoprotein E-deficient mice through a high-fat and high-choline diet. We utilized macrophages and vascular endothelial cells to investigate the effects of SZ-A on macrophage polarization and its anti-inflammatory properties on endothelial cells in vitro. The transcriptomic analyses were used to investigate the major molecule that mediates cell-cell interactions and the antiatherogenic mechanisms of SZ-A based on AS, subsequently validated in vivo and in vitro. RESULTS SZ-A demonstrated a significant inhibition in vascular inflammation and alleviation of AS severity by mitigating macrophage infiltration and modulating M1/M2 macrophage polarization in vitro and in vivo. Moreover, SZ-A effectively reduced the release of the proinflammatory mediator C-X-C motif chemokine ligand (CXCL)-10, predominantly secreted by M1 macrophages. This reduction in CXCL-10 contributed to improved endothelial cell function, reduced recruitment of additional macrophages, and inhibited the inflammatory amplification effect. This ultimately led to the suppression of atherogenesis. CONCLUSION SZ-A exhibited potent anti-inflammatory effects by inhibiting macrophage-mediated inflammation, providing a new therapeutic avenue against AS. This is the first study demonstrating the efficacy of SZ-A in alleviating AS severity and offers novel insights into its anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Dandan Peng
- Department of Endocrinology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China; Guizhou Medical University, Guiyang, Guizhou, China
| | - Fen Zhuge
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Mingwei Wang
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Binbin Zhang
- Department of Infectious Diseases and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhenjie Zhuang
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Zhejiang, China
| | - Run Zhou
- College of Nursing, Hangzhou Normal University, Zhejiang, China
| | - Yuanyuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University, Zhejiang, China
| | - Jie Li
- Department of Infectious Disease, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Zhenqiu Yu
- Guizhou Medical University, Guiyang, Guizhou, China; The Department of Hypertension, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.
| | - Junping Shi
- Department of Infectious Diseases and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Medical Epigenetics, Hangzhou, Zhejiang, China; Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Qian Z, Shaofang F, Chen C, Chunhua S, Nan W, Chao L. IL-33 Suppresses the Progression of Atherosclerosis via the ERK1/2-IRF1-VCAM-1 Pathway. Cardiovasc Drugs Ther 2024; 38:569-580. [PMID: 37957490 DOI: 10.1007/s10557-023-07523-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 11/15/2023]
Abstract
PURPOSE This study was designed to explore the effects of interleukin 33 (IL-33) on the progression of atherosclerosis and the possible mechanism. METHODS The adhesion assay was performed on isolated peripheral blood mononuclear cells (PBMCs) and human umbilical vein endothelial cells (HUVEC). The expression of proteins and messenger RNA (mRNA) were detected by western blot and quantitative real-time polymerase chain reaction (PCR), including intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and P-selectin. The effect of IL-33 on the interaction of growth stimulation expressed gene 2 (ST2) with myeloid differentiation factor 88 (MyD88) and interleukin-1 receptor-associated kinase (IRAK) 1/4 were investigated using co-immunoprecipitation assay. An apolipoprotein (Apo) E-/- mice model was used to confirm the effect of IL-33 on atherosclerosis progression. Area of plaques was recorded by hematoxylin-eosin (H&E) staining. The severity of atherosclerosis plaque was evaluated using immunohistochemistry assay, and lipid accumulation was measured by an oil red O staining. In contrast, western blot was performed to detect the expression levels of VCAM-1, extracellular signal-regulated kinase (ERK) 1/2, and interferon regulatory factor 1 (IRF1). RESULTS Our study observed that IL-33 suppressed cell adhesion and the expression of VCAM-1 in tumor necrosis factor-α (TNF-α) exposed HUVEC. Moreover, the addition of IL-33 significantly inhibited the expression of IRF1 and the binding level of IRF1 to VCAM-1 and also promoted the phosphorylation level of IRAK1/4 and ERK1/2 compared to TNF-α-stimulated HUVEC. The ST2 neutralizing antibody or ERK pathway inhibitor SCH772984 reversed the regulatory effects of IL-33 on HUVEC, suggesting that IL-33 suppressed IRF1 and VCAM-1 dependent on binding to ST2 and activating the ERK1/2 signaling pathway. Further investigation in vivo confirmed that IL-33 decreased the expressions of IRF1 and VCAM-1 by activating the phosphorylation of ERK1/2 in the thoracic aorta of Apo E-/- mice. CONCLUSION In conclusion, our results demonstrated that IL-33 plays a protective role in the progression of atherosclerosis by inhibiting cell adhesion via the ERK1/2-IRF1-VCAM-1 pathway. This study may provide a potential therapeutic way to prevent the development of atherosclerosis.
Collapse
Affiliation(s)
- Zhang Qian
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, 68 Changle Rd, Nanjing, 210006, Jiangsu, China
| | - Feng Shaofang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China
| | - Chen Chen
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, 68 Changle Rd, Nanjing, 210006, Jiangsu, China
| | - Shi Chunhua
- Medical Department, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Wang Nan
- Jinling Hospital, Medical School of Nanjing University, 22 Hankou Rd, Nanjing, 210093, Jiangsu, China.
| | - Liu Chao
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, 68 Changle Rd, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
6
|
Park Y, Korzun T, Moses AS, Singh P, Levasseur PR, Demessie AA, Sharma KS, Morgan T, Raitmayr CJ, Avila U, Sabei FY, Taratula OR, Marks DL, Taratula O. Targeted Nanocarriers for Systemic Delivery of IRAK4 Inhibitors to Inflamed Tissues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306270. [PMID: 37702136 PMCID: PMC10840923 DOI: 10.1002/smll.202306270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/01/2023] [Indexed: 09/14/2023]
Abstract
Persistent and uncontrolled inflammation is the root cause of various debilitating diseases. Given that interleukin-1 receptor-associated kinase 4 (IRAK4) is a critical modulator of inflammation, inhibition of its activity with selective drug molecules (IRAK4 inhibitors) represents a promising therapeutic strategy for inflammatory disorders. To exploit the full potential of this treatment approach, drug carriers for efficient delivery of IRAK4 inhibitors to inflamed tissues are essential. Herein, the first nanoparticle-based platform for the targeted systemic delivery of a clinically tested IRAK4 inhibitor, PF-06650833, with limited aqueous solubility (57 µg mL-1 ) is presented. The developed nanocarriers increase the intrinsic aqueous dispersibility of this IRAK4 inhibitor by 40 times. A targeting peptide on the surface of nanocarriers significantly enhances their accumulation after intravenous injection in inflamed tissues of mice with induced paw edema and ulcerative colitis when compared to non-targeted counterparts. The delivered IRAK4 inhibitor markedly abates inflammation and dramatically suppresses paw edema, mitigates colitis symptoms, and reduces proinflammatory cytokine levels in the affected tissues. Importantly, repeated injections of IRAK4 inhibitor-loaded nanocarriers have no acute toxic effect on major organs of mice. Therefore, the developed nanocarriers have the potential to significantly improve the therapeutic efficacy of IRAK4 inhibitors for different inflammatory diseases.
Collapse
Affiliation(s)
- Youngrong Park
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, Massachusetts, 02115, USA
| | - Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Mail Code L481, Portland, Oregon, 97239, USA
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Prem Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Mail Code L481, Portland, Oregon, 97239, USA
| | - Ananiya A Demessie
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Kongbrailatpam Shitaljit Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Terry Morgan
- Department of Pathology and Laboratory Medicine, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon, 97239, USA
| | - Constanze J Raitmayr
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Uriel Avila
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Fahad Y Sabei
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Al Maarefah Rd, Jazan, 88723, Kingdom of Saudi Arabia
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Mail Code L481, Portland, Oregon, 97239, USA
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 S Moody Avenue, Portland, Oregon, 97201, USA
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW Models of arterial thrombus formation represent a vital experimental tool to investigate platelet function and test novel antithrombotic drugs. This review highlights some of the recent advances in modelling thrombus formation in vitro and suggests potential future directions. RECENT FINDINGS Microfluidic devices and the availability of commercial chips in addition to enhanced accessibility of 3D printing has facilitated a rapid surge in the development of novel in-vitro thrombosis models. These include progression towards more sophisticated, 'vessel on a chip' models which incorporate vascular endothelial cells and smooth muscle cells. Other approaches include the addition of branches to the traditional single channel to yield an occlusive model; and developments in the adhesive coating of microfluidic chambers to better mimic the thrombogenic surface exposed following plaque rupture. Future developments in the drive to create more biologically relevant chambers could see a move towards the use of human placental vessels, perfused ex-vivo. However, further work is required to determine the feasibility and validity of this approach. SUMMARY Recent advances in thrombus formation models have significantly improved the pathophysiological relevance of in-vitro flow chambers to better reflect the in-vivo environment and provide a more translational platform to test novel antithrombotics.
Collapse
Affiliation(s)
- Amelia Drysdale
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | | | | |
Collapse
|
8
|
Echesabal-Chen J, Huang K, Vojtech L, Oladosu O, Esobi I, Sachdeva R, Vyavahare N, Jo H, Stamatikos A. Constructing Lipoparticles Capable of Endothelial Cell-Derived Exosome-Mediated Delivery of Anti-miR-33a-5p to Cultured Macrophages. Curr Issues Mol Biol 2023; 45:5631-5644. [PMID: 37504271 PMCID: PMC10378689 DOI: 10.3390/cimb45070355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/26/2023] [Accepted: 07/01/2023] [Indexed: 07/29/2023] Open
Abstract
Atherosclerosis is driven by intimal arterial macrophages accumulating cholesterol. Atherosclerosis also predominantly occurs in areas consisting of proinflammatory arterial endothelial cells. At time of writing, there are no available clinical treatments that precisely remove excess cholesterol from lipid-laden intimal arterial macrophages. Delivery of anti-miR-33a-5p to macrophages has been shown to increase apoAI-mediated cholesterol efflux via ABCA1 upregulation but delivering transgenes to intimal arterial macrophages is challenging due to endothelial cell barrier integrity. In this study, we aimed to test whether lipoparticles targeting proinflammatory endothelial cells can participate in endothelial cell-derived exosome exploitation to facilitate exosome-mediated transgene delivery to macrophages. We constructed lipoparticles that precisely target the proinflammatory endothelium and contain a plasmid that expresses XMOTIF-tagged anti-miR-33a-5p (LP-pXMoAntimiR33a5p), as XMOTIF-tagged small RNA demonstrates the capacity to be selectively shuttled into exosomes. The cultured cells used in our study were immortalized mouse aortic endothelial cells (iMAECs) and RAW 264.7 macrophages. From our results, we observed a significant decrease in miR-33a-5p expression in macrophages treated with exosomes released basolaterally by LPS-challenged iMAECs incubated with LP-pXMoAntimiR33a5p when compared to control macrophages. This decrease in miR-33a-5p expression in the treated macrophages caused ABCA1 upregulation as determined by a significant increase in ABCA1 protein expression in the treated macrophages when compared to the macrophage control group. The increase in ABCA1 protein also simulated ABCA1-dependent cholesterol efflux in treated macrophages-as we observed a significant increase in apoAI-mediated cholesterol efflux-when compared to the control group of macrophages. Based on these findings, strategies that involve combining proinflammatory-targeting lipoparticles and exploitation of endothelial cell-derived exosomes appear to be promising approaches for delivering atheroprotective transgenes to lipid-laden arterial intimal macrophages.
Collapse
Affiliation(s)
- Jing Echesabal-Chen
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (J.E.-C.); (K.H.); (O.O.); (I.E.)
| | - Kun Huang
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (J.E.-C.); (K.H.); (O.O.); (I.E.)
| | - Lucia Vojtech
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98109, USA;
| | - Olanrewaju Oladosu
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (J.E.-C.); (K.H.); (O.O.); (I.E.)
| | - Ikechukwu Esobi
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (J.E.-C.); (K.H.); (O.O.); (I.E.)
| | - Rakesh Sachdeva
- Department of Chemistry, Clemson University, Clemson, SC 29634, USA;
| | - Naren Vyavahare
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA;
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA;
| | - Alexis Stamatikos
- Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (J.E.-C.); (K.H.); (O.O.); (I.E.)
| |
Collapse
|
9
|
Huang K, Pitman M, Oladosu O, Echesabal-Chen J, Vojtech L, Esobi I, Larsen J, Jo H, Stamatikos A. The Impact of MiR-33a-5p Inhibition in Pro-Inflammatory Endothelial Cells. Diseases 2023; 11:88. [PMID: 37489440 PMCID: PMC10366879 DOI: 10.3390/diseases11030088] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/26/2023] Open
Abstract
Evidence suggests cholesterol accumulation in pro-inflammatory endothelial cells (EC) contributes to triggering atherogenesis and driving atherosclerosis progression. Therefore, inhibiting miR-33a-5p within inflamed endothelium may prevent and treat atherosclerosis by enhancing apoAI-mediated cholesterol efflux by upregulating ABCA1. However, it is not entirely elucidated whether inhibition of miR-33a-5p in pro-inflammatory EC is capable of increasing ABCA1-dependent cholesterol efflux. In our study, we initially transfected LPS-challenged, immortalized mouse aortic EC (iMAEC) with either pAntimiR33a5p plasmid DNA or the control plasmid, pScr. We detected significant increases in both ABCA1 protein expression and apoAI-mediated cholesterol efflux in iMAEC transfected with pAntimiR33a5p when compared to iMAEC transfected with pScr. We subsequently used polymersomes targeting inflamed endothelium to deliver either pAntimiR33a5p or pScr to cultured iMAEC and showed that the polymersomes were selective in targeting pro-inflammatory iMAEC. Moreover, when we exposed LPS-challenged iMAEC to these polymersomes, we observed a significant decrease in miR-33a-5p expression in iMAEC incubated with polymersomes containing pAntimR33a5p versus control iMAEC. We also detected non-significant increases in both ABCA1 protein and apoAI-mediated cholesterol in iMAEC exposed to polymersomes containing pAntimR33a5p when compared to control iMAEC. Based on our results, inhibiting miR-33a-5p in pro-inflammatory EC exhibits atheroprotective effects, and so precisely delivering anti-miR-33a-5p to these cells is a promising anti-atherogenic strategy.
Collapse
Affiliation(s)
- Kun Huang
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (O.O.); (J.E.-C.); (I.E.)
| | - Mark Pitman
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC 29634, USA; (M.P.); (J.L.)
| | - Olanrewaju Oladosu
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (O.O.); (J.E.-C.); (I.E.)
| | - Jing Echesabal-Chen
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (O.O.); (J.E.-C.); (I.E.)
| | - Lucia Vojtech
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98109, USA;
| | - Ikechukwu Esobi
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (O.O.); (J.E.-C.); (I.E.)
| | - Jessica Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC 29634, USA; (M.P.); (J.L.)
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
| | - Hanjoong Jo
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA;
| | - Alexis Stamatikos
- Department of Food, Nutrition, and Packaging Sciences, Clemson University, Clemson, SC 29634, USA; (K.H.); (O.O.); (J.E.-C.); (I.E.)
| |
Collapse
|
10
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
11
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
12
|
Huang H, Ren P, Zhao Y, Weng H, Jia C, Yu F, Nie Y. Low shear stress induces inflammatory response via CX3CR1/NF-κB signal pathway in human umbilical vein endothelial cells. Tissue Cell 2023; 82:102043. [PMID: 36827822 DOI: 10.1016/j.tice.2023.102043] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Low shear stress (LSS) has been reported to induce atherosclerosis. However, the molecular mechanisms underlying inflammation induced by LSS are still poorly understood. The objective of our study is the comprehensive identification of molecular circuitry involved in low shear stress-induced inflammation in human umbilical vein endothelial cells (HUVECs) through protein profiling and cell function experiment. In this study, Western blotting analyses revealed a significant increase in the expression of CX3CR1, nucleusP65, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and Interleukin-6 (IL-6), while the expression of cytosolic P65 and IκB has significantly decreased in HUVECs treated with low shear stress. CX3CR1 Sh-RNA was use to reveal its effect on LSS-induced inflammation. Further, specific NF-κB P65 inhibitors pyrrolidinedithiocarbamate (PDTC) were used to reveal the downstream NF-κB P65 exclusively involved in LSS-induced inflammation in HUVECs, this effect can be abrogated by CX3CR1 sh-RNA and NF-κB inhibitors. Monocyte adhesion assay and scratch test revealed low shear stress to promotes adhesion of monocytes and migration of cells, this effect can be abrogated by CX3CR1 sh-RNA and NF-κB inhibitors. LSS was involved in the expression of adhesion molecules and chemokines, which are important for the initiation of endothelial inflammation-related atherosclerosis. Therefore, the cell signaling pathways activated by LSS in endothelial cells may represent therapeutic targets of atherosclerosis.
Collapse
Affiliation(s)
- Haozhong Huang
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Peile Ren
- Department of Physiology, School of Medicine, Xinjiang Medical University, Urumqi 830011, Xinjiang, PR China.
| | - Yiwei Zhao
- The School of Medicine, Huanghe S&T College, Zhengzhou 450063, Henan, PR China.
| | - Huimin Weng
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Chunsen Jia
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China.
| | - Fengxu Yu
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China; Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, PR China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, PR China.
| | - Yongmei Nie
- Department of Cardiovascular Surgery, Affiliated Hospital of the Southwest Medical University, Luzhou 646000, Sichuan, PR China; Luzhou Key Laboratory of Cardiovascular and Metabolic Diseases, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, PR China; Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou 646000, Sichuan, PR China.
| |
Collapse
|
13
|
Nambo-Venegas R, Palacios-González B, Mas-Oliva J, Aurioles-Amozurrutia AK, Cruz-Rangel A, Moreno A, Hidalgo-Miranda A, Rodríguez-Dorantes M, Vadillo-Ortega F, Xicohtencatl-Cortes J, Ruiz-Olmedo MI, Reyes-Grajeda JP. Conversion of M1 Macrophages to Foam Cells: Transcriptome Differences Determined by Sex. Biomedicines 2023; 11:biomedicines11020490. [PMID: 36831031 PMCID: PMC9953229 DOI: 10.3390/biomedicines11020490] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND M1 macrophages involved in pro-inflammatory processes can be induced by low-density lipoproteins (LDL), giving rise to foam cells. In the atheroma plaque, it has been identified that males present more advanced lesions associated with infiltration. Therefore, our study aims to investigate sex-related changes in the transcriptome of M1 macrophages during the internalization process of LDL particles. METHODS Peripheral blood mononuclear cells (PBMCs) from healthy male and female subjects were separated using Hystopaque, and monocytes were isolated from PBMCs using a positive selection of CD14+ cells. Cells were stimulated with LDL 10 µg/mL, and the transcriptional profile of M1 macrophages performed during LDL internalization was determined using a Clariom D platform array. RESULTS Chromosome Y influences the immune system and inflammatory responses in males expressing 43% of transcripts in response to LDL treatment. Males and females share 15 transcripts, where most correspond to non-coding elements involved in oxidative stress and endothelial damage. CONCLUSIONS During LDL internalization, male monocyte-derived M1 macrophages display more marked proinflammatory gene expression. In contrast, female M1 macrophages display a more significant number of markers associated with cell damage.
Collapse
Affiliation(s)
- Rafael Nambo-Venegas
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | - Berenice Palacios-González
- Laboratorio de Envejecimiento Saludable, Centro de Investigación Sobre Envejecimiento (CIE-CINVESTAV Sur), Instituto Nacional de Medicina Genómica, Mexico City 14330, Mexico
| | - Jaime Mas-Oliva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Armando Cruz-Rangel
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | - Abel Moreno
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | | | - Felipe Vadillo-Ortega
- Unidad de Vinculación Científica de la Facultad de Medicina UNAM en INMEGEN, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
| | - Juan Xicohtencatl-Cortes
- Laboratorio de Bacteriología Intestinal, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | | | - Juan Pablo Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica, Mexico City 14600, Mexico
- Correspondence:
| |
Collapse
|
14
|
Zhao CR, Li J, Jiang ZT, Zhu JJ, Zhao JN, Yang QR, Yao W, Pang W, Li N, Yu M, Gan Y, Zhou J. Disturbed Flow-Facilitated Margination and Targeting of Nanodisks Protect against Atherosclerosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204694. [PMID: 36403215 DOI: 10.1002/smll.202204694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/22/2022] [Indexed: 06/16/2023]
Abstract
Disturbed blood flow induces endothelial pro-inflammatory responses that promote atherogenesis. Nanoparticle-based therapeutics aimed at treating endothelial inflammation in vasculature where disturbed flow occurs may provide a promising avenue to prevent atherosclerosis. By using a vertical-step flow apparatus and a microfluidic chip of vascular stenosis, herein, it is found that the disk-shaped versus the spherical nanoparticles exhibit preferential margination (localization and adhesion) to the regions with the pro-atherogenic disturbed flow. By employing a mouse model of carotid partial ligation, superior targeting and higher accumulation of the disk-shaped particles are also demonstrated within disturbed flow areas than that of the spherical particles. In hyperlipidemia mice, administration of disk-shaped particles loaded with hypomethylating agent decitabine (DAC) displays greater anti-inflammatory and anti-atherosclerotic effects compared with that of the spherical counterparts and exhibits reduced toxicity than "naked" DAC. The findings suggest that shaping nanoparticles to disk is an effective strategy for promoting their delivery to atheroprone endothelia.
Collapse
Affiliation(s)
- Chuan-Rong Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhi-Tong Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Juan-Juan Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Jia-Nan Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Qian-Ru Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| | - Weijuan Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Wei Pang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Ning Li
- Center for Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, 100190, China
- School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Miaorong Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Gan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Hemorheology Center, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, 100191, China
- National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University, Beijing, 100191, China
| |
Collapse
|
15
|
Liu H, Liu J, Long C, Chen L, Zhan W, Xiao W, Gong X, Liu M, Tian XL, Chen S. Lack of NPR1 Increases Vascular Endothelial Adhesion through Induction of Integrin Beta 4. Int J Mol Sci 2022; 23:ijms232012627. [PMID: 36293483 PMCID: PMC9604115 DOI: 10.3390/ijms232012627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 12/02/2022] Open
Abstract
Natriuretic peptide receptor 1 (NPR1) serves as a modulator of vascular endothelial homeostasis. Interactions between monocytes and endothelial cells may initiate endothelium dysfunction, which is known as an early hallmark of atherosclerosis. In this study, we performed RNA-sequencing analysis for the aorta of Npr1 knockout (Npr1+/−) mice and found that differentially expressed genes were significantly related to cell adhesion. This result was supported by an increased expression of intercellular adhesion molecule 1 (ICAM-1) in the aortic endothelium of Npr1+/− mice. Moreover, we observed that the knockdown of NPR1 increased ICAM-1 expression and promoted THP-1 monocyte adhesion to human umbilical vein endothelial cells (HUVECs). NPR1 overexpression decreased ICAM-1 expression and inhibited the adhesion of monocytes to HUVECs treated by TNF-α (a cell adhesion inducer). Further analysis showed that adhesion-related genes were enriched in the focal adhesion signaling pathway, in which integrin beta 4 (Itgb4) was determined as a key gene. Notably, ITGB4 expression increased in vascular endothelium of Npr1+/− mice and in NPR1-knockdown HUVECs. The deficiency of ITGB4 decreased ICAM-1 expression and attenuated monocyte adhesion to NPR1-knockdown endothelial cells. Additionally, a reduced NPR1 and an increased ITGB4 expression level were found in an atherosclerosis mouse model. In conclusion, our findings demonstrate that NPR1 deficiency increases vascular endothelial cell adhesion by stimulating ITGB4 expression, which may contribute to the development of atherosclerosis.
Collapse
Affiliation(s)
- Hongfei Liu
- Vascular Function Laboratory, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Jiankun Liu
- Aging and Vascular Diseases, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Changkun Long
- Vascular Function Laboratory, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Liping Chen
- Vascular Function Laboratory, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Wenxing Zhan
- Vascular Function Laboratory, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Wanli Xiao
- Metabolic Control and Aging, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Xueting Gong
- Aging and Vascular Diseases, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Man Liu
- Aging and Vascular Diseases, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
| | - Shenghan Chen
- Vascular Function Laboratory, Human Aging Research Institute, School of Life Science, Jiangxi Key Laboratory of Human Aging, Nanchang University, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
16
|
Zulkapli R, Yusof MYPM, Abd Muid S, Wang SM, Firus Khan AY, Nawawi H. A Systematic Review on Attenuation of PCSK9 in Relation to Atherogenesis Biomarkers Associated with Natural Products or Plant Bioactive Compounds in In Vitro Studies: A Critique on the Quality and Imprecision of Studies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:12878. [PMID: 36232177 PMCID: PMC9566180 DOI: 10.3390/ijerph191912878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
A systematic review was performed to identify all the related publications describing PCSK9 and atherogenesis biomarkers attenuation associated with a natural product and plant bioactive compounds in in vitro studies. This review emphasized the imprecision and quality of the included research rather than the detailed reporting of the results. Literature searches were conducted in Scopus, PubMed, and Science Direct from 2003 until 2021, following the Cochrane handbook. The screening of titles, abstracts, and full papers was performed by two independent reviewers, followed by data extraction and validity. Study quality and validity were assessed using the Imprecision Tool, Model, and Marker Validity Assessment that has been developed for basic science studies. A total of 403 articles were identified and 31 of those that met the inclusion criteria were selected. 13 different atherogenesis biomarkers in relation to PCSK9 were found, and the most studied biomarkers are LDLR, SREBP, and HNF1α. In terms of quality, our review suggests that the basic science study in investigating atherogenesis biomarkers is deficient in terms of imprecision and validity.
Collapse
Affiliation(s)
- Rahayu Zulkapli
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Dentistry, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
| | - Mohd Yusmiaidil Putera Mohd Yusof
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Dentistry, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
| | - Suhaila Abd Muid
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
| | - Seok Mui Wang
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
| | - Al’Aina Yuhainis Firus Khan
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
| | - Hapizah Nawawi
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
- Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Jalan Hospital, Sungai Buloh 47000, Selangor, Malaysia
| |
Collapse
|
17
|
Qiu D, Hu J, Wang P, Huang D, Lin Y, Tian H, Yi X, Zou Q, Zhu H. Synthesis of NaYF4:20% Yb3+,2% Er3+,2% Ce3+@NaYF4 nanorods and their size dependent uptake efficiency under flow condition. J RARE EARTH 2022. [DOI: 10.1016/j.jre.2021.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
Wu L, Yin Z, Zheng Z, Tang Y, Guo S. Comprehensive Relationship Analysis of the Long Noncoding RNAs (lncRNAs) and the Target mRNAs in Response to the Infection of Edwardsiella anguillarum in European eel (Anguilla anguilla) Inoculated with Freund's Adjuvant. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:956-968. [PMID: 35995892 DOI: 10.1007/s10126-022-10157-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Freund's complete adjuvant (FCA) and incomplete adjuvant (FIA), generally applied in subunit fishery vaccine, have not been explored on the molecular mechanism of the non-specific immune enhancement. As long noncoding RNAs (lncRNAs) play vital regulating roles in various biological activities, in this study, we examined the genome-wide expression of transcripts in the liver of European eel (Anguilla anguilla, Aa) inoculated with FCA and FIA (FCIA) to elucidate the regulators of lncRNAs in the process of Edwardsiella anguillarum (Ea) infection and Aa anti-Ea infection using strand-specific RNA-seq. After eels were challenged by Ea at 28 days post the first inoculation (dpi), compared to the control uninfected eels (Li group), the control infected eels (Con_Li group) showed severe bleeding, hepatocyte atrophy, and thrombi formed in the hepatic vessels of the liver, although eels inoculated with FCIA (FCIA_Li group) also formed slight thrombi in the hepatic vessels. Compared to the FCIA_Li group, there was about 10 times colony-forming unit (cfu) in the Con_Li group per 100 μg liver tissue, and the relative percent survival (RPS) of eels was 50% in FCIA_Li vs Con_Li. Using high-throughput transcriptomics, differential expressed genes (DEGs) and transcripts were identified and the results were verified using fluorescence real-time polymerase chain reaction (qRT-PCR). Interactions between the differential expressed lncRNAs (DE-lncRNAs) and the target DEGs were explored using Cytoscape according to their co-expression and co-location relationship. We found 13,499 lncRNAs (10,176 annotated and 3423 novel lncRNAs) between 3 comparisons of Con_Li vs Li, FCIA_Li vs Li, and FCIA_Li vs Con_Li, of which 111, 110, and 129 DE-lncRNAs were ascertained. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of DEGs targeted by DE-lncRNAs revealed these DEGs mainly involved in single-organism cellular process in BP, membrane in CC and binding in MF, and KEGG pathways showed that the target DEGs in co-expression and co-location enriched in cell adhesion molecules. Finally, 118 DE-lncRNAs target 1161 DEGs were involved in an interaction network of 8474 co-expression and 333 co-location-related links, of which 16 DE-lncRNAs play vital roles in anti-Ea infection. Taken together, the interaction networks revealed that DE-lncRNAs underlies the process of Ea infection and Aa anti-Ea infection.
Collapse
Affiliation(s)
- Liqun Wu
- College of Overseas Education, Jimei University, Xiamen, 361021, China
| | - Zhijie Yin
- Fisheries College, Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PRC, Jimei University, Xiamen, 361021, China
| | - Zhijin Zheng
- Fisheries College, Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PRC, Jimei University, Xiamen, 361021, China
| | - Yijun Tang
- Department of Chemistry, University of Wisconsin Oshkosh, 800 Algoma Blvd., Oshkosh, WI, USA
| | - Songlin Guo
- Fisheries College, Engineering Research Center of the Modern Industry Technology for Eel. Ministry of Education of PRC, Jimei University, Xiamen, 361021, China.
| |
Collapse
|
19
|
Kalinin RE, Korotkova NV, Suchkov IA, Mzhavanadze ND, Ryabkov AN. Selectins and their involvement in the pathogenesis of cardiovascular diseases. KAZAN MEDICAL JOURNAL 2022; 103:617-627. [DOI: 10.17816/kmj2022-617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The review presents current data on the structure and functional role of cell adhesion molecules belonging to the selectin family (selectins P, L and E), and their involvement in the pathogenesis of cardiovascular diseases. On the one hand, intercellular adhesion molecules of the vascular wall endothelium, platelets and leukocytes are an important link in the processes of vasculogenesis, development and regeneration of the vascular system. On the other hand, these molecules participate in the earliest stages of endothelial dysfunction with the subsequent development of pathology. For this reason, figuring out the mechanisms of activity of this group of molecules is very important for understanding the molecular basis of the cardiovascular diseases pathogenesis. The adhesion of molecules, both between cells and between cells and a component of the extracellular matrix, is the most important stage of physiological and biochemical processes. According to present knowledge, five classes of intercellular adhesion molecules are known: integrins, cadherins, immunoglobulins (including nectins), selectins and addressins. All of them are bonded to a cytoplasmic membrane and provide the interaction of cells with each other. Some of them are transmembrane and associated with the cytoskeleton of the cell. On the cell surface, intercellular adhesion molecules can be located in clusters, forming multipoint binding sites and thereby determining the degree of avidity. One of the most significant functions of selectins is participation in the initial stage of the leukocyte adhesion cascade, which results in their binding to the endothelium, rolling and further extravasation into tissues. The first stage of this process is mediated by specific non-covalent interactions between selectins and their glycan ligands, with the glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains one of the main strategies aimed at developing new methods of treating immune, inflammatory and oncological diseases.
Collapse
|
20
|
Muñoz-Ortiz T, Hu J, Sanz-Rodríguez F, Ortgies DH, Jaque D, Méndez-González D, Aguilar R, Alfonso F, Rivero F, Martín Rodríguez E, García Solé J. Optical detection of atherosclerosis at molecular level by optical coherence tomography: An in vitro study. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 43:102556. [PMID: 35390527 DOI: 10.1016/j.nano.2022.102556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/22/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Abstract
There is an urgent need for contrast agents to detect the first inflammation stage of atherosclerosis by cardiovascular optical coherence tomography (CV-OCT), the imaging technique with the highest spatial resolution and sensitivity of those used during coronary interventions. Gold nanoshells (GNSs) provide the strongest signal by CV-OCT. GNSs are functionalized with the cLABL peptide that binds specifically to the ICAM-1 molecule upregulated in the first stage of atherosclerosis. Dark field microscopy and CV-OCT are used to evaluate the specific adhesion of these functionalized GNSs to activated endothelial cells. This adhesion is investigated under static and dynamic conditions, for shear stresses comparable to those of physiological conditions. An increase in the scattering signal given by the functionalized GNSs attached to activated cells is observed compared to non-activated cells. Thus, cLABL-functionalized GNSs behave as excellent contrast agents for CV-OCT and promise a novel strategy for clinical molecular imaging of atherosclerosis.
Collapse
Affiliation(s)
- Tamara Muñoz-Ortiz
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Instituto Nicolás Cabrera, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jie Hu
- Xiamen Institute of Rare-earth Materials, Haixi Institutes Chinese Academy of Sciences, Xiamen, Fujian, China
| | - Francisco Sanz-Rodríguez
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Dirk H Ortgies
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Nanomaterials for Bioimaging Group (nanoBIG), Instituto Ramón y Cajal de Investigación, Sanitaria Hospital Ramón y Cajal, Madrid, Spain
| | - Daniel Jaque
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Instituto Nicolás Cabrera, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Nanomaterials for Bioimaging Group (nanoBIG), Instituto Ramón y Cajal de Investigación, Sanitaria Hospital Ramón y Cajal, Madrid, Spain
| | - Diego Méndez-González
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Nanomaterials for Bioimaging Group (nanoBIG), Instituto Ramón y Cajal de Investigación, Sanitaria Hospital Ramón y Cajal, Madrid, Spain
| | - Río Aguilar
- Cardiology Department, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), CIBER-CV, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fernando Alfonso
- Cardiology Department, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), CIBER-CV, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fernando Rivero
- Cardiology Department, Hospital Universitario de la Princesa, Instituto Investigación Sanitaria Princesa (IIS-IP), CIBER-CV, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emma Martín Rodríguez
- Instituto Nicolás Cabrera, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Nanomaterials for Bioimaging Group (nanoBIG), Instituto Ramón y Cajal de Investigación, Sanitaria Hospital Ramón y Cajal, Madrid, Spain; Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Física Aplicada, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.
| | - José García Solé
- Nanomaterials for Bioimaging Group (nanoBIG), Departamento de Física de Materiales, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain; Instituto Nicolás Cabrera, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
21
|
Cheraga N, Ye Z, Xu MJ, Zou L, Sun NC, Hang Y, Shan CJ, Yang ZZ, Chen LJ, Huang NP. Targeted therapy of atherosclerosis by pH-sensitive hyaluronic acid nanoparticles co-delivering all-trans retinal and rapamycin. NANOSCALE 2022; 14:8709-8726. [PMID: 35673987 DOI: 10.1039/d1nr06514a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Atherosclerosis, the leading cause of death in the elderly worldwide, is typically characterized by elevated reactive oxygen species (ROS) levels and a chronic inflammatory state at the arterial plaques. Herein, pH-sensitive nanoparticles (HRRAP NPs) co-delivering all-trans retinal (ATR), an antioxidant linked to hyaluronic acid (HA) through a pH-sensitive hydrazone bond, and rapamycin (RAP), an anti-atherosclerotic drug loaded into the nanoparticle core, are developed for targeted combination therapy of atherosclerosis. In this way, HRRAP NPs might simultaneously reduce ROS levels via ATR antioxidant activity and reduce inflammation via the anti-inflammatory effect of RAP. In response to mildly acidic conditions mimicking the lesional inflammation in vitro, HRRAP NPs dissociated and both ATR and RAP were effectively released. The developed HRRAP NPs effectively inhibited pro-inflammatory macrophage proliferation, and displayed dose- and time-dependent specific internalization by different cellular models of atherosclerosis. Also, HRRAP NP combination therapy showed an efficient synergetic anti-atherosclerotic effect in vitro by effectively inhibiting the inflammatory response and oxidative stress in inflammatory cells. More importantly, HR NPs specifically accumulated in the atherosclerotic plaques of apolipoprotein E-deficient (ApoE-/-) mice, by active interaction with HA receptors overexpressed by different cells of the plaque. The treatment with HRRAP NPs remarkably inhibited the progression of atherosclerosis in ApoE-/- mice which resulted in stable plaques with considerably smaller necrotic cores, lower matrix metalloproteinase-9, and decreased proliferation of macrophages and smooth muscle cells (SMCs). Furthermore, HRRAP NPs attenuated RAP adverse effects and exhibited a good safety profile after long-term treatment in mice. Consequently, the developed pH-sensitive HRRAP NP represent a promising nanoplatform for atherosclerosis combination therapy.
Collapse
Affiliation(s)
- Nihad Cheraga
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China.
| | - Zheng Ye
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China.
| | - Ming-Jie Xu
- Nanjing University Medical School, Nanjing, 210093, China
| | - Lin Zou
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China.
| | - Ning-Cong Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China.
| | - Yue Hang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China.
| | - Cong-Jia Shan
- Nanjing University Medical School, Nanjing, 210093, China
| | | | - Li-Juan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China.
| |
Collapse
|
22
|
Targeting vascular inflammation through emerging methods and drug carriers. Adv Drug Deliv Rev 2022; 184:114180. [PMID: 35271986 PMCID: PMC9035126 DOI: 10.1016/j.addr.2022.114180] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
Abstract
Acute inflammation is a common dangerous component of pathogenesis of many prevalent conditions with high morbidity and mortality including sepsis, thrombosis, acute respiratory distress syndrome (ARDS), COVID-19, myocardial and cerebral ischemia-reperfusion, infection, and trauma. Inflammatory changes of the vasculature and blood mediate the course and outcome of the pathology in the tissue site of insult, remote organs and systemically. Endothelial cells lining the luminal surface of the vasculature play the key regulatory functions in the body, distinct under normal vs. pathological conditions. In theory, pharmacological interventions in the endothelial cells might enable therapeutic correction of the overzealous damaging pro-inflammatory and pro-thrombotic changes in the vasculature. However, current agents and drug delivery systems (DDS) have inadequate pharmacokinetics and lack the spatiotemporal precision of vascular delivery in the context of acute inflammation. To attain this level of precision, many groups design DDS targeted to specific endothelial surface determinants. These DDS are able to provide specificity for desired tissues, organs, cells, and sub-cellular compartments needed for a particular intervention. We provide a brief overview of endothelial determinants, design of DDS targeted to these molecules, their performance in experimental models with focus on animal studies and appraisal of emerging new approaches. Particular attention is paid to challenges and perspectives of targeted therapeutics and nanomedicine for advanced management of acute inflammation.
Collapse
|
23
|
Zinellu A, Mangoni AA. A systematic review and meta-analysis of the effect of statin treatment on sVCAM-1 and sICAM-1. Expert Rev Clin Pharmacol 2022; 15:601-620. [PMID: 35485866 DOI: 10.1080/17512433.2022.2072294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND AND AIMS Statins might prevent cell adhesion to the endothelium, a key step in atherosclerosis. We conducted a systematic review and meta-analysis of the effect of statins on soluble vascular (sVCAM-1) and intercellular (sICAM-1) adhesion molecule 1. METHODS A systematic literature search was conducted in PubMed, Web of Science, and Scopus, from inception to July 2021. Risk of bias and certainty of evidence were assessed using the Joanna Briggs Institute Critical Appraisal Checklist for analytical studies and GRADE, respectively. RESULTS Statins significantly reduced both sVCAM-1 (standard mean difference, SMD=-0.28, 95% CI -0.44 to -0.12, p=0.001; 46 treatment arms; low certainty of evidence) and sICAM-1 (SMD=-0.75, 95% CI -1.00 to -0.50, p<0.001; 61 treatment arms; moderate certainty of evidence) concentrations. In sensitivity analysis, the SMD values were not modified when individual studies were sequentially removed. There were significant associations between SMD and publication year and, for sICAM-1, statin-induced changes in HDL-cholesterol. In subgroup analysis, the lowering effect was significant with liphophilic, but not hydrophilic, statins, and similar, for sICAM-1, in participants with or without clinically overt atherosclerosis. CONCLUSIONS Statins significantly lower sVCAM-1/sICAM-1. Prospective studies are required to determine whether this mediates their atheroprotective effects (PROSPERO registration number: CRD42021276825).
Collapse
Affiliation(s)
- Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| |
Collapse
|
24
|
Praveen TK, Gangadharappa HV, Abu Lila AS, Moin A, Mehmood K, Krishna KL, Hussain T, Alafanan A, Shakil S, Rizvi SMD. Inflammation targeted nanomedicines: patents and applications in cancer therapy. Semin Cancer Biol 2022; 86:645-663. [DOI: 10.1016/j.semcancer.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/08/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022]
|
25
|
Forouzandehmehr M, Ghoytasi I, Shamloo A, Ghosi S. Particles in coronary circulation: A review on modelling for drug carrier design. MATERIALS & DESIGN 2022; 216:110511. [DOI: 10.1016/j.matdes.2022.110511] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Bazban-Shotorbani S, Gavins F, Kant K, Dufva M, Kamaly N. A Biomicrofluidic Screening Platform for Dysfunctional Endothelium‐Targeted Nanoparticles and Therapeutics. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Salime Bazban-Shotorbani
- Department of Health Technology DTU Health Tech Technical University of Denmark Lyngby 2800 Kgs. Denmark
- Department of Chemistry Molecular Sciences Research Hub (MSRH) Imperial College London London W12 0BZ UK
| | - Felicity Gavins
- Department of Life Sciences Centre for Inflammation Research and Translational Medicine (CIRTM) Brunel University London London UB8 3PH UK
| | - Krishna Kant
- Department of Physical Chemistry Biomedical Research Center of Galicia (CINBIO) University of Vigo Vigo 36310 Spain
| | - Martin Dufva
- Department of Health Technology DTU Health Tech Technical University of Denmark Lyngby 2800 Kgs. Denmark
| | - Nazila Kamaly
- Department of Chemistry Molecular Sciences Research Hub (MSRH) Imperial College London London W12 0BZ UK
| |
Collapse
|
27
|
Hossaini Nasr S, Huang X. Nanotechnology for Targeted Therapy of Atherosclerosis. Front Pharmacol 2021; 12:755569. [PMID: 34867370 PMCID: PMC8633109 DOI: 10.3389/fphar.2021.755569] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/22/2021] [Indexed: 01/20/2023] Open
Abstract
Atherosclerosis is the major cause of heart attack and stroke that are the leading causes of death in the world. Nanomedicine is a powerful tool that can be engineered to target atherosclerotic plaques for therapeutic and diagnosis purposes. In this review, advances in designing nanoparticles with therapeutic effects on atherosclerotic plaques known as atheroprotective nanomedicine have been summarized to stimulate further development and future translation.
Collapse
Affiliation(s)
- Seyedmehdi Hossaini Nasr
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
28
|
Singh M, Thakur M, Mishra M, Yadav M, Vibhuti R, Menon AM, Nagda G, Dwivedi VP, Dakal TC, Yadav V. Gene regulation of intracellular adhesion molecule-1 (ICAM-1): A molecule with multiple functions. Immunol Lett 2021; 240:123-136. [PMID: 34715236 DOI: 10.1016/j.imlet.2021.10.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 01/04/2023]
Abstract
Intracellular adhesion molecule 1 (ICAM-1) is one of the most extensively studied inducible cell adhesion molecules which is responsible for several immune functions like T cell activation, extravasation, inflammation, etc. The molecule is constitutively expressed over the cell surface and is regulated up / down in response to inflammatory mediators like cellular stress, proinflammatory cytokines, viral infection. These stimuli modulate the expression of ICAM-1 primarily through regulating the ICAM-1 gene transcription. On account of the presence of various binding sites for NF-κB, AP-1, SP-1, and many other transcription factors, the architecture of the ICAM-1 promoter become complex. Transcription factors in union with other transcription factors, coactivators, and suppressors promote their assembly in a stereospecific manner on ICAM-1 promoter which mediates ICAM-1 regulation in response to different stimuli. Along with transcriptional regulation, epigenetic modifications also play a pivotal role in controlling ICAM-1 expression on different cell types. In this review, we summarize the regulation of ICAM-1 expression both at the transcriptional as well as post-transcriptional level with an emphasis on transcription factors and signaling pathways involved.
Collapse
Affiliation(s)
- Mona Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi-110067 India
| | - Mony Thakur
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| | - Manish Mishra
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research- Institute of Microbial Technology, Chandigarh-160036 India
| | - Manisha Yadav
- Division of Cell Biology and Immunology, Council of Scientific and Industrial Research- Institute of Microbial Technology, Chandigarh-160036 India
| | - Rajkamal Vibhuti
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| | - Athira M Menon
- Genome and computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001 India
| | - Girima Nagda
- Department of Zoology, Mohanlal Sukhadia University, Udaipur, Rajasthan-313001 India
| | - Ved Prakash Dwivedi
- International Centre for Genetic Engineering and Biotechnology, ICGEB Campus, Aruna Asaf Ali Marg, New Delhi-110067 India
| | - Tikam Chand Dakal
- Genome and computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001 India
| | - Vinod Yadav
- Department of Microbiology, Central University of Haryana, Mahendergarh, Haryana-123031 India
| |
Collapse
|
29
|
Zhang G, Ma L, Bai L, Li M, Guo T, Tian B, He Z, Fu Q. Inflammatory microenvironment-targeted nanotherapies. J Control Release 2021; 334:114-126. [PMID: 33887284 DOI: 10.1016/j.jconrel.2021.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022]
Abstract
Inflammatory microenvironments (IMEs) are common pathological characteristics and drive the development of multiple chronic diseases. Thus, IME-targeted therapies exhibit potential for the treatment of inflammatory diseases. Nanoplatforms have significant advantages in improving the efficiency of anti-inflammatory treatments. Owing to their improved therapeutic effects and reduced side effects, IME-targeted nanotherapies have recently drawn interest from the research community. This review introduces IMEs and discusses the application of IME-targeted nanotherapies for inflammatory diseases. The development of rational targeting strategies tailored to IMEs in damaged tissues can help promote therapies for chronic diseases.
Collapse
Affiliation(s)
- Guangshuai Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Lixue Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Lijun Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Tiange Guo
- Laboratory Animal Department, General Hospital of Northern Theater Command, No. 83, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
30
|
VCAM-1-targeted and PPARδ-agonist-loaded nanomicelles enhanced suppressing effects on apoptosis and migration of oxidized low-density lipoprotein-induced vascular smooth muscle cells. Biosci Rep 2021; 40:222727. [PMID: 32314783 PMCID: PMC7218220 DOI: 10.1042/bsr20200559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 11/29/2022] Open
Abstract
Purpose: Nanomicelles (NMs) have been widely used for various biomedical applications due to its unique physiochemical properties. The present study aims to investigate the effects of vascular cell adhesion molecule-1 (VCAM-1)-targeted and peroxisome proliferator-activated receptor δ (PPARδ) agonist (GW0742)-loaded NMs on apoptosis and migration in oxidized low-density lipoprotein (ox-LDL)-induced human aortic vascular smooth muscle cells (HAVSMCs). Methods: The GW0742-loaded NMs (M-GW) and VCAM-1-targeted NMs loaded with GW0742 (TM-GW) were prepared, and then the morphologies and the size distribution of M-GM and TM-GM were observed by transmission electron microscopy (TEM) and dynamic light scattering (DLS), respectively. In vitro drug release assay of M-GM and TM-GM were performed as well. Next, HAVSMCs were cultured in medium containing ox-LDL to mimic atherosclerotic environment, and the effects of free GW0742, M-GM and TM-GM on endocytosis, cell migration and apoptosis, as well as the expression of VCAM-1, and proteins associated with migration and apoptosis were measured in HAVSMCs treated with ox-LDL. Results: M-GM and TM-GM were successfully prepared. VCAM-1 was overexpressed in HAVSMCs treated with ox-LDL, and TM-GM had a strong targeting ability to HAVSMCs treated with ox-LDL compared with M-GM. In addition, compared with free GW0742, both M-GM and TM-GM significantly diminished cell apoptosis and migration in HAVSMCs treated with ox-LDL. Conclusions: TM-GM had a superior suppressing effect on apoptosis and migration of ox-LDL-induced HAVSMCs.
Collapse
|
31
|
Immunomodulatory biomaterials and their application in therapies for chronic inflammation-related diseases. Acta Biomater 2021; 123:1-30. [PMID: 33484912 DOI: 10.1016/j.actbio.2021.01.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/05/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
The degree of tissue injuries such as the level of scarring or organ dysfunction, and the immune response against them primarily determine the outcome and speed of healing process. The successful regeneration of functional tissues requires proper modulation of inflammation-producing immune cells and bioactive factors existing in the damaged microenvironment. In the tissue repair and regeneration processes, different types of biomaterials are implanted either alone or by combined with other bioactive factors, which will interact with the immune systems including immune cells, cytokines and chemokines etc. to achieve different results highly depending on this interplay. In this review article, the influences of different types of biomaterials such as nanoparticles, hydrogels and scaffolds on the immune cells and the modification of immune-responsive factors such as reactive oxygen species (ROS), cytokines, chemokines, enzymes, and metalloproteinases in tissue microenvironment are summarized. In addition, the recent advances of immune-responsive biomaterials in therapy of inflammation-associated diseases such as myocardial infarction, spinal cord injury, osteoarthritis, inflammatory bowel disease and diabetic ulcer are discussed.
Collapse
|
32
|
Petreski T, Piko N, Ekart R, Hojs R, Bevc S. Review on Inflammation Markers in Chronic Kidney Disease. Biomedicines 2021; 9:182. [PMID: 33670423 PMCID: PMC7917900 DOI: 10.3390/biomedicines9020182] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) is one of the major health problems of the modern age. It represents an important public health challenge with an ever-lasting rising prevalence, which reached almost 700 million by the year 2017. Therefore, it is very important to identify patients at risk for CKD development and discover risk factors that cause the progression of the disease. Several studies have tackled this conundrum in recent years, novel markers have been identified, and new insights into the pathogenesis of CKD have been gained. This review summarizes the evidence on markers of inflammation and their role in the development and progression of CKD. It will focus primarily on cytokines, chemokines, and cell adhesion molecules. Nevertheless, further large, multicenter studies are needed to establish the role of these markers and confirm possible treatment options in everyday clinical practice.
Collapse
Affiliation(s)
- Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Nejc Piko
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia;
| | - Robert Ekart
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia;
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia; (T.P.); (N.P.); (R.H.)
- Department of Internal Medicine and Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
| |
Collapse
|
33
|
Fleischmann D, Maslanka Figueroa S, Goepferich A. Steric Shielding of cRGD-Functionalized Nanoparticles from Premature Exposition to Off-Target Endothelial Cells under a Physiological Flow. ACS APPLIED BIO MATERIALS 2020. [DOI: 10.1021/acsabm.0c01193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Sara Maslanka Figueroa
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| |
Collapse
|
34
|
Zhang M, Du Y, Wang S, Chen B. A Review of Biomimetic Nanoparticle Drug Delivery Systems Based on Cell Membranes. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5495-5503. [PMID: 33363358 PMCID: PMC7753887 DOI: 10.2147/dddt.s282368] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
Cancers have always been an intractable problem because of recurrence and drug resistance. In the past few decades, nanoparticles have been explored intensely to diagnose, prevent and treat malignancy due to their good penetrability and better targeting. However, most nanocarriers have poor biodegradation and can be discharged out of the body quickly or cleared by immune cells while failing to obtain effective drug concentration at the specific sites. The emergence of biological membrane encapsulation technology relieves the fast clearance of antitumor drugs and reduces toxicity in vivo. This review will discuss the advantages and disadvantages of several blood cell membrane-coated nanoparticles and further introduce exosome-carried drugs to evidence the promising prospect of biomimetic nanoparticle drug delivery systems.
Collapse
Affiliation(s)
- Meilin Zhang
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Ying Du
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| | - Shujun Wang
- Jinling Hospital Department of Blood Transfusion, School of Medicine, Nanjing University, Nanjing, Jiangsu Province 210002, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province 210009, People's Republic of China
| |
Collapse
|
35
|
Utsunomiya A, Oyama N, Hasegawa M. Potential Biomarkers in Systemic Sclerosis: A Literature Review and Update. J Clin Med 2020; 9:E3388. [PMID: 33105647 PMCID: PMC7690387 DOI: 10.3390/jcm9113388] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by dysregulation of the immune system, vascular damage, and fibrosis of the skin and internal organs. Patients with SSc show a heterogeneous phenotype and a range of clinical courses. Therefore, biomarkers that are helpful for precise diagnosis, prediction of clinical course, and evaluation of the therapeutic responsiveness of disease are required in clinical practice. SSc-specific autoantibodies are currently used for diagnosis and prediction of clinical features, as other biomarkers have not yet been fully vetted. Krebs von den Lungen-6 (KL-6), surfactant protein-D (SP-D), and CCL18 have been considered as serum biomarkers of SSc-related interstitial lung disease. Moreover, levels of circulating brain natriuretic peptide (BNP) and N-terminal pro-brain natriuretic peptide (NT-proBNP) can provide diagnostic information and indicate the severity of pulmonary arterial hypertension. Assessment of several serum/plasma cytokines, chemokines, growth factors, adhesion molecules, and other molecules may also reflect the activity or progression of fibrosis and vascular involvement in affected organs. Recently, microRNAs have also been implicated as possible circulating indicators of SSc. In this review, we focus on several potential SSc biomarkers and discuss their clinical utility.
Collapse
Affiliation(s)
| | | | - Minoru Hasegawa
- Department of Dermatology, Divison of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; (A.U.); (N.O.)
| |
Collapse
|
36
|
Zhong M, Zhang X, Shi X, Zheng C. Halofuginone inhibits LPS-induced attachment of monocytes to HUVECs. Int Immunopharmacol 2020; 87:106753. [DOI: 10.1016/j.intimp.2020.106753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/17/2020] [Accepted: 06/25/2020] [Indexed: 01/20/2023]
|
37
|
Li J, Tan T, Zhao L, Liu M, You Y, Zeng Y, Chen D, Xie T, Zhang L, Fu C, Zeng Z. Recent Advancements in Liposome-Targeting Strategies for the Treatment of Gliomas: A Systematic Review. ACS APPLIED BIO MATERIALS 2020; 3:5500-5528. [PMID: 35021787 DOI: 10.1021/acsabm.0c00705] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant tumors represent some of the most intractable diseases that endanger human health. A glioma is a tumor of the central nervous system that is characterized by severe invasiveness, blurred boundaries between the tumor and surrounding normal tissue, difficult surgical removal, and high recurrence. Moreover, the blood-brain barrier (BBB) and multidrug resistance (MDR) are important factors that contribute to the lack of efficacy of chemotherapy in treating gliomas. A liposome is a biofilm-like drug delivery system with a unique phospholipid bilayer that exhibits high affinities with human tissues/organs (e.g., BBB). After more than five decades of development, classical and engineered liposomes consist of four distinct generations, each with different characteristics: (i) traditional liposomes, (ii) stealth liposomes, (iii) targeting liposomes, and (iv) biomimetic liposomes, which offer a promising approach to promote drugs across the BBB and to reverse MDR. Here, we review the history, preparatory methods, and physicochemical properties of liposomes. Furthermore, we discuss the mechanisms by which liposomes have assisted in the diagnosis and treatment of gliomas, including drug transport across the BBB, inhibition of efflux transporters, reversal of MDR, and induction of immune responses. Finally, we highlight ongoing and future clinical trials and applications toward further developing and testing the efficacies of liposomes in treating gliomas.
Collapse
Affiliation(s)
- Jie Li
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Tiantian Tan
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Liping Zhao
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Mengmeng Liu
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Yu You
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Yiying Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Dajing Chen
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Tian Xie
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| | - Lele Zhang
- School of Medicine, Chengdu University, Chengdu 610106, Sichuan, China
| | - Chaomei Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Zhaowu Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, Hangzhou 311121, Zhejiang, China.,Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, Zhejiang, China.,Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, Zhejiang, China
| |
Collapse
|
38
|
Fereydouni Z, Amirinezhad Fard E, Mansouri K, Mohammadi Motlagh HR, Mostafaie A. Saponins from Tribulus terrestris L. Extract Down-regulate the Expression of ICAM-1, VCAM-1 and E-selectin in Human Endothelial Cell Lines. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:73-83. [PMID: 32832486 PMCID: PMC7422852 DOI: 10.22088/ijmcm.bums.9.1.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Atherosclerosis is an inflammatory disease in which intracellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin (SELE) are consistently expressed in the vascular endothelium. Several evidence support the crucial role of adhesion molecules in the development of atherosclerosis and plaque instability. Due to the anti-inflammatory activity of Tribulus terrestris (TT), the present study investigated the effect of aqueous extract and saponin fraction of TT on the expression of ICAM-1, VCAM-1, and SELE genes in endothelial cells during normal and lipopolysaccharide (LPS) induced conditions. Human umbilical vein endothelial cells (HUVEC) and human bone marrow endothelial cells (HBMEC) were cultured, stimulated by LPS, and treated with aqueous extract and saponin fraction of TT. Finally, the expression of ICAM-1, VCAM-1, and SELE genes were measured using quantitative real-time polymerase chain reaction. LPS-induced HUVECs and HBMECs significantly increased the expression of ICAM-1, VCAM-1, and SELE in comparison with control groups (P<0.001). Treatment of LPS-induced HUVECs and HBMECs by aqueous extract and saponin fraction of TT decreased the expression of all three mentioned genes significantly (P<0.001) in comparison with LPS-induced cells. Taken together, our data suggest that TT has an anti-inflammatory effect. In vivo study about anti-inflammatory effect of this herb may provide new insights into the development of a herbal drug for the prevention/therapy of atherosclerosis.
Collapse
Affiliation(s)
- Zahra Fereydouni
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elahe Amirinezhad Fard
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hamid-Reza Mohammadi Motlagh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Mostafaie
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
39
|
Li W, Li Y, Zhao Y, Ren L. The protective effects of aloperine against ox-LDL-induced endothelial dysfunction and inflammation in HUVECs. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:107-115. [PMID: 31852304 DOI: 10.1080/21691401.2019.1699816] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is a potentially life-threatening cardiovascular disease characterized by chronic endothelial inflammation and the formation of atherosclerotic lesions. Circulating ox-LDL is known to induce atherosclerosis by triggering oxidative stress, the expression of inflammatory mediators and adhesion molecules, as well as downregulating the atheroprotective transcriptional factor KLF2. Aloperine is an alkaloid compound isolated from the plant Sophora alopecuroides. Here, we employed various experimental methods to determine the effects of aloperine on ox-LDL-induced markers of atherosclerosis. DHE staining revealed that aloperine may restore the oxidant/antioxidant balance in HUVECs by reducing the level of ROS and rescuing the reduction in NOQ-1 and GCLC induced by ox-LDL. Aloperine treatment reduced ox-LDL-induced expression of IL-6, MCP-1, VCAM-1, and E-selectin and rescued the reduction in KLF2. Aloperine also downregulated ox-LDL-induced expression of the LOX-1. We also demonstrate that aloperine improved cell viability and inhibited the adhesion of U937 monocytes to HUVECs. Finally, we demonstrate that the effects of aloperine are mediated through the rescue of KLF2 expression via suppression of the phosphorylation of p53 protein. Together, our results implicate the potential of aloperine as a safe and effective antiatherosclerosis treatment.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Obstetrics, First Hospital of China Medical University, Shenyang, China
| | - Yanshu Li
- Key Laboratory of Cell Biology of Ministry of Public Health, China Medical University, Shenyang, China
| | - Yi Zhao
- Department of Obstetrics, First Hospital of China Medical University, Shenyang, China
| | - Lina Ren
- Department of Obstetrics, First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Kheradmandi M, Ackers I, Burdick MM, Malgor R, Farnoud AM. Targeting Dysfunctional Vascular Endothelial Cells Using Immunoliposomes Under Flow Conditions. Cell Mol Bioeng 2020; 13:189-199. [PMID: 32426057 DOI: 10.1007/s12195-020-00616-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 04/24/2020] [Indexed: 02/08/2023] Open
Abstract
Introduction Atherosclerosis (ATH), the build up of fat in the arteries, is a principal cause of heart attack and stroke. Drug instability and lack of target specificity are major drawbacks of current clinical therapeutics. These undesirable effects can be eliminated by site-specific drug delivery. The endothelial surface over ATH lesions has been shown to overexpress vascular cell adhesion molecule1 (VCAM1), which can be used for targeted therapy. Methods Here, we report the synthesis, characterization, and development of anti VCAM1-functionalized liposomes to target cells overexpressing VCAM1 under static and flow conditions. Liposomes were composed of dioleoyl-phosphatidylcholine, sphingomyelin, cholesterol, and distearoyl-phosphatidylethanolamine-polyethylene glycol-cyanur (31.67:31.67:31.67:5 mol%). VCAM1 expression in endothelial cells was induced by lipopolysaccharide (LPS) treatment. Results Characterization study revealed that liposomes were negatively charged (- 7.7 ± 2.6 mV) with an average diameter of 201.3 ± 3.3 nm. Liposomes showed no toxicity toward THP-1 derived macrophages and endothelial cells. Liposomes were able to target both fixed and non-fixed endothelial cells, in vitro, with significantly higher localization observed in non-fixed conditions. To mimic biological and physiologically-relevant conditions, liposome targeting was also examined under flow (4 dyn/cm2) with or without erythrocytes (40% v/v hematocrit). Liposomes were able to target LPS-treated endothelial cells under dynamic culture, in the presence or absence of erythrocytes, although targeting efficiency was five-fold lower in flow compared to static conditions. Conclusions This liposomal delivery system showed a significant improvement in localization on dysfunctional endothelium after surface functionalization. We conclude that VCAM1-functionalized liposomes can target and potentially deliver therapeutic compounds to ATH regions.
Collapse
Affiliation(s)
- Mahsa Kheradmandi
- Department of Chemical and Biomolecular Engineering, Ohio University, 161 Stocker Center, Athens, OH 45701 USA
| | - Ian Ackers
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| | - Monica M Burdick
- Department of Chemical and Biomolecular Engineering, Ohio University, 161 Stocker Center, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| | - Ramiro Malgor
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| | - Amir M Farnoud
- Department of Chemical and Biomolecular Engineering, Ohio University, 161 Stocker Center, Athens, OH 45701 USA.,Translational Biomedical Science Program, Ohio University, Athens, OH 45701 USA
| |
Collapse
|
41
|
Wang S, Wang F, Yang L, Li Q, Huang Y, Cheng Z, Chu H, Song Y, Shang L, Hao W, Wei X. Effects of coal-fired PM 2.5 on the expression levels of atherosclerosis-related proteins and the phosphorylation level of MAPK in ApoE -/- mice. BMC Pharmacol Toxicol 2020; 21:34. [PMID: 32384920 PMCID: PMC7206822 DOI: 10.1186/s40360-020-00411-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
Background Air pollution increases the morbidity and mortality of cardiovascular disease (CVD). Atherosclerosis (AS) is the pathological basis of most CVD, and the progression of atherosclerosis and the increase of fragile plaque rupture are the mechanism basis of the relationship between atmospheric particulate pollution and CVD. The aim of the present study was to investigate the effects of coal-fired fine particulate matter (PM2.5) on the expression levels of atherosclerosis-related proteins (von Willebrand factor (vWF), Endothelin-1 (ET-1), intercellular adhesion molecule-1 (ICAM-1), and E-selectin, and to explore the role and mechanism of the progression of atherosclerosis induced by coal-fired PM2.5 via the mitogen-activated protein kinase (MAPK) signaling pathways. Methods Different concentrations of PM2.5 were given to apolipoprotein-E knockout (ApoE−/−) mice via intratracheal instillation for 8 weeks. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of vWF, ET-1 in serum of mice. Immunohistochemistry was used to observe the expression and distribution of ICAM-1 and E-selectin in the aorta of mice. Western blot was used to investigate the phosphoylation of proteins relevant to MAPK signaling pathways. Results Coal-fired PM2.5 exacerbated atherosclerosis induced by a high-fat diet. Fibrous cap formation, foam cells accumulation, and atherosclerotic lesions were observed in the aortas of PM2.5-treated mice. Coal-fired PM2.5 increased the protein levels of ET-1, ICAM-1, and E-selectin, but there was no significant difference in the vWF levels between the PM2.5-treatment mice and the HFD control mice. Coal-fired PM2.5 promoted the phosphorylation of p38, c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK) in aortic tissues of mice. Conclusion Coal-derived PM2.5 exacerbated the formation of atherosclerosis in mice, increased the expression levels of atherosclerosis-related proteins in mice serum, and promoted the phosphorylation of proteins relevant to MAPK signaling pathway. Thus, MAPK signaling pathway may play a role in the atherosclerosis pathogenesis induced by Coal-derived PM2.5.
Collapse
Affiliation(s)
- Siqi Wang
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Feifei Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, People's Republic of China
| | - Lixin Yang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, People's Republic of China
| | - Qin Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing, 100012, People's Republic of China
| | - Yao Huang
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Zhiyuan Cheng
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Hongqian Chu
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, People's Republic of China.,Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, People's Republic of China
| | - Yiming Song
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Lanqin Shang
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China.,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University Health Science Center, No.38 XueYuan Road, HaiDian District, Beijing, 100191, People's Republic of China. .,Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, 100191, People's Republic of China.
| |
Collapse
|
42
|
Zukerman H, Khoury M, Shammay Y, Sznitman J, Lotan N, Korin N. Targeting functionalized nanoparticles to activated endothelial cells under high wall shear stress. Bioeng Transl Med 2020; 5:e10151. [PMID: 32440559 PMCID: PMC7237145 DOI: 10.1002/btm2.10151] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/12/2019] [Accepted: 12/03/2019] [Indexed: 11/26/2022] Open
Abstract
Local inflammation of the endothelium is associated with a plethora of cardiovascular diseases. Vascular-targeted carriers (VTCs) have been advocated to provide focal effective therapeutics to these disease sites. Here, we examine the design of functionalized nanoparticles (NPs) as VTCs that can specifically localize at an inflamed vessel wall under pathological levels of high shear stress, associated for example with clinical (or in vivo) conditions of vascular narrowing and arteriogenesis. To test this, carboxylated fluorescent 200 nm polystyrene particles were functionalized with ligands to activated endothelium, that is, an E-selectin binding peptide (Esbp), an anti ICAM-1 antibody, or using a combination of both. The functionalized NPs were investigated in vitro using microfluidic models lined with inflamed (TNF-α stimulated) and control endothelial cells (EC). Specifically, their adhesion was monitored under different relevant wall shear stresses (i.e., 40-300 dyne/cm2) via real-time confocal microscopy. Experiments reveal a significantly higher specific adhesion of the examined functionalized NPs to activated EC for the window of examined wall shear stresses. Moreover, particle adhesion correlated with the surface coating density whereby under high surface coating (i.e., ~10,000 molecule/particle), shear-dependent particle adhesion increased significantly. Altogether, our results show that functionalized NPs can be designed to target inflamed endothelial cells under high shear stress. Such VTCs underscore the potential for attractive avenues in targeting drugs to vasoconstriction and arteriogenesis sites.
Collapse
Affiliation(s)
- Hila Zukerman
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Maria Khoury
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Yosi Shammay
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Josué Sznitman
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Noah Lotan
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| | - Netanel Korin
- Department of Biomedical EngineeringTechnion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
43
|
Emami N, Pakchin PS, Ferdousi R. Computational predictive approaches for interaction and structure of aptamers. J Theor Biol 2020; 497:110268. [PMID: 32311376 DOI: 10.1016/j.jtbi.2020.110268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/27/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Aptamers are short single-strand sequences that can bind to their specific targets with high affinity and specificity. Usually, aptamers are selected experimentally via systematic evolution of ligands by exponential enrichment (SELEX), an evolutionary process that consists of multiple cycles of selection and amplification. The SELEX process is expensive, time-consuming, and its success rates are relatively low. To overcome these difficulties, in recent years, several computational techniques have been developed in aptamer sciences that bring together different disciplines and branches of technologies. In this paper, a complementary review on computational predictive approaches of the aptamer has been organized. Generally, the computational prediction approaches of aptamer have been proposed to carry out in two main categories: interaction-based prediction and structure-based predictions. Furthermore, the available software packages and toolkits in this scope were reviewed. The aim of describing computational methods and tools in aptamer science is that aptamer scientists might take advantage of these computational techniques to develop more accurate and more sensitive aptamers.
Collapse
Affiliation(s)
- Neda Emami
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Samadi Pakchin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
44
|
Wang P, Kim T, Harada M, Contag C, Huang X, Smith BR. Nano-immunoimaging. NANOSCALE HORIZONS 2020; 5:628-653. [PMID: 32226975 DOI: 10.1039/c9nh00514e] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunoimaging is a rapidly growing field stoked in large part by the intriguing triumphs of immunotherapy. On the heels of immunotherapy's successes, there exists a growing need to evaluate tumor response to therapy particularly immunotherapy, stratify patients into responders vs. non-responders, identify inflammation, and better understand the fundamental roles of immune system components to improve both immunoimaging and immunotherapy. Innovative nanomaterials have begun to provide novel opportunities for immunoimaging, in part due to their sensitivity, modularity, capacity for many potentially varied ligands (high avidity), and potential for multifunctionality/multimodality imaging. This review strives to comprehensively summarize the integration of nanotechnology and immunoimaging, and the field's potential for clinical applications.
Collapse
Affiliation(s)
- Ping Wang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA
| | - Taeho Kim
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 488824, USA
| | - Xuefei Huang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Chemistry, Michigan State University, East Lansing, MI 488824, USA
| | - Bryan Ronain Smith
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Radiology, Stanford University, Stanford, CA 94306, USA
| |
Collapse
|
45
|
Camacho OM, Hedge A, Lowe F, Newland N, Gale N, McEwan M, Proctor C. Statistical analysis plan for "A randomised, controlled study to evaluate the effects of switching from cigarette smoking to using a tobacco heating product on health effect indicators in healthy subjects". Contemp Clin Trials Commun 2020; 17:100535. [PMID: 32072070 PMCID: PMC7013164 DOI: 10.1016/j.conctc.2020.100535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/10/2020] [Accepted: 01/26/2020] [Indexed: 02/06/2023] Open
Abstract
Tobacco harm reduction strategies aim to substitute smoking with potentially reduced risk products (PRRPs) such as e-cigarettes and tobacco-heating products (THPs). The health benefits of switching from smoking to PRRPs is unknown. A randomised controlled trial is being conducted to increase understanding of the health effects of switching from smoking to a THP in a 12-month long ambulatory study (ISRCTN81075760). Here we describe the study endpoints and the statistical analysis plan. Endpoints are divided into biomarkers of exposure (BoE) to tobacco smoke constituents and health effect indicators related to risk of lung cancer, cardiovascular and obstructive lung disease. These have been selected on the basis of extensive literature evidence. Three primary endpoints, augmentation index (risk factor for cardiovascular disease), total NNAL (linked to lung cancer) and 8-Epi-PGF2α type III (indicator of oxidative stress linked to various diseases), and multiple secondary endpoints will be analysed at 90, 180, and 360 days. Changes from baseline will be compared between study arms by specific contrasts in mixed models. Study wise multiple comparisons adjustments will be performed to account for multiplicity of timepoints and comparisons within timepoints. Generalisability of outcomes will be tested by a sensitivity analysis adjusting for age and gender. Importantly, an ancillary analysis will be performed to assess product compliance during the study based on plasma levels of CEVal, a surrogate marker for acrylonitrile exposure. The rationale underlying the selection of BoEs and health effect indicators, coupled with the statistical analysis plan will be central to understanding the potential health effects of replacing smoking with THP use for one year.
Collapse
Affiliation(s)
- Oscar M. Camacho
- British American Tobacco Investments Ltd, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| | - Andrew Hedge
- Covance Clinical Research Unit Ltd, Springfield House, Hyde Street, Leeds, Yorkshire, LS2 9LH, UK
| | - Frazer Lowe
- British American Tobacco Investments Ltd, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| | - Nik Newland
- British American Tobacco Investments Ltd, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| | - Nathan Gale
- British American Tobacco Investments Ltd, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| | - Mike McEwan
- British American Tobacco Investments Ltd, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| | - Christopher Proctor
- British American Tobacco Investments Ltd, Regents Park Road, Southampton, Hampshire, SO15 8TL, UK
| |
Collapse
|
46
|
Mohd Ariff A, Abu Bakar NA, Abd Muid S, Omar E, Ismail NH, Ali AM, Mohd Kasim NA, Mohd Nawawi H. Ficus deltoidea suppresses endothelial activation, inflammation, monocytes adhesion and oxidative stress via NF-κB and eNOS pathways in stimulated human coronary artery endothelial cells. BMC Complement Med Ther 2020; 20:56. [PMID: 32066426 PMCID: PMC7076839 DOI: 10.1186/s12906-020-2844-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/06/2020] [Indexed: 12/25/2022] Open
Abstract
Background Ficus deltoidea (FD) has been shown to have antidiabetic, anti-inflammatory, antinociceptive and antioxidant properties. However, its effects on key events in the pathogenesis of atherosclerosis are unknown. Aim To investigate the endothelial activation, inflammation, monocyte-endothelial cell binding and oxidative stress effects of four FD varieties. Methods Human coronary artery endothelial cells (HCAEC) were incubated with different concentrations of aqueous ethanolic extracts of FD var. trengganuensis (FDT), var. kunstleri (FDK), var. deltoidea (FDD) and var. intermedia (FDI), together with LPS. Protein and gene expression of vascular cell adhesion molecule-1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), endothelial-leukocyte adhesion molecule-1 (E-selectin), interleukin-6 (IL-6), Nuclear factor-κB (NF-κB) p50 and p65 and endothelial nitric oxide synthase (eNOS) were measured using ELISA and QuantiGene plex, respectively. Adhesion of monocyte to HCAEC and formation of reactive oxygen species (ROS) were detected by Rose Bengal staining and 2′-7′-dichlorofluorescein diacetate (DCFH-DA) assay. Results FDK exhibited the highest inhibition of biomarkers in relation to endothelial activation and inflammation, second in reducing monocyte binding (17.3%) compared to other varieties. FDK (25.6%) was also the most potent at decreasing ROS production. Conclusion FD has anti-atherogenic effects, possibly mediated by NF-κB and eNOS pathways; with FDK being the most potent variety. It is potentially beneficial in mitigating atherogenesis.
Collapse
Affiliation(s)
- Amirah Mohd Ariff
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia
| | - Nurul Ain Abu Bakar
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia
| | - Suhaila Abd Muid
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia.,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Effat Omar
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia.,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Nor Hadiani Ismail
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia
| | - Abdul Manaf Ali
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin (UniSZA), 20300, Kuala Terengganu, Terengganu, Malaysia
| | - Noor Alicezah Mohd Kasim
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia.,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Hapizah Mohd Nawawi
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia. .,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia.
| |
Collapse
|
47
|
A prospective study in women: açaí (Euterpe oleracea Martius) dietary intake affects serum p-selectin, leptin, and visfatin levels. NUTR HOSP 2020; 38:121-127. [PMID: 33319583 DOI: 10.20960/nh.03342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Introduction Background: açaí is the fruit of the palm tree Euterpe oleracea Martius, which is native to the Amazon region. This fruit has been extensively studied due to its potential effects on human health. Studies have also evaluated the potential effect of açaí on the inflammatory response, but there are still few studies that have assessed this property in humans. Objective: in this study we aimed to evaluate the effects of 200 g of açaí pulp consumption per day during four weeks on a rich panel of inflammatory biomarkers. Methods: a prospective nutritional intervention study was conducted on forty apparently healthy women who consumed 200 g of açaí pulp per day for four weeks. A panel of serum inflammatory markers were evaluated before and after the nutritional intervention, namely, cell adhesion molecules (ICAM-1, IVAM-1, P-selectin, MCP-1, and fractalkine), interleukins (IL-1β, IL-6, IL-8, IL-10, and IL-17) and adipokines (adiponectin, leptin, visfatin, and adipsin). The data were analyzed using paired Student's t-test to evaluate the effect of the intervention using PASW Statistics, version 17.0, and a p-value of < 0.05 was considered significant. Results: four weeks of açaí pulp consumption decreased p-selectin, leptin, and visfatin concentrations in the serum of the participating women. Conclusion: these results show that consumption of açaí pulp was able to modulate important biomarkers of the inflammatory process in apparently healthy women.
Collapse
|
48
|
Nurhidayah D, Maruf A, Zhang X, Liao X, Wu W, Wang G. Advanced drug-delivery systems: mechanoresponsive nanoplatforms applicable in atherosclerosis management. Nanomedicine (Lond) 2019; 14:3105-3122. [PMID: 31823682 DOI: 10.2217/nnm-2019-0172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Nanoplatforms have been used extensively as advanced carriers to enhance the effectiveness of drug delivery, mostly through passive aggregation provided by the enhanced permeability and retention effect. Mechanical stimuli provide a robust strategy to bolster drug delivery performance by increasing the accumulation of nanoplatforms at the lesion sites, facilitating on-demand cargo release and providing theranostic aims. In this review, we focus on recent advances of mechanoresponsive nanoplatforms that can accomplish targeted drug delivery, and subsequent drug release, under specific stimuli, either endogenous (shear stress) or exogenous (magnetic field and ultrasound), to synergistically combat atherosclerosis at the molecular level.
Collapse
Affiliation(s)
- Deti Nurhidayah
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Ali Maruf
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science & Technology, Chongqing 401331, China
| | - Wei Wu
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science & Technology of Ministry of Education, State & Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| |
Collapse
|
49
|
The Many Roles of Cell Adhesion Molecules in Hepatic Fibrosis. Cells 2019; 8:cells8121503. [PMID: 31771248 PMCID: PMC6952767 DOI: 10.3390/cells8121503] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 01/09/2023] Open
Abstract
Fibrogenesis is a progressive scarring event resulting from disrupted regular wound healing due to repeated tissue injury and can end in organ failure, like in liver cirrhosis. The protagonists in this process, either liver-resident cells or patrolling leukocytes attracted to the site of tissue damage, interact with each other by soluble factors but also by direct cell–cell contact mediated by cell adhesion molecules. Since cell adhesion molecules also support binding to the extracellular matrix, they represent excellent biosensors, which allow cells to modulate their behavior based on changes in the surrounding microenvironment. In this review, we focus on selectins, cadherins, integrins and members of the immunoglobulin superfamily of adhesion molecules as well as some non-classical cell adhesion molecules in the context of hepatic fibrosis. We describe their liver-specific contributions to leukocyte recruitment, cell differentiation and survival, matrix remodeling or angiogenesis and touch on their suitability as targets in antifibrotic therapies.
Collapse
|
50
|
Rashad NM, El-Shal AS, Abomandour HG, Aboelfath AMK, Rafeek MES, Badr MS, Ali AE, Yousef MS, Fathy MA, Sharaf El Din MTA. Intercellular adhesion molecule-1 expression and serum levels as markers of pre-clinical atherosclerosis in polycystic ovary syndrome. J Ovarian Res 2019; 12:97. [PMID: 31629408 PMCID: PMC6800507 DOI: 10.1186/s13048-019-0566-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/10/2019] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder characterized by obesity, hyperandrogenism, and insulin resistance. Intercellular adhesion molecule-1 (ICAM-1) is a proinflammatory and proatherogenic cytokine which is associated with atherosclerosis, insulin resistance, and cardiovascular disease (CVD). The pathogenesis of PCOS is not precisely known. Thus, the purpose of this study was to investigate the potential role of ICAM-1 expression and serum ICAM-1 concentrations in pathogenesis of PCOS. Moreover, we aimed to evaluate the possible relationship between ICAM-1 gene expression with carotid intima-media thickness as well as clinic-morphological features of PCOS. METHODS This case control study enrolled 180 patients with PCOS and 120 controls groups and they were stratified according to their fasting plasma glucose (FPG) into three subgroups; normal glucose tolerance (NGT) [n = 75], those with impaired glucose tolerance (IGT) [n = 65], and 40 patients with type 2 diabetes mellitus (T2DM). Circulating ICAM-1 expression levels were determined by real time polymerase chain reaction (RT-PCR). Serum ICAM-1 concentrations were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS Our results revealed that PCOS patients had higher values of ICAM-1expression and serum levels. Among PCOS patients, T2DM patients had the highest values of ICAM-1 expression and serum levels compared to IGT and NGT subgroups. The ICAM-1 expression and serum levels were significantly positive correlated with cardiovascular risk and PCOS phenotypes. Linear regression test showed that HOMA-IR was the main predictors of serum ICAM-1 levels in PCOS. Receiver operating characteristic curve (ROC) analysis revealed that, the power of ICAM-1 expression levels was higher than serum ICAM-1 in diagnosis of PCOS and in differentiating T2DM from IGT and NGT subgroups. Interestingly, combination of both ICAM-1 expression and serum levels improved the diagnostic role of serum ICAM-1. CONCLUSION ICAM-1 expression and serum levels were higher in women with PCOS compared to control group also, there was a strong independent association between higher ICAM-1 expression and serum levels with cardiovascular risks in PCOS group.
Collapse
Affiliation(s)
- Nearmeen M Rashad
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Amal S El-Shal
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hala G Abomandour
- Cardiology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Mohamed El Sayed Rafeek
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohammad Samir Badr
- Obstetrics and Gynecology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ayman E Ali
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohammed S Yousef
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | | |
Collapse
|