151
|
Wieczorek E, Ożyhar A. Transthyretin: From Structural Stability to Osteoarticular and Cardiovascular Diseases. Cells 2021; 10:1768. [PMID: 34359938 PMCID: PMC8307983 DOI: 10.3390/cells10071768] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/29/2021] [Accepted: 07/09/2021] [Indexed: 01/10/2023] Open
Abstract
Transthyretin (TTR) is a tetrameric protein transporting hormones in the plasma and brain, which has many other activities that have not been fully acknowledged. TTR is a positive indicator of nutrition status and is negatively correlated with inflammation. TTR is a neuroprotective and oxidative-stress-suppressing factor. The TTR structure is destabilized by mutations, oxidative modifications, aging, proteolysis, and metal cations, including Ca2+. Destabilized TTR molecules form amyloid deposits, resulting in senile and familial amyloidopathies. This review links structural stability of TTR with the environmental factors, particularly oxidative stress and Ca2+, and the processes involved in the pathogenesis of TTR-related diseases. The roles of TTR in biomineralization, calcification, and osteoarticular and cardiovascular diseases are broadly discussed. The association of TTR-related diseases and vascular and ligament tissue calcification with TTR levels and TTR structure is presented. It is indicated that unaggregated TTR and TTR amyloid are bound by vicious cycles, and that TTR may have an as yet undetermined role(s) at the crossroads of calcification, blood coagulation, and immune response.
Collapse
Affiliation(s)
- Elżbieta Wieczorek
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wroclaw, Poland;
| | | |
Collapse
|
152
|
Chen L, Dai L, Yan D, Zhou B, Zheng W, Yin J, Zhou T, Liu Z, Deng J, Wang R, Ding X, Chen J. Gleevec and Rapamycin Synergistically Reduce Cell Viability and Inhibit Proliferation and Angiogenic Function of Mouse Bone Marrow-Derived Endothelial Progenitor Cells. J Vasc Res 2021; 58:330-342. [PMID: 34247157 DOI: 10.1159/000515816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/08/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study investigates the synergistic effects of Gleevec (imatinib) and rapamycin on the proliferative and angiogenic properties of mouse bone marrow-derived endothelial progenitor cells (EPCs). MATERIALS AND METHODS EPCs were isolated from mouse bone marrow and treated with different concentrations of Gleevec or rapamycin individually or in combination. The cell viability and proliferation were examined using the MTT assay. An analysis of cell cycle and apoptosis was performed using flow cytometry. Formation of capillary-like tubes was examined in vitro, and the protein expression of cell differentiation markers was determined using Western blot analysis. RESULTS Gleevec significantly reduced cell viability, cell proliferation, and induced cell apoptosis in EPCs. Rapamycin had similar effects on EPCs, but it did not induce cell apoptosis. The combination of Gleevec and rapamycin reduced the cell proliferation but increased cell apoptosis. Although rapamycin had no demonstratable effect on tube formation, the combined therapy of Gleevec and rapamycin significantly reduced tube formation when compared with Gleevec alone. Mechanistically, Gleevec, but not rapamycin, induced a significant elevation in caspase-3 activity in EPCs, and it attenuated the expression of the endothelial protein marker platelet-derived growth factor receptor α. Functionally, rapamycin, but not Gleevec, significantly enhanced the expression of endothelial differentiation marker proteins, while attenuating the expression of mammalian target of rapamycin signaling-related proteins. CONCLUSIONS Gleevec and rapamycin synergistically suppress cell proliferation and tube formation of EPCs by inducing cell apoptosis and endothelial differentiation. Mechanistically, it is likely that rapamycin enhances the proapoptotic and antiangiogenic effects of Gleevec by promoting the endothelial differentiation of EPCs. Given that EPCs are involved in the pathogenesis of some cardiovascular diseases and critical to angiogenesis, pharmacological inhibition of EPC proliferation by combined Gleevec and rapamycin therapy may be a promising approach for suppressing cardiovascular disease pathologies associated with angiogenesis.
Collapse
Affiliation(s)
- Ling Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Luping Dai
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Dewen Yan
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Boya Zhou
- Department of Ultrasound, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Wei Zheng
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jia Yin
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| | - Tao Zhou
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Zehua Liu
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jianxin Deng
- Department of Endocrinology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center of Shenzhen University, Shenzhen, China
| | - Rehua Wang
- Department of Cardiology, Fujian Provincial Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaorong Ding
- Nursing Department, Peking University Shenzhen Hospital, Shenzhen, China
| | - Junhui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
153
|
Kai H, Wu Q, Yin R, Tang X, Shi H, Wang T, Zhang M, Pan C. LncRNA NORAD Promotes Vascular Endothelial Cell Injury and Atherosclerosis Through Suppressing VEGF Gene Transcription via Enhancing H3K9 Deacetylation by Recruiting HDAC6. Front Cell Dev Biol 2021; 9:701628. [PMID: 34307380 PMCID: PMC8301222 DOI: 10.3389/fcell.2021.701628] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/08/2021] [Indexed: 11/15/2022] Open
Abstract
Coronary artery disease (CAD) is a major atherosclerotic cardiovascular disease and the leading cause of mortality globally. Long non-coding RNAs (lncRNAs) play crucial roles in CAD development. To date, the effect of lncRNA non-coding RNA activated by DNA damage (NORAD) on atherosclerosis in CAD remains unclear. The primary aim of this study was to investigate the effect of lncRNA NORAD on vascular endothelial cell injury and atherosclerosis. Here, ox-LDL-treated human umbilical vein endothelial cells (HUVECs) and high-fat-diet (HFD)-fed ApoE–/– mice were utilized as in vitro and in vivo models. The present study found that lncRNA NORAD expression was increased in ox-LDL-treated HUVECs and thoracic aorta of atherosclerotic mice, and knockdown of lncRNA NORAD alleviated vascular endothelial cell injury and atherosclerosis development in vitro and in vivo. Knockdown of lncRNA NORAD aggravated ox-LDL-reduced or atherosclerosis-decreased vascular endothelial growth factor (VEGF) expression in HUVECs and thoracic aorta of mice to ameliorate vascular endothelial cell injury and atherosclerosis development. Moreover, nucleus lncRNA NORAD suppressed VEGF gene transcription through enhancing H3K9 deacetylation via recruiting HDAC6 to the VEGF gene promoter in ox-LDL-treated HUVECs. In addition, VEGF reduced FUS (FUS RNA binding protein) expression by a negative feedback regulation in HUVECs. In summary, lncRNA NORAD enhanced vascular endothelial cell injury and atherosclerosis through suppressing VEGF gene transcription via enhancing H3K9 deacetylation by recruiting HDAC6. The findings could facilitate discovering novel diagnostic markers and therapeutic targets for CAD.
Collapse
Affiliation(s)
- Huihua Kai
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Qiyong Wu
- Department of Thoracic and Cardiac Surgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ruohan Yin
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Xiaoqiang Tang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Tao Wang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ming Zhang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Changjie Pan
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
154
|
Steinkamp PJ, Vonk J, Huisman LA, Meersma GJ, Diercks GFH, Hillebrands JL, Nagengast WB, Zeebregts CJ, Slart RHJA, Boersma HH, van Dam GM. VEGF-Targeted Multispectral Optoacoustic Tomography and Fluorescence Molecular Imaging in Human Carotid Atherosclerotic Plaques. Diagnostics (Basel) 2021; 11:diagnostics11071227. [PMID: 34359310 PMCID: PMC8305003 DOI: 10.3390/diagnostics11071227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/15/2022] Open
Abstract
Vulnerable atherosclerotic carotid plaques are prone to rupture, resulting in ischemic strokes. In contrast to radiological imaging techniques, molecular imaging techniques have the potential to assess plaque vulnerability by visualizing diseases-specific biomarkers. A risk factor for rupture is intra-plaque neovascularization, which is characterized by overexpression of vascular endothelial growth factor-A (VEGF-A). Here, we study if administration of bevacizumab-800CW, a near-infrared tracer targeting VEGF-A, is safe and if molecular assessment of atherosclerotic carotid plaques in vivo is possible using multispectral optoacoustic tomography (MSOT). Healthy volunteers and patients with symptomatic carotid artery stenosis scheduled for carotid artery endarterectomy were imaged with MSOT. Secondly, patients were imaged two days after intravenous administration of 4.5 bevacizumab-800CW. Ex vivo fluorescence molecular imaging of the surgically removed plaque specimen was performed and correlated with histopathology. In this first-in-human MSOT and fluorescence molecular imaging study, we show that administration of 4.5 mg bevacizumab-800CW appeared to be safe in five patients and accumulated in the carotid atherosclerotic plaque. Although we could visualize the carotid bifurcation area in all subjects using MSOT, bevacizumab-800CW-resolved signal could not be detected with MSOT in the patients. Future studies should evaluate tracer safety, higher doses of bevacizumab-800CW or develop dedicated contrast agents for carotid atherosclerotic plaque assessment using MSOT.
Collapse
Affiliation(s)
- Pieter J. Steinkamp
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (P.J.S.); (L.A.H.); (C.J.Z.)
| | - Jasper Vonk
- Department of Oral & Maxillofacial Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Lydian A. Huisman
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (P.J.S.); (L.A.H.); (C.J.Z.)
| | - Gert-Jan Meersma
- Department of Pathology & Medical Biology, Pathology Division, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (G.-J.M.); (G.F.H.D.); (J.-L.H.)
| | - Gilles F. H. Diercks
- Department of Pathology & Medical Biology, Pathology Division, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (G.-J.M.); (G.F.H.D.); (J.-L.H.)
| | - Jan-Luuk Hillebrands
- Department of Pathology & Medical Biology, Pathology Division, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (G.-J.M.); (G.F.H.D.); (J.-L.H.)
| | - Wouter B. Nagengast
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands;
| | - Clark J. Zeebregts
- Department of Surgery, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (P.J.S.); (L.A.H.); (C.J.Z.)
| | - Riemer H. J. A. Slart
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (R.H.J.A.S.); (H.H.B.)
- Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, 7522 ND Enschede, The Netherlands
| | - Hendrikus H. Boersma
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (R.H.J.A.S.); (H.H.B.)
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Gooitzen M. van Dam
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands; (R.H.J.A.S.); (H.H.B.)
- AxelaRx/TRACER BV, 9700 RB Groningen, The Netherlands
- Correspondence: ; Tel.: +31-50-361-12283; Fax: +31-50-361-4873
| |
Collapse
|
155
|
Ullah K, Wu R. Hypoxia-Inducible Factor Regulates Endothelial Metabolism in Cardiovascular Disease. Front Physiol 2021; 12:670653. [PMID: 34290616 PMCID: PMC8287728 DOI: 10.3389/fphys.2021.670653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells (ECs) form a physical barrier between the lumens and vascular walls of arteries, veins, capillaries, and lymph vessels; thus, they regulate the extravasation of nutrients and oxygen from the circulation into the perivascular space and participate in mechanisms that maintain cardiovascular homeostasis and promote tissue growth and repair. Notably, their role in tissue repair is facilitated, at least in part, by their dependence on glycolysis for energy production, which enables them to resist hypoxic damage and promote angiogenesis in ischemic regions. ECs are also equipped with a network of oxygen-sensitive molecules that collectively activate the response to hypoxic injury, and the master regulators of the hypoxia response pathway are hypoxia-inducible factors (HIFs). HIFs reinforce the glycolytic dependence of ECs under hypoxic conditions, but whether HIF activity attenuates or exacerbates the progression and severity of cardiovascular dysfunction varies depending on the disease setting. This review summarizes how HIF regulates the metabolic and angiogenic activity of ECs under both normal and hypoxic conditions and in a variety of diseases that are associated with cardiovascular complications.
Collapse
Affiliation(s)
- Karim Ullah
- Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Rongxue Wu
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
156
|
Janaszak-Jasiecka A, Siekierzycka A, Płoska A, Dobrucki IT, Kalinowski L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021; 11:biom11070982. [PMID: 34356605 PMCID: PMC8301841 DOI: 10.3390/biom11070982] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. The initial stage of CVDs is characterized by endothelial dysfunction, defined as the limited bioavailability of nitric oxide (NO). Thus, any factors that interfere with the synthesis or metabolism of NO in endothelial cells are involved in CVD pathogenesis. It is well established that hypoxia is both the triggering factor as well as the accompanying factor in cardiovascular disease, and diminished tissue oxygen levels have been reported to influence endothelial NO bioavailability. In endothelial cells, NO is produced by endothelial nitric oxide synthase (eNOS) from L-Arg, with tetrahydrobiopterin (BH4) as an essential cofactor. Here, we discuss the mechanisms by which hypoxia affects NO bioavailability, including regulation of eNOS expression and activity. What is particularly important is the fact that hypoxia contributes to the depletion of cofactor BH4 and deficiency of substrate L-Arg, and thus elicits eNOS uncoupling-a state in which the enzyme produces superoxide instead of NO. eNOS uncoupling and the resulting oxidative stress is the major driver of endothelial dysfunction and atherogenesis. Moreover, hypoxia induces impairment in mitochondrial respiration and endothelial cell activation; thus, oxidative stress and inflammation, along with the hypoxic response, contribute to the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Laboratory of Trace Elements Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Iwona T. Dobrucki
- University of Illinois at Urbana-Champaign Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL 61801, USA;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
- Correspondence:
| |
Collapse
|
157
|
Yan B, Guo Y, Gui Y, Jiang ZS, Zheng XL. Multifunctional RNase MCPIP1 and its Role in Cardiovascular Diseases. Curr Med Chem 2021; 28:3385-3405. [PMID: 33191882 DOI: 10.2174/0929867327999201113100918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/20/2020] [Accepted: 10/09/2020] [Indexed: 11/22/2022]
Abstract
Monocyte chemoattractant protein-1 induced protein 1 (MCPIP1), one of the MCPIP family members, is characterized by the presence of both C-x8-C-x5-C-x3-H (CCCH)- type zinc finger and PilT-N-terminal domains. As a potent regulator of innate immunity, MCPIP1 exerts anti-inflammatory effects through its ribonuclease (RNase) and deubiquitinating enzyme activities to degrade cytokine mRNAs and inhibit nuclear factor- kappa B (NF-κB), respectively. MCPIP1 is expressed not only in immune cells but also in many other cell types, including cardiomyocytes, vascular endothelial cells (ECs) and smooth muscle cells (SMCs). Increasing evidence indicates that MCPIP1 plays a role in the regulation of cardiac functions and is involved in the processes of vascular diseases, such as ischemia-reperfusion (I/R) and atherosclerosis. To better understand the emerging roles of MCPIP1 in the cardiovascular system, we reviewed the current literature with respect to MCPIP1 functions and discussed its association with the pathogenesis of cardiovascular diseases and the implication as a therapeutic target.
Collapse
Affiliation(s)
- Binjie Yan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, China
| | - Yanan Guo
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary, ABT2N 4N1, Canada
| | - Yu Gui
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary, ABT2N 4N1, Canada
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, University of South China, Hengyang City, Hunan Province 421001, China
| | - Xi-Long Zheng
- Departments of Biochemistry & Molecular Biology and Physiology & Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary, ABT2N 4N1, Canada
| |
Collapse
|
158
|
The BMP Pathway in Blood Vessel and Lymphatic Vessel Biology. Int J Mol Sci 2021; 22:ijms22126364. [PMID: 34198654 PMCID: PMC8232321 DOI: 10.3390/ijms22126364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) were originally identified as the active components in bone extracts that can induce ectopic bone formation. In recent decades, their key role has broadly expanded beyond bone physiology and pathology. Nowadays, the BMP pathway is considered an important player in vascular signaling. Indeed, mutations in genes encoding different components of the BMP pathway cause various severe vascular diseases. Their signaling contributes to the morphological, functional and molecular heterogeneity among endothelial cells in different vessel types such as arteries, veins, lymphatic vessels and capillaries within different organs. The BMP pathway is a remarkably fine-tuned pathway. As a result, its signaling output in the vessel wall critically depends on the cellular context, which includes flow hemodynamics, interplay with other vascular signaling cascades and the interaction of endothelial cells with peri-endothelial cells and the surrounding matrix. In this review, the emerging role of BMP signaling in lymphatic vessel biology will be highlighted within the framework of BMP signaling in the circulatory vasculature.
Collapse
|
159
|
Boswell-Patterson CA, Hétu MF, Kearney A, Pang SC, Tse MY, Herr JE, Spence M, Zhou J, Johri AM. Vascularized Carotid Atherosclerotic Plaque Models for the Validation of Novel Methods of Quantifying Intraplaque Neovascularization. J Am Soc Echocardiogr 2021; 34:1184-1194. [PMID: 34129920 DOI: 10.1016/j.echo.2021.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Intraplaque neovascularization (IPN) in advanced lesions of the carotid artery has been linked to plaque progression and risk of rupture. Quantitative measurement of IPN may provide a more powerful tool for the detection of such "vulnerable" plaque than the current visual scoring method. The aim of this study was to develop a phantom platform of a neovascularized atherosclerotic plaque within a carotid artery to assess new methods of quantifying IPN. METHODS Ninety-two synthetic plaque models with various IPN architectures representing different ranges of IPN scoring were created and assessed using contrast-enhanced ultrasound. Intraplaque neovascularization volume was calculated from contrast infiltration in B mode. The plaque models were used to develop a testing platform for IPN quantification. A neovascularized enhancement ratio (NER) was calculated using commercially available software. The plaque model NERs were then compared to human plaque NERs (n = 42) to assess score relationship. Parametric mapping of dynamic intensity over time was used to differentiate IPN from calcified plaque regions. RESULTS A positive correlation between NER and IPN volume (rho = 0.45; P < .0001) was found in the plaque models. Enhancement of certain plaque model types showed that they resembled human plaques, with visual grade scores of 0 (NER mean difference = 1.05 ± SE 2.45; P = .67), 1 (NER mean difference = 0.22 ± SE 3.26; P = .95), and 2 (NER mean difference = -0.84 ± SE 3.33; P = .80). An optimal cutoff for NER (0.355) identified grade 2 human plaques with a sensitivity of 95% and specificity of 91%. CONCLUSIONS We developed a carotid artery model of neovascularized plaque and established a quantitative method for IPN using commercially available technology. We also developed an analysis method to quantify IPN in calcified plaques. This novel tool has the potential to improve clinical identification of vulnerable plaques, providing objective measures of IPN for cardiovascular risk assessment.
Collapse
Affiliation(s)
| | - Marie-France Hétu
- Department of Medicine, Cardiovascular Imaging Network at Queen's, Queen's University, Kingston, Ontario, Canada
| | - Abigail Kearney
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Stephen C Pang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - M Yat Tse
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Julia E Herr
- Department of Medicine, Cardiovascular Imaging Network at Queen's, Queen's University, Kingston, Ontario, Canada
| | - Michaela Spence
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jianhua Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Amer M Johri
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Medicine, Cardiovascular Imaging Network at Queen's, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
160
|
Amadio P, Cosentino N, Eligini S, Barbieri S, Tedesco CC, Sandrini L, Zarà M, Fabiocchi F, Niccoli G, Magnani G, Fracassi F, Crea F, Veglia F, Marenzi G, Barbieri SS. Potential Relation between Plasma BDNF Levels and Human Coronary Plaque Morphology. Diagnostics (Basel) 2021; 11:diagnostics11061010. [PMID: 34205863 PMCID: PMC8226920 DOI: 10.3390/diagnostics11061010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/30/2021] [Indexed: 01/13/2023] Open
Abstract
Coronary artery disease (CAD) patients are at high ischemic risk, and new biomarkers reflecting atherosclerotic disease severity and coronary plaque vulnerability are required. The Brain-Derived Neurotrophic Factor (BDNF) affects endothelial and macrophage activation suggesting its involvement in atherosclerotic plaque behavior. To investigate whether plasma BDNF is associated with in vivo coronary plaque features, assessed by optical coherence tomography (OCT), in both acute myocardial infarction (AMI) and stable angina (SA) patients, we enrolled 55 CAD patients (31 SA and 24 AMI), and 21 healthy subjects (HS). BDNF was lower in CAD patients than in HS (p < 0.0001), and it decreased with the presence, clinical acuity and severity of CAD. The greater BDNF levels were associated with OCT features of plaque vulnerability in overall CAD as well as in SA and AMI patients (p < 0.03). Specifically, in SA patients, BDNF correlated positively with macrophages’ infiltration within atherosclerotic plaque (p = 0.01) and inversely with minimal lumen area (p = 0.02). In AMI patients a negative correlation between BDNF and cap thickness was found (p = 0.02). Despite a small study population, our data suggest a relationship between BDNF and coronary plaque vulnerability, showing that vulnerable plaque is positively associated with plasma BDNF levels, regardless of the clinical CAD manifestation.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
| | - Nicola Cosentino
- Intensive Cardiac Care Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (N.C.); (G.M.)
| | - Sonia Eligini
- Unit of Metabolomics and Cellular Biochemistry of Atherothrombosis, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy;
| | - Simone Barbieri
- Unit of Biostatistics, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (S.B.); (C.C.T.); (F.V.)
| | - Calogero Claudio Tedesco
- Unit of Biostatistics, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (S.B.); (C.C.T.); (F.V.)
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
| | - Franco Fabiocchi
- Interventional Cardiology Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy;
| | - Giampaolo Niccoli
- Cardiology Unit, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (G.N.); (G.M.)
| | - Giulia Magnani
- Cardiology Unit, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy; (G.N.); (G.M.)
| | - Francesco Fracassi
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (F.F.); (F.C.)
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (F.F.); (F.C.)
| | - Fabrizio Veglia
- Unit of Biostatistics, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (S.B.); (C.C.T.); (F.V.)
| | - Giancarlo Marenzi
- Intensive Cardiac Care Unit, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (N.C.); (G.M.)
| | - Silvia Stella Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy; (P.A.); (L.S.); (M.Z.)
- Correspondence: ; Tel.: +39-02-58002021
| |
Collapse
|
161
|
Katsi V, Magkas N, Antonopoulos A, Trantalis G, Toutouzas K, Tousoulis D. Aortic valve: anatomy and structure and the role of vasculature in the degenerative process. Acta Cardiol 2021; 76:335-348. [PMID: 32602774 DOI: 10.1080/00015385.2020.1746053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aortic valve stenosis is a degenerative disease affecting increasing number of individuals and characterised by thickening, calcification and fibrosis of the valve resulting in restricted valve motion. Degeneration of the aortic valve is no longer considered a passive deposition of calcium, but an active process that involves certain mechanisms, that is endothelial dysfunction, inflammation, increased oxidative stress, calcification, bone formation, lipid deposition, extracellular matrix (ECM) remodelling and neoangiogenesis. Accumulating evidence indicates an important role for neoangiogenesis (i.e. formation of new vessels) in the pathogenesis of aortic valve stenosis. The normal aortic valve is generally an avascular tissue supplied with oxygen and nutrients via diffusion from the circulating blood. In contrast, presence of intrinsic micro-vasculature has been demonstrated in stenotic and calcified valves. Importantly, presence and density of neovessels have been associated with inflammation, calcification and bone formation. It remains unclear whether neoangiogenesis is a compensatory mechanism aiming to counteract hypoxia and increased metabolic demands of the thickened tissue or represents an active contributor to disease progression. Data extracted mainly from animal studies are supportive of a direct detrimental effect of neoangiogenesis, however, robust evidence from human studies is lacking. Thus, there is inadequate knowledge to assess whether neoangiogenesis could serve as a future therapeutic target for a disease that no effective medical therapy exists. In this review, we present basic aspects of anatomy and structure of the normal and stenotic aortic valve and we focus on the role of valve vasculature in the natural course of valve calcification and stenosis.
Collapse
Affiliation(s)
- Vasiliki Katsi
- First Department of Cardiology, ‘Hippokration’ Hospital, Medical School, University of Athens, Athens, Greece
| | - Nikolaos Magkas
- First Department of Cardiology, ‘Hippokration’ Hospital, Medical School, University of Athens, Athens, Greece
| | - Alexios Antonopoulos
- First Department of Cardiology, ‘Hippokration’ Hospital, Medical School, University of Athens, Athens, Greece
| | - Georgios Trantalis
- First Department of Cardiology, ‘Hippokration’ Hospital, Medical School, University of Athens, Athens, Greece
| | - Konstantinos Toutouzas
- First Department of Cardiology, ‘Hippokration’ Hospital, Medical School, University of Athens, Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, ‘Hippokration’ Hospital, Medical School, University of Athens, Athens, Greece
| |
Collapse
|
162
|
Lin D, Zhang X, Zhang C, Jin Q, Jiang L. LncRNA-TCONS_00034812 is upregulated in atherosclerosis and upregulates miR-21 through methylation in vascular smooth muscle cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1005. [PMID: 34277805 PMCID: PMC8267259 DOI: 10.21037/atm-21-2632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
Background LncRNA-TCONS_00034812 is a critical player in the proliferation of aortic smooth muscle cells. It is known that artery injury plays an important role in atherosclerosis. However, the potential implication of LncRNA-TCONS_00034812 in atherosclerosis remains unclear. In this study, we collected artery specimens from patients with atherosclerosis and healthy controls to investigate the involvement of LncRNA-TCONS_00034812 in atherosclerosis. Methods Sixty patients with atherosclerosis and 60 controls, admitted at The First Hospital of Changsha (Changsha, China), between March 2017 and March 2019, were included. An artery biopsy was performed on all participants to obtain the artery specimens. Real-time quantitative PCR were performed to quantify the relative expression level of LncRNA-TCONS_00034812. Its role in atherosclerotic lesion was evaluated in (high fat diet) HFD-induced ApoE−/− mice. Moreover, human aortic smooth muscle cells (HAOSMCs) was employed to study functional role of LncRNA-TCONS_00034812 overexpression and knockdown by methylation-specific PCR and cell proliferation assay. Results Overexpression of TCONS_00034812 resulted in miR-21 upregulation and a decrease of miR-21 gene methylation. In contrast, silencing of TCONS_00034812 caused miR-21 downregulation and an increase of miR-21 gene methylation. Cell proliferation analysis indicated that the overexpression of TCONS_00034812 and miR-21 promoted cell proliferation, while silencing of TCONS_00034812 played an opposite role. Moreover, miR-21 overexpression weakened the effects of silencing TCONS_00034812 on cell proliferation. Conclusions In summary, LncRNA-TCONS_00034812 is upregulated in atherosclerotic samples, and its overexpression upregulates miR-21 through methylation in human aortic smooth muscle cells (HAOSMCs). Our study indicates that LncRNA-TCONS_00034812 could serve as a potential biomarker for diagnosis of atherosclerosis.
Collapse
Affiliation(s)
- Dongsheng Lin
- Department of Cardiovascular Medicine, The First Hospital of Changsha, Changsha, China
| | - Xian Zhang
- Department of Cardiovascular Medicine, The People's Hospital of Zhangjiajie, Zhangjiajie, China
| | - Chiyuan Zhang
- Department of Cardiovascular Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qiao Jin
- Department of Cardiovascular Medicine, University of South China Affiliated Changsha Central Hospital, Changsha, China
| | - Luping Jiang
- Department of Cardiovascular Medicine, University of South China Affiliated Changsha Central Hospital, Changsha, China
| |
Collapse
|
163
|
Fleischmann D, Goepferich A. General sites of nanoparticle biodistribution as a novel opportunity for nanomedicine. Eur J Pharm Biopharm 2021; 166:44-60. [PMID: 34087354 DOI: 10.1016/j.ejpb.2021.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
The development of nanomedical devices has led to a considerable number of clinically applied nanotherapeutics. Yet, the overall poor translation of nanoparticular concepts into marketable systems has not met the initial expectations and led to increasing criticism in recent years. Most novel nano approaches thereby use highly refined formulations including a plethora of active targeting sequences, but ultimately fail to reach their target due to a generally high off-target deposition in organs such as the liver or kidney. In this context, we argue that initial nanoparticle (NP) development should not entirely become set on conventional formulation aspects. In contrast, we propose a change of focus towards a prior analysis of general sites of NP in vivo deposition and an assessment of how accumulation in these organs or tissues can be harnessed to develop therapies for site-related pathologies. We therefore give a comprehensive overview of existing nanotherapeutic targeting strategies for specific cell types within three of the usual suspects, i.e. the liver, kidney and the vascular system. We discuss the physiological surroundings and relevant pathologies of described tissues as well as the implications for NP-mediated drug delivery. Additionally, successful cell-selective NP concepts using active targeting strategies are assessed. By bringing together both (patho)physiological aspects and concepts for cell-selective NP formulations, we hope to show a novel opportunity for the development of more promising nanotherapeutic devices.
Collapse
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany.
| |
Collapse
|
164
|
Abstract
Dynamic remodeling of the actin cytoskeleton is an essential feature for virtually all actin-dependent cellular processes, including cell migration, cell cycle progression, chromatin remodeling and gene expression, and even the DNA damage response. An altered actin cytoskeleton is a structural hallmark associated with numerous pathologies ranging from cardiovascular diseases to immune disorders, neurological diseases and cancer. The actin cytoskeleton in cells is regulated through the orchestrated actions of a myriad of actin-binding proteins. In this Review, we provide a brief overview of the structure and functions of the actin-monomer-binding protein profilin-1 (Pfn1) and then discuss how dysregulated expression of Pfn1 contributes to diseases associated with the cardiovascular system.
Collapse
Affiliation(s)
| | - David Gau
- Bioengineering, University of Pittsburgh
| | - Partha Roy
- Bioengineering, University of Pittsburgh.,Pathology, University of Pittsburgh, 306 Center for Bioengineering, University of Pittsburgh, 300 Technology Drive, Pittsburgh, PA 15219, USA
| |
Collapse
|
165
|
Cai Y, Pan J, Li Z. Mathematical modeling of intraplaque neovascularization and hemorrhage in a carotid atherosclerotic plaque. Biomed Eng Online 2021; 20:42. [PMID: 33926451 PMCID: PMC8082657 DOI: 10.1186/s12938-021-00878-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/13/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Growing experimental evidence has identified neovascularization from the adventitial vasa vasorum and induced intraplaque hemorrhage (IPH) as critical indicators during the development of vulnerable atherosclerotic plaques. In this study, we propose a mathematical model incorporating intraplaque angiogenesis and hemodynamic calculation of the microcirculation, to obtain the quantitative evaluation of the influences of intraplaque neovascularization and hemorrhage on vulnerable plaque development. A two-dimensional nine-point model of angiogenic microvasculature is generated based on the histology of a patient's carotid plaque. The intraplaque angiogenesis model includes three key cells (endothelial cells, smooth muscle cells, and macrophages) and three key chemical factors (vascular endothelial growth factors, extracellular matrix, and matrix metalloproteinase), which densities and concentrations are described by a series of reaction-diffusion equations. The hemodynamic calculation by coupling the intravascular blood flow, the extravascular plasma flow, and the transvascular transport is carried out on the generated angiogenic microvessel network. We then define the IPH area by using the plasma concentration in the interstitial tissue, as well as the extravascular transport across the capillary wall. RESULTS The simulational results reproduce a series of pathophysiological phenomena during the atherosclerotic plaque progression. It is found that the high microvessel density region at the shoulder areas and the extravascular flow across the leaky wall of the neovasculature contribute to the IPH observed widely in vulnerable plaques. The simulational results are validated by both the in vivo MR imaging data and in vitro experimental observations and show significant consistency in quantity ground. Moreover, the sensitivity analysis of model parameters reveals that the IPH area and extent can be reduced significantly by decreasing the MVD and the wall permeability of the neovasculature. CONCLUSIONS The current quantitative model could help us to better understand the roles of microvascular and intraplaque hemorrhage during the carotid plaque progression.
Collapse
Affiliation(s)
- Yan Cai
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, China.
| | - Jichao Pan
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, China
| | - Zhiyong Li
- School of Biological Sciences & Medical Engineering, Southeast University, Nanjing, China
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD, 4001, Australia
| |
Collapse
|
166
|
Shen Z, Li H. Long non-coding RNA GAS5 knockdown facilitates proliferation and impedes apoptosis by regulating miR-128-3p/FBLN2 axis in ox-LDL-induced THP-1 cells. Clin Hemorheol Microcirc 2021; 77:153-164. [PMID: 33074219 DOI: 10.3233/ch-200897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are found to involve in modulating the development of atherosclerosis (AS). But the molecular mechanism of lncRNA growth-arrest specific transcript 5 (GAS5) in AS is not fully understood. METHODS QRT-PCR was performed to measure the abundances of GAS5, miR-128-3p and fibulin 2 (FBLN2). Oxidized low-density lipoprotein (ox-LDL)-treated THP-1 cells were employed as cell models of AS. The cell proliferation and apoptosis were analyzed using CCK-8 and Flow cytometry assays, respectively. Levels of all protein were examined by western blot. The interaction among GAS5, miR-128-3p and FBLN2 was confirmed via dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. RESULTS GAS5 was elevated and miR-128-3p was decreased in the serum of patients with AS and ox-LDL-stimulated THP-1 cells. Ox-LDL stimulation inhibited proliferation and induced apoptosis of THP-1 cells. Meanwhile, GAS5 directly targeted miR-128-3p and inversely modulated its expression. Importantly, GAS5 depletion facilitated cell proliferation and impaired apoptosis in ox-LDL-induced THP-1 cells. Additionally, GAS5 augmented FBLN2 expression through sponging miR-128-3p, and miR-128-3p facilitated proliferation and retarded apoptosis of ox-LDL-induced THP-1 cells by targeting FBLN2. CONCLUSION GAS5 knockdown promoted the growth of ox-LDL-induced THP-1 cells through down-modulating FBLN2 and increasing miR-128-3p, suggesting the potential value of GAS5 for treatment of AS.
Collapse
Affiliation(s)
- Zijian Shen
- Deparment of Vasculocardiology, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow, Jiangsu, China
| | - Haigang Li
- Deparment of Vasculocardiology, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow, Jiangsu, China
| |
Collapse
|
167
|
Manchanda R, Fernandez-Fernandez A, Paluri SLA, Smith BR. Nanomaterials to target immunity. ADVANCES IN PHARMACOLOGY 2021; 91:293-335. [PMID: 34099112 DOI: 10.1016/bs.apha.2021.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Critical advances have recently been made in the field of immunotherapy, contributing to an improved understanding of how to harness and balance the power of immune responses in the treatment of diseases such as cancer, cardiovascular disease, infectious diseases, and autoimmune diseases. Combining nanomedicine with immunotherapy provides the opportunity for customization, rational design, and targeting to minimize side effects and maximize efficacy. This review highlights current developments in the design and utilization of nano-based immunotherapy systems, including how rationally-designed nanosystems can target and modify immune cells to modulate immune responses in a therapeutic manner. We discuss the following topics: targeted immuno-engineered nanoformulations, commercial formulations, clinical applicability, challenges associated with current approaches, and future directions.
Collapse
Affiliation(s)
- Romila Manchanda
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Alicia Fernandez-Fernandez
- Dr. Pallavi Patel College of Health Care Sciences, Nova Southeastern University, Ft. Lauderdale, FL, United States
| | - Sesha Lakshmi Arathi Paluri
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Bryan Ronain Smith
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
168
|
Georgakis MK, van der Laan SW, Asare Y, Mekke JM, Haitjema S, Schoneveld AH, de Jager SCA, Nurmohamed NS, Kroon J, Stroes ESG, de Kleijn DPV, de Borst GJ, Maegdefessel L, Soehnlein O, Pasterkamp G, Dichgans M. Monocyte-Chemoattractant Protein-1 Levels in Human Atherosclerotic Lesions Associate With Plaque Vulnerability. Arterioscler Thromb Vasc Biol 2021; 41:2038-2048. [PMID: 33827260 DOI: 10.1161/atvbaha.121.316091] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marios K Georgakis
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Germany (M.K.G., Y.A., M.D.)
| | - Sander W van der Laan
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University of Utrecht, the Netherlands (S.W.v.d.L., S.C.A.d.J.)
| | - Yaw Asare
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Germany (M.K.G., Y.A., M.D.)
| | - Joost M Mekke
- Department of Vascular Surgery, Division of Surgical Specialties (J.M.M., D.P.V.d.K., G.J.d.B.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Saskia Haitjema
- Center Diagnostic Laboratory, Division Laboratories and Pharmacy (S.H., A.H.S., G.P.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Arjan H Schoneveld
- Center Diagnostic Laboratory, Division Laboratories and Pharmacy (S.H., A.H.S., G.P.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Saskia C A de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, University of Utrecht, the Netherlands (S.W.v.d.L., S.C.A.d.J.)
| | - Nick S Nurmohamed
- Department of Vascular Medicine (N.S.N., E.S.G.S.), Amsterdam University Medical Centers (UMC), University of Amsterdam, the Netherlands.,Department of Cardiology (N.S.N.), Amsterdam University Medical Centers (UMC), University of Amsterdam, the Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences (J.K.), Amsterdam University Medical Centers (UMC), University of Amsterdam, the Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine (N.S.N., E.S.G.S.), Amsterdam University Medical Centers (UMC), University of Amsterdam, the Netherlands
| | - Dominique P V de Kleijn
- Department of Vascular Surgery, Division of Surgical Specialties (J.M.M., D.P.V.d.K., G.J.d.B.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Gert J de Borst
- Department of Vascular Surgery, Division of Surgical Specialties (J.M.M., D.P.V.d.K., G.J.d.B.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum Rechts der Isar, Technical University Munich, Germany (L.M.).,German Center for Cardiovascular Research (DZHK partner site), Munich, Germany (L.M.)
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Klinikum LMU Munich, Germany (O.S.).,German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany (O.S.).,Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (O.S.).,Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation, University of Münster, Germany (O.S.)
| | - Gerard Pasterkamp
- Center Diagnostic Laboratory, Division Laboratories and Pharmacy (S.H., A.H.S., G.P.), University Medical Centre Utrecht, Utrecht University, the Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Germany (M.K.G., Y.A., M.D.).,Munich Cluster for Systems Neurology (SyNergy), Germany (M.D.)
| |
Collapse
|
169
|
Ong SWX, Fong SW, Young BE, Chan YH, Lee B, Amrun SN, Chee RSL, Yeo NKW, Tambyah P, Pada S, Tan SY, Ding Y, Renia L, Leo YS, Ng LFP, Lye DC. Persistent Symptoms and Association With Inflammatory Cytokine Signatures in Recovered Coronavirus Disease 2019 Patients. Open Forum Infect Dis 2021; 8:ofab156. [PMID: 34095336 PMCID: PMC8083585 DOI: 10.1093/ofid/ofab156] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022] Open
Abstract
Background The complications and sequelae of coronavirus disease 2019 (COVID-19) and their effect on long-term health are unclear, and the trajectory of associated immune dysregulation is poorly understood. Methods We conducted a prospective longitudinal multicenter cohort study at 4 public hospitals in Singapore. Patients with COVID-19 were monitored for a median of 6 months after recovery from acute infection. Clinical symptoms and radiologic data were collected, along with plasma samples for quantification of immune mediators. The relationship between clinical symptoms and immune cytokine profiles was investigated. Results Two hundred eighty-eight participants were recruited, and follow-up data were available for 183, 175, and 120 participants at days 30, 90, and 180 postsymptom onset, respectively. Symptoms related to COVID-19 were present in 31 (16.9%), 13 (7.4%), and 14 (11.7%) at days 30, 90, and 180. In a multivariable model, age >65 years, non-Chinese ethnicity, and the severity of acute infection were associated with increased likelihood of persistent symptoms. Recovered COVID-19 patients had elevated levels of proinflammatory interleukin (IL)-17A, stem cell factor, IL-12p70, and IL-1β and pro-angiogenic macrophage inflammatory protein 1β, brain-derived neurotrophic factor, and vascular endothelial growth factor at day 180 compared with healthy controls. Higher levels of monocyte chemoattractant protein-1 and platelet-derived growth factor-BB were detected in patients with persistent symptoms, versus symptom-free patients. Conclusions Approximately 10% of recovered patients had persistent symptoms 6 months after initial infection. Immune cytokine signatures of the recovered patients reflected ongoing chronic inflammation and angiogenesis. Patients with COVID-19 should be monitored closely for emerging long-term health consequences.
Collapse
Affiliation(s)
- Sean Wei Xiang Ong
- National Centre for Infectious Diseases, Singapore.,Tan Tock Seng Hospital, Singapore
| | - Siew-Wai Fong
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, Singapore.,Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Yi-Hao Chan
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Bernett Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Siti Naqiah Amrun
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Rhonda Sin-Ling Chee
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Nicholas Kim-Wah Yeo
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Paul Tambyah
- National University Health System, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | - Ying Ding
- National Centre for Infectious Diseases, Singapore
| | - Laurent Renia
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, Singapore.,Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lisa F P Ng
- ASTAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, United Kingdom
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore.,Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
170
|
In vitro angiogenesis inhibition with selective compounds targeting the key glycolytic enzyme PFKFB3. Pharmacol Res 2021; 168:105592. [PMID: 33813027 DOI: 10.1016/j.phrs.2021.105592] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 03/27/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022]
Abstract
Abnormal glycolytic metabolism contributes to angiogenic sprouting involved in atherogenesis. We investigated the potential anti-angiogenic properties of specific 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) inhibitors in endothelial cells (ECs). ECs were treated with PFKFB3 inhibitors (named PA-1 and PA-2) and their effects on metabolic and functional characteristics of ECs were investigated. The anti-glycolytic compound 3-(pyridinyl)- 1-(4-pyridinyl)- 2-propen-1-one (3PO) was used as reference compound. PFKFB3 expression and activity (IC50 about 3-21 nM) was inhibited upon treatment with both compounds. Glucose uptake and lactate export were measured using commercial assays and showed a partial reduction up to 40%. PFKFB3 inhibition increased intracellular lactate accumulation, and reduced expression of monocarboxylate transporters-1 (MCT1) and MCT4. Furthermore, endothelial cell migration and proliferation assays demonstrated significant reduction upon treatment with both compounds. Matrix- metalloproteinase (MMP) activity, measured by gelatin zymography, and expression was significantly reduced (up to 25%). In addition, PA compounds downregulated the expression of VCAM-1, VE-cadherin, VEGFa, VEGFR2, TGF-β, and IL-1β, in inflamed ECs. Finally, PA-1 and PA-2 treatment impaired the formation of angiogenic sprouts measured by both morphogenesis and spheroid-based angiogenesis assays. Our data demonstrate that the anti-glycolytic PA compounds may affect several steps involved in angiogenesis. Targeting the key glycolytic enzyme PFKFB3 might represent an attractive therapeutic strategy to improve the efficacy of cancer treatments, or to be applied in other pathologies where angiogenesis is a detrimental factor.
Collapse
|
171
|
Pan J, Cai Y, Wang L, Maehara A, Mintz GS, Tang D, Li Z. A prediction tool for plaque progression based on patient-specific multi-physical modeling. PLoS Comput Biol 2021; 17:e1008344. [PMID: 33780445 PMCID: PMC8057612 DOI: 10.1371/journal.pcbi.1008344] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 04/20/2021] [Accepted: 03/10/2021] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic plaque rupture is responsible for a majority of acute vascular syndromes and this study aims to develop a prediction tool for plaque progression and rupture. Based on the follow-up coronary intravascular ultrasound imaging data, we performed patient-specific multi-physical modeling study on four patients to obtain the evolutional processes of the microenvironment during plaque progression. Four main pathophysiological processes, i.e., lipid deposition, inflammatory response, migration and proliferation of smooth muscle cells (SMCs), and neovascularization were coupled based on the interactions demonstrated by experimental and clinical observations. A scoring table integrating the dynamic microenvironmental indicators with the classical risk index was proposed to differentiate their progression to stable and unstable plaques. The heterogeneity of plaque microenvironment for each patient was demonstrated by the growth curves of the main microenvironmental factors. The possible plaque developments were predicted by incorporating the systematic index with microenvironmental indicators. Five microenvironmental factors (LDL, ox-LDL, MCP-1, SMC, and foam cell) showed significant differences between stable and unstable group (p < 0.01). The inflammatory microenvironments (monocyte and macrophage) had negative correlations with the necrotic core (NC) expansion in the stable group, while very strong positive correlations in unstable group. The inflammatory microenvironment is strongly correlated to the NC expansion in unstable plaques, suggesting that the inflammatory factors may play an important role in the formation of a vulnerable plaque. This prediction tool will improve our understanding of the mechanism of plaque progression and provide a new strategy for early detection and prediction of high-risk plaques. Besides the traditional systematic factors, the influences of the local microenvironmental factors on atherosclerotic plaque progression have been demonstrated. Mathematical and computational modeling is an important tool to investigate the complex interplay between plaque progression and the microenvironment, and provides a potential way toward the prediction of plaque vulnerability according to the comprehensive evaluation of both morphological and/or biochemical factors in tissue level with microenvironmental factors in cellular level. We performed patient-specific multi-physical modeling study on four patients to obtain the evolutional processes of the microenvironment during plaque progression and predicted the possible plaque developments. A scoring table integrating the dynamic microenvironmental indicators with the classical risk index was proposed to differentiate their progression to stable and unstable plaques. Based on patient-specific imaging data, the mathematical model will provide a novel method to predict the changes of plaque microenvironment and improve ability to access the personal therapeutic strategy for atherosclerotic plaque.
Collapse
Affiliation(s)
- Jichao Pan
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
| | - Yan Cai
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
| | - Liang Wang
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
| | - Akiko Maehara
- The Cardiovascular Research Foundation, New York, New York, United States of America
| | - Gary S Mintz
- The Cardiovascular Research Foundation, New York, New York, United States of America
| | - Dalin Tang
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
- Mathematical Sciences Department, Worcester Polytechnic Institute, Massachusetts, United States of America
| | - Zhiyong Li
- School of Biological Sciences and Medical Engineering, Southeast University, Nanjing Jiangsu, China
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
172
|
Zhang Y, Wernly B, Cao X, Mustafa SJ, Tang Y, Zhou Z. Adenosine and adenosine receptor-mediated action in coronary microcirculation. Basic Res Cardiol 2021; 116:22. [PMID: 33755785 PMCID: PMC7987637 DOI: 10.1007/s00395-021-00859-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/08/2021] [Indexed: 12/20/2022]
Abstract
Adenosine is an ubiquitous extracellular signaling molecule and plays a fundamental role in the regulation of coronary microcirculation through activation of adenosine receptors (ARs). Adenosine is regulated by various enzymes and nucleoside transporters for its balance between intra- and extracellular compartments. Adenosine-mediated coronary microvascular tone and reactive hyperemia are through receptors mainly involving A2AR activation on both endothelial and smooth muscle cells, but also involving interaction among other ARs. Activation of ARs further stimulates downstream targets of H2O2, KATP, KV and KCa2+ channels leading to coronary vasodilation. An altered adenosine-ARs signaling in coronary microcirculation has been observed in several cardiovascular diseases including hypertension, diabetes, atherosclerosis and ischemic heart disease. Adenosine as a metabolite and its receptors have been studied for its both therapeutic and diagnostic abilities. The present review summarizes important aspects of adenosine metabolism and AR-mediated actions in the coronary microcirculation.
Collapse
Affiliation(s)
- Ying Zhang
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bernhard Wernly
- Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Xin Cao
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, USA
| | - Yong Tang
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
173
|
Lukitasari M, Saifur Rohman M, Nugroho DA, Widodo N, Nugrahini NIP. Cardiovascular protection effect of chlorogenic acid: focus on the molecular mechanism. F1000Res 2021; 9:1462. [PMID: 33708382 PMCID: PMC7927207 DOI: 10.12688/f1000research.26236.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial cells have a variety of functions such as the control of blood coagulation, vascular permeability, and tone regulation, as well as quiesce of immune cells. Endothelial dysfunction is a cardiovascular events predictor, which is considered the initial stage in atherosclerosis development. It is characterized by alterations in endothelium functions due to imbalanced vasodilators and vasoconstrictors, procoagulant and anticoagulant mediators, as well as growth inhibitor and promotor substances. Chlorogenic acid (CGA) is the primary polyphenol in coffee and some fruits. It has many health-promoting properties, especially in the cardiovascular system. Many studies investigated the efficacy and mechanism of this compound in vascular health. CGA has several vascular benefits such as anti-atherosclerosis, anti-thrombosis, and anti-hypertensive. This review focuses on the molecular mechanism of CGA in vascular health.
Collapse
Affiliation(s)
- Mifetika Lukitasari
- Department of Nursing, Faculty of Medicine, Brawijaya University, Malang, East java, +62, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University-Saiful Anwar General Hospital, Malang, East java, +62, Indonesia
| | - Dwi Adi Nugroho
- Department of Herbal Medicine, Cardiovascular research group, Faculty of Medicine, Brawijaya University, Malang, East java, +62, Indonesia
| | - Nashi Widodo
- Department of Biology, Faculty of Mathematics and Natural Science, Brawijaya University, Malang, East java, +62, Indonesia
| | - Nur Ida Panca Nugrahini
- Department Agricultural Product Technology, Brawijaya University, Malang, East java, +62, Indonesia
| |
Collapse
|
174
|
Dakroub A, Nasser SA, Kobeissy F, Yassine HM, Orekhov A, Sharifi-Rad J, Iratni R, El-Yazbi AF, Eid AH. Visfatin: An emerging adipocytokine bridging the gap in the evolution of cardiovascular diseases. J Cell Physiol 2021; 236:6282-6296. [PMID: 33634486 DOI: 10.1002/jcp.30345] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/24/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Visfatin/nicotinamide phosphoribosyltransferase (NAMPT) is an adipokine expressed predominately in visceral fat tissues. High circulating levels of visfatin/NAMPT have been implicated in vascular remodeling, vascular inflammation, and atherosclerosis, all of which pose increased risks of cardiovascular events. In this context, increased levels of visfatin have been correlated with several upregulated pro-inflammatory mediators, such as IL-1, IL-1Ra, IL-6, IL-8, and TNF-α. Furthermore, visfatin is associated with leukocyte recruitment by endothelial cells and the production of adhesion molecules such as vascular cell adhesion molecule 1, intercellular cell adhesion molecule 1, and E-selectin, which are well known to mediate the progression of atherosclerosis. Moreover, diverse angiogenic factors have been found to mediate visfatin-induced angiogenesis. These include matrix metalloproteinases, vascular endothelial growth factor, monocyte chemoattractant protein 1, and fibroblast growth factor 2. This review aims to provide a comprehensive overview of the pro-inflammatory and angiogenic actions of visfatin, with a focus on the pertinent signaling pathways whose dysregulation contributes to the pathogenesis of atherosclerosis. Most importantly, some hypotheses regarding the integration of the aforementioned factors with the plausible atherogenic effect of visfatin are put forth for consideration in future studies. The pharmacotherapeutic potential of modulating visfatin's roles could be important in the management of cardiovascular disease, which continues to be the leading cause of death worldwide.
Collapse
Affiliation(s)
- Ali Dakroub
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Lebanon
| | - Suzanne A Nasser
- Department of Pharmacology and Therapeutics, Beirut Arab University, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Alexander Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Moscow, Russia.,Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia.,Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | - Rabah Iratni
- Department of Biology, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Lebanon.,Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.,Faculty of Pharmacy, Faculty of Pharmacy, Alamein International University, Alamein, Egypt
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
175
|
Li YY, Zhang S, Wang H, Zhang SX, Xu T, Chen SW, Zhang Y, Chen Y. Identification of Crucial Genes and Pathways Associated with Atherosclerotic Plaque in Diabetic Patients. Pharmgenomics Pers Med 2021; 14:211-220. [PMID: 33568933 PMCID: PMC7869704 DOI: 10.2147/pgpm.s281705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/26/2020] [Indexed: 01/20/2023] Open
Abstract
Background Patients with diabetes have more calcification in atherosclerotic plaque and a higher occurrence of secondary cardiovascular events than patients without diabetes. The objective of this study was to identify crucial genes involved in the development of diabetic atherosclerotic plaque using a bioinformatics approach. Methods Microarray dataset GSE118481 was downloaded from the Gene Expression Omnibus (GEO) database; the dataset included 6 patients with diabetic atherosclerotic plaque (DBT) and 6 nondiabetic patients with atherosclerotic plaque (Ctrl). Differentially expressed genes (DEG) between the DBT and Ctrl groups were identified and then subjected to functional enrichment analysis. Based on the enriched pathways of DEGs, diabetic atherosclerotic plaque-related pathways were screened using the comparative toxicogenomics database (CTD). We then constructed a protein–protein interaction (PPI) network and transcription factor (TF)–miRNA–mRNA network. Results A total of 243 DEGs were obtained in the DBT group compared with the Ctrl group, including 85 up-regulated and 158 down-regulated DEGs. Functional enrichment analysis showed that up-regulated DEGs were mainly enriched in isoprenoid metabolic process, DNA-binding TF activity, and response to virus. Additionally, DEGs participating in the toll-like receptor signaling pathway were closely related to diabetes, carotid stenosis, and insulin resistance. The TF–miRNA–mRNA network showed that toll-like receptor 4 (TLR4), BCL2-like 11 (BCL2L11), and glutamate-cysteine ligase catalytic subunit (GCLC) were hub genes. Furthermore, TLR4 was regulated by TF signal transducer and activator of transcription 6 (STAT6); BCL2L11 was targeted by hsa-miR-24-3p; and GCLC was regulated by nuclear factor, erythroid 2 like 2 (NFE2L2). Conclusion Identification of hub genes and pathways increased our understanding of the molecular mechanisms underlying the atherosclerotic plaque in patients with or without diabetes. These crucial genes (TLR4, BC2L11, and GCLC) might function as molecular biomarkers for diabetic atherosclerotic plaque.
Collapse
Affiliation(s)
- Yuan-Yuan Li
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Sheng Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Hua Wang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Shun-Xiao Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Ting Xu
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Shu-Wen Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Yan Zhang
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| | - Yue Chen
- Department of Endocrinology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, People's Republic of China
| |
Collapse
|
176
|
Huisman LA, Steinkamp PJ, Hillebrands JL, Zeebregts CJ, Linssen MD, Jorritsma-Smit A, Slart RHJA, van Dam GM, Boersma HH. Feasibility of ex vivo fluorescence imaging of angiogenesis in (non-) culprit human carotid atherosclerotic plaques using bevacizumab-800CW. Sci Rep 2021; 11:2899. [PMID: 33536498 PMCID: PMC7858611 DOI: 10.1038/s41598-021-82568-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/11/2021] [Indexed: 01/30/2023] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A) is assumed to play a crucial role in the development and rupture of vulnerable plaques in the atherosclerotic process. We used a VEGF-A targeted fluorescent antibody (bevacizumab-IRDye800CW [bevacizumab-800CW]) to image and visualize the distribution of VEGF-A in (non-)culprit carotid plaques ex vivo. Freshly endarterectomized human plaques (n = 15) were incubated in bevacizumab-800CW ex vivo. Subsequent NIRF imaging showed a more intense fluorescent signal in the culprit plaques (n = 11) than in the non-culprit plaques (n = 3). A plaque received from an asymptomatic patient showed pathologic features similar to the culprit plaques. Cross-correlation with VEGF-A immunohistochemistry showed co-localization of VEGF-A over-expression in 91% of the fluorescent culprit plaques, while no VEGF-A expression was found in the non-culprit plaques (p < 0.0001). VEGF-A expression was co-localized with CD34, a marker for angiogenesis (p < 0.001). Ex vivo near-infrared fluorescence (NIRF) imaging by incubation with bevacizumab-800CW shows promise for visualizing VEGF-A overexpression in culprit atherosclerotic plaques in vivo.
Collapse
Affiliation(s)
- Lydian A. Huisman
- grid.4494.d0000 0000 9558 4598Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Pieter J. Steinkamp
- grid.4494.d0000 0000 9558 4598Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- grid.4494.d0000 0000 9558 4598Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Clark J. Zeebregts
- grid.4494.d0000 0000 9558 4598Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Matthijs D. Linssen
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annelies Jorritsma-Smit
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Riemer H. J. A. Slart
- grid.4494.d0000 0000 9558 4598Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.6214.10000 0004 0399 8953Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Gooitzen M. van Dam
- grid.4494.d0000 0000 9558 4598Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Department of Surgery, Nuclear Medicine and Molecular Imaging and Intensive Care, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hendrikus H. Boersma
- grid.4494.d0000 0000 9558 4598Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands ,grid.4494.d0000 0000 9558 4598Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
177
|
Wijeratne T, Gillard Crewther S, Sales C, Karimi L. COVID-19 Pathophysiology Predicts That Ischemic Stroke Occurrence Is an Expectation, Not an Exception-A Systematic Review. Front Neurol 2021; 11:607221. [PMID: 33584506 PMCID: PMC7876298 DOI: 10.3389/fneur.2020.607221] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/30/2020] [Indexed: 12/27/2022] Open
Abstract
Clinical reports of neurological manifestations associated with severe coronavirus disease 2019 (COVID-19), such as acute ischemic stroke (AIS), encephalopathy, seizures, headaches, acute necrotizing encephalitis, cerebral microbleeds, posterior reversible leukoencephalopathy syndrome, hemophagocytic lymphohistiocytosis, peripheral neuropathy, cranial nerve palsies, transverse myelitis, and demyelinating disorders, are increasing rapidly. However, there are comparatively few studies investigating the potential impact of immunological responses secondary to hypoxia, oxidative stress, and excessive platelet-induced aggregation on the brain. This scoping review has focused on the pathophysiological mechanisms associated with peripheral and consequential neural (central) inflammation leading to COVID-19-related ischemic strokes. It also highlights the common biological processes shared between AIS and COVID-19 infection and the importance of the recognition that severe respiratory dysfunction and neurological impairments associated with COVID and chronic inflammation [post-COVID-19 neurological syndrome (PCNS)] may significantly impact recovery and ability to benefit from neurorehabilitation. This study provides a comprehensive review of the pathobiology of COVID-19 and ischemic stroke. It also affirms that the immunological contribution to the pathophysiology of COVID-19 is predictive of the neurological sequelae particularly ischemic stroke, which makes it the expectation rather than the exception. This work is of fundamental significance to the neurorehabilitation community given the increasing number of COVID-related ischemic strokes, the current limited knowledge regarding the risk of reinfection, and recent reports of a PCNS. It further highlights the need for global collaboration and research into new pathobiology-based neurorehabilitation treatment strategies and more integrated evidence-based care.
Collapse
Affiliation(s)
- Tissa Wijeratne
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, University of Rajarata, Anuradhapura, Sri Lanka
| | - Sheila Gillard Crewther
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
| | - Carmela Sales
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
- Department of Medicine, Faculty of Medicine, University of Rajarata, Anuradhapura, Sri Lanka
| | - Leila Karimi
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Neurology, Western Health and University Melbourne, Australian Institute of Muscular Skeletal Sciences (AIMSS), Level Three, Western Health Centre for Research and Education (WHCRE), Sunshine Hospital, Melbourne, VIC, Australia
- Faculty of Social and Political Sciences, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| |
Collapse
|
178
|
Yang Z, Huang Y, Zhu L, Yang K, Liang K, Tan J, Yu B. SIRT6 promotes angiogenesis and hemorrhage of carotid plaque via regulating HIF-1α and reactive oxygen species. Cell Death Dis 2021; 12:77. [PMID: 33436551 PMCID: PMC7804142 DOI: 10.1038/s41419-020-03372-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/15/2022]
Abstract
As a member of Sirtuins family, SIRT6 participates in the physiological and pathological progress of DNA repair, anti-aging, metabolism, and so on. Several studies have demonstrated that knockdown of SIRT6 inhibited the development of atherosclerosis (AS), indicated SIRT6 as a protective factor for AS. However, we confirmed SIRT6 was significantly overexpressed in human unstable carotid plaques compared with stable carotid plaques. This result indicated a more complex role of SIRT6 in AS. Furthermore, we constructed mice model with unstable carotid plaque and injected them with SIRT6 overexpressed adeno-associated virus (AAV-SIRT6). AAV-SIRT6 significantly promoted angiogenesis as well as hemorrhage in plaques. In vitro, we demonstrated overexpression of SIRT6 prevented HIF-1α from degradation by deubiquitination at K37 and K532 of HIF-1α, thus promoted the expression of HIF-1α under both normoxia and hypoxia in human umbilical vein endothelial cells (HUVECs). Through regulating HIF-1α, overexpression of SIRT6 promoted invasion, migration, proliferation, as well as tube formation ability of HUVECs. Interestingly, under different conditions, SIRT6 played different roles in the function of HUVECs. Under oxidative stress, another important pathological environment for AS, SIRT6 bound to the promoter of Catalase, a main reactive oxygen species scavenger, and depleted H3K56 acetylation, thus inhibited expression and activity of Catalase at the transcriptional level. Subsequently, inhibited Catalase promoted reactive oxygen species (ROS) under oxidative stress. Accumulated ROS further aggravated oxidative stress injury of HUVECs. On one hand, SIRT6 promoted angiogenesis in plaque via HIF-1α under hypoxia. On the other hand, SIRT6 promoted injury of neovascular via ROS under oxidative stress. It is this process of continuous growth and damage that leads to hemorrhage in carotid plaque. In conclusion, we innovatively confirmed SIRT6 promoted the angiogenesis and IPH via promoting HIF-1α and ROS in different environments, thus disclosed the unknowing danger of SIRT6.
Collapse
Affiliation(s)
- Zhou Yang
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Yijun Huang
- Department of General Surgery, Huashan Hospital North, Fudan University, Shanghai, 201907, China
| | - Lei Zhu
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Kai Yang
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Kun Liang
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jinyun Tan
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Bo Yu
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
- Department of Vascular Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
179
|
Mercier C, Rousseau M, Geraldes P. Growth Factor Deregulation and Emerging Role of Phosphatases in Diabetic Peripheral Artery Disease. Front Cardiovasc Med 2021; 7:619612. [PMID: 33490120 PMCID: PMC7817696 DOI: 10.3389/fcvm.2020.619612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/10/2020] [Indexed: 01/25/2023] Open
Abstract
Peripheral artery disease is caused by atherosclerosis of lower extremity arteries leading to the loss of blood perfusion and subsequent critical ischemia. The presence of diabetes mellitus is an important risk factor that greatly increases the incidence, the progression and the severity of the disease. In addition to accelerated disease progression, diabetic patients are also more susceptible to develop serious impairment of their walking abilities through an increased risk of lower limb amputation. Hyperglycemia is known to alter the physiological development of collateral arteries in response to ischemia. Deregulation in the production of several critical pro-angiogenic factors has been reported in diabetes along with vascular cell unresponsiveness in initiating angiogenic processes. Among the multiple molecular mechanisms involved in the angiogenic response, protein tyrosine phosphatases are potent regulators by dephosphorylating pro-angiogenic tyrosine kinase receptors. However, evidence has indicated that diabetes-induced deregulation of phosphatases contributes to the progression of several micro and macrovascular complications. This review provides an overview of growth factor alterations in the context of diabetes and peripheral artery disease, as well as a description of the role of phosphatases in the regulation of angiogenic pathways followed by an analysis of the effects of hyperglycemia on the modulation of protein tyrosine phosphatase expression and activity. Knowledge of the role of phosphatases in diabetic peripheral artery disease will help the development of future therapeutics to locally regulate phosphatases and improve angiogenesis.
Collapse
Affiliation(s)
- Clément Mercier
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marina Rousseau
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
180
|
Silva H, Francisco R, Saraiva A, Francisco S, Carrascosa C, Raposo A. The Cardiovascular Therapeutic Potential of Propolis-A Comprehensive Review. BIOLOGY 2021; 10:biology10010027. [PMID: 33406745 PMCID: PMC7823408 DOI: 10.3390/biology10010027] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
Simple Summary Propolis, also described as bee glue, is a natural component made up of a resinous mixture of honeybee compounds from multiple botanical sources. The literature has demonstrated a variety of medicinal properties attributed to propolis due to its chemical complexity. However, the positive effects of propolis on cardiovascular health have gained little coverage. Therefore, we aimed to provide an accurate and up-to-date review of the main cardiovascular health benefits of propolis. In particular, we intend to establish the key varieties of propolis and pharmacological compounds with the therapeutic effects that are most encouraging, as well as the physiological processes by which those advantages are accomplished. The Brazilian green and red varieties reveal the greatest number of beneficial activities among the varieties of propolis studied. While much of the cardiovascular beneficial effects appear to derive from the cumulative actions of several compounds working via multiple signaling mechanisms, some individual compounds that may enhance the existing therapeutic arsenal have also shown significant results. It is also worth exploring the prospect of using propolis as food supplements. Abstract Owing to its chemical richness, propolis has a myriad of therapeutic properties. To the authors’ knowledge, this is the first comprehensive review paper on propolis to focus exclusively on its major effects for cardiovascular health. The propolis compound varieties with the most promising therapeutic benefits and their respective physiological mechanisms will be discussed. Propolis displays an anti-atherosclerotic activity, attained through modulation of the plasma lipid profile and through stabilization of the fatty plaque by inhibiting macrophage apoptosis, vascular smooth muscle proliferation and metalloproteinase activity. The antihypertensive effects of propolis probably arise through the combination of several mechanisms, including the suppression of catecholamine synthesis, stimulation of endothelium-dependent vasorelaxation and vascular anti-inflammatory activity. The anti-hemostatic activity of propolis is attributed to the inhibition of platelet plug formation and antifibrinolytic activity. By inhibiting the secretion of proangiogenic factors, propolis suppresses endothelial cell migration and tubulogenesis, exerting antiangiogenic activity. The antioxidant and anti-inflammatory activities are responsible for protection against vascular endothelial and cardiomyocyte dysfunction, mostly by the prevention of oxidative stress. Among the reviewed propolis varieties, the Brazilian green and red varieties show the largest number of beneficial activities. Further research, especially preclinical, should be conducted to assess the cardiovascular benefits of the given varieties with different compositions.
Collapse
Affiliation(s)
- Henrique Silva
- Informetrics Research Group, Ton Duc Thang University, Ho Chi Minh City 758307, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 758307, Vietnam
- Correspondence: (H.S.); (A.R.)
| | - Rafaela Francisco
- Pharmacological Sciences Department, Faculty of Pharmacy, Universidade de Lisboa, Av Prof Gama Pinto, 1649-003 Lisboa, Portugal;
| | - Ariana Saraiva
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain; (A.S.); (C.C.)
| | - Simone Francisco
- Faculty of Medicine, Nutrition Lab—Universidade de Lisboa, 1649-028 Lisboa, Portugal;
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain; (A.S.); (C.C.)
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
- Correspondence: (H.S.); (A.R.)
| |
Collapse
|
181
|
Ou LC, Zhong S, Ou JS, Tian JW. Application of targeted therapy strategies with nanomedicine delivery for atherosclerosis. Acta Pharmacol Sin 2021; 42:10-17. [PMID: 32457416 DOI: 10.1038/s41401-020-0436-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/09/2020] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis (AS) is the main pathological cause of coronary heart disease (CHD). Current clinical interventions including statin drugs can effectively reduce acute myocardial infarction and stroke to some extent, but residual risk remains high. The current clinical treatment regimens are relatively effective for early atherosclerotic plaques and can even reverse their progression. However, the effectiveness of these treatments for advanced AS is not ideal, and advanced atherosclerotic plaques-the pathological basis of residual risk-can still cause a recurrence of acute cardiovascular and cerebrovascular events. Recently, nanomedicine-based treatment strategies have been extensively used in antitumor therapy, and also shown great potential in anti-AS therapy. There are many microstructures in late-stage atherosclerotic plaques, such as neovascularization, micro-calcification, and cholesterol crystals, and these have become important foci for targeted nanomedicine delivery. The use of targeted nanoparticles has become an important strategy for the treatment of advanced AS to further reduce the residual risk of cardiovascular events. Furthermore, the feasibility and safety of nanotechnology in clinical treatment have been preliminarily confirmed. In this review, we summarize the application of nanomedicine delivery in the treatment of advanced AS and the clinical value of several promising nanodrugs.
Collapse
|
182
|
Liu W, Zong M, Gong HY, Ling LJ, Ye XH, Wang S, Li CY. Comparison of Diagnostic Efficacy Between Contrast-Enhanced Ultrasound and DCE-MRI for Mass- and Non-Mass-Like Enhancement Types in Breast Lesions. Cancer Manag Res 2020; 12:13567-13578. [PMID: 33408526 PMCID: PMC7781362 DOI: 10.2147/cmar.s283656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/16/2020] [Indexed: 01/19/2023] Open
Abstract
Background Contrast-enhanced ultrasound (CEUS) can provide angiogenesis information about breast lesions; however, its diagnostic performance in comparison with that of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) has not been systematically investigated. This study aimed to evaluate the diagnostic efficacy of CEUS and DCE-MRI in mass-like and non-mass-like enhancement types of breast lesions. Material and Methods A retrospective study was conducted on 252 patients with breast lesions who underwent CEUS and DCE-MRI before surgery between January 2016 and February 2020. Histopathological results were used as reference standards. All patients were classified into mass-like and non-mass-like enhancement lesion groups. The mass-like lesion group was further divided into three categories according to different sizes (group 1: <10 mm, group 2: 10-20 mm, and group 3: >20 mm). Sensitivity, specificity, positive predictive value, negative predictive value, and receiver operating characteristic curve were analyzed to assess the diagnostic performance of these two modalities. Results For mass-like breast lesions, DCE-MRI (Az=0.981) manifested better diagnostic performance than CEUS (Az=0.940) in medium-sized (10-20 mm) tumors (Z=2.018, P=0.043), but both had similar diagnostic performance in smaller (<10 mm) and larger (>20 mm) tumors (P=0.717, P=0.394). For non-mass-like enhancement lesions, CEUS and DCE-MRI showed no significant difference (Z=1.590, P=0.119) and revealed good diagnostic performance (Az=0.859, Az=0.947) in differentiating the two groups. Conclusion For mass-like breast lesions, DCE-MRI showed better diagnostic performance than CEUS in differentiating benign and malignant tumors of medium-sizes (10-20mm) but not of smaller (<10mm) and larger (>20 mm) sizes. For non-mass-like lesions, both modalities showed similar diagnostic performance.
Collapse
Affiliation(s)
- Wei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Min Zong
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Hai-Yan Gong
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Li-Jun Ling
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Xin-Hua Ye
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Shui Wang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Cui-Ying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| |
Collapse
|
183
|
Vazquez-Prada KX, Lam J, Kamato D, Xu ZP, Little PJ, Ta HT. Targeted Molecular Imaging of Cardiovascular Diseases by Iron Oxide Nanoparticles. Arterioscler Thromb Vasc Biol 2020; 41:601-613. [PMID: 33356385 DOI: 10.1161/atvbaha.120.315404] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease is one of the major contributors to global disease burden. Atherosclerosis is an inflammatory process that involves the accumulation of lipids and fibrous elements in the large arteries, forming an atherosclerotic plaque. Rupture of unstable plaques leads to thrombosis that triggers life-threatening complications such as myocardial infarction. Current diagnostic methods are invasive as they require insertion of a catheter into the coronary artery. Molecular imaging techniques, such as magnetic resonance imaging, have been developed to image atherosclerotic plaques and thrombosis due to its high spatial resolution and safety. The sensitivity of magnetic resonance imaging can be improved with contrast agents, such as iron oxide nanoparticles. This review presents the most recent advances in atherosclerosis, thrombosis, and myocardial infarction molecular imaging using iron oxide-based nanoparticles. While some studies have shown their effectiveness, many are yet to undertake comprehensive testing of biocompatibility. There are still potential hazards to address and complications to diagnosis, therefore strategies for overcoming these challenges are required.
Collapse
Affiliation(s)
- Karla X Vazquez-Prada
- Australian Institute for Bioengineering and Nanotechnology (K.X.V.-P., Z.P.X., H.T.T.), the University of Queensland, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence (K.X.V.-P., J.L., D.K., P.J.L.), the University of Queensland, Australia.,Queensland Micro- and Nanotechnology (K.X.V.-P., H.T.T.), Griffith University, Brisbane, Australia
| | - Jacinta Lam
- School of Pharmacy, Pharmacy Australia Centre of Excellence (K.X.V.-P., J.L., D.K., P.J.L.), the University of Queensland, Australia
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence (K.X.V.-P., J.L., D.K., P.J.L.), the University of Queensland, Australia
| | - Zhi Ping Xu
- Australian Institute for Bioengineering and Nanotechnology (K.X.V.-P., Z.P.X., H.T.T.), the University of Queensland, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence (K.X.V.-P., J.L., D.K., P.J.L.), the University of Queensland, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, China (P.J.L.)
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology (K.X.V.-P., Z.P.X., H.T.T.), the University of Queensland, Australia.,Queensland Micro- and Nanotechnology (K.X.V.-P., H.T.T.), Griffith University, Brisbane, Australia.,School of Environment and Science (H.T.T.), Griffith University, Brisbane, Australia
| |
Collapse
|
184
|
Joshkon A, Heim X, Dubrou C, Bachelier R, Traboulsi W, Stalin J, Fayyad-Kazan H, Badran B, Foucault-Bertaud A, Leroyer AS, Bardin N, Blot-Chabaud M. Role of CD146 (MCAM) in Physiological and Pathological Angiogenesis-Contribution of New Antibodies for Therapy. Biomedicines 2020; 8:biomedicines8120633. [PMID: 33352759 PMCID: PMC7767164 DOI: 10.3390/biomedicines8120633] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
The fundamental role of cell adhesion molecules in mediating various biological processes as angiogenesis has been well-documented. CD146, an adhesion molecule of the immunoglobulin superfamily, and its soluble form, constitute major players in both physiological and pathological angiogenesis. A growing body of evidence shows soluble CD146 to be significantly elevated in the serum or interstitial fluid of patients with pathologies related to deregulated angiogenesis, as autoimmune diseases, obstetric and ocular pathologies, and cancers. To block the undesirable effects of this molecule, therapeutic antibodies have been developed. Herein, we review the multifaceted functions of CD146 in physiological and pathological angiogenesis and summarize the interest of using monoclonal antibodies for therapeutic purposes.
Collapse
Affiliation(s)
- Ahmad Joshkon
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
- Correspondence:
| | - Xavier Heim
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Service d’immunologie, Pôle de Biologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Cléa Dubrou
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Richard Bachelier
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Wael Traboulsi
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Jimmy Stalin
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Science, Lebanese University, Hadath 1104, Lebanon; (H.F.-K.); (B.B.)
| | - Alexandrine Foucault-Bertaud
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Aurelie S. Leroyer
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| | - Nathalie Bardin
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
- Service d’immunologie, Pôle de Biologie, Hôpital de la Conception, Assistance Publique Hôpitaux de Marseille (AP-HM), 13005 Marseille, France
| | - Marcel Blot-Chabaud
- Hematology Department, Center for CardioVascular and Nutrition Research C2VN, Faculty of Pharmacy, Timone Campus, Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de Recherche Pour L’agriculture, L’alimentation et L’environnement (INRAE), 13005 Marseille, France; (X.H.); (C.D.); (R.B.); (W.T.); (J.S.); (A.F.-B.); (A.S.L.); (N.B.); (M.B.-C.)
| |
Collapse
|
185
|
Circulating Exosomal miRNAs as Novel Biomarkers for Stable Coronary Artery Disease. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3593962. [PMID: 33381550 PMCID: PMC7748912 DOI: 10.1155/2020/3593962] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/24/2020] [Accepted: 11/27/2020] [Indexed: 12/16/2022]
Abstract
Exosomal miRNAs are currently being explored as a novel class of biomarkers in cardiovascular diseases. However, few reports have focused on the value of circulating exosomal miRNAs as biomarkers for stable coronary artery disease (SCAD). Here, we aimed to investigate whether miRNAs involved in cardiovascular diseases in circulating exosomes could serve as novel diagnostic biomarkers for SCAD. Firstly, the serum exosomes were isolated and purified by the ExoQuick reagent and identified by transmission electron microscopy, western blot, and nanoparticle tracking analysis. Then, the purified exosomes were quantified by measuring the exosome protein concentration and calculating the total protein amount. Next, eight miRNAs involved in cardiovascular diseases, miR-192-5p, miR-148b-3p, miR-125a-3p, miR-942-5p, miR-149-5p, miR-32-5p, miR-144-3p, and miR-142-5p, were quantified in circulating exosomes from the control group (n = 20) and the SCAD group (n = 20) by quantitative real-time polymerase chain reaction (qPCR). Finally, the gene targets of the differentially expressed miRNAs were predicted, and the functions and signaling pathways of these targets were analyzed using an online database. The isolated exosomes had a bilayer membrane with a diameter of about 100 nm and expressed exosomal markers including CD63, Tsg101, and Flotillin but negatively expressed Calnexin. Both the exosome protein concentration and total protein amount exhibited no significant differences between the two groups. The qPCR assay demonstrated that among the eight miRNAs, the expression levels of miR-942-5p, miR-149-5p, and miR-32-5p in the serum exosomes from the SCAD group were significantly higher than that from the control group. And the three miRNAs for SCAD diagnosis exhibited AUC values of 0.693, 0.702, and 0.691, respectively. GO categories and signaling pathways analysis showed that some of the predictive targets of these miRNAs were involved in the pathophysiology processes of SCAD. In conclusion, our findings suggest that serum exosomal miR-942-5p, miR-149-5p, and miR-32-5p may serve as potential diagnostic biomarkers for SCAD.
Collapse
|
186
|
Li H, Zou J, Yu XH, Ou X, Tang CK. Zinc finger E-box binding homeobox 1 and atherosclerosis: New insights and therapeutic potential. J Cell Physiol 2020; 236:4216-4230. [PMID: 33275290 DOI: 10.1002/jcp.30177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/07/2020] [Accepted: 11/12/2020] [Indexed: 12/29/2022]
Abstract
Zinc finger E-box binding homeobox 1 (ZEB1), an important transcription factor belonging to the ZEB family, plays a crucial role in regulating gene expression required for both normal physiological and pathological processes. Accumulating evidence has shown that ZEB1 participates in the initiation and progression of atherosclerotic cardiovascular disease. Recent studies suggest that ZEB1 protects against atherosclerosis by regulation of endothelial cell angiogenesis, endothelial dysfunction, monocyte-endothelial cell interaction, macrophage lipid accumulation, macrophage polarization, monocyte-vascular smooth muscle cell (VSMC) interaction, VSMC proliferation and migration, and T cell proliferation. In this review, we summarize the recent progress of ZEB1 in the pathogenesis of atherosclerosis and provide insights into the prevention and treatment of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Instrument and Equipment Technology Laboratory of Hengyang Medical College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| | - Jin Zou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Instrument and Equipment Technology Laboratory of Hengyang Medical College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China.,Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiang Ou
- Department of Endocrinology, The First Hospital of Changsha, Changsha, Hunan, China
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Medical Instrument and Equipment Technology Laboratory of Hengyang Medical College, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan, 421001, China
| |
Collapse
|
187
|
Du J, Han R, Li Y, Liu X, Liu S, Cai Z, Xu Z, Li Y, Yuan X, Guo X, Lu B, Sun K. LncRNA HCG11/miR-26b-5p/QKI5 feedback loop reversed high glucose-induced proliferation and angiogenesis inhibition of HUVECs. J Cell Mol Med 2020; 24:14231-14246. [PMID: 33128346 PMCID: PMC7753996 DOI: 10.1111/jcmm.16040] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/23/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Acute coronary syndrome caused by the rupture of atherosclerotic plaques is one of the primary causes of cerebrovascular and cardiovascular events. Neovascularization within the plaque is closely associated with its stability. Long non-coding RNA (lncRNA) serves a crucial role in regulating vascular endothelial cells (VECs) proliferation and angiogenesis. In this study, we identified lncRNA HCG11, which is highly expressed in patients with vulnerable plaque compared with stable plaque. Then, functional experiments showed that HCG11 reversed high glucose-induced vascular endothelial injury through increased cell proliferation and tube formation. Meanwhile, vascular-related RNA-binding protein QKI5 was greatly activated. Luciferase reporter assays and RNA-binding protein immunoprecipitation (RIP) assays verified interaction between them. Interestingly, HCG11 can also positively regulated by QKI5. Bioinformatics analysis and luciferase reporter assays showed HCG11 can worked as a competing endogenous RNA by sponging miR-26b-5p, and QKI5 was speculated as the target of miR-26b-5p. Taken together, our findings revered that the feedback loop of lncRNA HCG11/miR-26b-5p/QKI-5 played a vital role in the physiological function of HUVECs, and this also provide a potential target for therapeutic strategies of As.
Collapse
Affiliation(s)
- Jiao Du
- Department of RadiologyState Key Laboratory of Cardiovascular DiseaseFu Wai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
- Department of RadiologyBayannur HospitalBayannurChina
| | - Ruijuan Han
- Department of RadiologyState Key Laboratory of Cardiovascular DiseaseFu Wai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
| | - Yihua Li
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
| | - Xiaolin Liu
- Department of RadiologyBaotou Central HospitalBaotouChina
| | - Shurong Liu
- Department of RadiologyBaotou Central HospitalBaotouChina
| | - Zhenyu Cai
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
| | - Zhaolong Xu
- Institute of cardiovascular diseasethe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Ya Li
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
| | - Xuchun Yuan
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
| | - Xiuhai Guo
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Bin Lu
- Department of RadiologyState Key Laboratory of Cardiovascular DiseaseFu Wai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Kai Sun
- Department of RadiologyState Key Laboratory of Cardiovascular DiseaseFu Wai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of RadiologyFuwai Hospital Chinese Academy of Medical SciencesShenzhenChina
| |
Collapse
|
188
|
Silva H, Lopes NMF. Cardiovascular Effects of Caffeic Acid and Its Derivatives: A Comprehensive Review. Front Physiol 2020; 11:595516. [PMID: 33343392 PMCID: PMC7739266 DOI: 10.3389/fphys.2020.595516] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Caffeic acid (CA) and its phenethyl ester (CAPE) are naturally occurring hydroxycinnamic acids with an interesting array of biological activities; e.g., antioxidant, anti-inflammatory, antimicrobial and cytostatic. More recently, several synthetic analogs have also shown similar properties, and some with the advantage of added stability. The actions of these compounds on the cardiovascular system have not been thoroughly explored despite presenting an interesting potential. Indeed the mechanisms underlying the vascular effects of these compounds particularly need clarifying. The aim of this paper is to provide a comprehensive and up-to-date review on current knowledge about CA and its derivatives in the cardiovascular system. Caffeic acid, CAPE and the synthetic caffeic acid phenethyl amide (CAPA) exhibit vasorelaxant activity by acting on the endothelial and vascular smooth muscle cells. Vasorelaxant mechanisms include the increased endothelial NO secretion, modulation of calcium and potassium channels, and modulation of adrenergic receptors. Together with a negative chronotropic effect, vasorelaxant activity contributes to lower blood pressure, as several preclinical studies show. Their antioxidant, anti-inflammatory and anti-angiogenic properties contribute to an important anti-atherosclerotic effect, and protect tissues against ischemia/reperfusion injuries and the cellular dysfunction caused by different physico-chemical agents. There is an obvious shortage of in vivo studies to further explore these compounds' potential in vascular physiology. Nevertheless, their favorable pharmacokinetic profile and overall lack of toxicity make these compounds suitable for clinical studies.
Collapse
Affiliation(s)
- Henrique Silva
- CBIOS – Universidade Lusófona’s Research Center for Biosciences and Health Technologies, Lisboa, Portugal
- Department of Pharmacological Sciences, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno Miguel F. Lopes
- Department of Pharmacological Sciences, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
189
|
Yan H, Wu X, He Y, Staub D, Wen X, Luo Y. Carotid Intraplaque Neovascularization on Contrast-Enhanced Ultrasound Correlates with Cardiovascular Events and Poor Prognosis: A Systematic Review and Meta-analysis. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 47:167-176. [PMID: 33213970 DOI: 10.1016/j.ultrasmedbio.2020.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/15/2020] [Accepted: 10/23/2020] [Indexed: 02/08/2023]
Abstract
The goal of this meta-analysis is to investigate whether carotid intraplaque neovascularization (IPN) on contrast-enhanced ultrasound (CEUS) correlates with past cardiovascular events (CVEs) and prognosis. The present meta-analysis included 22 studies involving 3232 patients. The pooled analysis revealed that the presence of IPN was significantly associated with a higher incidence of future CVEs (pooled relative risk = 3.28, 95% confidence interval [CI]: 2.28-4.73) and a lower event-free probability (pooled hazard ratio = 2.51, 95% CI: 1.48-4.27). The presence of IPN was significantly associated with higher rates of past cardiac events (odds ratio = 4.25, 95% CI: 2.48-7.29) and past cerebrovascular accidents (odds ratio = 4.83, 95% CI: 2.66-8.78). Our results suggest that carotid IPN on CEUS significantly correlates with past cardiac events and cerebrovascular accidents and can predict future CVEs. Carotid CEUS is useful in CVE risk stratification.
Collapse
Affiliation(s)
- Hualin Yan
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Wu
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Ying He
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Daniel Staub
- Department of Angiology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Xiaorong Wen
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Luo
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
190
|
Activating CD137 Signaling Promotes Sprouting Angiogenesis via Increased VEGFA Secretion and the VEGFR2/Akt/eNOS Pathway. Mediators Inflamm 2020; 2020:1649453. [PMID: 33162828 PMCID: PMC7604604 DOI: 10.1155/2020/1649453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/13/2020] [Accepted: 08/03/2020] [Indexed: 11/18/2022] Open
Abstract
Combination of antiangiogenesis and immunotherapy may be an effective strategy for treatment of solid tumors. Our previous work reported that activation of CD137 signaling promotes intraplaque angiogenesis. A number of studies have demonstrated that vascular endothelial growth factor receptor 2 (VEGFR2) is a key target for angiogenesis. However, it is unknown whether CD137-mediated angiogenesis is related to VEGFR2. In this study, we investigated the effect of CD137 on the VEGFR2 expression and explored the underlying mechanisms of CD137-mediated angiogenesis. Knock-out of CD137 in ApoE−/− mice significantly decreased neovessel density in atherosclerotic plaques. CD137 silencing or inhibition attenuated endothelial cell (ECs) proliferation, migration, and tube formation. We found activation of CD137 signaling for increased VEGFR2 transcription and translation steadily. Moreover, CD137 signaling activated phosphorylated VEGFR2 (Tyr1175) and the downstream Akt/eNOS pathway, whereas neutralizing CD137 signaling weakened the activation of VEGFR2 and the downstream Akt/eNOS pathway. The aortic ring assay further demonstrated that CD137 signaling promoted ECc sprouting. Inhibition of VEGFR2 by siRNA or XL184 (cabozantinib) and inhibition of downstream signaling by LY294002 (inhibits AKT activation) and L-NAME (eNOS inhibitor) remarkably abolished proangiogenic effects of CD137 signaling both in vitro and ex vivo. In addition, the condition medium from CD137-activated ECs and vascular endothelial growth factor A (VEGFA) had similar effects on ECs that expressed high VEGFR2. Additionally, activating CD137 signaling promoted endothelial secretion of VEGFA, while blocking CD137 signaling attenuated VEGFA secretion. In conclusion, activation of CD137 signaling promoted sprouting angiogenesis by increased VEGFA secretion and the VEGFR2/Akt/eNOS pathway. These findings provide a basis for stabilizing intraplaque angiogenesis through VEGFR2 intervatioin, as well as cancer treatment via combination of CD137 agonists and specific VEGFR2 inhibitors.
Collapse
|
191
|
Abstract
Healthy vascular endothelial cells regulate vascular tone and permeability, prevent vessel wall inflammation, enhance thromboresistance, and contribute to general vascular health. Furthermore, they perform important functions including the production of vasoactive substances such as nitric oxide (NO) and endothelium-derived hyperpolarizing factors, as well as the regulation of smooth muscle cell functions. Conversely, vascular endothelial dysfunction leads to atherosclerosis, thereby enhancing the risk of stroke, myocardial infarction, and other cardiovascular diseases (CVDs). Observational studies and randomized trials showed that green tea intake was inversely related to CVD risk. Furthermore, evidence indicates that epigallocatechin gallate (EGCG) found in green tea might exert a preventive effect against CVDs. EGCG acts as an antioxidant, inducing NO release and reducing endothelin-1 production in endothelial cells. EGCG enhances the bioavailability of normal NO by reducing levels of the endogenous NO inhibitor asymmetric dimethylarginine. Furthermore, it inhibits the enhanced expression of adhesion molecules such as vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 and attenuates monocyte adhesion. In addition, EGCG prevents enhanced oxidative stress through the Nrf2/HO-1 pathway. These effects indicate that it might prevent the production of reactive oxygen species, inhibit inflammation, and reduce endothelial cell apoptosis during the initial stages of atherosclerosis. The current review summarizes recent research in this area and discusses novel findings regarding the protective effect of EGCG on endothelial dysfunction and CVDs in general.
Collapse
|
192
|
Meng Q, Pu L, Luo X, Wang B, Li F, Liu B. Regulatory Roles of Related Long Non-coding RNAs in the Process of Atherosclerosis. Front Physiol 2020; 11:564604. [PMID: 33192561 PMCID: PMC7604474 DOI: 10.3389/fphys.2020.564604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis (AS) is the main cause of coronary heart disease, cerebral infarction, and peripheral vascular disease, which comprise serious hazards to human health. Atherosclerosis is characterized by the deposition of lipids on the interior walls of blood vessels, causing an inflammatory response of immune cells, endothelial cells, and smooth muscle cells, and a proliferation cascade reaction. Despite years of research, the underlying pathogenesis of AS is not fully defined. Recent advances in our understanding of the molecular mechanisms by which non-coding RNA influences the initiation and progression of AS have shown that long non-coding RNAs (lncRNAs) regulate important stages in the atherosclerotic process. In this review, we summarize current knowledge of lncRNAs, which influence the development of AS. We review the regulatory processes of lncRNAs on core stages of atherosclerotic progression, including lipid metabolism, inflammation, vascular cell proliferation, apoptosis, adhesion and migration, and angiogenesis. A growing body of evidence suggests that lncRNAs have great potential as new therapeutic targets for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Qingyu Meng
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Luya Pu
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Xizi Luo
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Baisen Wang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, China.,The Key Laboratory for Bionics Engineering, Ministry of Education, Jilin University, Changchun, China.,Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, China.,Key Laboratory for Health Biomedical Materials of Jilin Province, Jilin University, Changchun, China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang, China
| | - Bin Liu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
193
|
Puchenkova OA, Nadezhdin SV, Soldatov VO, Zhuchenko MA, Korshunova DS, Kubekina MV, Korshunov EN, Korokina LV, Golubinskaya PA, Kulikov AL, Gureev VV, Pokrovskiy VM, Patrakhanov EA, Lebedev PR, Denisyuk TA, Belyaeva VS, Movchan EA, Lepetukha EI, Pokrovskiy MV. STUDY OF ANTIATHEROSCLEROTIC AND ENDOTHELIOPROTECTIVE ACTIVITY OF PEPTIDE AGONISTS OF EPOR/CD131 HETERORECEPTOR. PHARMACY & PHARMACOLOGY 2020. [DOI: 10.19163/2307-9266-2020-8-2-100-111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Introduction. The drugs affecting a mitochondrial dysfunction, oxidative stresses, apoptosis and inflammation of the vascular wall, have a high potential for the prevention and treatment of atherosclerotic lesions. In this regard, the use of EPOR/CD131 heteroreceptor agonists which have a similar spectrum of pharmacological effects, is one of the promising strategies in the treatment of cardiovascular diseases.Materials and Methods. The study was carried out on 68 C57Bl/6J male mice. Atherosclerosis was simulated in transgenic animals with an endotheliospecific knockdown of the Polg gene by simulating a balloon injury and keeping on a Western diet. Then, the studied drugs were injected once every 3 days at the dose of 20 μg/kg for 27 days. On the 28-th day, the animals were euthanized and the area of atherosclerotic plaques was assessed. The gene expression associated with the processes of inflammation, antioxidant protection, apoptosis, and angiogenesis was also determined in the aortic tissues. In addition, the endothelium protective effect of peptides on primary cultures of endothelial cells of wild and transgenic Polg-D257A mice was studied.Results. No statistically significant effect of drugs on the area of lipid infiltration have been found. However, the studied peptides have significantly reduced the expression of proinflammatory genes (iNos, Icam1, Vcam1, Sele, Il6, Tnfa), the genes associated with angiogenesis (Vegfa, Kdr, and Hif1a), the expression of proapoptic factors; they decreased the Bax/Bcl-2 ratio by more than 1.5 times. In addition, when supplemented with H2 O2 in vitro, peptides dose-dependently increased endothelial cell survival.Conclusion. The erythropoietin-based peptides can be used to improve the functional state of the vascular wall against the background of atherosclerotic lesions and have a depressing effect on pathobiological processes associated with a mitochondrial dysfunction. In addition, the studied peptides have a significant endothelial protective effect in the induction of oxidative stress in vitro.
Collapse
Affiliation(s)
| | | | - Vladislav O. Soldatov
- Federal Publicly Funded Institution of Science “Institute of Gene Biology of the Russian Academy of Sciences”
| | - Maxim A. Zhuchenko
- Russian Research Center “Kurchatov Institute” – State Science Research Institute of Genetics
| | - Diana S. Korshunova
- Federal Publicly Funded Institution of Science “Institute of Gene Biology of the Russian Academy of Sciences”
| | - Marina V. Kubekina
- Federal Publicly Funded Institution of Science “Institute of Gene Biology of the Russian Academy of Sciences”
| | - Evgeny N. Korshunov
- Federal Publicly Funded Institution of Science “Institute of Gene Biology of the Russian Academy of Sciences”
| | | | - Polina A. Golubinskaya
- Clinical diagnostic laboratory, Budgetary institution of public healthcare of the Voronezh Region “Voronezh Regional Clinical Ophthalmological Hospital”
| | | | | | | | | | | | - Tatyana A. Denisyuk
- Federal State Budgetary Educational Institution of Higher Education “Kursk State Medical University”
| | | | | | | | | |
Collapse
|
194
|
Wijeratne T, Menon R, Sales C, Karimi L, Crewther S. Carotid artery stenosis and inflammatory biomarkers: the role of inflammation-induced immunological responses affecting the vascular systems. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1276. [PMID: 33178808 PMCID: PMC7607082 DOI: 10.21037/atm-20-4388] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The death, disability and economic cost of stroke are enormous. Indeed, among the 16 million people worldwide who suffer a stroke' annually, nearly six million die, and another five million are left permanently disabled making prevention of stroke one of the most important priorities in healthcare. Currently carotid artery stenosis (CS) or narrowing of the common carotid artery (CCA) or internal carotid artery (ICA) due to atherosclerotic plaque, accounts for 20-30% of all ischemic strokes. Atherosclerosis is now regarded as a chronic inflammatory disease in response to vascular compromise especially from hypertension. This has long been known to lead to inflammation and atherosclerotic plaque formation in the blood vessels. This mini-review aims to highlight the role of inflammation and neuro-immunological processes in carotid artery disease. Various cellular elements of inflammation and advanced imaging techniques have been identified as potential markers of plaque progression. Therapies related to decreasing and modulating immune-responsive inflammation in the carotid vessels have been shown to translate into decreased occurrence of acute neurologic events and improvement of clinical outcomes.
Collapse
Affiliation(s)
- Tissa Wijeratne
- Department of Neurology, AIMSS, WHCRE level three, Sunshine Hospital and Melbourne Medical School, St Albans, Victoria, Australia.,School of Public health and Psychology, La Trobe University, Bundoora, Victoria, Australia.,Department of Medicine, Faculty of Medicine, Rajarata University, Saliyapura, Anuradhapura, Sri Lanka
| | - Rohit Menon
- Department of Neurology and Stroke Service, Western Health, AIMSS, level 3, WHCRE, Sunshine Hospital, St Albans, Victoria, Australia
| | - Carmela Sales
- Department of Neurology and Stroke Service, Western Health, AIMSS, level 3, WHCRE, Sunshine Hospital, St Albans, Victoria, Australia
| | - Leila Karimi
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia.,Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Sheila Crewther
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| |
Collapse
|
195
|
Mensah SA, Nersesyan AA, Ebong EE. Endothelial Glycocalyx-Mediated Intercellular Interactions: Mechanisms and Implications for Atherosclerosis and Cancer Metastasis. Cardiovasc Eng Technol 2020; 12:72-90. [PMID: 33000443 PMCID: PMC7904750 DOI: 10.1007/s13239-020-00487-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Purpose The endothelial glycocalyx (GCX) plays a critical role in the health of the vascular system. Degradation of the GCX has been implicated in the onset of diseases like atherosclerosis and cancer because it disrupts endothelial cell (EC) function that is meant to protect from atherosclerosis and cancer. Examples of such EC function include interendothelial cell communication via gap junctions and receptor-mediated interactions between endothelial and tumor cells. This review focuses on GCX-dependent regulation of these intercellular interactions in healthy and diseased states. The ultimate goal is to build new knowledge that can be applied to developing GCX regeneration strategies that can control intercellular interaction in order to combat the progression of diseases such as atherosclerosis and cancer. Methods In vitro and in vivo studies were conducted to determine the baseline expression of GCX in physiologically relevant conditions. Chemical and mechanical GCX degradation approaches were employed to degrade the GCX. The impact of intact versus degraded GCX on intercellular interactions was assessed using cytochemistry, histochemistry, a Lucifer yellow dye transfer assay, and confocal, intravital, and scanning electron microscopy techniques. Results Relevant to atherosclerosis, we found that GCX stability determines the expression and functionality of Cx43 in gap junction-mediated EC-to-EC communication. Relevant to cancer metastasis, we found that destabilizing the GCX through either disturbed flow-induced or enzyme induced GCX degradation results in increased E-selectin receptor-mediated EC-tumor cell interactions. Conclusion Our findings lay a foundation for future endothelial GCX-targeted therapy, to control intercellular interactions and limit the progression of atherosclerosis and cancer.
Collapse
Affiliation(s)
- Solomon A Mensah
- Department of Bioengineering, Northeastern University, Boston, MA, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Alina A Nersesyan
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Eno E Ebong
- Department of Bioengineering, Northeastern University, Boston, MA, USA. .,Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 335 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA. .,Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
196
|
Biomimetic 3D Models for Investigating the Role of Monocytes and Macrophages in Atherosclerosis. Bioengineering (Basel) 2020; 7:bioengineering7030113. [PMID: 32947976 PMCID: PMC7552756 DOI: 10.3390/bioengineering7030113] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis, the inflammation of artery walls due to the accumulation of lipids, is the most common underlying cause for cardiovascular diseases. Monocytes and macrophages are major cells that contribute to the initiation and progression of atherosclerotic plaques. During this process, an accumulation of LDL-laden macrophages (foam cells) and an alteration in the extracellular matrix (ECM) organization leads to a local vessel stiffening. Current in vitro models are carried out onto two-dimensional tissue culture plastic and cannot replicate the relevant microenvironments. To bridge the gap between in vitro and in vivo conditions, we utilized three-dimensional (3D) collagen matrices that allowed us to mimic the ECM stiffening during atherosclerosis by increasing collagen density. First, human monocytic THP-1 cells were embedded into 3D collagen matrices reconstituted at low and high density. Cells were subsequently differentiated into uncommitted macrophages (M0) and further activated into pro- (M1) and anti-inflammatory (M2) phenotypes. In order to mimic atherosclerotic conditions, cells were cultured in the presence of oxidized LDL (oxLDL) and analyzed in terms of oxLDL uptake capability and relevant receptors along with their cytokine secretomes. Although oxLDL uptake and larger lipid size could be observed in macrophages in a matrix dependent manner, monocytes showed higher numbers of oxLDL uptake cells. By analyzing major oxLDL uptake receptors, both monocytes and macrophages expressed lectin-like oxidized low-density lipoprotein receptor-1 (LOX1), while enhanced expression of scavenger receptor CD36 could be observed only in M2. Notably, by analyzing the secretome of macrophages exposed to oxLDL, we demonstrated that the cells could, in fact, secrete adipokines and growth factors in distinct patterns. Besides, oxLDL appeared to up-regulate MHCII expression in all cells, while an up-regulation of CD68, a pan-macrophage marker, was found only in monocytes, suggesting a possible differentiation of monocytes into a pro-inflammatory macrophage. Overall, our work demonstrated that collagen density in the plaque could be one of the major factors driving atherosclerotic progression via modulation of monocyte and macrophages behaviors.
Collapse
|
197
|
Wang W, Liu Y, You L, Sun M, Qu C, Dong X, Yin X, Ni J. Inhibitory effects of Paris saponin I, II, Ⅵ and Ⅶ on HUVEC cells through regulation of VEGFR2, PI3K/AKT/mTOR, Src/eNOS, PLCγ/ERK/MERK, and JAK2-STAT3 pathways. Biomed Pharmacother 2020; 131:110750. [PMID: 32942160 DOI: 10.1016/j.biopha.2020.110750] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023] Open
Abstract
Rhizoma Paris is a popular Chinese medicine in clinics. It contains four main saponins which are its major bioactive compounds. These saponins are Paris saponin I, II, VI and VII (PSI, PSII, PSVI and PSVII, respectively). Up to now, the research using HUVEC cells to evaluate the anti-angiogenic activity of four saponins is blank. The purpose of this study was to evaluate the anti-angiogenic properties (also known as angiotoxicity) of the four saponins in Rhizoma Paris on vascular endothelial cells-HUVEC cells, and to investigate the underlying mechanism, which has not been studied before. In this study, MTT assay, Lactate dehydrogenase (LDH) assay, wound healing experiments, transwell cell invasion assay, tubule formation experiment, DAPI staining, AV-PI double staining, and cell cycle analysis were used to determine the effects of Paris saponins. The results showed that, with increases in concentrations of PSI, PSII, PSVI and PSVII, the viability of HUVEC cells decreased significantly. In addition, four saponins dose-dependent enhanced LDH release and inhibited HUVEC cell migration, invasion, and angiogenesis. In terms of mechanism, PSI significantly inhibited protein expression in multiple signaling pathways. In particular, with the VEGF2 as the target, it activate the downstream PI3K / AKT / mTOR, SRC / eNOS, P38, PLCγ / ERK / MERK and JAK2/STAT3 signaling pathways. In conclusion, PSI, PSII, PSVI and PSVII can inhibit endothelial cell proliferation, migration and invasion, block endothelial cell cycle, induce endothelial cell apoptosis, act on protein expression in several anti-angiogenic signaling pathways, and finally inhibit angiogenesis in vitro. This study provides further data support for the clinical application of Paris saponins as antiangiogenic drugs.
Collapse
Affiliation(s)
- Wenping Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yi Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Longtai You
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingyi Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Changhai Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - XiaoXv Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jian Ni
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
198
|
Rezaei ZS, Shahangian SS, Hasannia S, Sajedi RH. Development of a phage display-mediated immunoassay for the detection of vascular endothelial growth factor. Anal Bioanal Chem 2020; 412:7639-7648. [PMID: 32876721 DOI: 10.1007/s00216-020-02901-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 01/08/2023]
Abstract
Because of the critical role of vascular endothelial growth factor (VEGF) in angiogenesis and its significantly increased serum levels in early stages of cancer, VEGF is considered an important prognostic biomarker in different cancers. Herein, the amplification power of PCR combined with phage displaying anti-VEGF VHH, a sensitive real-time immunoassay, was precisely designed based on phage display-mediated immuno-PCR (PD-IPCR) for the detection of VEGF. This system benefits from strong and specific binding of antigen and antibody in a sandwich immunosorbent assay platform using avastin (anti-VEGF monoclonal antibody) as the capture antibody. The anti-VEGF phage particles were used as both anti-VEGF agent and DNA template in the PD-IPCR. Anti-VEGF phage ELISA showed a linear range of 3-250 ng/ml and a limit of detection (LOD) of 1.1 ng/ml. Using the PD-IPCR method, the linear range of VEGF detection was found to be 0.06-700 ng/ml, with a detection limit of 3 pg/ml. The recovery rate in serum ranged from 83% to 99%, with a relative standard deviation of 1.2-4.9%. These values indicate that the method has good sensitivity for use in clinical analysis. The proposed method was successfully applied to the clinical determination of VEGF in human serum samples, and the results showed excellent correlation with conventional ELISA (R2 = 0.995). The novel immunoassay provides a specific and sensitive immunoassay protocol for VEGF detection at very low levels. Graphical abstract.
Collapse
Affiliation(s)
- Zahra S Rezaei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran, 14115-154, Iran
| | - S Shirin Shahangian
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Guilan, 4199613776, Iran
| | - Sadegh Hasannia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran, 14115-154, Iran
| | - Reza H Sajedi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran, 14115-154, Iran.
| |
Collapse
|
199
|
Enriquez J, Mims BMD, Trasti S, Furr KL, Grisham MB. Genomic, microbial and environmental standardization in animal experimentation limiting immunological discovery. BMC Immunol 2020; 21:50. [PMID: 32878597 PMCID: PMC7464063 DOI: 10.1186/s12865-020-00380-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Background The use of inbred mice housed under standardized environmental conditions has been critical in identifying immuno-pathological mechanisms in different infectious and inflammatory diseases as well as revealing new therapeutic targets for clinical trials. Unfortunately, only a small percentage of preclinical intervention studies using well-defined mouse models of disease have progressed to clinically-effective treatments in patients. The reasons for this lack of bench-to-bedside transition are not completely understood; however, emerging data suggest that genetic diversity and housing environment may greatly influence muring immunity and inflammation. Results Accumulating evidence suggests that certain immune responses and/or disease phenotypes observed in inbred mice may be quite different than those observed in their outbred counterparts. These differences have been thought to contribute to differing immune responses to foreign and/or auto-antigens in mice vs. humans. There is also a growing literature demonstrating that mice housed under specific pathogen free conditions possess an immature immune system that remarkably affects their ability to respond to pathogens and/or inflammation when compared with mice exposed to a more diverse spectrum of microorganisms. Furthermore, recent studies demonstrate that mice develop chronic cold stress when housed at standard animal care facility temperatures (i.e. 22–24 °C). These temperatures have been shown alter immune responses to foreign and auto-antigens when compared with mice housed at their thermo-neutral body temperature of 30–32 °C. Conclusions Exposure of genetically diverse mice to a spectrum of environmentally-relevant microorganisms at housing temperatures that approximate their thermo-neutral zone may improve the chances of identifying new and more potent therapeutics to treat infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Josue Enriquez
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Brianyell Mc Daniel Mims
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Scott Trasti
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.,Laboratory Animal Research Center, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Kathryn L Furr
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA
| | - Matthew B Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street STOP 6591, Lubbock, TX, 79430-6591, USA.
| |
Collapse
|
200
|
Wang P, Liu S, Wang Z, Zhao H, Zhang X. Altered levels of circulating natural antibodies against VEGFR1-derived peptide in atherosclerosis. J Int Med Res 2020; 48:300060520948750. [PMID: 32811267 PMCID: PMC7513417 DOI: 10.1177/0300060520948750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/20/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Several lines of evidence have pointed to a protective role of natural antibodies in chronic diseases like atherosclerosis and cancer. Vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2 are important regulators of angiogenesis and may be involved in the development of atherosclerosis. In this retrospective study, we developed an in-house enzyme-linked immunosorbent assay to assess whether natural IgG levels against VEGFR1 and the regulatory T cell markers CD25 and FOXP3 were associated with atherosclerosis. METHODS A total of 218 patients with atherosclerosis and 200 healthy controls were enrolled. All patients had atherosclerotic carotid plaques. Carotid intima-media thickness was analyzed using a diagnostic ultrasound system. RESULTS Plasma anti-VEGFR1 IgG levels were significantly lower in patients with atherosclerosis than control subjects. Decreased anti-VEGFR1 IgG levels were more obvious in male patients. Spearman correlation analysis showed no significant correlation between natural IgG levels and carotid intima-media thickness. CONCLUSIONS Decreased levels of anti-VEGFR1 IgG may be involved in development of atherosclerosis and related conditions.
Collapse
Affiliation(s)
- Peng Wang
- Second Hospital of Jilin University, Changchun, China
| | - Siqi Liu
- Second Hospital of Jilin University, Changchun, China
| | - Zhenqi Wang
- School of Public Health, Jilin University, Changchun,
China
| | - Huan Zhao
- Second Hospital of Jilin University, Changchun, China
| | - Xuan Zhang
- Second Hospital of Jilin University, Changchun, China
| |
Collapse
|