1
|
Deng H, Eichmann A, Schwartz MA. Fluid Shear Stress-Regulated Vascular Remodeling: Past, Present, and Future. Arterioscler Thromb Vasc Biol 2025; 45:882-900. [PMID: 40207366 DOI: 10.1161/atvbaha.125.322557] [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] [Indexed: 04/11/2025]
Abstract
The vascular system remodels throughout life to ensure adequate perfusion of tissues as they grow, regress, or change metabolic activity. Angiogenesis, the sprouting of new blood vessels to expand the capillary network, versus regression, in which endothelial cells die or migrate away to remove unneeded capillaries, controls capillary density. In addition, upstream arteries adjust their diameters to optimize blood flow to downstream vascular beds, which is controlled primarily by vascular endothelial cells sensing fluid shear stress (FSS) from blood flow. Changes in capillary density and small artery tone lead to changes in the resistance of the vascular bed, which leads to changes in flow through the arteries that feed these small vessels. The resultant decreases or increases in FSS through these vessels then stimulate their inward or outward remodeling, respectively. This review summarizes our knowledge of endothelial FSS-dependent vascular remodeling, offering insights into potential therapeutic interventions. We first provide a historical overview, then discuss the concept of set point and mechanisms of low-FSS-mediated and high-FSS-mediated inward and outward remodeling. We then cover in vivo animal models, molecular mechanisms, and clinical implications. Understanding the mechanisms underlying physiological endothelial FSS-mediated vascular remodeling and their failure due to mutations or chronic inflammatory and metabolic stresses may lead to new therapeutic strategies to prevent or treat vascular diseases.
Collapse
Affiliation(s)
- Hanqiang Deng
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Anne Eichmann
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
| | - Martin A Schwartz
- Yale Cardiovascular Research Center CT (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Section of Cardiovascular Medicine, Department of Internal Medicine (H.D., A.E., M.A.S.), Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale School of Medicine, New Haven, CT (M.A.S.)
- Department of Biomedical Engineering, Yale School of Engineering, New Haven, CT (M.A.S.)
| |
Collapse
|
2
|
Chen LJ, Li JYS, Nguyen P, Norwich G, Wang Y, Teng D, Shiu YT, Shyy JYJ, Chien S. Pulsatile flow induces chromatin interaction with lamin-associated proteins to enrich H3K9 methylation in endothelial cells. Proc Natl Acad Sci U S A 2025; 122:e2424566122. [PMID: 40117319 PMCID: PMC11962468 DOI: 10.1073/pnas.2424566122] [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: 11/26/2024] [Accepted: 02/19/2025] [Indexed: 03/23/2025] Open
Abstract
Endothelial cells (ECs) are constantly exposed to hemodynamic forces, which play a crucial role in regulating EC functions. Pulsatile laminar shear stress (PS), representing atheroprotective flow, maintains the anti-inflammation and homeostatic phenotype of ECs, but the comprehensive mechanism underlying the PS-repression of inflammatory genes remains to be determined. In this study, we investigated the role of chromatin organization in mediating the effects of PS on inflammatory gene expression in ECs. We demonstrated that PS induced the expression of histone methyltransferase SUV39H1 to promote heterochromatin formation via H3K9 trimethylation (H3K9me3) enrichment, a hallmark gene repression mechanism. SUV39H1 interacts with lamin-associated proteins and facilitates the perinuclear localization of the H3K9me3-enrichment. Silencing the lamin-associated protein emerin (EMD) not only led to the reductions of cytoskeletal F-actin formation and perinuclear H3K9me3 enrichment; but also the impairment of PS-induced SUV39H1 expression, H3K9me3 enrichment at E-selectin and vascular cell adhesion molecule 1 loci to revert their PS-repressed expression. Hence, EMD acts as a hub to transmit mechanical cues from the cytoskeleton to the nucleus and recruits SUV39H1, which regulate nuclear organization, chromatin state, and gene expression. These results accentuate the critical role of nuclear architecture in mechanotransduction and EC responses to mechanical stimuli.
Collapse
Affiliation(s)
- Li-Jing Chen
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA92093
| | - Julie Yi-Shuan Li
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA92093
| | - Phu Nguyen
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA92093
| | - Gerard Norwich
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA92093
| | - Yingxiao Wang
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA90089
| | - Dayu Teng
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA92093
| | - Yan-Ting Shiu
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah, Salt Lake City, UT84112
- Veterans Affairs Medical Center, University of Utah, Salt Lake City, UT84112
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT84112
| | - John Y. J. Shyy
- Department of Medicine, University of California at San Diego, La Jolla, CA92093
| | - Shu Chien
- Department of Bioengineering, University of California at San Diego, La Jolla, CA92093
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA92093
- Department of Medicine, University of California at San Diego, La Jolla, CA92093
| |
Collapse
|
3
|
Tiberio F, Coda ARD, Tosi DD, Luzi D, Polito L, Liso A, Lattanzi W. Mechanobiology and Primary Cilium in the Pathophysiology of Bone Marrow Myeloproliferative Diseases. Int J Mol Sci 2024; 25:8860. [PMID: 39201546 PMCID: PMC11354938 DOI: 10.3390/ijms25168860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Philadelphia-Negative Myeloproliferative neoplasms (MPNs) are a diverse group of blood cancers leading to excessive production of mature blood cells. These chronic diseases, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), can significantly impact patient quality of life and are still incurable in the vast majority of the cases. This review examines the mechanobiology within a bone marrow niche, emphasizing the role of mechanical cues and the primary cilium in the pathophysiology of MPNs. It discusses the influence of extracellular matrix components, cell-cell and cell-matrix interactions, and mechanosensitive structures on hematopoietic stem cell (HSC) behavior and disease progression. Additionally, the potential implications of the primary cilium as a chemo- and mechanosensory organelle in bone marrow cells are explored, highlighting its involvement in signaling pathways crucial for hematopoietic regulation. This review proposes future research directions to better understand the dysregulated bone marrow niche in MPNs and to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Federica Tiberio
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Domiziano Dario Tosi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
| | - Debora Luzi
- S.C. Oncoematologia, Azienda Ospedaliera di Terni, 05100 Terni, Italy;
| | - Luca Polito
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
| | - Arcangelo Liso
- Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
4
|
Brown PA. Genes Differentially Expressed Across Major Arteries Are Enriched in Endothelial Dysfunction-Related Gene Sets: Implications for Relative Inter-artery Atherosclerosis Risk. Bioinform Biol Insights 2024; 18:11779322241251563. [PMID: 38765020 PMCID: PMC11100403 DOI: 10.1177/11779322241251563] [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: 09/05/2023] [Accepted: 04/13/2024] [Indexed: 05/21/2024] Open
Abstract
Atherosclerosis differs across major arteries. Although the biological basis is not fully understood, limited evidence of genetic differences has been documented. This study, therefore, was aimed to identify differentially expressed genes between clinically relevant major arteries and investigate their enrichment in endothelial dysfunction-related gene sets. A bioinformatic analysis of publicly available gene-level read counts for coronary, aortic, and tibial arteries was performed. Differential gene expression was conducted with DeSeq2 at a false discovery rate of 0.05. Differentially expressed genes were then subjected to over-representation analysis and active-subnetwork-oriented enrichment analysis, both at a false discovery rate of 0.005. Enriched terms common to both analyses were categorized for each contrast into immunity/inflammation-, membrane biology-, lipid metabolism-, and coagulation-related terms, and the top differentially expressed genes validated against Swiss Institute of Bioinformatics' Bgee database. There was mostly upregulation of differentially expressed genes for the coronary/tibial and aorta/tibial contrasts, but milder changes for the coronary/aorta contrast. Transcriptomic differences between coronary or aortic versus tibial samples largely involved immunity/inflammation-, membrane biology-, lipid metabolism-, and coagulation-related genes, suggesting potential to modulate endothelial dysfunction and atherosclerosis. These results imply atheroprone coronary and aortic environments compared with tibial artery tissue, which may explain observed relative inter-artery atherosclerosis risk.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Kingston, Jamaica
| |
Collapse
|
5
|
Lang MB, Leung KY, Greene ND, Malone KM, Saginc G, Randi AM, Kiprianos A, Maughan RT, Pericleous C, Mason JC. The actions of methotrexate on endothelial cells are dependent on the shear stress-induced regulation of one carbon metabolism. Front Immunol 2023; 14:1209490. [PMID: 37457690 PMCID: PMC10349526 DOI: 10.3389/fimmu.2023.1209490] [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: 04/20/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Objectives The disease-modifying anti-rheumatic drug methotrexate (MTX) is recognized to reduce cardiovascular risk in patients with systemic inflammatory diseases. However, the molecular basis for these cardioprotective effects remains incompletely understood. This study evaluated the actions of low-dose MTX on the vascular endothelium. Methods Human endothelial cells (EC) were studied under in vitro conditions relevant to inflammatory arthritis. These included culture in a pro-inflammatory microenvironment and exposure to fluid shear stress (FSS) using a parallel plate model. Respectively treated cells were analyzed by RNA sequencing and quantitative real-time PCR for gene expression, by immunoblotting for protein expression, by phosphokinase activity arrays, by flow cytometry for cell cycle analyses and by mass spectrometry to assess folate metabolite levels. Results In static conditions, MTX was efficiently taken up by EC and caused cell cycle arrest concurrent with modulation of cell signaling pathways. These responses were reversed by folinic acid (FA), suggesting that OCM is a predominant target of MTX. Under FSS, MTX did not affect cell proliferation or pro-inflammatory gene expression. Exposure to FSS downregulated endothelial one carbon metabolism (OCM) as evidenced by decreased expression of key OCM genes and metabolites. Conclusion We found that FSS significantly downregulated OCM and thereby rendered EC less susceptible to the effects of MTX treatment. The impact of shear stress on OCM suggested that MTX does not directly modulate endothelial function. The cardioprotective actions of MTX likely reflect direct actions on inflammatory cells and indirect benefit on the vascular endothelium.
Collapse
Affiliation(s)
- Marie B. Lang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kit-Yi Leung
- Developmental Biology & Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nicholas D.E. Greene
- Developmental Biology & Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Kerri M. Malone
- European Bioinformatics Institute, Cambridge, United Kingdom
| | - Gaye Saginc
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Anna M. Randi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Allan Kiprianos
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Robert T. Maughan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Charis Pericleous
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Justin C. Mason
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Banerjee P, Rosales JE, Chau K, Nguyen MTH, Kotla S, Lin SH, Deswal A, Dantzer R, Olmsted-Davis EA, Nguyen H, Wang G, Cooke JP, Abe JI, Le NT. Possible molecular mechanisms underlying the development of atherosclerosis in cancer survivors. Front Cardiovasc Med 2023; 10:1186679. [PMID: 37332576 PMCID: PMC10272458 DOI: 10.3389/fcvm.2023.1186679] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Cancer survivors undergone treatment face an increased risk of developing atherosclerotic cardiovascular disease (CVD), yet the underlying mechanisms remain elusive. Recent studies have revealed that chemotherapy can drive senescent cancer cells to acquire a proliferative phenotype known as senescence-associated stemness (SAS). These SAS cells exhibit enhanced growth and resistance to cancer treatment, thereby contributing to disease progression. Endothelial cell (EC) senescence has been implicated in atherosclerosis and cancer, including among cancer survivors. Treatment modalities for cancer can induce EC senescence, leading to the development of SAS phenotype and subsequent atherosclerosis in cancer survivors. Consequently, targeting senescent ECs displaying the SAS phenotype hold promise as a therapeutic approach for managing atherosclerotic CVD in this population. This review aims to provide a mechanistic understanding of SAS induction in ECs and its contribution to atherosclerosis among cancer survivors. We delve into the mechanisms underlying EC senescence in response to disturbed flow and ionizing radiation, which play pivotal role in atherosclerosis and cancer. Key pathways, including p90RSK/TERF2IP, TGFβR1/SMAD, and BH4 signaling are explored as potential targets for cancer treatment. By comprehending the similarities and distinctions between different types of senescence and the associated pathways, we can pave the way for targeted interventions aim at enhancing the cardiovascular health of this vulnerable population. The insights gained from this review may facilitate the development of novel therapeutic strategies for managing atherosclerotic CVD in cancer survivors.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Julia Enterría Rosales
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- School of Medicine, Instituto Tecnológico de Monterrey, Guadalajara, Mexico
| | - Khanh Chau
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
- Department of Life Science, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth A. Olmsted-Davis
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Hung Nguyen
- Cancer Division, Burnett School of Biomedical Science, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
7
|
Afshar Y, Ma F, Quach A, Jeong A, Sunshine HL, Freitas V, Jami-Alahmadi Y, Helaers R, Li X, Pellegrini M, Wohlschlegel JA, Romanoski CE, Vikkula M, Iruela-Arispe ML. Transcriptional drifts associated with environmental changes in endothelial cells. eLife 2023; 12:e81370. [PMID: 36971339 PMCID: PMC10168696 DOI: 10.7554/elife.81370] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/26/2023] [Indexed: 03/29/2023] Open
Abstract
Environmental cues, such as physical forces and heterotypic cell interactions play a critical role in cell function, yet their collective contributions to transcriptional changes are unclear. Focusing on human endothelial cells, we performed broad individual sample analysis to identify transcriptional drifts associated with environmental changes that were independent of genetic background. Global gene expression profiling by RNA sequencing and protein expression by liquid chromatography-mass spectrometry directed proteomics distinguished endothelial cells in vivo from genetically matched culture (in vitro) samples. Over 43% of the transcriptome was significantly changed by the in vitro environment. Subjecting cultured cells to long-term shear stress significantly rescued the expression of approximately 17% of genes. Inclusion of heterotypic interactions by co-culture of endothelial cells with smooth muscle cells normalized approximately 9% of the original in vivo signature. We also identified novel flow dependent genes, as well as genes that necessitate heterotypic cell interactions to mimic the in vivo transcriptome. Our findings highlight specific genes and pathways that rely on contextual information for adequate expression from those that are agnostic of such environmental cues.
Collapse
Affiliation(s)
- Yalda Afshar
- Department of Obstetrics and Gynecology, University of California, Los AngelesLos AngelesUnited States
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
| | - Feyiang Ma
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Austin Quach
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - Anhyo Jeong
- Department of Obstetrics and Gynecology, University of California, Los AngelesLos AngelesUnited States
| | - Hannah L Sunshine
- Department of Molecular, Cellular and Integrative Physiology, University of California, Los AngelesLos AngelesUnited States
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of MedicineChicagoUnited States
| | - Vanessa Freitas
- Departament of Cell and Developmental Biology, Institute of Biomedical Science, University of Sao PauloLos AngelesUnited States
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of CaliforniaLos AngelesUnited States
| | - Raphael Helaers
- Human Molecular Genetics, de Duve Institute, University of LouvainBrusselsBelgium
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, University of CaliforniaLos AngelesUnited States
| | - Matteo Pellegrini
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of CaliforniaLos AngelesUnited States
| | - Casey E Romanoski
- Department of Cellular and Molecular Medicine, University of ArizonaTucsonUnited States
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of LouvainBrusselsBelgium
- WELBIO department, WEL Research InstituteWavreBelgium
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology, University of California, Los AngelesLos AngelesUnited States
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of MedicineChicagoUnited States
| |
Collapse
|
8
|
Becker LM, Chen SH, Rodor J, de Rooij LPMH, Baker AH, Carmeliet P. Deciphering endothelial heterogeneity in health and disease at single-cell resolution: progress and perspectives. Cardiovasc Res 2023; 119:6-27. [PMID: 35179567 PMCID: PMC10022871 DOI: 10.1093/cvr/cvac018] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/16/2021] [Accepted: 02/16/2022] [Indexed: 11/14/2022] Open
Abstract
Endothelial cells (ECs) constitute the inner lining of vascular beds in mammals and are crucial for homeostatic regulation of blood vessel physiology, but also play a key role in pathogenesis of many diseases, thereby representing realistic therapeutic targets. However, it has become evident that ECs are heterogeneous, encompassing several subtypes with distinct functions, which makes EC targeting and modulation in diseases challenging. The rise of the new single-cell era has led to an emergence of studies aimed at interrogating transcriptome diversity along the vascular tree, and has revolutionized our understanding of EC heterogeneity from both a physiological and pathophysiological context. Here, we discuss recent landmark studies aimed at teasing apart the heterogeneous nature of ECs. We cover driving (epi)genetic, transcriptomic, and metabolic forces underlying EC heterogeneity in health and disease, as well as current strategies used to combat disease-enriched EC phenotypes, and propose strategies to transcend largely descriptive heterogeneity towards prioritization and functional validation of therapeutically targetable drivers of EC diversity. Lastly, we provide an overview of the most recent advances and hurdles in single EC OMICs.
Collapse
Affiliation(s)
| | | | | | | | - Andrew H Baker
- Corresponding authors. Tel: +32 16 32 62 47, E-mail: (P.C.); Tel: +44 (0)131 242 6774, E-mail: (A.H.B.)
| | - Peter Carmeliet
- Corresponding authors. Tel: +32 16 32 62 47, E-mail: (P.C.); Tel: +44 (0)131 242 6774, E-mail: (A.H.B.)
| |
Collapse
|
9
|
Meng F, Cheng H, Qian J, Dai X, Huang Y, Fan Y. In vitro fluidic systems: Applying shear stress on endothelial cells. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
10
|
Langston JC, Rossi MT, Yang Q, Ohley W, Perez E, Kilpatrick LE, Prabhakarpandian B, Kiani MF. Omics of endothelial cell dysfunction in sepsis. VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:R15-R34. [PMID: 35515704 PMCID: PMC9066943 DOI: 10.1530/vb-22-0003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022]
Abstract
During sepsis, defined as life-threatening organ dysfunction due to dysregulated host response to infection, systemic inflammation activates endothelial cells and initiates a multifaceted cascade of pro-inflammatory signaling events, resulting in increased permeability and excessive recruitment of leukocytes. Vascular endothelial cells share many common properties but have organ-specific phenotypes with unique structure and function. Thus, therapies directed against endothelial cell phenotypes are needed to address organ-specific endothelial cell dysfunction. Omics allow for the study of expressed genes, proteins and/or metabolites in biological systems and provide insight on temporal and spatial evolution of signals during normal and diseased conditions. Proteomics quantifies protein expression, identifies protein-protein interactions and can reveal mechanistic changes in endothelial cells that would not be possible to study via reductionist methods alone. In this review, we provide an overview of how sepsis pathophysiology impacts omics with a focus on proteomic analysis of mouse endothelial cells during sepsis/inflammation and its relationship with the more clinically relevant omics of human endothelial cells. We discuss how omics has been used to define septic endotype signatures in different populations with a focus on proteomic analysis in organ-specific microvascular endothelial cells during sepsis or septic-like inflammation. We believe that studies defining septic endotypes based on proteomic expression in endothelial cell phenotypes are urgently needed to complement omic profiling of whole blood and better define sepsis subphenotypes. Lastly, we provide a discussion of how in silico modeling can be used to leverage the large volume of omics data to map response pathways in sepsis.
Collapse
Affiliation(s)
- Jordan C Langston
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania, USA
| | | | - Qingliang Yang
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| | - William Ohley
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Edwin Perez
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Laurie E Kilpatrick
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Balabhaskar Prabhakarpandian
- Center for Inflammation and Lung Research, Department of Microbiology, Immunology and Inflammation, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Mohammad F Kiani
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania, USA
- Department of Mechanical Engineering, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
11
|
Chen ZB, Liu X, Chen AT. "Enhancing" mechanosensing: Enhancers and enhancer-derived long non-coding RNAs in endothelial response to flow. CURRENT TOPICS IN MEMBRANES 2021; 87:153-169. [PMID: 34696884 DOI: 10.1016/bs.ctm.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Endothelial cells (ECs), uniquely localized and strategically forming the inner lining of vascular wall, constitute the largest cell surface by area in the human body. The dynamic sensing and response of ECs to mechanical cues, especially shear stress, is crucial for maintenance of vascular homeostasis. It is well recognized that different flow patterns associated with atheroprotective vs atheroprone regions in the arterial tree, result in distinct EC functional phenotypes with differential transcriptome profiles. Mounting evidence has demonstrated an integrative and essential regulatory role of non-coding genome in EC biology. In particular, recent studies have begun to reveal the importance of enhancers and enhancer-derived transcripts in flow-regulated EC gene expression and function. In this minireview, we summarize studies in this area and discuss examples in support of the emerging importance of enhancers and enhancer(-derived) long non-coding RNAs (elncRNAs) in EC mechanosensing, with a focus on flow-responsive EC transcription. Finally, we will provide perspective and discuss standing questions to elucidate the role of these novel regulators in EC mechanobiology.
Collapse
Affiliation(s)
- Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Duarte, CA, United States; Irell and Manella Graduate School of Biological Sciences, Duarte, CA, United States.
| | - Xuejing Liu
- Department of Diabetes Complications and Metabolism, Duarte, CA, United States
| | - Aleysha T Chen
- Department of Bioengineering, University of California, Berkeley, CA, United States
| |
Collapse
|
12
|
Li H, Luo Q, Shan W, Cai S, Tie R, Xu Y, Lin Y, Qian P, Huang H. Biomechanical cues as master regulators of hematopoietic stem cell fate. Cell Mol Life Sci 2021; 78:5881-5902. [PMID: 34232331 PMCID: PMC8316214 DOI: 10.1007/s00018-021-03882-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/02/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem cells (HSCs) perceive both soluble signals and biomechanical inputs from their microenvironment and cells themselves. Emerging as critical regulators of the blood program, biomechanical cues such as extracellular matrix stiffness, fluid mechanical stress, confined adhesiveness, and cell-intrinsic forces modulate multiple capacities of HSCs through mechanotransduction. In recent years, research has furthered the scientific community's perception of mechano-based signaling networks in the regulation of several cellular processes. However, the underlying molecular details of the biomechanical regulatory paradigm in HSCs remain poorly elucidated and researchers are still lacking in the ability to produce bona fide HSCs ex vivo for clinical use. This review presents an overview of the mechanical control of both embryonic and adult HSCs, discusses some recent insights into the mechanisms of mechanosensing and mechanotransduction, and highlights the application of mechanical cues aiming at HSC expansion or differentiation.
Collapse
Affiliation(s)
- Honghu Li
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Qian Luo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Wei Shan
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Shuyang Cai
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Yulin Xu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Yu Lin
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, China.
| |
Collapse
|
13
|
Zhou S, Jin J, Wang J, Zhang Z, Huang S, Zheng Y, Cai L. Effects of Breast Cancer Genes 1 and 2 on Cardiovascular Diseases. Curr Probl Cardiol 2021; 46:100421. [PMID: 31558344 DOI: 10.1016/j.cpcardiol.2019.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/06/2019] [Indexed: 12/20/2022]
Abstract
Carriers of mutations of breast cancer gene 1 and/or 2 (BRCA1/2) have a higher risk of developing breast and ovarian cancers at a relatively young age. Recently, a causative role for BRCA1/2 in cardiovascular diseases has been emerging. In this review, we summarize currently available evidence obtained from studies on animal models and human BRCA1/2 mutation carriers that shows a correlation of BRCA1/2 deficiency with various cardiovascular diseases, including ischemic heart disease, atherosclerosis, and chemotherapy-linked cardiac muscle disorders. We also discuss one of the major mechanisms by which BRCA1/2 protects the heart against oxidative stress, ie mediating the activity of Nrf2 and its downstream targets that govern antioxidant signaling. More research is needed to elucidate whether the carriers of the BRCA1/2 mutations with ovarian and breast cancers have increased susceptibility to chemotherapy-induced cardiac functional impairment.
Collapse
|
14
|
Kulthong K, Hooiveld GJEJ, Duivenvoorde L, Miro Estruch I, Marin V, van der Zande M, Bouwmeester H. Transcriptome comparisons of in vitro intestinal epithelia grown under static and microfluidic gut-on-chip conditions with in vivo human epithelia. Sci Rep 2021; 11:3234. [PMID: 33547413 PMCID: PMC7864925 DOI: 10.1038/s41598-021-82853-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Gut-on-chip devices enable exposure of cells to a continuous flow of culture medium, inducing shear stresses and could thus better recapitulate the in vivo human intestinal environment in an in vitro epithelial model compared to static culture methods. We aimed to study if dynamic culture conditions affect the gene expression of Caco-2 cells cultured statically or dynamically in a gut-on-chip device and how these gene expression patterns compared to that of intestinal segments in vivo. For this we applied whole genome transcriptomics. Dynamic culture conditions led to a total of 5927 differentially expressed genes (3280 upregulated and 2647 downregulated genes) compared to static culture conditions. Gene set enrichment analysis revealed upregulated pathways associated with the immune system, signal transduction and cell growth and death, and downregulated pathways associated with drug metabolism, compound digestion and absorption under dynamic culture conditions. Comparison of the in vitro gene expression data with transcriptome profiles of human in vivo duodenum, jejunum, ileum and colon tissue samples showed similarities in gene expression profiles with intestinal segments. It is concluded that both the static and the dynamic gut-on-chip model are suitable to study human intestinal epithelial responses as an alternative for animal models.
Collapse
Affiliation(s)
- Kornphimol Kulthong
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700 EA, Wageningen, The Netherlands.
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands.
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand.
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Loes Duivenvoorde
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Ignacio Miro Estruch
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700 EA, Wageningen, The Netherlands
| | - Victor Marin
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Meike van der Zande
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700 EA, Wageningen, The Netherlands.
| |
Collapse
|
15
|
Lee SH, Kim SW, Kim JM, Son WC. Histopathologic findings of vascular damage after mechanical thrombectomy using stent retriever in canine models. J Thromb Thrombolysis 2021; 51:1094-1100. [PMID: 33387206 DOI: 10.1007/s11239-020-02353-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2020] [Indexed: 10/22/2022]
Abstract
Although mechanical thrombectomy is a powerful predictor of stroke outcome, it induces vessel wall injury in the acute phase. This study aimed to analyze the degree and the condition of recovery of wall injury after the acute phase via angiography and histopathological analysis of autopsied canine models. Digital subtraction angiography (DSA) and embolization with autologous thrombus were performed in six canines. The model of arterial occlusion was effective in all target vessels. Mechanical thrombectomy was performed in completely occluded vessels using stent retriever. Follow-up angiographic and histopathologic evaluations were performed 1 month later. Complete recanalization using stent retriever was achieved in four cases. Slight residual vessel narrowing after recanalization and moderate narrowing was observed in one case each. Histopathological analysis showed that inflammation, hemorrhage, and device-induced medial injury were not observed in any of the cases. Severe intimal proliferation (grade 4), marked diffuse thrombosis (grade 4), and weak vascular endothelial cell loss (grade 1) were observed in one case and weak endovascular proliferation was observed in one case. Although successful complete recanalization was achieved with a single mechanical thrombectomy attempt and no change was observed in the follow-up DSA, special attention should be paid to postoperative follow-up, as device-induced intimal proliferation, diffuse thrombosis, and endothelial cell loss may remain after 1 month.
Collapse
Affiliation(s)
- Sang-Hun Lee
- Department of Neurology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Republic of Korea.
| | - Sang Woo Kim
- Laboratory Animal Center, Osong Medical Innovation Foundation (K-BIO HEALTH), Cheonju, Republic of Korea
| | - Jong Min Kim
- Laboratory Animal Center, Osong Medical Innovation Foundation (K-BIO HEALTH), Cheonju, Republic of Korea
| | - Woo Chan Son
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
16
|
Shafi O. Switching of vascular cells towards atherogenesis, and other factors contributing to atherosclerosis: a systematic review. Thromb J 2020; 18:28. [PMID: 33132762 PMCID: PMC7592591 DOI: 10.1186/s12959-020-00240-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
Background Onset, development and progression of atherosclerosis are complex multistep processes. Many aspects of atherogenesis are not yet properly known. This study investigates the changes in vasculature that contribute to switching of vascular cells towards atherogenesis, focusing mainly on ageing. Methods Databases including PubMed, MEDLINE and Google Scholar were searched for published articles without any date restrictions, involving atherogenesis, vascular homeostasis, aging, gene expression, signaling pathways, angiogenesis, vascular development, vascular cell differentiation and maintenance, vascular stem cells, endothelial and vascular smooth muscle cells. Results Atherogenesis is a complex multistep process that unfolds in a sequence. It is caused by alterations in: epigenetics and genetics, signaling pathways, cell circuitry, genome stability, heterotypic interactions between multiple cell types and pathologic alterations in vascular microenvironment. Such alterations involve pathological changes in: Shh, Wnt, NOTCH signaling pathways, TGF beta, VEGF, FGF, IGF 1, HGF, AKT/PI3K/ mTOR pathways, EGF, FOXO, CREB, PTEN, several apoptotic pathways, ET - 1, NF-κB, TNF alpha, angiopoietin, EGFR, Bcl - 2, NGF, BDNF, neurotrophins, growth factors, several signaling proteins, MAPK, IFN, TFs, NOs, serum cholesterol, LDL, ephrin, its receptor pathway, HoxA5, Klf3, Klf4, BMPs, TGFs and others.This disruption in vascular homeostasis at cellular, genetic and epigenetic level is involved in switching of the vascular cells towards atherogenesis. All these factors working in pathologic manner, contribute to the development and progression of atherosclerosis. Conclusion The development of atherosclerosis involves the switching of gene expression towards pro-atherogenic genes. This happens because of pathologic alterations in vascular homeostasis. When pathologic alterations in epigenetics, genetics, regulatory genes, microenvironment and vascular cell biology accumulate beyond a specific threshold, then the disease begins to express itself phenotypically. The process of biological ageing is one of the most significant factors in this aspect as it is also involved in the decline in homeostasis, maintenance and integrity.The process of atherogenesis unfolds sequentially (step by step) in an interconnected loop of pathologic changes in vascular biology. Such changes are involved in 'switching' of vascular cells towards atherosclerosis.
Collapse
Affiliation(s)
- Ovais Shafi
- Sindh Medical College - Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
17
|
McGurk KA, Owen B, Watson WD, Nethononda RM, Cordell HJ, Farrall M, Rider OJ, Watkins H, Revell A, Keavney BD. Heritability of haemodynamics in the ascending aorta. Sci Rep 2020; 10:14356. [PMID: 32873833 PMCID: PMC7463029 DOI: 10.1038/s41598-020-71354-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/25/2020] [Indexed: 01/27/2023] Open
Abstract
Blood flow in the vasculature can be characterised by dimensionless numbers commonly used to define the level of instabilities in the flow, for example the Reynolds number, Re. Haemodynamics play a key role in cardiovascular disease (CVD) progression. Genetic studies have identified mechanosensitive genes with causal roles in CVD. Given that CVD is highly heritable and abnormal blood flow may increase risk, we investigated the heritability of fluid metrics in the ascending aorta calculated using patient-specific data from cardiac magnetic resonance (CMR) imaging. 341 participants from 108 British Caucasian families were phenotyped by CMR and genotyped for 557,124 SNPs. Flow metrics were derived from the CMR images to provide some local information about blood flow in the ascending aorta, based on maximum values at systole at a single location, denoted max, and a 'peak mean' value averaged over the area of the cross section, denoted pm. Heritability was estimated using pedigree-based (QTDT) and SNP-based (GCTA-GREML) methods. Estimates of Reynolds number based on spatially averaged local flow during systole showed substantial heritability ([Formula: see text], [Formula: see text]), while the estimated heritability for Reynolds number calculated using the absolute local maximum velocity was not statistically significant (12-13%; [Formula: see text]). Heritability estimates of the geometric quantities alone; e.g. aortic diameter ([Formula: see text], [Formula: see text]), were also substantially heritable, as described previously. These findings indicate the potential for the discovery of genetic factors influencing haemodynamic traits in large-scale genotyped and phenotyped cohorts where local spatial averaging is used, rather than instantaneous values. Future Mendelian randomisation studies of aortic haemodynamic estimates, which are swift to derive in a clinical setting, will allow for the investigation of causality of abnormal blood flow in CVD.
Collapse
Affiliation(s)
- Kathryn A McGurk
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Benjamin Owen
- Department of Mechanical, Aerospace and Civil Engineering, Faculty of Science and Engineering, University of Manchester, Manchester, UK
- School of Engineering, Multiscale Thermofluids Institute, University of Edinburgh, Edinburgh, UK
| | - William D Watson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Richard M Nethononda
- Division of Cardiology, Chris Hani Baragwanath Hospital, Soweto and the University of Witwatersrand, Johannesburg, South Africa
| | - Heather J Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Newcastle upon Tyne, UK
| | - Martin Farrall
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alistair Revell
- Department of Mechanical, Aerospace and Civil Engineering, Faculty of Science and Engineering, University of Manchester, Manchester, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
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: 30] [Impact Index Per Article: 6.0] [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
|
Bittner KR, Jiménez JM, Peyton SR. Vascularized Biomaterials to Study Cancer Metastasis. Adv Healthc Mater 2020; 9:e1901459. [PMID: 31977160 PMCID: PMC7899188 DOI: 10.1002/adhm.201901459] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/07/2019] [Indexed: 12/15/2022]
Abstract
Cancer metastasis, the spread of cancer cells to distant organs, is responsible for 90% of cancer-related deaths. Cancer cells need to enter and exit circulation in order to form metastases, and the vasculature and endothelial cells are key regulators of this process. While vascularized 3D in vitro systems have been developed, few have been used to study cancer, and many lack key features of vessels that are necessary to study metastasis. This review focuses on current methods of vascularizing biomaterials for the study of cancer, and three main factors that regulate intravasation and extravasation: endothelial cell heterogeneity, hemodynamics, and the extracellular matrix of the perivascular niche.
Collapse
Affiliation(s)
- Katharine R Bittner
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
| | - Juan M Jiménez
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Shelly R Peyton
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
20
|
Souilhol C, Serbanovic-Canic J, Fragiadaki M, Chico TJ, Ridger V, Roddie H, Evans PC. Endothelial responses to shear stress in atherosclerosis: a novel role for developmental genes. Nat Rev Cardiol 2020; 17:52-63. [PMID: 31366922 DOI: 10.1038/s41569-019-0239-5] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 01/04/2023]
Abstract
Flowing blood generates a frictional force called shear stress that has major effects on vascular function. Branches and bends of arteries are exposed to complex blood flow patterns that exert low or low oscillatory shear stress, a mechanical environment that promotes vascular dysfunction and atherosclerosis. Conversely, physiologically high shear stress is protective. Endothelial cells are critical sensors of shear stress but the mechanisms by which they decode complex shear stress environments to regulate physiological and pathophysiological responses remain incompletely understood. Several laboratories have advanced this field by integrating specialized shear-stress models with systems biology approaches, including transcriptome, methylome and proteome profiling and functional screening platforms, for unbiased identification of novel mechanosensitive signalling pathways in arteries. In this Review, we describe these studies, which reveal that shear stress regulates diverse processes and demonstrate that multiple pathways classically known to be involved in embryonic development, such as BMP-TGFβ, WNT, Notch, HIF1α, TWIST1 and HOX family genes, are regulated by shear stress in arteries in adults. We propose that mechanical activation of these pathways evolved to orchestrate vascular development but also drives atherosclerosis in low shear stress regions of adult arteries.
Collapse
Affiliation(s)
- Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jovana Serbanovic-Canic
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Maria Fragiadaki
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Timothy J Chico
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Bateson Centre for Lifecourse Biology, University of Sheffield, Sheffield, UK
| | - Victoria Ridger
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Hannah Roddie
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
- Bateson Centre for Lifecourse Biology, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK.
| |
Collapse
|
21
|
Allahverdian S, Chaabane C, Boukais K, Francis GA, Bochaton-Piallat ML. Smooth muscle cell fate and plasticity in atherosclerosis. Cardiovasc Res 2019; 114:540-550. [PMID: 29385543 DOI: 10.1093/cvr/cvy022] [Citation(s) in RCA: 356] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/22/2018] [Indexed: 12/21/2022] Open
Abstract
Current knowledge suggests that intimal smooth muscle cells (SMCs) in native atherosclerotic plaque derive mainly from the medial arterial layer. During this process, SMCs undergo complex structural and functional changes giving rise to a broad spectrum of phenotypes. Classically, intimal SMCs are described as dedifferentiated/synthetic SMCs, a phenotype characterized by reduced expression of contractile proteins. Intimal SMCs are considered to have a beneficial role by contributing to the fibrous cap and thereby stabilizing atherosclerotic plaque. However, intimal SMCs can lose their properties to such an extent that they become hard to identify, contribute significantly to the foam cell population, and acquire inflammatory-like cell features. This review highlights mechanisms of SMC plasticity in different stages of native atherosclerotic plaque formation, their potential for monoclonal or oligoclonal expansion, as well as recent findings demonstrating the underestimated deleterious role of SMCs in this disease.
Collapse
Affiliation(s)
- Sima Allahverdian
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, Room 166 Burrard Building, St Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Chiraz Chaabane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel Servet-1, 1211 Geneva 4, Switzerland
| | - Kamel Boukais
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, Room 166 Burrard Building, St Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Gordon A Francis
- Department of Medicine, Centre for Heart Lung Innovation, Providence Health Care Research Institute, University of British Columbia, Room 166 Burrard Building, St Paul's Hospital, 1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada
| | - Marie-Luce Bochaton-Piallat
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Rue Michel Servet-1, 1211 Geneva 4, Switzerland
| |
Collapse
|
22
|
He M, Huang TS, Li S, Hong HC, Chen Z, Martin M, Zhou X, Huang HY, Su SH, Zhang J, Wang WT, Kang J, Huang HD, Zhang J, Chien S, Shyy JYJ. Atheroprotective Flow Upregulates ITPR3 (Inositol 1,4,5-Trisphosphate Receptor 3) in Vascular Endothelium via KLF4 (Krüppel-Like Factor 4)-Mediated Histone Modifications. Arterioscler Thromb Vasc Biol 2019; 39:902-914. [PMID: 30917677 PMCID: PMC6536300 DOI: 10.1161/atvbaha.118.312301] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective- The topographical distribution of atherosclerosis in vasculature underscores the importance of shear stress in regulating endothelium. With a systems approach integrating sequencing data, the current study aims to explore the link between shear stress-regulated master transcription factor and its regulation of endothelial cell (EC) function via epigenetic modifications. Approach and Results- H3K27ac (acetylation of histone 3 lysine 27)-ChIP-seq (chromatin immunoprecipitation followed by high throughput sequencing), ATAC-seq (an assay for transposase-accessible chromatin-sequencing), and RNA-seq (RNA-sequencing) were performed to investigate the genome-wide epigenetic regulations in ECs in response to atheroprotective pulsatile shear stress (PS). In silico prediction revealed that KLF4 binding motifs were enriched in the PS-enhanced H3K27ac regions. By integrating PS- and KLF4-modulated H3K27ac, we identified 18 novel PS-upregulated genes. The promoter regions of these genes showed an overlap between the KLF4-enhanced assay for transposase-accessible chromatin signals and the PS-induced H3K27ac peaks. Experiments using ECs isolated from mouse aorta, lung ECs from EC-KLF4-TG versus EC-KLF4-KO mice, and atorvastatin-treated ECs showed that ITPR3 (inositol 1,4,5-trisphosphate receptor 3) was robustly activated by KLF4 and statins. KLF4 ATAC-qPCR (quantitative polymerase chain reaction) and ChIP-qPCR further demonstrated that a specific locus in the promoter region of the ITPR3 gene was essential for KLF4 binding, H3K27ac enrichment, chromatin accessibility, RNA polymerase II recruitment, and ITPR3 transcriptional activation. Deletion of this KLF4 binding locus in ECs by using CRISPR-Cas9 resulted in blunted calcium influx, reduced expression of endothelial nitric oxide synthase, and diminished nitric oxide bioavailability. Conclusions- These results from a novel multiomics study suggest that KLF4 is crucial for PS-modulated H3K27ac that allow the transcriptional activation of ITPR3. This novel mechanism contributes to the Ca2+-dependent eNOS (endothelial nitric oxide synthase) activation and EC homeostasis.
Collapse
Affiliation(s)
| | | | - Shuai Li
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Hsiao-Chin Hong
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Zhen Chen
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Marcy Martin
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Xin Zhou
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Hsi-Yuan Huang
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Shu-Han Su
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Jiao Zhang
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Wei-Ting Wang
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Jian Kang
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Hsien-Da Huang
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Jin Zhang
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - Shu Chien
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| | - John Y-J. Shyy
- Department of Medicine (M.H., S.L., H.-C.H., Z.C., M.M., H.-Y.H., S.-H.S., Jiao Zhang, W.-T.W., J.K., S.C., J.Y-J.S.), Department of Bioengineering and Institute of Engineering in Medicine (T.-S.H., S.C.), Department of Pharmacology (X.Z., Jin Zhang.), University of California, San Diego, and Institute of Bioinformatics and Systems Biology (H.-C.H., H.-Y.H., H.-D.H.), Department of Biological Science and Technology (W.-T.W.), National Chiao Tung University, Hsin-Chu, Taiwan
| |
Collapse
|
23
|
Pfeiffer D, Wankhammer K, Stefanitsch C, Hingerl K, Huppertz B, Dohr G, Desoye G, Lang I. Amnion-derived mesenchymal stem cells improve viability of endothelial cells exposed to shear stress in ePTFE grafts. Int J Artif Organs 2018; 42:80-87. [PMID: 30585116 DOI: 10.1177/0391398818815470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE: Blood vessel reconstruction is an increasing need of patients suffering from cardiovascular diseases. For the development of microvascular prostheses, efficient endothelialization is mandatory to prevent graft occlusion. Here, we assessed the impact of amnion-derived mesenchymal stem/stromal cells (hAMSC), known for their important angiogenic potential, on the integrity and stability of endothelial cells exposed to shear stress in vascular grafts. METHODS: Human placental endothelial cells (hPEC) were cultured at the inner surface of an expanded polytetrafluoroethylene (ePTFE) graft positioned within a bioreactor and exposed to a minimal shear stress of 0.015 dyne/cm2 or a physiological shear stress of 0.92 dyne/cm2. hAMSC attached to the outer graft surface were able to interact with human placental endothelial cells by paracrine factors. RESULTS: Microscopical analysis and evaluation of glucose/lactate metabolism evidenced successful cell seeding of the graft: hPEC formed a stable monolayer, hAMSC showed a continuous growth during 72 h incubation. hAMSC improved the viability of hPEC exposed to 0.015 dyne/cm2 as shown by a decreased lactate dehydrogenase release of 13% after 72 h compared to hPEC single culture. The viability-enhancing effect of hAMSC on hPEC was further improved by 13% under physiological shear stress. Angiogenesis array analysis revealed that hPEC exposed to physiological shear stress and hAMSC co-culture reduced the secretion of angiogenin, GRO, MCP-1, and TIMP-2. CONCLUSION: hAMSC exerted best survival-enhancing effects on hPEC under exposure to physiological shear stress and modulated endothelial function by paracrine factors. Our data support further studies on the development of grafts functionalized with hAMSC-derived secretomes to enable fast clinical application.
Collapse
Affiliation(s)
- Dagmar Pfeiffer
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Karin Wankhammer
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Christina Stefanitsch
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Kerstin Hingerl
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Berthold Huppertz
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Gottfried Dohr
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- 2 Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Ingrid Lang
- 1 Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
24
|
Computing of Low Shear Stress-Driven Endothelial Gene Network Involved in Early Stages of Atherosclerotic Process. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5359830. [PMID: 30356351 PMCID: PMC6176299 DOI: 10.1155/2018/5359830] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/03/2018] [Indexed: 12/02/2022]
Abstract
Background In the pathogenesis of atherosclerosis, a central role is represented by endothelial inflammation with influx of chemokine-mediated leukocytes in the vascular wall. Aim of this study was to analyze the effect of different shear stresses on endothelial gene expression and compute gene network involved in atherosclerotic disease, in particular to homeostasis, inflammatory cell migration, and apoptotic processes. Methods HUVECs were subjected to shear stress of 1, 5, and 10 dyne/cm2 in a Flow Bioreactor for 24 hours to compare gene expression modulation. Total RNA was analyzed by Affymetrix technology and the expression of two specific genes (CXCR4 and ICAM-1) was validated by RT-PCR. To highlight possible regulations between genes and as further validation, a bioinformatics analysis was performed. Results At low shear stress (1 dyne/cm2) we observed the following: (a) strong upregulation of CXCR4; (b) mild upregulation of Caspase-8; (c) mild downregulation of ICAM-1; (d) marked downexpression of TNFAIP3. Bioinformatics analysis showed the presence of network composed by 59 new interactors (14 transcription factors and 45 microRNAs) appearing strongly related to shear stress. Conclusions The significant modulation of these genes at low shear stress and their close relationships through transcription factors and microRNAs suggest that all may promote an initial inflamed endothelial cell phenotype, favoring the atherosclerotic disease.
Collapse
|
25
|
Pulmonary Vascular Platform Models the Effects of Flow and Pressure on Endothelial Dysfunction in BMPR2 Associated Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19092561. [PMID: 30158434 PMCID: PMC6164056 DOI: 10.3390/ijms19092561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Endothelial dysfunction is a known consequence of bone morphogenetic protein type II receptor (BMPR2) mutations seen in pulmonary arterial hypertension (PAH). However, standard 2D cell culture models fail to mimic the mechanical environment seen in the pulmonary vasculature. Hydrogels have emerged as promising platforms for 3D disease modeling due to their tunable physical and biochemical properties. In order to recreate the mechanical stimuli seen in the pulmonary vasculature, we have created a novel 3D hydrogel-based pulmonary vasculature model (“artificial arteriole”) that reproduces the pulsatile flow rates and pressures seen in the human lung. Using this platform, we studied both Bmpr2R899X and WT endothelial cells to better understand how the addition of oscillatory flow and physiological pressure influenced gene expression, cell morphology, and cell permeability. The addition of oscillatory flow and pressure resulted in several gene expression changes in both WT and Bmpr2R899X cells. However, for many pathways with relevance to PAH etiology, Bmpr2R899X cells responded differently when compared to the WT cells. Bmpr2R899X cells were also found not to elongate in the direction of flow, and instead remained stagnant in morphology despite mechanical stimuli. The increased permeability of the Bmpr2R899X layer was successfully reproduced in our artificial arteriole, with the addition of flow and pressure not leading to significant changes in permeability. Our artificial arteriole is the first to model many mechanical properties seen in the lung. Its tunability enables several new opportunities to study the endothelium in pulmonary vascular disease with increased control over environmental parameters.
Collapse
|
26
|
Li W, Khan M, Mao S, Feng S, Lin JM. Advances in tumor-endothelial cells co-culture and interaction on microfluidics. J Pharm Anal 2018; 8:210-218. [PMID: 30140484 PMCID: PMC6104288 DOI: 10.1016/j.jpha.2018.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 01/10/2023] Open
Abstract
The metastasis in which the cancer cells degrade the extracellular matrix (ECM) and invade to the surrounding and far tissues of the body is the leading cause of mortality in cancer patients. With a lot of advancement in the field, yet the biological cause of metastasis are poorly understood. The microfluidic system provides advanced technology to reconstruct a variety of in vivo-like environment for studying the interactions between tumor cells (TCs) and endothelial cells (ECs). This review gives a brief account of both two-dimensional models and three-dimensional microfluidic systems for the analysis of TCs-ECs co-culture as well as their applications to anti-cancer drug screening. Furthermore, the advanced methods for analyzing cell-to-cell interactions at single-cell level were also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Jin-Ming Lin
- Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and Instrumentation, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
27
|
Merna N, Wong AK, Barahona V, Llanos P, Kunar B, Palikuqi B, Ginsberg M, Rafii S, Rabbany SY. Laminar shear stress modulates endothelial luminal surface stiffness in a tissue-specific manner. Microcirculation 2018; 25:e12455. [PMID: 29665185 DOI: 10.1111/micc.12455] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 04/09/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Endothelial cells form vascular beds in all organs and are exposed to a range of mechanical forces that regulate cellular phenotype. We sought to determine the role of endothelial luminal surface stiffness in tissue-specific mechanotransduction of laminar shear stress in microvascular mouse cells and the role of arachidonic acid in mediating this response. METHODS Microvascular mouse endothelial cells were subjected to laminar shear stress at 4 dynes/cm2 for 12 hours in parallel plate flow chambers that enabled real-time optical microscopy and atomic force microscopy measurements of cell stiffness. RESULTS Lung endothelial cells aligned parallel to flow, while cardiac endothelial cells did not. This rapid alignment was accompanied by increased cell stiffness. The addition of arachidonic acid to cardiac endothelial cells increased alignment and stiffness in response to shear stress. Inhibition of arachidonic acid in lung endothelial cells and embryonic stem cell-derived endothelial cells prevented cellular alignment and decreased cell stiffness. CONCLUSIONS Our findings suggest that increased endothelial luminal surface stiffness in microvascular cells may facilitate mechanotransduction and alignment in response to laminar shear stress. Furthermore, the arachidonic acid pathway may mediate this tissue-specific process. An improved understanding of this response will aid in the treatment of organ-specific vascular disease.
Collapse
Affiliation(s)
- Nick Merna
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Andrew K Wong
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Victor Barahona
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Pierre Llanos
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA
| | - Balvir Kunar
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| | - Brisa Palikuqi
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Shahin Rafii
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sina Y Rabbany
- Bioengineering Program, Fred DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, USA.,Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
28
|
Wang HJ, Lo WY. Identification of Basic Fibroblast Growth Factor as the Dominant Protector of Laminar Shear Medium from the Modified Shear Device in Tumor Necrosis Factor-α Induced Endothelial Dysfunction. Front Physiol 2018; 8:1095. [PMID: 29354066 PMCID: PMC5760543 DOI: 10.3389/fphys.2017.01095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
Background and Aims: Endothelial dysfunction is a hallmark of cardiovascular diseases. The straight region of an artery is protected from atherosclerosis via its laminar blood flow and high shear stress. This study investigated the cytoprotective effects of a new laminar shear medium (LSM) derived from a modified cone-and-plate shear device and identified basic fibroblast growth factor (bFGF) secreted by human aortic endothelial cells (HAECs) as the dominant protective factor in the LSM. Methods: Based on a modified cone-and-plate shear device system, HAECs were exposed to laminar shear (15 dynes/cm2) and static control for 24 h to produce a new supernatant LSM and static medium (SM). Evaluation of the protective effects of LSM and SM on endothelial dysfunction induced by tumor necrosis factor (TNF)-α (10 ng/mL), which leads to production of reactive oxygen species (ROS), inflammatory monocyte adhesion, and tissue factor activity. ROS induction-, inflammation-, and thrombosis-related genes and protein expression were evaluated by quantitative-PCR and western blotting. To identify the cytokines that played a key role in the cytoprotective action of the LSM, we used cytokine antibody arrays, selected an abundant marker cytokine, bFGF, and validated the different cytoprotective effects of recombinant bFGF (rbFGF) and neutralization by monoclonal antibody (rbFGF+Ab) co-treatment. Aortic and lung tissues from different groups of C57BL/6J mice were examined by immunohistochemistry. SB203580 (specific inhibitor of p38) and BIX02189 (specific inhibitor of MEK5) were used to identify bFGF as the main cytoprotective factor acting via p38/MAPK and MEK5-KLF2 pathways. Results: Compared with traditional LSM, the new LSM not only significantly decreased TNF-α-induced intracellular adhesion molecule 1 and plasminogen activator inhibitor type 1 gene expression, but also significantly increased heme oxygenase 1 gene expression. The new LSM and bFGF attenuated TNF-α-induced ROS induction, inflammation, and tissue factor activity and inhibited the inflammatory- and thrombosis-related gene/protein overexpression both in vitro and in vivo. Mechanistically, the cytoprotective action of bFGF was mediated via the p38/MAPK and MEK5-KLF2 pathways. Conclusion: bFGF was identified as the critical factor mediating the cytoprotective effects of LSM derived from the modified laminar shear system.
Collapse
Affiliation(s)
- Huang-Joe Wang
- Department of Internal Medicine, School of Medicine, China Medical University, Taichung, Taiwan.,Cardiovascular Research Laboratory, Division of Cardiovascular Medicine, Department of Internal Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Wan-Yu Lo
- Cardiovascular and Translational Medicine Laboratory, Department of Biotechnology, Hungkuang University, Taichung, Taiwan.,Bachelor Degree Program in Animal Healthcare, Hungkuang University, Taichung, Taiwan
| |
Collapse
|
29
|
Wang F, Jia J, Lal N, Zhang D, Chiu APL, Wan A, Vlodavsky I, Hussein B, Rodrigues B. High glucose facilitated endothelial heparanase transfer to the cardiomyocyte modifies its cell death signature. Cardiovasc Res 2017; 112:656-668. [PMID: 27979811 DOI: 10.1093/cvr/cvw211] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 09/21/2016] [Accepted: 09/16/2016] [Indexed: 01/26/2023] Open
Abstract
AIMS The secretion of enzymatically active heparanase (HepA) has been implicated as an essential metabolic adaptation in the heart following diabetes. However, the regulation and function of the enzymatically inactive heparanase (HepL) remain poorly understood. We hypothesized that in response to high glucose (HG) and secretion of HepL from the endothelial cell (EC), HepL uptake and function can protect the cardiomyocyte by modifying its cell death signature. METHODS AND RESULTS HG promoted both HepL and HepA secretion from microvascular (rat heart micro vessel endothelial cells, RHMEC) and macrovascular (rat aortic endothelial cells, RAOEC) EC. However, only RAOEC were capable of HepL reuptake. This occurred through a low-density lipoprotein receptor-related protein 1 (LRP1) dependent mechanism, as LRP1 inhibition using small interfering RNA (siRNA), receptor-associated protein, or an LRP1 neutralizing antibody significantly reduced uptake. In cardiomyocytes, which have a negligible amount of heparanase gene expression, LRP1 also participated in the uptake of HepL. Exogenous addition of HepL to rat cardiomyocytes produced a dramatically altered expression of apoptosis-related genes, and protection against HG and H2O2 induced cell death. Cardiomyocytes from acutely diabetic rats demonstrated a robust increase in LRP1 expression and levels of heparanase, a pro-survival gene signature, and limited evidence of cell death, observations that were not apparent following chronic and progressive diabetes. CONCLUSION Our results highlight EC-to-cardiomyocyte transfer of heparanase to modulate the cardiomyocyte cell death signature. This mechanism was observed in the acutely diabetic heart, and its interruption following chronic diabetes may contribute towards the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Fulong Wang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Jocelyn Jia
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Nathaniel Lal
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Dahai Zhang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Amy Pei-Ling Chiu
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Andrea Wan
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | - Bahira Hussein
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC, Canada V6T 1Z3
| |
Collapse
|
30
|
Acute effects of focused ultrasound-induced increases in blood-brain barrier permeability on rat microvascular transcriptome. Sci Rep 2017; 7:45657. [PMID: 28374753 PMCID: PMC5379491 DOI: 10.1038/srep45657] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/02/2017] [Indexed: 02/07/2023] Open
Abstract
Therapeutic treatment options for central nervous system diseases are greatly limited by the blood-brain barrier (BBB). Focused ultrasound (FUS), in conjunction with circulating microbubbles, can be used to induce a targeted and transient increase in BBB permeability, providing a unique approach for the delivery of drugs from the systemic circulation into the brain. While preclinical research has demonstrated the utility of FUS, there remains a large gap in our knowledge regarding the impact of sonication on BBB gene expression. This work is focused on investigating the transcriptional changes in dorsal hippocampal rat microvessels in the acute stages following sonication. Microarray analysis of microvessels was performed at 6 and 24 hrs post-FUS. Expression changes in individual genes and bioinformatic analysis suggests that FUS may induce a transient inflammatory response in microvessels. Increased transcription of proinflammatory cytokine genes appears to be short-lived, largely returning to baseline by 24 hrs. This observation may help to explain some previously observed bioeffects of FUS and may also be a driving force for the angiogenic processes and reduced drug efflux suggested by this work. While further studies are necessary, these results open up intriguing possibilities for novel FUS applications and suggest possible routes for pharmacologically modifying the technique.
Collapse
|
31
|
Mansilla Alvarez L, Blanco P, Bulant C, Dari E, Veneziani A, Feijóo R. Transversally enriched pipe element method (TEPEM): An effective numerical approach for blood flow modeling. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2017; 33:e2808. [PMID: 27302372 DOI: 10.1002/cnm.2808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 06/06/2023]
Abstract
In this work, we present a novel approach tailored to approximate the Navier-Stokes equations to simulate fluid flow in three-dimensional tubular domains of arbitrary cross-sectional shape. The proposed methodology is aimed at filling the gap between (cheap) one-dimensional and (expensive) three-dimensional models, featuring descriptive capabilities comparable with the full and accurate 3D description of the problem at a low computational cost. In addition, this methodology can easily be tuned or even adapted to address local features demanding more accuracy. The numerical strategy employs finite (pipe-type) elements that take advantage of the pipe structure of the spatial domain under analysis. While low order approximation is used for the longitudinal description of the physical fields, transverse approximation is enriched using high order polynomials. Although our application of interest is computational hemodynamics and its relevance to pathological dynamics like atherosclerosis, the approach is quite general and can be applied in any internal fluid dynamics problem in pipe-like domains. Numerical examples covering academic cases as well as patient-specific coronary arterial geometries demonstrate the potentialities of the developed methodology and its performance when compared against traditional finite element methods. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Luis Mansilla Alvarez
- National Laboratory for Scientific Computing, LNCC/MCTI, Av. Getúlio Vargas 333, 25651-075, Petrópolis, Brazil
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| | - Pablo Blanco
- National Laboratory for Scientific Computing, LNCC/MCTI, Av. Getúlio Vargas 333, 25651-075, Petrópolis, Brazil
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| | - Carlos Bulant
- National Laboratory for Scientific Computing, LNCC/MCTI, Av. Getúlio Vargas 333, 25651-075, Petrópolis, Brazil
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| | - Enzo Dari
- CONICET, Centro Atómico Bariloche-CNEA, Instituto Balseiro-UNCUYO, Av. Bustillo 9500, 8400, S.C. de Bariloche, Argentina
| | - Alessandro Veneziani
- Department of Mathematics and Computer Science, Emory University, Atlanta, GA, USA
| | - Raúl Feijóo
- National Laboratory for Scientific Computing, LNCC/MCTI, Av. Getúlio Vargas 333, 25651-075, Petrópolis, Brazil
- National Institute of Science and Technology in Medicine Assisted by Scientific Computing, INCT-MACC, Petrópolis, Brazil
| |
Collapse
|
32
|
Roth CC, Glickman RD, Martens SL, Echchgadda I, Beier HT, Barnes RA, Ibey BL. Adult human dermal fibroblasts exposed to nanosecond electrical pulses exhibit genetic biomarkers of mechanical stress. Biochem Biophys Rep 2017; 9:302-309. [PMID: 28956017 PMCID: PMC5614618 DOI: 10.1016/j.bbrep.2017.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 11/17/2016] [Accepted: 01/24/2017] [Indexed: 11/29/2022] Open
Abstract
Background Exposure of cells to very short (<1 µs) electric pulses in the megavolt/meter range have been shown to cause a multitude of effects, both physical and molecular in nature. Physically, nanosecond electrical pulses (nsEP) can cause disruption of the plasma membrane, cellular swelling, shrinking and blebbing. Molecularly, nsEP have been shown to activate signaling pathways, produce oxidative stress, stimulate hormone secretion and induce both apoptotic and necrotic death. We hypothesize that studying the genetic response of primary human dermal fibroblasts exposed to nsEP, will gain insight into the molecular mechanism(s) either activated directly by nsEP, or indirectly through electrophysiology interactions. Methods Microarray analysis in conjunction with quantitative real time polymerase chain reaction (qRT-PCR) was used to screen and validate genes selectively upregulated in response to nsEP exposure. Results Expression profiles of 486 genes were found to be significantly changed by nsEP exposure. 50% of the top 20 responding genes coded for proteins located in two distinct cellular locations, the plasma membrane and the nucleus. Further analysis of five of the top 20 upregulated genes indicated that the HDFa cells’ response to nsEP exposure included many elements of a mechanical stress response. Conclusions We found that several genes, some of which are mechanosensitive, were selectively upregulated due to nsEP exposure. This genetic response appears to be a primary response to the stimuli and not a secondary response to cellular swelling. General significance This work provides strong evidence that cells exposed to nsEP interpret the insult as a mechanical stress. Global gene expression analysis was performed on primary cells exposed to nsEP. The bioeffects of nsEP on adult human dermal fibroblasts were investigated. Microarray analysis suggests nsEP imparts a mechanical stress on cells. FOS, NR4A2, ITPKB, KLHL24, and SOD2 were upregulated in response to nsEP.
Collapse
Affiliation(s)
- Caleb C Roth
- University of Texas Health Science Center San Antonio, School of Medicine, Dept. of Radiological Sciences, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.,General Dynamics IT, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA.,Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory, Radio Frequency Bioeffects Branch, Bioeffects Division, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Randolph D Glickman
- University of Texas Health Science Center San Antonio, School of Medicine, Dept. of Ophthalmology, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Stacey L Martens
- Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory, Radio Frequency Bioeffects Branch, Bioeffects Division, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Ibtissam Echchgadda
- Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory, Radio Frequency Bioeffects Branch, Bioeffects Division, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Hope T Beier
- Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory, Optical Radiation Bioeffects Branch, Bioeffects Division, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Ronald A Barnes
- Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory, Radio Frequency Bioeffects Branch, Bioeffects Division, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| | - Bennett L Ibey
- Human Effectiveness Directorate, 711th Human Performance Wing, Air Force Research Laboratory, Radio Frequency Bioeffects Branch, Bioeffects Division, 4141 Petroleum Road, JBSA Fort Sam Houston, TX 78234, USA
| |
Collapse
|
33
|
Unthank JL, Sheridan KM, Dalsing MC. Collateral Growth in the Peripheral Circulation: A Review. Vasc Endovascular Surg 2016; 38:291-313. [PMID: 15306947 DOI: 10.1177/153857440403800401] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Arterial occlusive diseases are a major cause of morbidity and death in the United States. The enlargement of pre-existing vessels, which bypass the site of arterial occlusion, provide a natural way for the body to compensate for such obstructions. Individuals differ in their capacity to develop collateral vessels. In recent years much attention has been focused upon therapy to promote collateral development, primarily using individual growth factors. Such studies have had mixed results. Persistent controversies exist regarding the initiating stimuli, the processes involved in enlargement, the specific vessels that should be targeted, and the most appropriate terminology. Consequently, it is now recognized that more research is needed to extend our knowledge of the complex process of collateral growth. This basic science review addresses five questions essential in understanding current problems in collateral growth research and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Joseph L Unthank
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | |
Collapse
|
34
|
Qiao C, Meng F, Jang I, Jo H, Chen YE, Zhang J. Deep transcriptomic profiling reveals the similarity between endothelial cells cultured under static and oscillatory shear stress conditions. Physiol Genomics 2016; 48:660-6. [PMID: 27449656 DOI: 10.1152/physiolgenomics.00025.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 07/17/2016] [Indexed: 01/26/2023] Open
Abstract
Atherosclerosis is a multifactorial disease that preferentially develops in specific regions in the arterial tree. This characteristic is mainly attributed to the unique pattern of hemodynamic shear stress in vivo. High laminar shear stress (LS) found in straight lumen exerts athero-protective effects. Low or oscillatory shear stress (OS) present in regions of lesser curvature and arterial bifurcations predisposes arterial intima to atherosclerosis. Shear stress-regulated endothelial function plays an important role in the process of atherosclerosis. Most in vitro research studies focusing on the molecular mechanisms of endothelial function are performed in endothelial cells (ECs) under cultured static (ST) condition. Some findings, however, are not recapitulated in subsequent translational studies, mostly likely due to the missing biomechanical milieu. Here, we profiled the whole transcriptome of primary human coronary arterial endothelial cells (HCAECs) under different shear stress conditions with RNA sequencing. Among 16,313 well-expressed genes, we detected 8,177 that were differentially expressed in OS vs. LS conditions and 9,369 in ST vs. LS conditions. Notably, only 1,618 were differentially expressed in OS vs. ST conditions. Hierarchical clustering of ECs demonstrated a strong similarity between ECs under OS and ST conditions at the transcriptome level. Subsequent pairwise heat mapping and principal component analysis gave further weight to the similarity. At the individual gene level, expressional analysis of representative well-known genes as well as novel genes showed a comparable amount at mRNA and protein levels in ECs under ST and OS conditions. In conclusion, the present work compared the whole transcriptome of HCAECs under different shear stress conditions at the transcriptome level as well as at the individual gene level. We found that cultured ECs are significantly different from those under LS conditions. Thus using cells under ST conditions is unlikely to elucidate endothelial physiology. Given the revealed high similarities of the endothelial transcriptome under OS and ST conditions, it may be helpful to understand the underlying mechanisms of OS-induced endothelial dysfunction from static cultured endothelial studies.
Collapse
Affiliation(s)
- Congzhen Qiao
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan
| | - Fan Meng
- Department of Psychiatry and Molecular and Behavioral Neuroscience Institute, University of Michigan Medical Center, Ann Arbor, Michigan; and
| | - Inhwan Jang
- Wallace H. Coulter Department of Biomedical Engineering, Division of Cardiology, Department of Medicine, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Division of Cardiology, Department of Medicine, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Y Eugene Chen
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan
| | - Jifeng Zhang
- Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan;
| |
Collapse
|
35
|
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
|
36
|
Simmons RD, Kumar S, Thabet SR, Sur S, Jo H. Omics-based approaches to understand mechanosensitive endothelial biology and atherosclerosis. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:378-401. [PMID: 27341633 DOI: 10.1002/wsbm.1344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is a multifactorial disease that preferentially occurs in arterial regions exposed to d-flow can be used to indicate disturbed flow or disturbed blood flow. The mechanisms by which d-flow induces atherosclerosis involve changes in the transcriptome, methylome, proteome, and metabolome of multiple vascular cells, especially endothelial cells. Initially, we begin with the pathogenesis of atherosclerosis and the changes that occur at multiple levels owing to d-flow, especially in the endothelium. Also, there are a variety of strategies used for the global profiling of the genome, transcriptome, miRNA-ome, DNA methylome, and metabolome that are important to define the biological and pathophysiological mechanisms of endothelial dysfunction and atherosclerosis. Finally, systems biology can be used to integrate these 'omics' datasets, especially those that derive data based on a single animal model, in order to better understand the pathophysiology of atherosclerosis development in a holistic manner and how this integrative approach could be used to identify novel molecular diagnostics and therapeutic targets to prevent or treat atherosclerosis. WIREs Syst Biol Med 2016, 8:378-401. doi: 10.1002/wsbm.1344 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Rachel D Simmons
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandeep Kumar
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Salim Raid Thabet
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sanjoli Sur
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hanjoong Jo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
37
|
Evaluation of the Genetic Response of U937 and Jurkat Cells to 10-Nanosecond Electrical Pulses (nsEP). PLoS One 2016; 11:e0154555. [PMID: 27135944 PMCID: PMC4852903 DOI: 10.1371/journal.pone.0154555] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/21/2016] [Indexed: 12/21/2022] Open
Abstract
Nanosecond electrical pulse (nsEP) exposure activates signaling pathways, produces oxidative stress, stimulates hormone secretion, causes cell swelling and induces apoptotic and necrotic death. The underlying biophysical connection(s) between these diverse cellular reactions and nsEP has yet to be elucidated. Using global genetic analysis, we evaluated how two commonly studied cell types, U937 and Jurkat, respond to nsEP exposure. We hypothesized that by studying the genetic response of the cells following exposure, we would gain direct insight into the stresses experienced by the cell and in turn better understand the biophysical interaction taking place during the exposure. Using Ingenuity Systems software, we found genes associated with cell growth, movement and development to be significantly up-regulated in both cell types 4 h post exposure to nsEP. In agreement with our hypothesis, we also found that both cell lines exhibit significant biological changes consistent with mechanical stress induction. These results advance nsEP research by providing strong evidence that the interaction of nsEPs with cells involves mechanical stress.
Collapse
|
38
|
Tian Y, Gawlak G, O'Donnell JJ, Mambetsariev I, Birukova AA. Modulation of Endothelial Inflammation by Low and High Magnitude Cyclic Stretch. PLoS One 2016; 11:e0153387. [PMID: 27128976 PMCID: PMC4851399 DOI: 10.1371/journal.pone.0153387] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 03/29/2016] [Indexed: 11/29/2022] Open
Abstract
Excessive mechanical ventilation exerts pathologic mechanical strain on lung vascular endothelium and promotes endothelial cell (EC) inflammatory activation; however, the specific mechanisms underlying EC inflammatory response caused by mechanical ventilation related cyclic stretch (CS) remain unclear. This study investigated the effects of chronic exposure to CS at physiologic (5%) and pathologic (18%) magnitude on pulmonary EC inflammatory status in control conditions and bacterial lipopolysacharide (LPS)-stimulated conditions. EC exposure to high or low CS magnitudes for 28–72 hrs had distinct effects on EC inflammatory activation. 18% CS increased surface expression of endothelial adhesion molecule ICAM1 and release of its soluble form (sICAM1) and inflammatory cytokine IL-8 by CS-stimulated pulmonary endothelial cells (EC). EC inflammatory activation was not observed in EC exposed to 5% CS. Chronic exposure to 18% CS, but not to 5% CS, augmented ICAM1 and IL-8 production and EC monolayer barrier disruption induced by LPS. 18% CS, but not 5% CS, stimulated expression of RhoA GTPase-specific guanine nucleotide exchange factor GEF-H1. GEF-H1 knockdown using gene-specific siRNA abolished 18% CS-induced ICAM1 expression and sICAM1 and IL-8 release by EC. GEF-H1 knockdown also prevented disruption of EC monolayer integrity and attenuated sICAM1 and IL-8 release in the two-hit model of EC barrier dysfunction caused by combined stimulation with 18% CS and LPS. These data demonstrate that exacerbation of inflammatory response by pulmonary endothelium exposed to excessive mechanical stretch is mediated by CS-induced induction of Rho activating protein GEF-H1.
Collapse
Affiliation(s)
- Yufeng Tian
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, United States of America
| | - Grzegorz Gawlak
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, United States of America
| | - James J. O'Donnell
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, United States of America
| | - Isa Mambetsariev
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, United States of America
| | - Anna A. Birukova
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, Illinois 60637, United States of America
- * E-mail:
| |
Collapse
|
39
|
Childs PG, Boyle CA, Pemberton GD, Nikukar H, Curtis AS, Henriquez FL, Dalby MJ, Reid S. Use of nanoscale mechanical stimulation for control and manipulation of cell behaviour. Acta Biomater 2016; 34:159-168. [PMID: 26612418 DOI: 10.1016/j.actbio.2015.11.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/25/2015] [Accepted: 11/19/2015] [Indexed: 02/06/2023]
Abstract
The ability to control cell behaviour, cell fate and simulate reliable tissue models in vitro remains a significant challenge yet is crucial for various applications of high throughput screening e.g. drug discovery. Mechanotransduction (the ability of cells to convert mechanical forces in their environment to biochemical signalling) represents an alternative mechanism to attain this control with such studies developing techniques to reproducibly control the mechanical environment in techniques which have potential to be scaled. In this review, the use of techniques such as finite element modelling and precision interferometric measurement are examined to provide context for a novel technique based on nanoscale vibration, also known as "nanokicking". Studies have shown this stimulus to alter cellular responses in both endothelial and mesenchymal stem cells (MSCs), particularly in increased proliferation rate and induced osteogenesis respectively. Endothelial cell lines were exposed to nanoscale vibration amplitudes across a frequency range of 1-100 Hz, and MSCs primarily at 1 kHz. This technique provides significant potential benefits over existing technologies, as cellular responses can be initiated without the use of expensive engineering techniques and/or chemical induction factors. Due to the reproducible and scalable nature of the apparatus it is conceivable that nanokicking could be used for controlling cell behaviour within a wide array of high throughput procedures in the research environment, within drug discovery, and for clinical/therapeutic applications. STATEMENT OF SIGNIFICANCE The results discussed within this article summarise the potential benefits of using nanoscale vibration protocols for controlling cell behaviour. There is a significant need for reliable tissue models within the clinical and pharma industries, and the control of cell behaviour and stem cell differentiation would be highly beneficial. The full potential of this method of controlling cell behaviour has not yet been realised.
Collapse
|
40
|
Berg C. Quantitative analysis of nanoparticle transport through in vitro blood-brain barrier models. Tissue Barriers 2016; 4:e1143545. [PMID: 27141425 PMCID: PMC4836482 DOI: 10.1080/21688370.2016.1143545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/08/2016] [Accepted: 01/12/2016] [Indexed: 01/11/2023] Open
Abstract
Nanoparticle transport through the blood-brain barrier has received much attention of late, both from the point of view of nano-enabled drug delivery, as well as due to concerns about unintended exposure of nanomaterials to humans and other organisms. In vitro models play a lead role in efforts to understand the extent of transport through the blood-brain barrier, but unique features of the nanoscale challenge their direct adaptation. Here we highlight some of the differences compared to molecular species when utilizing in vitro blood-brain barrier models for nanoparticle studies. Issues that may arise with transwell systems are discussed, together with some potential alternative methodologies. We also briefly review the biomolecular corona concept and its importance for how nanoparticles interact with the blood-brain barrier. We end with considering future directions, including indirect effects and application of shear and fluidics-technologies.
Collapse
Affiliation(s)
- Christoffer Berg
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen ; Groningen, The Netherlands
| |
Collapse
|
41
|
Endothelial Plasticity: Shifting Phenotypes through Force Feedback. Stem Cells Int 2016; 2016:9762959. [PMID: 26904133 PMCID: PMC4745942 DOI: 10.1155/2016/9762959] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/31/2015] [Indexed: 12/28/2022] Open
Abstract
The endothelial lining of the vasculature is exposed to a large variety of biochemical and hemodynamic stimuli with different gradients throughout the vascular network. Adequate adaptation requires endothelial cells to be highly plastic, which is reflected by the remarkable heterogeneity of endothelial cells in tissues and organs. Hemodynamic forces such as fluid shear stress and cyclic strain are strong modulators of the endothelial phenotype and function. Although endothelial plasticity is essential during development and adult physiology, proatherogenic stimuli can induce adverse plasticity which contributes to disease. Endothelial-to-mesenchymal transition (EndMT), the hallmark of endothelial plasticity, was long thought to be restricted to embryonic development but has emerged as a pathologic process in a plethora of diseases. In this perspective we argue how shear stress and cyclic strain can modulate EndMT and discuss how this is reflected in atherosclerosis and pulmonary arterial hypertension.
Collapse
|
42
|
Lee WJ, Jung KH, Lee KJ, Kim JM, Lee ST, Chu K, Lee SK, Roh JK. Sonographic findings associated with stenosis progression and vascular complications in moyamoya disease. J Neurosurg 2015; 125:689-97. [PMID: 26654185 DOI: 10.3171/2015.6.jns15933] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT The progression of arterial stenosis in patients with moyamoya disease (MMD) has variable courses and an unclear mechanism. The authors hypothesized that elevated wall shear stress (WSS) at the terminal internal carotid artery (ICA) and proximal middle cerebral artery (MCA) may facilitate MMD progression. They indirectly evaluated the relative magnitude of WSS (WSS value [WSSV]) with MR angiography (MRA) and transcranial Doppler to determine its predictive value for stenosis progression (SP) and the development of vascular complications. METHODS Thirty-one medically treated patients (58 hemispheres and 95 nonoccluded vessels) were analyzed with serial MRA (median follow-up 23 months). The parameters studied were SP, SP rates (SPRs) for individual ICAs/MCAs, and their mean values from the ipsilateral hemispheres as mean SP (MSP) and MSP rates (MSPRs). Significant progression was defined as decrements of ≥ 20% for SP and MSP and ≥ 10%/year for SPR and MSPR. The development of vascular complications in relevant hemispheres was also recorded. The WSSV (dyne/cm(2)) was defined as the shear rate multiplied by blood viscosity. RESULTS After adjusting the initial stenosis degree and MRA stage of MMD, an SP of ≥ 20% and an SPR of ≥ 10%/year were associated with the highest-quartile WSSVs for all individual vessels and for MCAs and ICAs separately. For each hemisphere, an MSP of ≥ 20% and an MSPR of ≥ 10%/year were associated with the highest-quartile mean WSSVs. Furthermore, significant SP was highly correlated with vascular complications, and the highest-quartile mean WSSV was independently associated with vascular complications in relevant hemispheres. CONCLUSIONS An elevated WSSV is an independent predictor for SP and vascular complications in nonoccluded MMD.
Collapse
Affiliation(s)
- Woo-Jin Lee
- Department of Neurology, Seoul National University Hospital
| | - Keun-Hwa Jung
- Department of Neurology, Seoul National University Hospital;,Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University
| | - Keon-Joo Lee
- Department of Neurology, Seoul National University Hospital
| | - Jeong-Min Kim
- Department of Neurology, Chung-Ang University Hospital, Seoul; and
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital;,Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University
| | - Kon Chu
- Department of Neurology, Seoul National University Hospital;,Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital;,Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University
| | - Jae-Kyu Roh
- Department of Neurology, Seoul National University Hospital;,Department of Neurology, The Armed Forces Capital Hospital, Sungnam, South Korea
| |
Collapse
|
43
|
The role of endothelial mechanosensitive genes in atherosclerosis and omics approaches. Arch Biochem Biophys 2015; 591:111-31. [PMID: 26686737 DOI: 10.1016/j.abb.2015.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/29/2015] [Accepted: 11/04/2015] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is the leading cause of morbidity and mortality in the U.S., and is a multifactorial disease that preferentially occurs in regions of the arterial tree exposed to disturbed blood flow. The detailed mechanisms by which d-flow induces atherosclerosis involve changes in the expression of genes, epigenetic patterns, and metabolites of multiple vascular cells, especially endothelial cells. This review presents an overview of endothelial mechanobiology and its relation to the pathogenesis of atherosclerosis with special reference to the anatomy of the artery and the underlying fluid mechanics, followed by a discussion of a variety of experimental models to study the role of fluid mechanics and atherosclerosis. Various in vitro and in vivo models to study the role of flow in endothelial biology and pathobiology are discussed in this review. Furthermore, strategies used for the global profiling of the genome, transcriptome, miR-nome, DNA methylome, and metabolome, as they are important to define the biological and pathophysiological mechanisms of atherosclerosis. These "omics" approaches, especially those which derive data based on a single animal model, provide unprecedented opportunities to not only better understand the pathophysiology of atherosclerosis development in a holistic and integrative manner, but also to identify novel molecular and diagnostic targets.
Collapse
|
44
|
Buchanan CF, Verbridge SS, Vlachos PP, Rylander MN. Flow shear stress regulates endothelial barrier function and expression of angiogenic factors in a 3D microfluidic tumor vascular model. Cell Adh Migr 2015; 8:517-24. [PMID: 25482628 DOI: 10.4161/19336918.2014.970001] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Endothelial cells lining blood vessels are exposed to various hemodynamic forces associated with blood flow. These include fluid shear, the tangential force derived from the friction of blood flowing across the luminal cell surface, tensile stress due to deformation of the vessel wall by transvascular flow, and normal stress caused by the hydrodynamic pressure differential across the vessel wall. While it is well known that these fluid forces induce changes in endothelial morphology, cytoskeletal remodeling, and altered gene expression, the effect of flow on endothelial organization within the context of the tumor microenvironment is largely unknown. Using a previously established microfluidic tumor vascular model, the objective of this study was to investigate the effect of normal (4 dyn/cm(2)), low (1 dyn/cm(2)), and high (10 dyn/cm(2)) microvascular wall shear stress (WSS) on tumor-endothelial paracrine signaling associated with angiogenesis. It is hypothesized that high WSS will alter the endothelial phenotype such that vascular permeability and tumor-expressed angiogenic factors are reduced. Results demonstrate that endothelial permeability decreases as a function of increasing WSS, while co-culture with tumor cells increases permeability relative to mono-cultures. This response is likely due to shear stress-mediated endothelial cell alignment and tumor-VEGF-induced permeability. In addition, gene expression analysis revealed that high WSS (10 dyn/cm(2)) significantly down-regulates tumor-expressed MMP9, HIF1, VEGFA, ANG1, and ANG2, all of which are important factors implicated in tumor angiogenesis. This result was not observed in tumor mono-cultures or static conditioned media experiments, suggesting a flow-mediated paracrine signaling mechanism exists with surrounding tumor cells that elicits a change in expression of angiogenic factors. Findings from this work have significant implications regarding low blood velocities commonly seen in the tumor vasculature, suggesting high shear stress-regulation of angiogenic activity is lacking in many vessels, thereby driving tumor angiogenesis.
Collapse
Affiliation(s)
- Cara F Buchanan
- a Virginia Tech - Wake Forest University , School of Biomedical Engineering and Sciences; Virginia Tech ; Blacksburg , VA USA
| | | | | | | |
Collapse
|
45
|
Brask JB, Singla-Buxarrais G, Uroz M, Vincent R, Trepat X. Compressed sensing traction force microscopy. Acta Biomater 2015; 26:286-94. [PMID: 26300336 DOI: 10.1016/j.actbio.2015.08.023] [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] [Received: 04/10/2015] [Revised: 07/30/2015] [Accepted: 08/18/2015] [Indexed: 01/22/2023]
Abstract
Adherent cells exert traction forces on their substrate, and these forces play important roles in biological functions such as mechanosensing, cell differentiation and cancer invasion. The method of choice to assess these active forces is traction force microscopy (TFM). Despite recent advances, TFM remains highly sensitive to measurement noise and exhibits limited spatial resolution. To improve the resolution and noise robustness of TFM, here we adapt techniques from compressed sensing (CS) to the reconstruction of the traction field from the substrate displacement field. CS enables the recovery of sparse signals at higher resolution from lower resolution data. Focal adhesions (FAs) of adherent cells are spatially sparse implying that traction fields are also sparse. Here we show, by simulation and by experiment, that the CS approach enables circumventing the Nyquist-Shannon sampling theorem to faithfully reconstruct the traction field at a higher resolution than that of the displacement field. This allows reaching state-of-the-art resolution using only a medium magnification objective. We also find that CS improves reconstruction quality in the presence of noise. STATEMENT OF SIGNIFICANCE A great scientific advance of the past decade is the recognition that physical forces determine an increasing list of biological processes. Traction force microscopy which measures the forces that cells exert on their surroundings has seen significant recent improvements, however the technique remains sensitive to measurement noise and severely limited in spatial resolution. We exploit the fact that the force fields are sparse to boost the spatial resolution and noise robustness by applying ideas from compressed sensing. The novel method allows high resolution on a larger field of view. This may in turn allow better understanding of the cell forces at the multicellular level, which are known to be important in wound healing and cancer invasion.
Collapse
|
46
|
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
|
47
|
Dunn J, Thabet S, Jo H. Flow-Dependent Epigenetic DNA Methylation in Endothelial Gene Expression and Atherosclerosis. Arterioscler Thromb Vasc Biol 2015; 35:1562-9. [PMID: 25953647 DOI: 10.1161/atvbaha.115.305042] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/27/2015] [Indexed: 12/31/2022]
Abstract
Epigenetic mechanisms that regulate endothelial cell gene expression are now emerging. DNA methylation is the most stable epigenetic mark that confers persisting changes in gene expression. Not only is DNA methylation important in rendering cell identity by regulating cell type-specific gene expression throughout differentiation, but it is becoming clear that DNA methylation also plays a key role in maintaining endothelial cell homeostasis and in vascular disease development. Disturbed blood flow causes atherosclerosis, whereas stable flow protects against it by differentially regulating gene expression in endothelial cells. Recently, we and others have shown that flow-dependent gene expression and atherosclerosis development are regulated by mechanisms dependent on DNA methyltransferases (1 and 3A). Disturbed blood flow upregulates DNA methyltransferase expression both in vitro and in vivo, which leads to genome-wide DNA methylation alterations and global gene expression changes in a DNA methyltransferase-dependent manner. These studies revealed several mechanosensitive genes, such as HoxA5, Klf3, and Klf4, whose promoters were hypermethylated by disturbed blood flow, but rescued by DNA methyltransferases inhibitors such as 5Aza-2-deoxycytidine. These findings provide new insight into the mechanism by which flow controls epigenomic DNA methylation patterns, which in turn alters endothelial gene expression, regulates vascular biology, and modulates atherosclerosis development.
Collapse
Affiliation(s)
- Jessilyn Dunn
- From the Wallace H. Coulter Department of Biomedical Engineering (J.D., S.T., H.J.) and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta
| | - Salim Thabet
- From the Wallace H. Coulter Department of Biomedical Engineering (J.D., S.T., H.J.) and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta
| | - Hanjoong Jo
- From the Wallace H. Coulter Department of Biomedical Engineering (J.D., S.T., H.J.) and Division of Cardiology, Georgia Institute of Technology and Emory University, Atlanta.
| |
Collapse
|
48
|
Ehnes DD, Price FD, Shrive NG, Hart DA, Rancourt DE, zur Nieden NI. Embryonic stem cell-derived osteocytes are capable of responding to mechanical oscillatory hydrostatic pressure. J Biomech 2015; 48:1915-21. [PMID: 25936968 DOI: 10.1016/j.jbiomech.2015.04.015] [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] [Received: 01/07/2015] [Revised: 04/04/2015] [Accepted: 04/08/2015] [Indexed: 11/28/2022]
Abstract
Osteoblasts can be derived from embryonic stem cells (ESCs) by a 30 day differentiation process, whereupon cells spontaneously differentiate upon removal of LIF and respond to exogenously added 1,25α(OH)2 vitamin D3 with enhanced matrix mineralization. However, bone is a load-bearing tissue that has to perform under dynamic pressure changes during daily movement, a capacity that is executed by osteocytes. At present, it is unclear whether ESC-derived osteogenic cultures contain osteocytes and whether these are capable of responding to a relevant cyclic hydrostatic compression stimulus. Here, we show that ESC-osteoblastogenesis is followed by the generation of osteocytes and then mechanically load ESC-derived osteogenic cultures in a compression chamber using a cyclic loading protocol. Following mechanical loading of the cells, iNOS mRNA was upregulated 31-fold, which was consistent with a role for iNOS as an immediate early mechanoresponsive gene. Further analysis of matrix and bone-specific genes suggested a cellular response in favor of matrix remodeling. Immediate iNOS upregulation also correlated with a concomitant increase in Ctnnb1 and Tcf7l2 mRNAs along with increased nuclear TCF transcriptional activity, while the mRNA for the repressive Tcf7l1 was downregulated, providing a possible mechanistic explanation for the noted matrix remodeling. We conclude that ESC-derived osteocytes are capable of responding to relevant mechanical cues, at least such that mimic oscillatory compression stress, which not only provides new basic understanding, but also information that likely will be important for their use in cell-based regenerative therapies.
Collapse
Affiliation(s)
- D D Ehnes
- University of California Riverside, Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, 1113 Biological Sciences Building, Riverside, CA 92521, USA
| | - F D Price
- The Alberta Children's Hospital Research Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - N G Shrive
- McCaig Institute for Bone and Joint Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - D A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - D E Rancourt
- The Alberta Children's Hospital Research Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1
| | - N I zur Nieden
- University of California Riverside, Department of Cell Biology & Neuroscience and Stem Cell Center, College of Natural and Agricultural Sciences, 1113 Biological Sciences Building, Riverside, CA 92521, USA; The Alberta Children's Hospital Research Institute, University of Calgary, Heritage Medical Research Building, 3330 Hospital Drive NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|
49
|
Teng D, Pannell JS, Rennert RC, Li J, Li YS, Wong VW, Chien S, Khalessi AA. Endothelial Trauma From Mechanical Thrombectomy in Acute Stroke. Stroke 2015; 46:1099-106. [PMID: 25712942 DOI: 10.1161/strokeaha.114.007494] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Dayu Teng
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Jeffrey Scott Pannell
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Robert C. Rennert
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Jieying Li
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Yi-Shuan Li
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Victor W. Wong
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Shu Chien
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| | - Alexander A. Khalessi
- From the Division of Neurosurgery (J.S.P., R.C.R., V.W.W., A.A.K.), Department of Bioengineering and Institute of Engineering in Medicine (D.T., J.L., Y.-S., S.C.), University of California, San Diego
| |
Collapse
|
50
|
Jeon H, Tsui JH, Jang SI, Lee JH, Park S, Mun K, Boo YC, Kim DH. Combined effects of substrate topography and stiffness on endothelial cytokine and chemokine secretion. ACS APPLIED MATERIALS & INTERFACES 2015; 7:4525-4532. [PMID: 25658848 PMCID: PMC4937831 DOI: 10.1021/acsami.5b00554] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Endothelial physiology is regulated not only by humoral factors, but also by mechanical factors such as fluid shear stress and the underlying cellular matrix microenvironment. The purpose of the present study was to examine the effects of matrix topographical cues on the endothelial secretion of cytokines/chemokines in vitro. Human endothelial cells were cultured on nanopatterned polymeric substrates with different ratios of ridge to groove widths (1:1, 1:2, and 1:5) and with different stiffnesses (6.7 MPa and 2.5 GPa) in the presence and absence of 1.0 ng/mL TNF-α. The levels of cytokines/chemokines secreted into the conditioned media were analyzed with a multiplexed bead-based sandwich immunoassay. Of the nanopatterns tested, the 1:1 and 1:2 type patterns were found to induce the greatest degree of endothelial cell elongation and directional alignment. The 1:2 type nanopatterns lowered the secretion of inflammatory cytokines such as IL-1β, IL-3, and MCP-1, compared to unpatterned substrates. Additionally, of the two polymers tested, it was found that the stiffer substrate resulted in significant decreases in the secretion of IL-3 and MCP-1. These results suggest that substrates with specific extracellular nanotopographical cues or stiffnesses may provide anti-atherogenic effects like those seen with laminar shear stresses by suppressing the endothelial secretion of cytokines and chemokines involved in vascular inflammation and remodeling.
Collapse
Affiliation(s)
- Hyeona Jeon
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, 700-422, Republic of Korea
| | - Jonathan H. Tsui
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Sue Im Jang
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, 700-422, Republic of Korea
| | - Justin H. Lee
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Soojin Park
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, 700-422, Republic of Korea
| | - Kevin Mun
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Yong Chool Boo
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu, 700-422, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, 700-422, Republic of Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|