1
|
Tamargo IA, Baek KI, Kim Y, Park C, Jo H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat Rev Cardiol 2023; 20:738-753. [PMID: 37225873 PMCID: PMC10206587 DOI: 10.1038/s41569-023-00883-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/26/2023]
Abstract
Atherosclerotic diseases such as myocardial infarction, ischaemic stroke and peripheral artery disease continue to be leading causes of death worldwide despite the success of treatments with cholesterol-lowering drugs and drug-eluting stents, raising the need to identify additional therapeutic targets. Interestingly, atherosclerosis preferentially develops in curved and branching arterial regions, where endothelial cells are exposed to disturbed blood flow with characteristic low-magnitude oscillatory shear stress. By contrast, straight arterial regions exposed to stable flow, which is associated with high-magnitude, unidirectional shear stress, are relatively well protected from the disease through shear-dependent, atheroprotective endothelial cell responses. Flow potently regulates structural, functional, transcriptomic, epigenomic and metabolic changes in endothelial cells through mechanosensors and mechanosignal transduction pathways. A study using single-cell RNA sequencing and chromatin accessibility analysis in a mouse model of flow-induced atherosclerosis demonstrated that disturbed flow reprogrammes arterial endothelial cells in situ from healthy phenotypes to diseased ones characterized by endothelial inflammation, endothelial-to-mesenchymal transition, endothelial-to-immune cell-like transition and metabolic changes. In this Review, we discuss this emerging concept of disturbed-flow-induced reprogramming of endothelial cells (FIRE) as a potential pro-atherogenic mechanism. Defining the flow-induced mechanisms through which endothelial cells are reprogrammed to promote atherosclerosis is a crucial area of research that could lead to the identification of novel therapeutic targets to combat the high prevalence of atherosclerotic disease.
Collapse
Affiliation(s)
- Ian A Tamargo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA
| | - Kyung In Baek
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Yerin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Christian Park
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Molecular and Systems Pharmacology Program, Emory University, Atlanta, GA, USA.
- Department of Medicine, Emory University School, Atlanta, GA, USA.
| |
Collapse
|
2
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
3
|
Pekas EJ, Allen MF, Park SY. Prolonged sitting and peripheral vascular function: potential mechanisms and methodological considerations. J Appl Physiol (1985) 2023; 134:810-822. [PMID: 36794688 PMCID: PMC10042610 DOI: 10.1152/japplphysiol.00730.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Sitting time is associated with increased risks for subclinical atherosclerosis and cardiovascular disease development, and this is thought to be partially due to sitting-induced disturbances in macro- and microvascular function as well as molecular imbalances. Despite surmounting evidence supporting these claims, contributing mechanisms to these phenomena remain largely unknown. In this review, we discuss evidence for potential mechanisms of sitting-induced perturbations in peripheral hemodynamics and vascular function and how these potential mechanisms may be targeted using active and passive muscular contraction methods. Furthermore, we also highlight concerns regarding the experimental environment and population considerations for future studies. Optimizing prolonged sitting investigations may allow us to not only better understand the hypothesized sitting-induced transient proatherogenic environment but to also enhance methods and devise mechanistic targets to salvage sitting-induced attenuations in vascular function, which may ultimately play a role in averting atherosclerosis and cardiovascular disease development.
Collapse
Affiliation(s)
- Elizabeth J Pekas
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska, United States
| | - Michael F Allen
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska, United States
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska at Omaha, Omaha, Nebraska, United States
| |
Collapse
|
4
|
Reolizo LM, Williams H, Wadey K, Frankow A, Li Z, Gaston K, Jayaraman PS, Johnson JL, George SJ. Inhibition of Intimal Thickening By PRH (Proline-Rich Homeodomain) in Mice. Arterioscler Thromb Vasc Biol 2023; 43:456-473. [PMID: 36700427 PMCID: PMC9944393 DOI: 10.1161/atvbaha.122.318367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/05/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Late vein graft failure is caused by intimal thickening resulting from endothelial cell (EC) damage and inflammation which promotes vascular smooth muscle cell (VSMC) dedifferentiation, migration, and proliferation. Nonphosphorylatable PRH (proline-rich homeodomain) S163C:S177C offers enhanced stability and sustained antimitotic effect. Therefore, we investigated whether adenovirus-delivered PRH S163C:S177C protein attenuates intimal thickening via VSMC phenotype modification without detrimental effects on ECs. METHODS PRH S163C:S177C was expressed in vitro (human saphenous vein-VSMCs and human saphenous vein-ECs) and in vivo (ligated mouse carotid arteries) by adenoviruses. Proliferation, migration, and apoptosis were quantified and phenotype was assessed using Western blotting for contractile filament proteins and collagen gel contraction. EC inflammation was quantified using VCAM (vascular cell adhesion protein)-1, ICAM (intercellular adhesion molecule)-1, interleukin-6, and monocyte chemotactic factor-1 measurement and monocyte adhesion. Next Generation Sequencing was utilized to identify novel downstream mediators of PRH action and these and intimal thickening were investigated in vivo. RESULTS PRH S163C:S177C inhibited proliferation, migration, and apoptosis and promoted contractile phenotype (enhanced contractile filament proteins and collagen gel contraction) compared with virus control in human saphenous vein-VSMCs. PRH S163C:S177C expression in human saphenous vein-ECs significantly reduced apoptosis, without affecting cell proliferation and migration, while reducing TNF (tumor necrosis factor)-α-induced VCAM-1 and ICAM-1 and monocyte adhesion and suppressing interleukin-6 and monocyte chemotactic factor-1 protein levels. PRH S163C:S177C expression in ligated murine carotid arteries significantly impaired carotid artery ligation-induced neointimal proliferation and thickening without reducing endothelial coverage. Next Generation Sequencing revealed STAT-1 (signal transducer and activator of transcription 1) and HDAC-9 (histone deacetylase 9) as mediators of PRH action and was supported by in vitro and in vivo analyses. CONCLUSIONS We observed PRH S163C:S177C attenuated VSMC proliferation, and migration and enhanced VSMC differentiation at least in part via STAT-1 and HDAC-9 signaling while promoting endothelial repair and anti-inflammatory properties. These findings highlight the potential for PRH S163C:S177C to preserve endothelial function whilst suppressing intimal thickening, and reducing late vein graft failure.
Collapse
Affiliation(s)
- Lien M. Reolizo
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Helen Williams
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Kerry Wadey
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Aleksandra Frankow
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Ze Li
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Kevin Gaston
- School of Medicine and Biodiscovery Institute, Faculty of Medicine & Health Sciences, University of Nottingham, UK (K.G., P.-S.J.)
| | - Padma-Sheela Jayaraman
- School of Medicine and Biodiscovery Institute, Faculty of Medicine & Health Sciences, University of Nottingham, UK (K.G., P.-S.J.)
| | - Jason L. Johnson
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| | - Sarah J. George
- Bristol Heart Institute, University of Bristol, UK (L.M.R., H.W., K.W., A.F., Z.L., J.L.J., S.J.G.)
| |
Collapse
|
5
|
Peyronnel C, Totoson P, Martin H, Demougeot C. Relevance of circulating markers of endothelial activation for cardiovascular risk assessment in rheumatoid arthritis: a narrative review. Life Sci 2023; 314:121264. [PMID: 36470540 DOI: 10.1016/j.lfs.2022.121264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Rheumatoid arthritis (RA) is associated with excessive cardiovascular mortality secondary to premature atherosclerosis, in which endothelial activation (EA) plays a central role. EA is characterized by loss of vascular integrity, expression of leucocyte adhesion molecules, transition from antithrombotic to prothrombotic phenotype, cytokines production, shedding of membrane microparticles and recruitment of endothelial progenitor cells. As EA is an early event in atherogenesis, circulating markers of EA are putative markers of vascular pathology and cardiovascular (CV) risk. After a presentation of biology of EA, the present review analyzed the available data regarding changes in EA markers in RA in link with the vascular pathology and CV events, discussed their relevance as biomarkers of CV risk and proposed future directions.
Collapse
Affiliation(s)
- Célian Peyronnel
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Perle Totoson
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Hélène Martin
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France
| | - Céline Demougeot
- PEPITE EA 4267, Université de Franche-Comté, F-25000 Besançon, France.
| |
Collapse
|
6
|
Kolluru GK, Shackelford RE, Shen X, Dominic P, Kevil CG. Sulfide regulation of cardiovascular function in health and disease. Nat Rev Cardiol 2023; 20:109-125. [PMID: 35931887 PMCID: PMC9362470 DOI: 10.1038/s41569-022-00741-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2022] [Indexed: 01/21/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as a gaseous signalling molecule with crucial implications for cardiovascular health. H2S is involved in many biological functions, including interactions with nitric oxide, activation of molecular signalling cascades, post-translational modifications and redox regulation. Various preclinical and clinical studies have shown that H2S and its synthesizing enzymes - cystathionine γ-lyase, cystathionine β-synthase and 3-mercaptosulfotransferase - can protect against cardiovascular pathologies, including arrhythmias, atherosclerosis, heart failure, myocardial infarction and ischaemia-reperfusion injury. The bioavailability of H2S and its metabolites, such as hydropersulfides and polysulfides, is substantially reduced in cardiovascular disease and has been associated with single-nucleotide polymorphisms in H2S synthesis enzymes. In this Review, we highlight the role of H2S, its synthesizing enzymes and metabolites, their roles in the cardiovascular system, and their involvement in cardiovascular disease and associated pathologies. We also discuss the latest clinical findings from the field and outline areas for future study.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Paari Dominic
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Medicine, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Center of Excellence for Cardiovascular Diseases & Sciences, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
7
|
Yanikoglu A. The Probable Role of Chlamydia pneumoniae Infection in Acute Stroke. Infect Dis (Lond) 2023. [DOI: 10.5772/intechopen.109582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cardiovascular diseases are the most leading cause of worldwide mortality. According to USA statistics, about 1 of 6 cardiovascular deaths is due to stroke. Stroke is the second most common cause of death and a chief cause of disability due to EU data. Treatment, care providing, rehabilitation costs and with the labor loss, the overall cost in EU due to stroke was estimated about €45 billion in year 2017. Acute stroke due to infectious diseases via several possible mechanisms with various clinical presentations were previously reported in the literature. Chlamydia pneumoniae is an obligate intracellular bacteria and extremely common in adult individuals. Besides it being a major cause of pneumonia in adults, association between atherosclerosis and vascular diseases was demonstrated by several sero-epidemiological studies and by direct detection of organism in atherosclerotic lesions by electron microscopy, immunohistochemistry, polymerase chain reaction. Also, several sero-epidemiological studies have demonstrated a link between Chlamydia pneumoniae infection and acute stroke. In this chapter, we will summarize the data in literature regarding the association between Chlamydia pneumoniae infection and acute stroke and we will try to explain the possible mechanisms that could be responsible in pathophysiology of stroke in these patients.
Collapse
|
8
|
Cui Y, Shi W, Zhang K, Hou Z, Wang Y, Yan W, Ma Q, He S, Huang J, Lu C, Wang Y, Wang G, Qiu J. Temporal-spatial low shear stress induces heterogenous distribution of hematopoietic stem cell budding in zebrafish. Cell Mol Life Sci 2022; 79:399. [PMID: 35792959 PMCID: PMC11073138 DOI: 10.1007/s00018-022-04411-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/22/2022] [Accepted: 06/01/2022] [Indexed: 11/03/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) originate from endothelial cells (ECs) localized on the ventral side of the dorsal aorta (DA), and hemodynamic parameters may suffer sharp changes in DA at HSPCs development stage for intersegmental vessel formation. However, the temporal-spatial shear stress parameters and biomechanics mechanisms of HSPC budding remain unknown. Here, we found that the hematopoietic endothelium (HE) in the aorta-gonad-mesonephros was heterogeneous; that is, HEs were mainly distributed at the ventral side of the vascular bifurcation in zebrafish embryos, which was found to show low shear stress (LSS) through numerical simulation analysis. Furthermore, HSPCs localized in the posterior somite of aorta-gonad-mesonephros with slow velocity. On the temporal scale, there was a slow velocity and LSS during HE budding from 36 h post-fertilization and decreased shear stress with drug expanded HSPC numbers. Mechanistically, matrix metalloproteinase (MMP) expression and macrophage chemotaxis were significantly increased in HEs by RNA-seq. After treatment with an MMP13 inhibitor, HSPCs were significantly reduced in both the aorta-gonad-mesonephros and caudal hematopoietic tissue in embryos. Our results show that HSPC budding is heterogeneous, and the mechanism is that physiological LSS controls the emergence of HSPCs by promoting the accumulation of macrophages and subsequent MMP expression.
Collapse
Affiliation(s)
- Yuliang Cui
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wenpeng Shi
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Zhengjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yanyun Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - WenHua Yan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Qinfeng Ma
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Shicheng He
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Junli Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Chenfei Lu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
9
|
Salvador J, Iruela-Arispe ML. Nuclear Mechanosensation and Mechanotransduction in Vascular Cells. Front Cell Dev Biol 2022; 10:905927. [PMID: 35784481 PMCID: PMC9247619 DOI: 10.3389/fcell.2022.905927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/31/2022] [Indexed: 11/24/2022] Open
Abstract
Vascular cells are constantly subjected to physical forces associated with the rhythmic activities of the heart, which combined with the individual geometry of vessels further imposes oscillatory, turbulent, or laminar shear stresses on vascular cells. These hemodynamic forces play an important role in regulating the transcriptional program and phenotype of endothelial and smooth muscle cells in different regions of the vascular tree. Within the aorta, the lesser curvature of the arch is characterized by disturbed, oscillatory flow. There, endothelial cells become activated, adopting pro-inflammatory and athero-prone phenotypes. This contrasts the descending aorta where flow is laminar and endothelial cells maintain a quiescent and atheroprotective phenotype. While still unclear, the specific mechanisms involved in mechanosensing flow patterns and their molecular mechanotransduction directly impact the nucleus with consequences to transcriptional and epigenetic states. The linker of nucleoskeleton and cytoskeleton (LINC) protein complex transmits both internal and external forces, including shear stress, through the cytoskeleton to the nucleus. These forces can ultimately lead to changes in nuclear integrity, chromatin organization, and gene expression that significantly impact emergence of pathology such as the high incidence of atherosclerosis in progeria. Therefore, there is strong motivation to understand how endothelial nuclei can sense and respond to physical signals and how abnormal responses to mechanical cues can lead to disease. Here, we review the evidence for a critical role of the nucleus as a mechanosensor and the importance of maintaining nuclear integrity in response to continuous biophysical forces, specifically shear stress, for proper vascular function and stability.
Collapse
Affiliation(s)
| | - M. Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
10
|
Eton D, Zhou G, He TC, Bartholomew A, Patil R. Filgrastim, fibrinolysis, and neovascularization. J Tissue Eng Regen Med 2022; 16:496-510. [PMID: 35175691 PMCID: PMC9302657 DOI: 10.1002/term.3284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 12/17/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
Abstract
Segmental recanalization of chronically occluded arteries was observed in patients with chronic limb-threatening ischemia (CLTI) treated with Filgrastim, a granulocyte colony stimulating factor, every 72 h for up to a month, and an infra-geniculate programmed compression pump (PCP) for 3 h daily. Molecular evidence for fibrinolysis and neovascularization was sought. CLTI patients were treated with PCP alone (N = 19), or with Filgrastim and PCP (N = 8 and N = 6, at two institutions). Enzyme-Linked Immunosorbent Assay was used to measure the plasma concentration of plasmin and of fibrin degradation products (FDP), and the serum concentration of proteins associated with neovascularization. In the PCP-alone group, blood was sampled on Day 1 (baseline) and after 30 days of daily PCP. In the Filgrastim and PCP group, blood was drawn on Day 1, and 1 day after the 5th and the 10th Filgrastim doses. Each blood draw occurred before and after 2 h of supervised PCP. Significant (p < 0.01) PCP independent increases in the plasma concentration of plasmin (>10-fold) and FDP (>5-fold) were observed 1 day after both the 5th and the 10th Filgrastim doses, compared to Day 1. Significant (p < 0.05) increases in the concentration of pro-angiogenic proteins (e.g., HGF, MMP-9, VEGF A) were also observed. Filgrastim at this novel dosimetry induced fibrinolysis without causing acute hemorrhage, in addition to inducing a pro-angiogenic milieu conducive to NV. Further clinical testing is warranted at this novel dosimetry in CLTI, as well as in other chronically ischemic tissue beds. Trial registration. https://clinicaltrials.gov/ct2/show/NCT02802852.
Collapse
Affiliation(s)
- Darwin Eton
- Department of Surgery, University of Illinois Chicago, Chicago, Illinois, USA
| | - Guolin Zhou
- GCIS, University of Chicago, Chicago, Illinois, USA
| | - Tong-Chuan He
- Department of Orthopedic Surgery and Rehabilitation Medicine, University of Chicago, Chicago, Illinois, USA
| | - Amelia Bartholomew
- Department of Surgery, College of Medicine Research, University of Illinois Chicago, Chicago, Illinois, USA
| | - Rachana Patil
- Department of Pediatrics, Division of Stem Cell Transplant and Regenerative Medicine, Stanford University, Palo Alto, California, USA
| |
Collapse
|
11
|
Atcha H, Meli VS, Davis CT, Brumm KT, Anis S, Chin J, Jiang K, Pathak MM, Liu WF. Crosstalk Between CD11b and Piezo1 Mediates Macrophage Responses to Mechanical Cues. Front Immunol 2021; 12:689397. [PMID: 34630381 PMCID: PMC8493066 DOI: 10.3389/fimmu.2021.689397] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/12/2021] [Indexed: 11/15/2022] Open
Abstract
Macrophages are versatile cells of the innate immune system that perform diverse functions by responding to dynamic changes in their microenvironment. While the effects of soluble cues, including cytokines and chemokines, have been widely studied, the effects of physical cues, including mechanical stimuli, in regulating macrophage form and function are less well understood. In this study, we examined the effects of static and cyclic uniaxial stretch on macrophage inflammatory and healing activation. We found that cyclic stretch altered macrophage morphology and responses to IFNγ/LPS and IL4/IL13. Interestingly, we found that both static and cyclic stretch suppressed IFNγ/LPS induced inflammation. In contrast, IL4/IL13 mediated healing responses were suppressed with cyclic but enhanced with static stretch conditions. Mechanistically, both static and cyclic stretch increased expression of the integrin CD11b (αM integrin), decreased expression of the mechanosensitive ion channel Piezo1, and knock down of either CD11b or Piezo1 through siRNA abrogated stretch-mediated changes in inflammatory responses. Moreover, we found that knock down of CD11b enhanced the expression of Piezo1, and conversely knock down of Piezo1 enhanced CD11b expression, suggesting the potential for crosstalk between integrins and ion channels. Finally, stretch-mediated differences in macrophage activation were also dependent on actin, since pharmacological inhibition of actin polymerization abrogated the changes in activation with stretch. Together, this study demonstrates that the physical environment synergizes with biochemical cues to regulate macrophage morphology and function, and suggests a role for CD11b and Piezo1 crosstalk in mechanotransduction in macrophages.
Collapse
Affiliation(s)
- Hamza Atcha
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Vijaykumar S. Meli
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Chase T. Davis
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Kyle T. Brumm
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Sara Anis
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Jessica Chin
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Kevin Jiang
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
| | - Medha M. Pathak
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States
| | - Wendy F. Liu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, United States
- Department of Chemical Engineering and Materials Science, University of California, Irvine, Irvine, CA, United States
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
12
|
Zhao J, Yang M, Wu J. CXCL16 may be a predisposing factor to atherosclerosis: An animal study. Mol Med Rep 2021; 24:716. [PMID: 34396447 DOI: 10.3892/mmr.2021.12355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/29/2020] [Indexed: 11/06/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory process initiated when lipoprotein is retained in the arterial wall. Leukocyte recruitment accelerates this process. CXC chemokine ligand 16 (CXCL16) acts as a chemokine to attract immune cells and also facilitates the phagocytosis process of modified low‑density lipoprotein. Whether CXCL16 promotes or inhibits the pathological process of AS remains to be elucidated. To clarify this, CXCL16 gene was introduced into C57BL/6J wild‑type mice to establish a stable CXCL16 overexpression mouse model. The initial changes of AS in mice were induced by high‑fat diet (HFD). To study how the interaction of HFD and CXCL16 affected fatty acid metabolism and deposition, body weight and plasma lipid profile were assessed. Soluble CXCL16, matrix metalloproteinase‑9, monocyte chemoattractant protein‑1 and intercellular adhesion molecule‑1 were detected by immunohistochemistry and ELISA to identify how CXCL16 affects AS lesion formation. The present study suggested that overexpression of CXCL16 combined with HFD lead to atherogenesis by upregulating the aforementioned inflammatory related genes at a protein level. The present study was the first, to the best of the authors' knowledge, to build a CXCL16 homozygous transgenic mice model to study how overexpressed CXCL16 is associated with AS for intervening in the occurrence and development of AS.
Collapse
Affiliation(s)
- Junbi Zhao
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Menglin Yang
- Department of Burns, Plastic Surgery and Dermatology, No. 922 Hospital of Joint Support Unit of the People's Liberation Army Hengyang, Hunan 421002, P.R. China
| | - Jie Wu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
13
|
Engler AJ, Wang Y. Editorial: Understanding molecular interactions that underpin vascular mechanobiology. APL Bioeng 2021; 5:030401. [PMID: 34258496 PMCID: PMC8253597 DOI: 10.1063/5.0058611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 02/04/2023] Open
Abstract
Cells are exposed to a variety of mechanical forces in their daily lives, especially endothelial cells that are stretched from vessel distention and are exposed to hemodynamic shear stress from a blood flow. Exposure to excessive forces can induce a disease, but the molecular details on how these cells perceive forces, transduce them into biochemical signals and genetic events, i.e., mechanotransduction, and integrate them into physiological or pathological changes remain unclear. However, seminal studies in endothelial cells over the past several decades have begun to elucidate some of these signals. These studies have been highlighted in APL Bioengineering and elsewhere, describing a complex temporal pattern where forces are sensed immediately by ion channels and force-dependent conformational changes in surface proteins, followed by biochemical cascades, cytoskeletal contraction, and nuclear remodeling that can affect long-term changes in endothelial morphology and fate. Key examples from the endothelial literature that have established these pathways include showing that integrins and Flk-1 or VE-cadherin act as shear stress transducers, activating downstream proteins such as Cbl and Nckβ or Src, respectively. In this Editorial, we summarize a recent literature highlighting these accomplishments, noting the engineering tools and analysis methods used in these discoveries while also highlighting unanswered questions.
Collapse
|
14
|
Pello Lázaro AM, Blanco-Colio LM, Franco Peláez JA, Tuñón J. Anti-Inflammatory Drugs in Patients with Ischemic Heart Disease. J Clin Med 2021; 10:2835. [PMID: 34198968 PMCID: PMC8268779 DOI: 10.3390/jcm10132835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/13/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation has long been known to play a role in atherogenesis and plaque complication, as well as in some drugs used in therapy for atherosclerotic disease, such as statins, acetylsalicylic acid, and modulators of the renin-angiotensin system, which also have anti-inflammatory effects. Furthermore, inflammatory biomarkers have been demonstrated to predict the incidence of cardiovascular events. In spite of this, and with the exception of acetylsalicylic acid, non-steroidal anti-inflammatory drugs are unable to decrease the incidence of cardiovascular events and may even be harmful to the cardiovascular system. In recent years, other anti-inflammatory drugs, such as canakinumab and colchicine, have shown an ability to reduce the incidence of cardiovascular events in secondary prevention. Colchicine could be a potential candidate for use in clinical practice given its safety and low price, although the results of temporary studies require confirmation in large randomized clinical trials. In this paper, we discuss the evidence linking inflammation with atherosclerosis and review the results from various clinical trials performed with anti-inflammatory drugs. We also discuss the potential use of these drugs in routine clinical settings.
Collapse
Affiliation(s)
- Ana María Pello Lázaro
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (J.A.F.P.)
- Department of Medicine, Autónoma University, 28029 Madrid, Spain
| | - Luis M. Blanco-Colio
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain;
- CIBERCV, 28029 Madrid, Spain
| | - Juan Antonio Franco Peláez
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (J.A.F.P.)
- Department of Medicine, Autónoma University, 28029 Madrid, Spain
| | - José Tuñón
- Department of Cardiology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (A.M.P.L.); (J.A.F.P.)
- Department of Medicine, Autónoma University, 28029 Madrid, Spain
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain;
- CIBERCV, 28029 Madrid, Spain
| |
Collapse
|
15
|
Ku KH, Dubinsky MK, Sukumar AN, Subramaniam N, Feasson MYM, Nair R, Tran E, Steer BM, Knight BJ, Marsden PA. In Vivo Function of Flow-Responsive Cis-DNA Elements of eNOS Gene: A Role for Chromatin-Based Mechanisms. Circulation 2021; 144:365-381. [PMID: 33910388 DOI: 10.1161/circulationaha.120.051078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND eNOS (endothelial nitric oxide synthase) is an endothelial cell (EC)-specific gene predominantly expressed in medium- to large-sized arteries where ECs experience atheroprotective laminar flow with high shear stress. Disturbed flow with lower average shear stress decreases eNOS transcription, which leads to the development of atherosclerosis, especially at bifurcations and curvatures of arteries. This prototypic arterial EC gene contains 2 distinct flow-responsive cis-DNA elements in the promoter, the shear stress response element (SSRE) and the KLF (Krüppel-like factor) element. Previous in vitro studies suggested their positive regulatory functions on flow-induced transcription of EC genes including eNOS. However, the in vivo function of these cis-DNA elements remains unknown. METHODS Insertional transgenic mice with a mutation at each flow-responsive cis-DNA element were generated using a murine eNOS promoter-β-galactosidase reporter by linker-scanning mutagenesis and compared with episomal-based mutations in vitro. DNA methylation at the eNOS proximal promoter in mouse ECs was assessed by bisulfite sequencing or pyrosequencing. RESULTS Wild type mice with a functional eNOS promoter-reporter transgene exhibited reduced endothelial reporter expression in the atheroprone regions of disturbed flow (n=5). It is surprising that the SSRE mutation abrogated reporter expression in ECs and was associated with aberrant hypermethylation at the eNOS proximal promoter (n=7). Reporter gene silencing was independent of transgene copy number and integration position, indicating that the SSRE is a critical cis-element necessary for eNOS transcription in vivo. The KLF mutation demonstrated an integration site-specific decrease in eNOS transcription, again with marked promoter methylation (n=8), suggesting that the SSRE alone is not sufficient for eNOS transcription in vivo. In wild type mice, the native eNOS promoter was significantly hypermethylated in ECs from the atheroprone regions where eNOS expression was markedly repressed by chronic disturbed flow, demonstrating that eNOS expression is regulated by flow-dependent DNA methylation that is region-specific in the arterial endothelium in vivo. CONCLUSIONS We report, for the first time, that the SSRE and KLF elements are critical flow sensors necessary for a transcriptionally permissive, hypomethylated eNOS promoter in ECs under chronic shear stress in vivo. Moreover, eNOS expression is regulated by flow-dependent epigenetic mechanisms, which offers novel mechanistic insight on eNOS gene regulation in atherogenesis.
Collapse
Affiliation(s)
- Kyung Ha Ku
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Michelle K Dubinsky
- Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Aravin N Sukumar
- Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Noeline Subramaniam
- Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Manon Y M Feasson
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Ranju Nair
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Eileen Tran
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.)
| | - Brent M Steer
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.)
| | - Britta J Knight
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.)
| | - Philip A Marsden
- Department of Laboratory Medicine and Pathobiology (K.H.K., M.Y.M.F., R.N., E.T., B.J.K., P.A.M.).,Institute of Medical Science (M.K.D., A.N.S., N.S., P.A.M.) University of Toronto, Ontario, Canada.,Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute (K.H.K., M.K.D., A.N.S., N.S., M.Y.M.F., R.N., B.M.B., P.A.M.).,Department of Medicine (P.A.M.), St Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Altered Vascular Extracellular Matrix in the Pathogenesis of Atherosclerosis. J Cardiovasc Transl Res 2021; 14:647-660. [PMID: 33420681 DOI: 10.1007/s12265-020-10091-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular disease continues to grow as a massive global health burden, with coronary artery disease being one of its most lethal varieties. The pathogenesis of atherosclerosis induces changes in the blood vessel and its extracellular matrix (ECM) in each vascular layer. The alteration of the ECM homeostasis has significant modulatory effects on the inflammatory response, the proliferation and migration of vascular smooth muscle cells, neointimal formation, and vascular fibrosis seen in atherosclerosis. In this literature review, the role of the ECM, the multitude of components, and alterations to these components in the pathogenesis of atherosclerosis are discussed with a focus on versatile cellular phenotypes in the structure of blood vessel. An understanding of the various effects of ECM alterations opens up a plethora of therapeutic options that would mitigate the substantial health toll of atherosclerosis on the global population.
Collapse
|
17
|
Mechtouff L, Nighoghossian N. Response by Mechtouff and Nighoghossian to Letter Regarding Article "Matrix Metalloproteinase-9 and Monocyte Chemoattractant Protein-1 Are Associated With Collateral Status in Acute Ischemic Stroke With Large Vessel Occlusion". Stroke 2020; 52:e16. [PMID: 33370182 DOI: 10.1161/strokeaha.120.032518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France (L.M., N.N.).,INSERM U1060, CarMeN laboratory, University Lyon 1, France (L.M., N.N.)
| | - Norbert Nighoghossian
- Stroke Department, Hospices Civils de Lyon, France (L.M., N.N.).,INSERM U1060, CarMeN laboratory, University Lyon 1, France (L.M., N.N.)
| |
Collapse
|
18
|
Recruitment and maturation of the coronary collateral circulation: Current understanding and perspectives in arteriogenesis. Microvasc Res 2020; 132:104058. [PMID: 32798552 DOI: 10.1016/j.mvr.2020.104058] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/09/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
Abstract
The coronary collateral circulation is a rich anastomotic network of primitive vessels which have the ability to augment in size and function through the process of arteriogenesis. In this review, we evaluate the current understandings of the molecular and cellular mechanisms by which this process occurs, specifically focussing on elevated fluid shear stress (FSS), inflammation, the redox state and gene expression along with the integrative, parallel and simultaneous process by which this occurs. The initiating step of arteriogenesis occurs following occlusion of an epicardial coronary artery, with an increase in FSS detected by mechanoreceptors within the endothelium. This must occur within a 'redox window' where an equilibrium of oxidative and reductive factors are present. These factors initially result in an inflammatory milieu, mediated by neutrophils as well as lymphocytes, with resultant activation of a number of downstream molecular pathways resulting in increased expression of proteins involved in monocyte attraction and adherence; namely vascular cell adhesion molecule 1 (VCAM-1), monocyte chemoattractant protein 1 (MCP-1) and transforming growth factor beta (TGF-β). Once monocytes and other inflammatory cells adhere to the endothelium they enter the extracellular matrix and differentiate into macrophages in an effort to create a favourable environment for vessel growth and development. Activated macrophages secrete inflammatory cytokines such as tumour necrosis factor-α (TNF-α), growth factors such as fibroblast growth factor-2 (FGF-2) and matrix metalloproteinases. Finally, vascular smooth muscle cells proliferate and switch to a contractile phenotype, resulting in an increased diameter and functionality of the collateral vessel, thereby allowing improved perfusion of the distal myocardium subtended by the occluded vessel. This simultaneously reduces FSS within the collateral vessel, inhibiting further vessel growth.
Collapse
|
19
|
Mechtouff L, Bochaton T, Paccalet A, Crola Da Silva C, Buisson M, Amaz C, Derex L, Ong E, Berthezene Y, Eker OF, Dufay N, Mewton N, Ovize M, Cho TH, Nighoghossian N. Matrix Metalloproteinase-9 and Monocyte Chemoattractant Protein-1 Are Associated With Collateral Status in Acute Ischemic Stroke With Large Vessel Occlusion. Stroke 2020; 51:2232-2235. [PMID: 32568655 DOI: 10.1161/strokeaha.120.029395] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE In ischemic stroke, inflammatory status may condition the development of collateral circulation. Here we assessed the relationship between systemic inflammatory biomarkers and collateral status in large vessel occlusion before mechanical thrombectomy. METHODS HIBISCUS-STROKE is a cohort study including acute ischemic stroke patients with large vessel occlusion treated with mechanical thrombectomy following admission magnetic resonance imaging. MMP-9 (matrix metalloproteinase-9) and MCP-1 (monocyte chemoattractant protein-1) were measured on blood sampling collected at admission. Collateral status was assessed on pretreatment Digital subtraction angiography and categorized into poor (Higashida score, 0-2) and good (Higashida score, 3-4). A multiple logistic regression model was performed to detect independent markers of good collateral status. RESULTS One hundred and twenty-two patients were included, of them 71 patients (58.2%) had a good collateral status. In univariate analysis, low MMP-9 levels (P=0.01), high MCP-1 levels (P<0.01), a low National Institute of Health Stroke Score (P=0.046), a high diastolic blood pressure (P=0.049), the absence of tandem occlusion (P=0.046), a high Alberta Stroke Program Early CT Score (P<0.01) and a low volume on the diffusion-weighted imaging (P<0.01) were associated with good collateral status. Following multivariate analysis, low MMP-9 levels (P=0.02) and high MCP-1 levels (P<0.01) remained associated with good collateral status. CONCLUSIONS Low MMP-9 and high MCP-1 levels were associated with good pretreatment collateral status in patients with acute ischemic stroke with large vessel occlusion. These results might suggest a relationship between collateral status and inflammation.
Collapse
Affiliation(s)
- Laura Mechtouff
- Stroke Department (L.M., L.D., E.O., T.-H.C., N.N.), Hospices Civils de Lyon, France.,INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Thomas Bochaton
- Cardiac Intensive Care Unit (T.B.), Hospices Civils de Lyon, France.,INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Alexandre Paccalet
- INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Claire Crola Da Silva
- INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Marielle Buisson
- Clinical Investigation Center, INSERM 1407 (M.B., C.A., N.M., M.O.), Hospices Civils de Lyon, France
| | - Camille Amaz
- Clinical Investigation Center, INSERM 1407 (M.B., C.A., N.M., M.O.), Hospices Civils de Lyon, France
| | - Laurent Derex
- Stroke Department (L.M., L.D., E.O., T.-H.C., N.N.), Hospices Civils de Lyon, France
| | - Elodie Ong
- Stroke Department (L.M., L.D., E.O., T.-H.C., N.N.), Hospices Civils de Lyon, France.,INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Yves Berthezene
- Neuroradiology Department (Y.B., O.F.E.), Hospices Civils de Lyon, France.,CREATIS, CNRS UMR 5220, INSERM U1044 (Y.B.), University Lyon 1, France
| | - Omer Faruk Eker
- Neuroradiology Department (Y.B., O.F.E.), Hospices Civils de Lyon, France
| | - Nathalie Dufay
- NeuroBioTec, CRB (N.D.), Hospices Civils de Lyon, France
| | - Nathan Mewton
- Clinical Investigation Center, INSERM 1407 (M.B., C.A., N.M., M.O.), Hospices Civils de Lyon, France
| | - Michel Ovize
- Clinical Investigation Center, INSERM 1407 (M.B., C.A., N.M., M.O.), Hospices Civils de Lyon, France.,INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Tae-Hee Cho
- Stroke Department (L.M., L.D., E.O., T.-H.C., N.N.), Hospices Civils de Lyon, France.,INSERM U1060, CarMeN laboratory (L.M., T.B., A.P., C.C.D.S., E.O., M.O., T.-H.C.), University Lyon 1, France
| | - Norbert Nighoghossian
- Stroke Department (L.M., L.D., E.O., T.-H.C., N.N.), Hospices Civils de Lyon, France
| |
Collapse
|
20
|
He M, Martin M, Marin T, Chen Z, Gongol B. Endothelial mechanobiology. APL Bioeng 2020; 4:010904. [PMID: 32095737 PMCID: PMC7032971 DOI: 10.1063/1.5129563] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/19/2020] [Indexed: 12/11/2022] Open
Abstract
Lining the luminal surface of the vasculature, endothelial cells (ECs) are in direct
contact with and differentially respond to hemodynamic forces depending on their anatomic
location. Pulsatile shear stress (PS) is defined by laminar flow and is predominantly
located in straight vascular regions, while disturbed or oscillatory shear stress (OS) is
localized to branch points and bifurcations. Such flow patterns have become a central
focus of vascular diseases, such as atherosclerosis, because the focal distribution of
endothelial dysfunction corresponds to regions exposed to OS, whereas endothelial
homeostasis is maintained in regions defined by PS. Deciphering the mechanotransduction
events that occur in ECs in response to differential flow patterns has required the
innovation of multidisciplinary approaches in both in vitro and
in vivo systems. The results from these studies have identified a
multitude of shear stress-regulated molecular networks in the endothelium that are
implicated in health and disease. This review outlines the significance of scientific
findings generated in collaboration with Dr. Shu Chien.
Collapse
Affiliation(s)
- Ming He
- Department of Medicine, University of California, San Diego, California 92093, USA
| | - Marcy Martin
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Traci Marin
- Department of Health Sciences, Victor Valley College, Victorville, California 92395, USA
| | - Zhen Chen
- Department of Diabetes Complications and Metabolism, Beckman Research Institute, City of Hope, California 91010, USA
| | - Brendan Gongol
- Department of Medicine, University of California, San Diego, California 92093, USA
| |
Collapse
|
21
|
Wang G, Kostidis S, Tiemeier GL, Sol WMPJ, de Vries MR, Giera M, Carmeliet P, van den Berg BM, Rabelink TJ. Shear Stress Regulation of Endothelial Glycocalyx Structure Is Determined by Glucobiosynthesis. Arterioscler Thromb Vasc Biol 2019; 40:350-364. [PMID: 31826652 DOI: 10.1161/atvbaha.119.313399] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Endothelial cells exposed to laminar shear stress express a thick glycocalyx on their surface that plays an important role in reducing vascular permeability and endothelial anti-inflammatory, antithrombotic, and antiangiogenic properties. Production and maintenance of this glycocalyx layer is dependent on cellular carbohydrate synthesis, but its regulation is still unknown. Approach and Results: Here, we show that biosynthesis of the major structural component of the endothelial glycocalyx, hyaluronan, is regulated by shear. Both in vitro as well as in in vivo, hyaluronan expression on the endothelial surface is increased on laminar shear and reduced when exposed to oscillatory flow, which is regulated by KLF2 (Krüppel-like Factor 2). Using a CRISPR-CAS9 edited small tetracysteine tag to endogenous HAS2 (hyaluronan synthase 2), we demonstrated increased translocation of HAS2 to the endothelial cell membrane during laminar shear. Hyaluronan production by HAS2 was shown to be further driven by availability of the hyaluronan substrates UDP-glucosamine and UDP-glucuronic acid. KLF2 inhibits endothelial glycolysis and allows for glucose intermediates to shuttle into the hexosamine- and glucuronic acid biosynthesis pathways, as measured using nuclear magnetic resonance analysis in combination with 13C-labeled glucose. CONCLUSIONS These data demonstrate how endothelial glycocalyx function and functional adaptation to shear is coupled to KLF2-mediated regulation of endothelial glycolysis.
Collapse
Affiliation(s)
- Gangqi Wang
- From the Division of Nephrology, Department of Internal Medicine (G.W., G.L.T., W.M.P.J.S., B.M.v.d.B., T.J.R.), The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, the Netherlands (S.K., M.G.)
| | - Gesa L Tiemeier
- From the Division of Nephrology, Department of Internal Medicine (G.W., G.L.T., W.M.P.J.S., B.M.v.d.B., T.J.R.), The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Wendy M P J Sol
- From the Division of Nephrology, Department of Internal Medicine (G.W., G.L.T., W.M.P.J.S., B.M.v.d.B., T.J.R.), The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Margreet R de Vries
- Department of Surgery (M.R.d.V.), The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, the Netherlands (S.K., M.G.)
| | - Peter Carmeliet
- Department of Oncology, Laboratory of Angiogenesis and Vascular Metabolism, KU Leuven, Vesalius Research Center, VIB, Belgium (P.C.).,Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, VIB, Leuven, Belgium (P.C.)
| | - Bernard M van den Berg
- From the Division of Nephrology, Department of Internal Medicine (G.W., G.L.T., W.M.P.J.S., B.M.v.d.B., T.J.R.), The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Ton J Rabelink
- From the Division of Nephrology, Department of Internal Medicine (G.W., G.L.T., W.M.P.J.S., B.M.v.d.B., T.J.R.), The Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| |
Collapse
|
22
|
Hollander MR, Jansen MF, Hopman LHGA, Dolk E, van de Ven PM, Knaapen P, Horrevoets AJ, Lutgens E, van Royen N. Stimulation of Collateral Vessel Growth by Inhibition of Galectin 2 in Mice Using a Single-Domain Llama-Derived Antibody. J Am Heart Assoc 2019; 8:e012806. [PMID: 31594443 PMCID: PMC6818022 DOI: 10.1161/jaha.119.012806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background In the presence of arterial stenosis, collateral artery growth (arteriogenesis) can alleviate ischemia and preserve tissue function. In patients with poorly developed collateral arteries, Gal‐2 (galectin 2) expression is increased. In vivo administration of Gal‐2 inhibits arteriogenesis. Blocking of Gal‐2 potentially stimulates arteriogenesis. This study aims to investigate the effect of Gal‐2 inhibition on arteriogenesis and macrophage polarization using specific single‐domain antibodies. Methods and Results Llamas were immunized with Gal‐2 to develop anti–Gal‐2 antibodies. Binding of Gal‐2 to monocytes and binding inhibition of antibodies were quantified. To test arteriogenesis in vivo, Western diet‐fed LDLR.(low‐density lipoprotein receptor)–null Leiden mice underwent femoral artery ligation and received treatment with llama antibodies 2H8 or 2C10 or with vehicle. Perfusion restoration was measured with laser Doppler imaging. In the hind limb, arterioles and macrophage subtypes were characterized by histology, together with aortic atherosclerosis. Llama‐derived antibodies 2H8 and 2C10 strongly inhibited the binding of Gal‐2 to monocytes (93% and 99%, respectively). Treatment with these antibodies significantly increased perfusion restoration at 14 days (relative to sham, vehicle: 41.3±2.7%; 2H8: 53.1±3.4%, P=0.016; 2C10: 52.0±3.8%, P=0.049). In mice treated with 2H8 or 2C10, the mean arteriolar diameter was larger compared with control (vehicle: 17.25±4.97 μm; 2H8: 17.71±5.01 μm; 2C10: 17.84±4.98 μm; P<0.001). Perivascular macrophages showed a higher fraction of the M2 phenotype in both antibody‐treated animals (vehicle: 0.49±0.24; 2H8: 0.73±0.15, P=0.007; 2C10: 0.75±0.18, P=0.006). In vitro antibody treatment decreased the expression of M1‐associated cytokines compared with control (P<0.05 for each). Atherosclerotic lesion size was comparable between groups (overall P=0.59). Conclusions Inhibition of Gal‐2 induces a proarteriogenic M2 phenotype in macrophages, improves collateral artery growth, and increases perfusion restoration in a murine hind limb model.
Collapse
Affiliation(s)
- Maurits R Hollander
- Department of Cardiology VU University Medical Centre Amsterdam The Netherlands
| | - Matthijs F Jansen
- Department of Cardiology VU University Medical Centre Amsterdam The Netherlands.,Department of Medical Biochemistry Academic Medical Centre Amsterdam The Netherlands
| | - Luuk H G A Hopman
- Department of Cardiology VU University Medical Centre Amsterdam The Netherlands
| | | | - Peter M van de Ven
- Department of Epidemiology and Biostatistics VU University Amsterdam The Netherlands
| | - Paul Knaapen
- Department of Cardiology VU University Medical Centre Amsterdam The Netherlands
| | - Anton J Horrevoets
- Department of Molecular Cell Biology and Immunology VU Medical Center Amsterdam The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry Academic Medical Centre Amsterdam The Netherlands.,Institute for Cardiovascular Prevention (IPEK) Ludwig Maximilian's University Munich Germany
| | - Niels van Royen
- Department of Cardiology VU University Medical Centre Amsterdam The Netherlands.,Department of Cardiology Radboud University Medical Center Nijmegen The Netherlands
| |
Collapse
|
23
|
Rajendran S, Shen X, Glawe J, Kolluru GK, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Growth and Remodeling. Compr Physiol 2019; 9:1213-1247. [PMID: 31187898 DOI: 10.1002/cphy.c180026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro- and micro-circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213-1247, 2019.
Collapse
Affiliation(s)
| | - Xinggui Shen
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - John Glawe
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Gopi K Kolluru
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Christopher G Kevil
- Departments of Pathology, LSU Health Sciences Center, Shreveport.,Departments of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport.,Departments of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport
| |
Collapse
|
24
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
25
|
Harrison DL, Fang Y, Huang J. T-Cell Mechanobiology: Force Sensation, Potentiation, and Translation. FRONTIERS IN PHYSICS 2019; 7:45. [PMID: 32601597 PMCID: PMC7323161 DOI: 10.3389/fphy.2019.00045] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
A T cell is a sensitive self-referential mechanical sensor. Mechanical forces influence the recognition, activation, differentiation, and function throughout the lifetime of a T cell. T cells constantly perceive and respond to physical stimuli through their surface receptors, cytoskeleton, and subcellular structures. Surface receptors receive physical cues in the form of forces generated through receptor-ligand binding events, which are dynamically regulated by contact tension, shear stress, and substrate rigidity. The resulting mechanotransduction not only influences T-cell recognition and signaling but also possibly modulates cell metabolism and gene expression. Moreover, forces also dynamically regulate the deformation, organization, and translocation of cytoskeleton and subcellular structures, leading to changes in T-cell mobility, migration, and infiltration. However, the roles and mechanisms of how mechanical forces modulate T-cell recognition, signaling, metabolism, and gene expression, are largely unknown and underappreciated. Here, we review recent technological and scientific advances in T-cell mechanobiology, discuss possible roles and mechanisms of T-cell mechanotransduction, and propose new research directions of this emerging field in health and disease.
Collapse
Affiliation(s)
- Devin L. Harrison
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
| | - Yun Fang
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
- Section of Pulmonary and Critical Care, Department of Medicine, The University of Chicago, Chicago, IL, United States
| | - Jun Huang
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, United States
- Institute for Molecular Engineering, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Faienza MF, D'Amato G, Chiarito M, Colaianni G, Colucci S, Grano M, Corbo F, Brunetti G. Mechanisms Involved in Childhood Obesity-Related Bone Fragility. Front Endocrinol (Lausanne) 2019; 10:269. [PMID: 31130918 PMCID: PMC6509993 DOI: 10.3389/fendo.2019.00269] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/11/2019] [Indexed: 01/11/2023] Open
Abstract
Childhood obesity is one of the major health problems in western countries. The excessive accumulation of adipose tissue causes inflammation, oxidative stress, apoptosis, and mitochondrial dysfunctions. Thus, obesity leads to the development of severe co-morbidities including type 2 diabetes mellitus, liver steatosis, cardiovascular, and neurodegenerative diseases which can develop early in life. Furthermore, obese children have low bone mineral density and a greater risk of osteoporosis and fractures. The knowledge about the interplay bone tissue and between adipose is still growing, although recent findings suggest that adipose tissue activity on bone can be fat-depot specific. Obesity is associated to a low-grade inflammation that alters the expression of adiponectin, leptin, IL-6, Monocyte Chemotactic Protein 1 (MCP1), TRAIL, LIGHT/TNFSF14, OPG, and TNFα. These molecules can affect bone metabolism, thus resulting in osteoporosis. The purpose of this review was to deepen the cellular mechanisms by which obesity may facilitate osteoporosis and bone fractures.
Collapse
Affiliation(s)
- Maria Felicia Faienza
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | | | - Mariangela Chiarito
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Graziana Colaianni
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Silvia Colucci
- Department of Basic and Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari Aldo Moro, Bari, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Filomena Corbo
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, Bari, Italy
| | - Giacomina Brunetti
- Department of Basic and Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari Aldo Moro, Bari, Italy
- *Correspondence: Giacomina Brunetti
| |
Collapse
|
27
|
Yu B, Chen Q, Le Bras A, Zhang L, Xu Q. Vascular Stem/Progenitor Cell Migration and Differentiation in Atherosclerosis. Antioxid Redox Signal 2018; 29:219-235. [PMID: 28537424 DOI: 10.1089/ars.2017.7171] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Atherosclerosis is a major cause for the death of human beings, and it takes place in large- and middle-sized arteries. The pathogenesis of the disease has been widely investigated, and new findings on vascular stem/progenitor cells could have an impact on vascular regeneration. Recent Advances: Recent studies have shown that abundant stem/progenitor cells present in the vessel wall are mainly responsible for cell accumulation in the intima during vascular remodeling. It has been demonstrated that the mobilization and recruitment of tissue-resident stem/progenitor cells give rise to endothelial and smooth muscle cells (SMCs) that participate in vascular repair and remodeling such as neointimal hyperplasia and arteriosclerosis. Interestingly, cell lineage tracing studies indicate that a large proportion of SMCs in neointimal lesions is derived from adventitial stem/progenitor cells. CRITICAL ISSUES The influence of stem/progenitor cell behavior on the development of atherosclerosis is crucial. An understanding of the regulatory mechanisms that control stem/progenitor cell migration and differentiation is essential for stem/progenitor cell therapy for vascular diseases and regenerative medicine. FUTURE DIRECTIONS Identification of the detailed process driving the migration and differentiation of vascular stem/progenitor cells during the development of atherosclerosis, discovery of the environmental cues, and signaling pathways that control cell fate within the vasculature will facilitate the development of new preventive and therapeutic strategies to combat atherosclerosis. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Baoqi Yu
- 1 Department of Emergency, Guangdong General Hospital , Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qishan Chen
- 2 Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China
| | - Alexandra Le Bras
- 3 Cardiovascular Division, King's College London BHF Centre , London, United Kingdom
| | - Li Zhang
- 2 Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, China
| | - Qingbo Xu
- 3 Cardiovascular Division, King's College London BHF Centre , London, United Kingdom
| |
Collapse
|
28
|
Nakatsumi H, Matsumoto M, Nakayama KI. Noncanonical Pathway for Regulation of CCL2 Expression by an mTORC1-FOXK1 Axis Promotes Recruitment of Tumor-Associated Macrophages. Cell Rep 2018; 21:2471-2486. [PMID: 29186685 DOI: 10.1016/j.celrep.2017.11.014] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/26/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022] Open
Abstract
C-C chemokine ligand 2 (CCL2) plays pivotal roles in tumor formation, progression, and metastasis. Although CCL2 expression has been found to be dependent on the nuclear factor (NF)-κB signaling pathway, the regulation of CCL2 production in tumor cells has remained unclear. We have identified a noncanonical pathway for regulation of CCL2 production that is mediated by mammalian target of rapamycin complex 1 (mTORC1) but independent of NF-κB. Multiple phosphoproteomics approaches identified the transcription factor forkhead box K1 (FOXK1) as a downstream target of mTORC1. Activation of mTORC1 induces dephosphorylation of FOXK1, resulting in transactivation of the CCL2 gene. Inhibition of the mTORC1-FOXK1 axis attenuated insulin-induced CCL2 production as well as the accumulation of tumor-associated monocytes-macrophages and tumor progression in mice. Our results suggest that FOXK1 directly links mTORC1 signaling and CCL2 expression in a manner independent of NF-κB and that CCL2 produced by this pathway contributes to tumor progression.
Collapse
Affiliation(s)
- Hirokazu Nakatsumi
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Masaki Matsumoto
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan.
| |
Collapse
|
29
|
|
30
|
Bates DO, Beazley-Long N, Benest AV, Ye X, Ved N, Hulse RP, Barratt S, Machado MJ, Donaldson LF, Harper SJ, Peiris-Pages M, Tortonese DJ, Oltean S, Foster RR. Physiological Role of Vascular Endothelial Growth Factors as Homeostatic Regulators. Compr Physiol 2018; 8:955-979. [PMID: 29978898 DOI: 10.1002/cphy.c170015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vascular endothelial growth factor (VEGF) family of proteins are key regulators of physiological systems. Originally linked with endothelial function, they have since become understood to be principal regulators of multiple tissues, both through their actions on vascular cells, but also through direct actions on other tissue types, including epithelial cells, neurons, and the immune system. The complexity of the five members of the gene family in terms of their different splice isoforms, differential translation, and specific localizations have enabled tissues to use these potent signaling molecules to control how they function to maintain their environment. This homeostatic function of VEGFs has been less intensely studied than their involvement in disease processes, development, and reproduction, but they still play a substantial and significant role in healthy control of blood volume and pressure, interstitial volume and drainage, renal and lung function, immunity, and signal processing in the peripheral and central nervous system. The widespread expression of VEGFs in healthy adult tissues, and the disturbances seen when VEGF signaling is inhibited support this view of the proteins as endogenous regulators of normal physiological function. This review summarizes the evidence and recent breakthroughs in understanding of the physiology that is regulated by VEGF, with emphasis on the role they play in maintaining homeostasis. © 2017 American Physiological Society. Compr Physiol 8:955-979, 2018.
Collapse
Affiliation(s)
- David O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | | | - Andrew V Benest
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Xi Ye
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Nikita Ved
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Richard P Hulse
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Shaney Barratt
- Academic Respiratory Unit, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Maria J Machado
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom
| | - Lucy F Donaldson
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Steven J Harper
- School of Physiology, Pharmacology & Neuroscience, Medical School, University of Bristol, Bristol, United Kingdom
| | - Maria Peiris-Pages
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Domingo J Tortonese
- Centre for Comparative and Clinical Anatomy, University of Bristol, Bristol, United Kingdom
| | - Sebastian Oltean
- Institute of Biomedical & Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Rebecca R Foster
- Bristol Renal, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
31
|
Induction of extracranial arteriogenesis by an arteriovenous fistula in a pig model. Atherosclerosis 2018; 272:87-93. [PMID: 29579672 DOI: 10.1016/j.atherosclerosis.2018.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/20/2018] [Accepted: 03/02/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS Arteriogenesis, the positive outward remodeling and growth of pre-existent collateral vessels, holds potential as a novel treatment for ischemic vascular disease. An extracranial arteriogenesis model in a pig will allow us to study molecular changes in a complex arteriolar network in a more clinically relevant large-animal model. To increase fluid shear stress in the brain, an experimental carotid arteriovenous fistula (AVF model) in minipigs was established, providing high flow through the extracranial rete mirabile. The aim of the study was to examine whether creation of a carotid AVF can induce extracranial arteriogenesis in the pig. METHODS Angiography was performed to demonstrate blood flow diversion. Animals were sacrificed after 0, 3 and 14 days post-surgery and both retia mirabilia were removed. Immunohistochemical analysis was performed to analyze cell proliferation and accumulation of mononuclear cells in the vessel wall. RESULTS After 3 days of high-flow conditions, increases in vascular cell proliferation (approximately 1.5-fold; p = 0.143) and monocyte invasion (approximately 6-fold; p = 0.057) were observed when compared to animals sacrificed immediately after AVF formation. Quantitative PCR (RT-qPCR) analysis from rete mirabile tissue samples 3 days post-surgery revealed that monocyte chemoattractant protein (MCP)-1 and tissue inhibitor of metalloproteinases (TIMP)-1 were highly upregulated. Expression of the pro-arteriogenic marker, CD44, reached maximum expression level 14 days post-surgery. CONCLUSIONS In response to high levels of shear stress produced in the pig AVF model, the onset of the arteriogenic process can be induced. This was demonstrated by enhanced cell proliferation, monocyte invasion and vascular remodeling.
Collapse
|
32
|
Protective effects of continuous positive airway pressure on a nonventilated lung during one-lung ventilation: A prospective laboratory study in rats. Eur J Anaesthesiol 2018; 33:776-83. [PMID: 27139568 DOI: 10.1097/eja.0000000000000460] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The use of one-lung ventilation (OLV) to facilitate intrathoracic surgery is a cause of lung injury. OBJECTIVE We hypothesised that application of continuous positive airway pressure (CPAP) to a nonventilated lung during OLV would prevent alveolar hypoxia and blood flow shift from the nonventilated to the ventilated lung, thereby attenuating lung injury. DESIGN Controlled animal study. SETTINGS University laboratory. STUDY PARTICIPANTS Adult male Sprague-Dawley rats (n = 4 to 8 per group, depending on experiments). INTERVENTIONS Rats were alternately assigned to one of two ventilation protocol groups: control and CPAP groups. Rats received 240 min of OLV followed by 240 min of two-lung reventilation (re-TLV). The nonventilated lungs of rats in the control group were collapsed during OLV whereas rats in the CPAP group received CPAP (5 cmH2O with 100% oxygen) to the nonventilated lungs. MAIN OUTCOME MEASURES Pulmonary blood flow during OLV was measured by quantification of lung radioactivity after intravenous infusion of indium-labelled macroaggregated albumin. Inflammatory cytokines in the lungs after 240 min of OLV, and after the subsequent 240 min of re-TLV were measured. Additionally, we measured lung wet-to-dry weight ratios after re-TLV. We also measured lung malondialdehyde levels after re-TLV as an indicator of reactive oxygen species produced by reoxygenation. RESULTS Application of CPAP attenuated the pulmonary blood flow shift from the nonventilated to the ventilated lung. CPAP decreased the levels of IL-6, CXC chemokine ligand-1 and CC chemokine ligand-2 in both lungs after 240 min of OLV. CPAP also decreased CXC chemokine ligand-1 in the nonventilated lung and CC chemokine ligand-2 in both lungs after re-TLV. Moreover, wet-to-dry weight ratios of both lungs were decreased by application of CPAP. However, lung malondialdehyde concentrations were not affected by CPAP. CONCLUSIONS CPAP applied to the nonventilated lung during OLV suppresses blood flow shift and decreases inflammatory cytokines and water content in both lungs. Application of CPAP may attenuate lung injury during and after OLV.
Collapse
|
33
|
Ihalainen JK, Peltonen H, Paulsen G, Ahtiainen JP, Taipale RS, Hämäläinen M, Moilanen E, Mero AA. Inflammation status of healthy young men: initial and specific responses to resistance training. Appl Physiol Nutr Metab 2017; 43:252-258. [PMID: 29053943 DOI: 10.1139/apnm-2017-0315] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Our primary aim was to study the effects of a 4-week preparatory resistance-training (RT) period followed by 12 weeks of 2 specific RT protocols (either hypertrophic-strength (HS) or strength-hypertrophy-power training) on inflammation markers and the possible relationship of the changes in abdominal fat and lean mass to the changes in inflammation status. A total of 82 healthy men were included in the study. Maximal concentric leg press strength (1-repetition maximum), total body lean mass, total body and abdominal fat mass, circulating high-sensitivity C-reactive protein, interleukin-6, interleukin-1 receptor antagonist (IL-1ra), monocyte chemoattractant protein 1 (MCP-1), and selected adipocytokines (resistin, adiponectin, and leptin) concentrations were measured before and after 4 (wk4) and 16 weeks (wk16) of RT. After the initial phase of RT, on wk4, abdominal and total fat mass as well as plasma leptin concentrations were significantly reduced (p < 0.05), whereas muscle mass, IL-1ra, resistin, and MCP-1 concentrations were significantly increased (p < 0.05). During specialized training phase, at wk16, only HS led to further reduction in abdominal and total fat mass, resistin, and leptin (p < 0.05), whereas both training modes led to lower MCP-1 concentrations (p < 0.05). Abdominal fat mass and circulating leptin were reduced already after 4 weeks of RT. Simultaneously, circulating MCP-1 and resistin concentrations increased, possibly as markers of muscle adaptation and regeneration. The present findings also suggest that RT with hypertrophic focus is beneficial for further reductions in abdominal fat mass and to decrease circulating inflammatory markers.
Collapse
Affiliation(s)
- Johanna K Ihalainen
- a Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä FI-40014, Finland
| | - Heikki Peltonen
- a Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä FI-40014, Finland
| | - Göran Paulsen
- b The Norwegian Olympic and Paralympic Committee and Confederation of Sports, Oslo N-0130, Norway.,c Norwegian School of Sport Sciences, Oslo, Norway
| | - Juha P Ahtiainen
- a Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä FI-40014, Finland
| | - Ritva S Taipale
- a Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä FI-40014, Finland.,d Kajaani University of Applied Sciences, Kajaani FI-87101, Finland
| | - Mari Hämäläinen
- e The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere FI-33014, Finland
| | - Eeva Moilanen
- e The Immunopharmacology Research Group, Faculty of Medicine and Life Sciences, University of Tampere and Tampere University Hospital, Tampere FI-33014, Finland
| | - Antti A Mero
- a Biology of Physical Activity, Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä FI-40014, Finland
| |
Collapse
|
34
|
Guan Y, Cai B, Wu X, Peng S, Gan L, Huang D, Liu G, Dong L, Xiao L, Liu J, Zhang B, Cai WJ, Schaper J, Schaper W. microRNA-352 regulates collateral vessel growth induced by elevated fluid shear stress in the rat hind limb. Sci Rep 2017; 7:6643. [PMID: 28751690 PMCID: PMC5532297 DOI: 10.1038/s41598-017-06910-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/20/2017] [Indexed: 12/17/2022] Open
Abstract
Although collateral vessel growth is distinctly enhanced by elevated fluid shear stress (FSS), the underlying regulatory mechanism of this process remains incompletely understood. Recent studies have shown that microRNAs (miRNAs) play a pivotal role in vascular development, homeostasis and a variety of diseases. Therefore, this study was designed to identify miRNAs involved in elevated FSS-induced collateral vessel growth in rat hind limbs. A side-to-side arteriovenous (AV) shunt was created between the distal stump of one of the bilaterally occluded femoral arteries and the accompanying vein. The miRNA array profile showed 94 differentially expressed miRNAs in FSS-stressed collaterals including miRNA-352 which was down-regulated. Infusion of antagomir-352 increased the number and proliferation of collateral vessels and promoted collateral flow restoration in a model of rat hind limb ligation. In cell culture studies, the miR-352 inhibitor increased endothelial proliferation, migration and tube formation. In addition, antagomir-352 up-regulated the expression of insulin-like growth factor II receptor (IGF2R), which may play a part in the complex pathway leading to arterial growth. We conclude that enhanced collateral vessel growth is controlled by miRNAs, among which miR-352 is a novel candidate that negatively regulates arteriogenesis, meriting additional studies to unravel the pathways leading to improved collateral circulation.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.,Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, 77204, Houston, TX, United States
| | - Baizhen Cai
- Department of Intensive Care Unit, the 3rd Xiangya Hospital, Central South University Changsha, 410013, Hunan, P.R. China
| | - Xiaoqiong Wu
- Department of Anatomy & Neurobiology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Song Peng
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.,Department of Radiology, the 3rd Xiangya Hospital, Central South University Changsha, 410078, Hunan, P.R. China
| | - Liaoying Gan
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Da Huang
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Guangmin Liu
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Liping Dong
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Lin Xiao
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Junwen Liu
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Bin Zhang
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.
| | - Wei-Jun Cai
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.
| | - Jutta Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany.
| | - Wolfgang Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany
| |
Collapse
|
35
|
Experimental Study on the Efficacy of Site-Specific PEGylated Human Serum Albumins in Resuscitation From Hemorrhagic Shock. Crit Care Med 2017; 44:e1090-e1096. [PMID: 27760056 DOI: 10.1097/ccm.0000000000001825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To evaluate the resuscitative efficacy and the effect on reperfusion injury of two site-specific PEGylated human serum albumins modified with linear or branched PEG20kDa, compared with saline, 8% human serum albumin and 25% human serum albumin, in a hemorrhagic shock model. SETTING Laboratory. SUBJECTS Male Wistar rats. DESIGN Prospective study. INTERVENTIONS Rats were bled to hemorrhagic hypovolemic shock and resuscitated with different resuscitation fluids. MEASUREMENTS AND MAIN RESULTS The mean arterial pressure and blood gas variables were measured. Hemorheology analysis was performed to evaluate the influence of resuscitation on RBCs and blood viscosity. The microvascular state was indirectly characterized in terms of monocyte chemotactic protein-1 and endothelial nitric oxide synthase that related to shear stress and vasodilation, respectively. The levels of inflammation-related factors and apoptosis-related proteins were used to evaluate the reperfusion injury in lungs. The results showed that PEGylated human serum albumin could improve the level of mean arterial pressure and blood gas variables more effectively at the end of resuscitation. poly(ethylene glycol) modification was able to increase the viscosity of human serum albumin to the level of effectively enhancing the expression of monocyte chemotactic protein-1 and endothelial nitric oxide synthase, which could promote microvascular perfusion. The hyperosmotic resuscitative agents including both 25% human serum albumin and PEGylated human serum albumins could greatly attenuate lung injury. No significant therapeutic advantages but some disadvantages were found for Y shaped poly(ethylene glycol) modification over linear poly(ethylene glycol) modification, such as causing the decrease of erythrocyte deformability. CONCLUSIONS Linear high molecular weight site-specific PEGylated human serum albumin is recommended to be used as a hyperosmotic resuscitative agent.
Collapse
|
36
|
The role of chemokines in hypertension and consequent target organ damage. Pharmacol Res 2017; 119:404-411. [PMID: 28279813 DOI: 10.1016/j.phrs.2017.02.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/25/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Immune cells infiltrate the kidney, vasculature, and central nervous system during hypertension, consequently amplifying tissue damage and/or blood pressure elevation. Mononuclear cell motility depends partly on chemokines, which are small cytokines that guide cells through an increasing concentration gradient via ligation of their receptors. Tissue expression of several chemokines is elevated in clinical and experimental hypertension. Likewise, immune cells have enhanced chemokine receptor expression during hypertension, driving immune cell infiltration and inappropriate inflammation in cardiovascular control centers. T lymphocytes and monocytes/macrophages are pivotal mediators of hypertensive inflammation, and these cells migrate in response to several chemokines. As powerful drivers of diapedesis, the chemokines CCL2 and CCL5 have long been implicated in hypertension, but experimental data highlight divergent, context-specific effects of these chemokines on blood pressure and tissue injury. Several other chemokines, particularly those of the CXC family, contribute to blood pressure elevation and target organ damage. Given the significant interplay and chemotactic redundancy among chemokines during disease, future work must not only describe the actions of individual chemokines in hypertension, but also characterize how manipulating a single chemokine modulates the expression and/or function of other chemokines and their cognate receptors. This information will facilitate the design of precise chemotactic immunotherapies to limit cardiovascular and renal morbidity in hypertensive patients.
Collapse
|
37
|
Cheang WS, Wong WT, Zhao L, Xu J, Wang L, Lau CW, Chen ZY, Ma RCW, Xu A, Wang N, Tian XY, Huang Y. PPARδ Is Required for Exercise to Attenuate Endoplasmic Reticulum Stress and Endothelial Dysfunction in Diabetic Mice. Diabetes 2017; 66:519-528. [PMID: 27856609 DOI: 10.2337/db15-1657] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 11/11/2016] [Indexed: 11/13/2022]
Abstract
Physical activity has profound benefits on health, especially on cardiometabolic wellness. Experiments in rodents with trained exercise have shown that exercise improves vascular function and reduces vascular inflammation by modulating the balance between nitric oxide (NO) and oxidative stress. However, the upstream regulator of exercise-induced vascular benefits is unclear. We aimed to investigate the involvement of peroxisome proliferator-activated receptor δ (PPARδ) in exercise-induced vascular functional improvement. We show that PPARδ is a crucial mediator for exercise to exert a beneficial effect on the vascular endothelium in diabetic mice. In db/db mice and high-fat diet-induced obese mice, 4 weeks of treadmill exercise restored endothelium-dependent vasodilation of aortas and flow-mediated vasodilation in mesenteric resistance arteries, whereas genetic ablation of Ppard abolished such improvements. Exercise induces AMPK activation and subsequent PPARδ activation, which help to reduce endoplasmic reticulum (ER) and oxidative stress, thus increasing NO bioavailability in endothelial cells and vascular tissues. Chemical chaperones 4-phenylbutyric acid and tauroursodeoxycholic acid decrease ER stress and protect against endothelial dysfunction in diabetic mice. The results demonstrate that PPARδ-mediated inhibition of ER stress contributes to the vascular benefits of exercise and provides potentially effective targets for treating diabetic vasculopathy.
Collapse
Affiliation(s)
- Wai San Cheang
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau
| | - Wing Tak Wong
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong
| | - Lei Zhao
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Jian Xu
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Li Wang
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Chi Wai Lau
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Zhen Yu Chen
- School of Life Sciences, Chinese University of Hong Kong, Hong Kong
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong
| | - Aimin Xu
- Departments of Medicine and Pharmacology and Pharmacy, State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong
| | - Nanping Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiao Yu Tian
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| | - Yu Huang
- Institute of Vascular Medicine, Shenzhen Research Institute, Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
38
|
Kamphuis VP, Westenberg JJM, van der Palen RLF, Blom NA, de Roos A, van der Geest R, Elbaz MSM, Roest AAW. Unravelling cardiovascular disease using four dimensional flow cardiovascular magnetic resonance. Int J Cardiovasc Imaging 2016; 33:1069-1081. [PMID: 27888419 PMCID: PMC5489572 DOI: 10.1007/s10554-016-1031-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/21/2016] [Indexed: 11/29/2022]
Abstract
Knowledge of normal and abnormal flow patterns in the human cardiovascular system increases our understanding of normal physiology and may help unravel the complex pathophysiological mechanisms leading to cardiovascular disease. Four-dimensional (4D) flow cardiovascular magnetic resonance (CMR) has emerged as a suitable technique that enables visualization of in vivo blood flow patterns and quantification of parameters that could potentially be of prognostic value in the disease process. In this review, current image processing tools that are used for comprehensive visualization and quantification of blood flow and energy distribution in the heart and great vessels will be discussed. Also, imaging biomarkers extracted from 4D flow CMR will be reviewed that have been shown to distinguish between normal and abnormal flow patterns. Furthermore, current applications of 4D flow CMR in the heart and great vessels will be discussed, showing its potential as an additional diagnostic modality which could aid in disease management and timing of surgical intervention.
Collapse
Affiliation(s)
- Vivian P Kamphuis
- Division of Pediatric Cardiology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Jos J M Westenberg
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Roel L F van der Palen
- Division of Pediatric Cardiology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Nico A Blom
- Division of Pediatric Cardiology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Albert de Roos
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob van der Geest
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mohammed S M Elbaz
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arno A W Roest
- Division of Pediatric Cardiology, Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
39
|
Wong WT, Ma S, Tian XY, Gonzalez AB, Ebong EE, Shen H. Targeted Delivery of Shear Stress-Inducible Micrornas by Nanoparticles to Prevent Vulnerable Atherosclerotic Lesions. Methodist Debakey Cardiovasc J 2016; 12:152-156. [PMID: 27826369 DOI: 10.14797/mdcj-12-3-152] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular wall disease, and endothelial cell dysfunction plays an important role in its development and progression. Under the influence of laminar shear stress, however, the endothelium releases homeostatic factors such as nitric oxide and expresses of vasoprotective microRNAs that are resistant to atherosclerosis. Adhesion molecules such as E-selectin, exhibited on the endothelial surface, recruit monocytes that enter the vessel wall to form foam cells. Accumulation of these foam cells form fatty streaks that may progress to atherosclerotic plaques in the blood vessel wall. Interestingly, E-selectin may also serve as an affinity moiety for targeted drug delivery against atherosclerosis. We have recently developed an E-selectin-targeted platform that enriches therapeutic microRNAs in the inflamed endothelium to inhibit formation of vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Wing Tak Wong
- Houston Methodist Research Institute, Houston, Texas
| | - Shuangtao Ma
- Houston Methodist Research Institute, Houston, Texas
| | - Xiao Yu Tian
- Houston Methodist Research Institute, Houston, Texas
| | | | - Eno E Ebong
- Northeastern University, Boston, Massachusetts
| | - Haifa Shen
- Houston Methodist Research Institute, Houston, Texas
| |
Collapse
|
40
|
Abstract
Dysfunction of the endothelial lining of lesion-prone areas of the arterial vasculature is an important contributor to the pathobiology of atherosclerotic cardiovascular disease. Endothelial cell dysfunction, in its broadest sense, encompasses a constellation of various nonadaptive alterations in functional phenotype, which have important implications for the regulation of hemostasis and thrombosis, local vascular tone and redox balance, and the orchestration of acute and chronic inflammatory reactions within the arterial wall. In this review, we trace the evolution of the concept of endothelial cell dysfunction, focusing on recent insights into the cellular and molecular mechanisms that underlie its pivotal roles in atherosclerotic lesion initiation and progression; explore its relationship to classic, as well as more recently defined, clinical risk factors for atherosclerotic cardiovascular disease; consider current approaches to the clinical assessment of endothelial cell dysfunction; and outline some promising new directions for its early detection and treatment.
Collapse
Affiliation(s)
- Michael A Gimbrone
- From the Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| | - Guillermo García-Cardeña
- From the Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
41
|
Abstract
Objective: To review the physiological mechanisms determining venous return to the heart and the pathophysiological events culminating in chronic venous insufficiency (CVI), focusing primarily on the role of alterations in nitric oxide (NO) production by the vascular endothelium. Background: Congenital valve incompetence, thrombotic damage or venous outflow obstruction result in the development of chronic venous hypertension which frequently leads to ulceration. One major aetiological factor of trophic changes in the skin of patients with CVI is the phenomenon of leucocyte trapping. Hypothesis: It has been suggested that endothelial dysfunction, effectively resulting in a decrease in cellular levels of NO, is a key event in the initiation of enhanced adhesion molecule expression. Data: P-selectin, monocyte chemoattractant protein-1 and vascular cell adhesion molecule-1 expression can be enhanced by attenuating endothelial NO production. The mechanism by which NO alters the expression of genes encoding these adhesion molecules would appear to involve an interaction with transcription factors, in particular NF*** xB. Conclusion: Impaired endothelial NO synthesis associated with CVI may enhance the expression of adhesion molecules and chemotactic factors and lead to leucocyte adhesion and extravasation.
Collapse
Affiliation(s)
- J. Bauersachs
- Zentrum der Physiologie, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | - I. Fleming
- Zentrum der Physiologie, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| | - R. Busse
- Zentrum der Physiologie, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
42
|
MAP-Kinase Activated Protein Kinase 2 Links Endothelial Activation and Monocyte/macrophage Recruitment in Arteriogenesis. PLoS One 2015; 10:e0138542. [PMID: 26431421 PMCID: PMC4592267 DOI: 10.1371/journal.pone.0138542] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/01/2015] [Indexed: 12/17/2022] Open
Abstract
Arteriogenesis, the growth of natural bypass arteries, is triggered by hemodynamic forces within vessels and requires a balanced inflammatory response, involving induction of the chemokine MCP-1 and recruitment of leukocytes. However, little is known how these processes are coordinated. The MAP-kinase-activated-proteinkinase-2 (MK2) is a critical regulator of inflammatory processes and might represent an important link between cytokine production and cell recruitment during postnatal arteriogenesis. Therefore, the present study investigated the functional role of MK2 during postnatal arteriogenesis. In a mouse model of hindlimb ischemia (HLI) MK2-deficiency (MK2KO) significantly impaired ischemic blood flow recovery and growth of collateral arteries as well as perivascular recruitment of mononuclear cells and macrophages. This was accompanied by induction of endothelial MCP-1 expression in wildtype (WT) but not in MK2KO collateral arteries. Following HLI, MK2 activation rapidly occured in the endothelium of growing WT arteries in vivo. In vitro, inflammatory cytokines and cyclic stretch activated MK2 in endothelial cells, which was required for stretch- and cytokine-induced release of MCP-1. In addition, a monocyte cell autonomous function of MK2 was uncovered potentially regulating MCP-1-dependent monocyte recruitment to vessels: MCP-1 stimulation of WT monocytes induced MK2 activation and monocyte migration in vitro. The latter was reduced in MK2KO monocytes, while in vivo MK2 was activated in monocytes recruited to collateral arteries. In conclusion, MK2 regulates postnatal arteriogenesis by controlling vascular recruitment of monocytes/macrophages in a dual manner: regulation of endothelial MCP-1 expression in response to hemodynamic and inflammatory forces as well as MCP-1 dependent monocyte migration.
Collapse
|
43
|
Passacquale G, Di Giosia P, Ferro A. The role of inflammatory biomarkers in developing targeted cardiovascular therapies: lessons from the cardiovascular inflammation reduction trials. Cardiovasc Res 2015; 109:9-23. [PMID: 26410367 DOI: 10.1093/cvr/cvv227] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/06/2015] [Indexed: 01/04/2023] Open
Abstract
Anti-inflammatory add-on therapy to conventional cardiovascular prophylaxis has been proposed as a novel therapeutic approach to potentially reduce residual cardiovascular risk. This hypothesis has been challenged by a series of unsuccessful Phase III studies testing the impact on clinical outcomes of novel agents with immunomodulatory actions. Specifically, the apparent ability of phospholipase A2 (PLA2) inhibitors and of antioxidants to ameliorate inflammation and to reduce coronary disease in Phase II trials did not translate into improved secondary cardiovascular prevention in larger population-based studies. Other anti-inflammatory agents are still under scrutiny. However, studies to date have lacked information on the inflammatory profile of the participants, both at baseline and at follow-up, thereby limiting the possibility of identifying subgroups of patients in whom 'residual inflammation' can be detected despite optimal conventional therapy, and who could therefore benefit from a cardiovascular prevention strategy specifically targeting inflammation. This has also rendered it difficult to interpret the results as a conclusive demonstration of inefficacy of the tested anti-inflammatory strategies in the treatment of atherosclerosis. We here discuss the importance of better patient characterization to minimize heterogeneity of the study population, so that effectiveness of different anti-inflammatory strategies can be evaluated in targeted subgroups of patients. We also illustrate how specific inflammatory biomarkers could assist in this process, with particular emphasis on the roles of high-sensitivity C-reactive protein and circulating monocyte phenotype.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Paolo Di Giosia
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Albert Ferro
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
44
|
Hung OY, Brown AJ, Ahn SG, Veneziani A, Giddens DP, Samady H. Association of Wall Shear Stress with Coronary Plaque Progression and Transformation. Interv Cardiol Clin 2015; 4:491-502. [PMID: 28581935 DOI: 10.1016/j.iccl.2015.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Coronary endothelial function regulation by wall shear stress (WSS), the frictional force of blood exerted against the vessel wall, can help explain the focal propensity of plaque development in an environment of systemic atherosclerosis risk factors. Sustained abnormal pathologic WSS leads to a proatherogenic endothelial cell phenotype, plaque progression and transformation, and adaptive vascular remodeling in site-specific areas. Assessing dynamic coronary plaque compositional changes in vivo remains challenging; however, recent advances in intravascular image acquisition and processing may provide swifter WSS calculations and make possible larger prospective investigations on the prognostic value of WSS in patients with coronary atherosclerosis.
Collapse
Affiliation(s)
- Olivia Y Hung
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road NE, Atlanta, GA 30322, USA
| | - Adam J Brown
- Department of Cardiovascular Medicine, University of Cambridge, ACCI Level 6, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Sung Gyun Ahn
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road NE, Atlanta, GA 30322, USA; Division of Cardiology, Yonsei University, Wonju College of Medicine, 20 Ilsan-ro, Wonju 220-701, South Korea
| | - Alessandro Veneziani
- Department of Mathematics and Computer Science, Emory University, 400 Dowman Drive, Atlanta, GA 30322, USA
| | - Don P Giddens
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr, Atlanta, GA 30332, USA
| | - Habib Samady
- Interventional Cardiology, Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road, Suite F606, Atlanta, GA 30322, USA.
| |
Collapse
|
45
|
Remus EW, Sayeed I, Won S, Lyle AN, Stein DG. Progesterone protects endothelial cells after cerebrovascular occlusion by decreasing MCP-1- and CXCL1-mediated macrophage infiltration. Exp Neurol 2015; 271:401-8. [PMID: 26188381 DOI: 10.1016/j.expneurol.2015.07.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 06/29/2015] [Accepted: 07/13/2015] [Indexed: 12/16/2022]
Abstract
The neuroprotective effects of progesterone after ischemic stroke have been established, but the role of progesterone in promoting cerebrovascular repair remains under-explored. Male Sprague-Dawley rats underwent transient middle cerebral artery occlusion (tMCAO) for 90 min followed by reperfusion for 3 days. Progesterone (8 mg/kg/day) was administered intraperitoneally at 1h after initial occlusion followed by subcutaneous injections at 6, 24 and 48 h post-occlusion. Rats were euthanized after 72 h and brain endothelial cell density and macrophage infiltration were evaluated within the cerebral cortex. We also assessed progesterone's ability to induce macrophage migration toward hypoxic/reoxygenated cultured endothelial cells. We found that progesterone treatment post-tMCAO protects ischemic endothelial cells from macrophage infiltration. We further demonstrate that infiltration of monocytes/macrophages can be induced by potent chemotactic factors such as monocyte chemoattractant protein-1 (MCP-1) and the chemokine ligand 1 (CXCL1), secreted by hypoxic/reoxygenated endothelial cells. Progesterone blunts secretion of MCP-1 and CXCL1 from endothelial cells after hypoxia/reoxygenation injury and decreases leukocyte infiltration. The treatment protects ischemic endothelial cells from macrophage infiltration and thus preserves vascularization after ischemic injury.
Collapse
Affiliation(s)
- Ebony Washington Remus
- Department of Emergency Medicine Brain Research Laboratory, Emory University, Atlanta, GA, USA
| | - Iqbal Sayeed
- Department of Emergency Medicine Brain Research Laboratory, Emory University, Atlanta, GA, USA
| | - Soonmi Won
- Department of Emergency Medicine Brain Research Laboratory, Emory University, Atlanta, GA, USA
| | - Alicia N Lyle
- Department of Cardiology, Emory University Atlanta, GA, USA
| | - Donald G Stein
- Department of Emergency Medicine Brain Research Laboratory, Emory University, Atlanta, GA, USA.
| |
Collapse
|
46
|
Yang B, Cai B, Deng P, Wu X, Guan Y, Zhang B, Cai W, Schaper J, Schaper W. Nitric Oxide Increases Arterial Endotheial Permeability through Mediating VE-Cadherin Expression during Arteriogenesis. PLoS One 2015; 10:e0127931. [PMID: 26133549 PMCID: PMC4489889 DOI: 10.1371/journal.pone.0127931] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/13/2015] [Indexed: 11/18/2022] Open
Abstract
Macrophage invasion is an important event during arteriogenesis, but the underlying mechanism is still only partially understood. The present study tested the hypothesis that nitric oxide (NO) and VE-cadherin, two key mediators for vascular permeability, contribute to this event in a rat ischemic hindlimb model. In addition, the effect of NO on expression of VE-caherin and endothelial permeability was also studied in cultured HUVECs. We found that: 1) in normal arteriolar vessels (NAV), eNOS was moderately expressed in endothelial cells (EC) and iNOS was rarely detected. In contrast, in collateral vessels (CVs) induced by simple femoral artery ligation, both eNOS and iNOS were significantly upregulated (P<0.05). Induced iNOS was found mainly in smooth muscle cells, but also in other vascular cells and macrophages; 2) in NAV VE-cadherin was strongly expressed in EC. In CVs, VE-cadherin was significantly downregulated, with a discontinuous and punctate pattern. Administration of nitric oxide donor DETA NONOate (NONOate) further reduced the amounts of Ve-cadherin in CVs, whereas NO synthase inhibitor L-NAME inhibited downregulation of VE-cadherin in CVs; 3) in normal rats Evans blue extravasation (EBE) was low in the musculus gracilis, FITC-dextron leakage was not detected in the vascular wall and few macrophages were observed in perivascular space. In contrast, EBE was significantly increased in femoral artery ligation rats, FITC-dextron leakage and increased amounts of macrophages were detected in CVs, which were further enhanced by administration of NONOate, but inhibited by L-NAME supplement; 4) in vitro experiments confirmed that an increase in NO production reduced VE-cadherin expression, correlated with increases in the permeability of HUVECs. In conclusion, our data for the first time reveal the expression profile of VE-cadherin and alterations of vascular permeability in CVs, suggesting that NO-mediated VE-cadherin pathway may be one important mechanism responsible, at least in part, for macrophage invasion during arteriogenesis.
Collapse
Affiliation(s)
- Baolin Yang
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
- Department of Anatomy, School of Basic Medicine, Nanchang Univ., Nanchang, 330006, Jiangxi, P.R. China
| | - Baizhen Cai
- Dept. of Intensive Care Unit, the 3rd Xiangya Hospital, Central South Univ., Changsha, 410013, Hunan, P.R. China
| | - Panyue Deng
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
- * E-mail: (WC); (PD); (WS); (JS)
| | - Xiaoqiong Wu
- Department of Anatomy & Neurobiology, School of Basic Medicine, Central South Univ., Changsha, 410013, Hunan, P.R. China
| | - Yinglu Guan
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
| | - Bin Zhang
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
| | - Weijun Cai
- Department of Histology & Embryology, School of Basic Medicine, Central South Univ., Changsha, 410078, Hunan, P.R. China
- * E-mail: (WC); (PD); (WS); (JS)
| | - Jutta Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany
- * E-mail: (WC); (PD); (WS); (JS)
| | - Wolfgang Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany
- * E-mail: (WC); (PD); (WS); (JS)
| |
Collapse
|
47
|
Dunn J, Simmons R, Thabet S, Jo H. The role of epigenetics in the endothelial cell shear stress response and atherosclerosis. Int J Biochem Cell Biol 2015; 67:167-76. [PMID: 25979369 DOI: 10.1016/j.biocel.2015.05.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 04/30/2015] [Accepted: 05/02/2015] [Indexed: 12/15/2022]
Abstract
Currently in the field of vascular biology, the role of epigenetics in endothelial cell biology and vascular disease has attracted more in-depth study. Using both in vitro and in vivo models of blood flow, investigators have recently begun to reveal the underlying epigenetic regulation of endothelial gene expression. Recently, our group, along with two other independent groups, have demonstrated that blood flow controls endothelial gene expression by DNA methyltransferases (DNMT1 and 3A). Disturbed flow (d-flow), characterized by low and oscillating shear stress (OS), is pro-atherogenic and induces expression of DNMT1 both in vivo and in vitro. D-flow regulates genome-wide DNA methylation patterns in a DNMT-dependent manner. The DNMT inhibitor 5-Aza-2'deoxycytidine (5Aza) or DNMT1 siRNA reduces OS-induced endothelial inflammation. Moreover, 5Aza inhibits the development of atherosclerosis in ApoE(-/-) mice. Through a systems biological analysis of genome-wide DNA methylation patterns and gene expression data, we found 11 mechanosensitive genes which were suppressed by d-flow in vivo, experienced hypermethylation in their promoter region in response to d-flow, and were rescued by 5Aza treatment. Interestingly, among these mechanosensitive genes, the two transcription factors HoxA5 and Klf3 contain cAMP-response-elements (CRE), which may indicate that methylation of CRE sites could serve as a mechanosensitive master switch in gene expression. These findings provide new insight into the mechanism by which flow controls epigenetic DNA methylation patterns, which in turn alters endothelial gene expression, regulates vascular biology, and induces atherosclerosis. These novel findings have broad implications for understanding the biochemical mechanisms of atherogenesis and provide a basis for identifying potential therapeutic targets for atherosclerosis. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
Affiliation(s)
- Jessilyn Dunn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Rachel Simmons
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Salim Thabet
- Division of Cardiology, Georgia Institute of Technology and Emory University, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA; Division of Cardiology, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
48
|
He J, Li YL. Ginsenoside Rg1 Downregulates the Shear Stress Induced MCP-1 Expression by Inhibiting MAPK Signaling Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:305-17. [DOI: 10.1142/s0192415x15500202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ginsenoside Rg1 has been reported to possess anti-inflammatory activities, but the effects of Rg1 on the shear induced MCP-1 upregulation mechanism on endothelial cells (ECs) remain to be determined. In this study, we show that Rg1 down modulates shear induced pro-inflammatory cytokine MCP-1 gene expression and monocytes adhesion without potential cell toxicity. The negative effects on monocytes adhesion is due to a decrease in MCP-1 protein release. Furthermore, the inhibitory effect of Rg1 on the phosphorylation level of ERK, p38, and JNK mitogen-activated protein kinase (MAPK) induced by shear stress (SS) is similar with that of specific chemical inhibitors for MAPK pathways activation. These results demonstrate that ginsenoside Rg1 inhibits the shear induced inflammation by suppressing the MAPK pathway. This suggests that Rg1 may serve as a novel anti-inflammatory agent for inflammation-induced cardiovascular diseases treatment.
Collapse
Affiliation(s)
- Jian He
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning 116023, P. R. China
- Departments of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92037, USA
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, P. R. China
| | - Yu-Lin Li
- The Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
49
|
Davis CA, Zambrano S, Anumolu P, Allen ACB, Sonoqui L, Moreno MR. Device-Based In Vitro Techniques for Mechanical Stimulation of Vascular Cells: A Review. J Biomech Eng 2015; 137:040801. [DOI: 10.1115/1.4029016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/07/2014] [Indexed: 01/19/2023]
Abstract
The most common cause of death in the developed world is cardiovascular disease. For decades, this has provided a powerful motivation to study the effects of mechanical forces on vascular cells in a controlled setting, since these cells have been implicated in the development of disease. Early efforts in the 1970 s included the first use of a parallel-plate flow system to apply shear stress to endothelial cells (ECs) and the development of uniaxial substrate stretching techniques (Krueger et al., 1971, “An in Vitro Study of Flow Response by Cells,” J. Biomech., 4(1), pp. 31–36 and Meikle et al., 1979, “Rabbit Cranial Sutures in Vitro: A New Experimental Model for Studying the Response of Fibrous Joints to Mechanical Stress,” Calcif. Tissue Int., 28(2), pp. 13–144). Since then, a multitude of in vitro devices have been designed and developed for mechanical stimulation of vascular cells and tissues in an effort to better understand their response to in vivo physiologic mechanical conditions. This article reviews the functional attributes of mechanical bioreactors developed in the 21st century, including their major advantages and disadvantages. Each of these systems has been categorized in terms of their primary loading modality: fluid shear stress (FSS), substrate distention, combined distention and fluid shear, or other applied forces. The goal of this article is to provide researchers with a survey of useful methodologies that can be adapted to studies in this area, and to clarify future possibilities for improved research methods.
Collapse
Affiliation(s)
- Caleb A. Davis
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Steve Zambrano
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Pratima Anumolu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Alicia C. B. Allen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712-1801 e-mail:
| | - Leonardo Sonoqui
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3120 e-mail:
| | - Michael R. Moreno
- Department of Mechanical Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843-3123 e-mail:
| |
Collapse
|
50
|
Rennier K, Ji JY. Shear stress attenuates apoptosis due to TNFα, oxidative stress, and serum depletion via death-associated protein kinase (DAPK) expression. BMC Res Notes 2015; 8:85. [PMID: 25890206 PMCID: PMC4374420 DOI: 10.1186/s13104-015-1037-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 01/13/2023] Open
Abstract
Background Misdirected apoptosis in endothelial cells participates in the development of pathological conditions such as atherosclerosis. Tight regulation of apoptosis is necessary to ensure normal cell function. The rate of cell turnover is increased at sites prone to lesion development. Laminar shear stress is protective against atherosclerosis, and helps suppress apoptosis induced by cytokines, oxidative stress, and serum depletion. Current Studies have shown that the pro-apoptotic DAPK expression and function to be regulated in part by shear stress, and that shearing cells already treated with cytokine tumor necrosis factor (TNF) α significantly reduced apoptosis. We investigate further the suppression of endothelial apoptosis by shear stress with other apoptotic triggers, and the involvement of DAPK and caspase 3/7. Results We have shown that exposure to shear stress (12 dynes/cm2 for 6 hrs) suppressed endothelial apoptosis triggered by cytokine (TNFα), oxidative stress (H2O2), and serum depletion, either before or after a long term (18 hr) induction. This is correlated with a parallel decrease of DAPK expression and caspase activity compared to non-sheared cells. We found similar modulation of DAPK and apoptosis by shear stress with other pro-apoptotic signals. Changes in DAPK and caspase 3/7 are directly correlated to changes in apoptosis. Interestingly, shear stress applied to cells prior to induction with apoptosis agents resulted in a higher suppression of apoptosis and DAPK and caspase activity, compared to applying shear stress post induction. This is correlated with a higher expression and activation of DAPK in cells sheared at the end of 24-hr experiment. Also, shear stress alone also induced higher apoptosis and DAPK expression, and the effect is sustained even after 18 hrs incubation in static condition, compared to non-sheared cells. Conclusions Overall, we show that laminar shear stress inhibits various apoptosis pathways by modulating DAPK activity, as well as caspase activation, in a time-dependent manner. Shear stress could target DAPK as a converging point to exert its effects of suppressing endothelial apoptosis. The temporal shear stress stimulation of DAPK and its role in different apoptosis pathways may help identify key mechanisms of the endothelial mechanotransduction pathway.
Collapse
Affiliation(s)
- Keith Rennier
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL-220 J, Indianapolis, IN, 46202, USA.
| | - Julie Y Ji
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL-220 J, Indianapolis, IN, 46202, USA.
| |
Collapse
|