651
|
Baldwin WM, Halushka MK, Valujskikh A, Fairchild RL. B cells in cardiac transplants: from clinical questions to experimental models. Semin Immunol 2011; 24:122-30. [PMID: 21937238 DOI: 10.1016/j.smim.2011.08.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 08/24/2011] [Indexed: 12/31/2022]
Abstract
After many years of debate, there is now general agreement that B cells can participate in the immune response to cardiac transplants. Acute antibody-mediated rejection (AMR) is the best defined manifestation of B cell responses, but diagnostic and mechanistic questions still surround AMR. Many complement dependent mechanisms of antibody-mediated injury have been elucidated. C5 has become a therapeutic target that may not just truncate complement activation, but also may tip the balance away from inflammation by altering macrophage function. Additional complement independent effects have been identified. These may escape diagnosis and progress to chronic graft injury. The function of B cell infiltrates in cardiac transplants is even more enigmatic. Nodular endocardial infiltrates that contain B cells and plasma cells have been described in protocol biopsies of cardiac transplants for decades, but an understanding of their significance is still evolving based on more critical morphological and molecular evaluation of these infiltrates. A range of infiltrates containing B cells has also been described in the epicardial fat in transplants with advanced chronic rejection. B cells have been observed in endocardial and epicardial tertiary lymphoid nodules, but their impact on antigen presentation or antibody production remains to be determined. Experimental models in small and large animals suggest that B cells could be essential for the formation of lymphoid nodules through cytokine production. Similarly, the role of proinflammatory adipokines in the formation or function of epicardial lymphoid nodules has not been studied. These clinical observations provide critical questions to be addressed in experimental models.
Collapse
Affiliation(s)
- William M Baldwin
- Department of Immunology and the Glickman Urological and Kidney Disease Institute, The Cleveland Clinic, Cleveland, OH, USA.
| | | | | | | |
Collapse
|
652
|
Raines SM, Richards OC, Schneider LR, Schueler KL, Rabaglia ME, Oler AT, Stapleton DS, Genové G, Dawson JA, Betsholtz C, Attie AD. Loss of PDGF-B activity increases hepatic vascular permeability and enhances insulin sensitivity. Am J Physiol Endocrinol Metab 2011; 301:E517-26. [PMID: 21673305 PMCID: PMC3174531 DOI: 10.1152/ajpendo.00241.2011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hepatic vasculature is not thought to pose a permeability barrier for diffusion of macromolecules from the bloodstream to hepatocytes. In contrast, in extrahepatic tissues, the microvasculature is critically important for insulin action, because transport of insulin across the endothelial cell layer is rate limiting for insulin-stimulated glucose disposal. However, very little is known concerning the role in this process of pericytes, the mural cells lining the basolateral membrane of endothelial cells. PDGF-B is a growth factor involved in the recruitment and function of pericytes. We studied insulin action in mice expressing PDGF-B lacking the proteoglycan binding domain, producing a protein with a partial loss of function (PDGF-B(ret/ret)). Insulin action was assessed through measurements of insulin signaling and insulin and glucose tolerance tests. PDGF-B deficiency enhanced hepatic vascular transendothelial transport. One outcome of this change was an increase in hepatic insulin signaling. This correlated with enhanced whole body glucose homeostasis and increased insulin clearance from the circulation during an insulin tolerance test. In obese mice, PDGF-B deficiency was associated with an 80% reduction in fasting insulin and drastically reduced insulin secretion. These mice did not have significantly higher glucose levels, reflecting a dramatic increase in insulin action. Our findings show that, despite already having a high permeability, hepatic transendothelial transport can be further enhanced. To the best of our knowledge, this is the first study to connect PDGF-B-induced changes in hepatic sinusoidal transport to changes in insulin action, demonstrating a link between PDGF-B signaling and insulin sensitivity.
Collapse
|
653
|
Atkins GB, Jain MK, Hamik A. Endothelial differentiation: molecular mechanisms of specification and heterogeneity. Arterioscler Thromb Vasc Biol 2011; 31:1476-84. [PMID: 21677290 DOI: 10.1161/atvbaha.111.228999] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
A complex and diverse vascular system is requisite for the survival of higher organisms. The process of vascular development is highly regulated, involving the de novo formation of vessels (vasculogenesis), followed by expansion and remodeling of the primitive vasculature (angiogenesis), culminating in differentiation of endothelial phenotypes, as found in the mature vascular system. Over the last decade, significant advances have been made in understanding the molecular regulation of endothelial cell development and differentiation. Endothelial development, in particular the mechanisms in play during vasculogenesis and angiogenesis, is discussed in a sister review to this article. This review highlights the key pathways governing in endothelial differentiation, with a focus on the major molecular mechanisms of endothelial specification and heterogeneity.
Collapse
Affiliation(s)
- G Brandon Atkins
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University School of Medicine, University Hospitals Case Medical Center, Harrington-McLaughlin Heart and Vascular Institute, Cleveland, OH, USA
| | | | | |
Collapse
|
654
|
van der Heijden M, van Nieuw Amerongen GP, van Bezu J, Paul MA, Groeneveld ABJ, van Hinsbergh VWM. Opposing effects of the angiopoietins on the thrombin-induced permeability of human pulmonary microvascular endothelial cells. PLoS One 2011; 6:e23448. [PMID: 21858121 PMCID: PMC3156229 DOI: 10.1371/journal.pone.0023448] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 07/18/2011] [Indexed: 01/25/2023] Open
Abstract
Background Angiopoietin-2 (Ang-2) is associated with lung injury in ALI/ARDS. As endothelial activation by thrombin plays a role in the permeability of acute lung injury and Ang-2 may modulate the kinetics of thrombin-induced permeability by impairing the organization of vascular endothelial (VE-)cadherin, and affecting small Rho GTPases in human pulmonary microvascular endothelial cells (HPMVECs), we hypothesized that Ang-2 acts as a sensitizer of thrombin-induced hyperpermeability of HPMVECs, opposed by Ang-1. Methodology/Principal Findings Permeability was assessed by measuring macromolecule passage and transendothelial electrical resistance (TEER). Angiopoietins did not affect basal permeability. Nevertheless, they had opposing effects on the thrombin-induced permeability, in particular in the initial phase. Ang-2 enhanced the initial permeability increase (passage, P = 0.010; TEER, P = 0.021) in parallel with impairment of VE-cadherin organization without affecting VE-cadherin Tyr685 phosphorylation or increasing RhoA activity. Ang-2 also increased intercellular gap formation. Ang-1 preincubation increased Rac1 activity, enforced the VE-cadherin organization, reduced the initial thrombin-induced permeability (TEER, P = 0.027), while Rac1 activity simultaneously normalized, and reduced RhoA activity at 15 min thrombin exposure (P = 0.039), but not at earlier time points. The simultaneous presence of Ang-2 largely prevented the effect of Ang-1 on TEER and macromolecule passage. Conclusions/Significance Ang-1 attenuated thrombin-induced permeability, which involved initial Rac1 activation-enforced cell-cell junctions, and later RhoA inhibition. In addition to antagonizing Ang-1, Ang-2 had also a direct effect itself. Ang-2 sensitized the initial thrombin-induced permeability accompanied by destabilization of VE-cadherin junctions and increased gap formation, in the absence of increased RhoA activity.
Collapse
Affiliation(s)
- Melanie van der Heijden
- Department of Intensive Care, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - Geerten P. van Nieuw Amerongen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
- * E-mail:
| | - Jan van Bezu
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - Marinus A. Paul
- Department of Cardiothoracic Surgery, VU University Medical Centre, Amsterdam, The Netherlands
| | - A. B. Johan Groeneveld
- Department of Intensive Care, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - Victor W. M. van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
655
|
van Hinsbergh VWM. Endothelium--role in regulation of coagulation and inflammation. Semin Immunopathol 2011; 34:93-106. [PMID: 21845431 PMCID: PMC3233666 DOI: 10.1007/s00281-011-0285-5] [Citation(s) in RCA: 354] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 07/20/2011] [Indexed: 02/07/2023]
Abstract
By its strategic position at the interface between blood and tissues, endothelial cells control blood fluidity and continued tissue perfusion while simultaneously they direct inflammatory cells to areas in need of defense or repair. The endothelial response depends on specific tissue needs and adapts to local stresses. Endothelial cells counteract coagulation by providing tissue factor and thrombin inhibitors and receptors for protein C activation. The receptor PAR-1 is differentially activated by thrombin and the activated protein C/EPCR complex, resulting in antithrombotic and anti-inflammatory effects. Thrombin and vasoactive agents release von Willebrand factor as ultra-large platelet-binding multimers, which are cleaved by ADAMTS13. Platelets can also facilitate leukocyte-endothelium interaction. Platelet activation is prevented by nitric oxide, prostacyclin, and exonucleotidases. Thrombin-cleaved ADAMTS18 induces disintegration of platelet aggregates while tissue-type plasminogen activator initiates fibrinolysis. Fibrin and products of platelets and inflammatory cells modulate the angiogenic response of endothelial cells and contribute to tissue repair.
Collapse
Affiliation(s)
- Victor W M van Hinsbergh
- Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
656
|
Khan OF, Sefton MV. Endothelialized biomaterials for tissue engineering applications in vivo. Trends Biotechnol 2011; 29:379-87. [PMID: 21549438 PMCID: PMC3140588 DOI: 10.1016/j.tibtech.2011.03.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 01/20/2023]
Abstract
Rebuilding tissues involves the creation of a vasculature to supply nutrients and this in turn means that the endothelial cells (ECs) of the resulting endothelium must be a quiescent non-thrombogenic blood contacting surface. Such ECs are deployed on biomaterials that are composed of natural materials such as extracellular matrix proteins or synthetic polymers in the form of vascular grafts or tissue-engineered constructs. Because EC function is influenced by their origin, biomaterial surface chemistry and hemodynamics, these issues must be considered to optimize implant performance. In this review, we examine the recent in vivo use of endothelialized biomaterials and discuss the fundamental issues that must be considered when engineering functional vasculature.
Collapse
Affiliation(s)
- Omar F Khan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario, M5S 3E5, Canada
| | | |
Collapse
|
657
|
Barrett EJ, Wang H, Upchurch CT, Liu Z. Insulin regulates its own delivery to skeletal muscle by feed-forward actions on the vasculature. Am J Physiol Endocrinol Metab 2011; 301:E252-63. [PMID: 21610226 PMCID: PMC3154531 DOI: 10.1152/ajpendo.00186.2011] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Insulin, at physiological concentrations, regulates the volume of microvasculature perfused within skeletal and cardiac muscle. It can also, by relaxing the larger resistance vessels, increase total muscle blood flow. Both of these effects require endothelial cell nitric oxide generation and smooth muscle cell relaxation, and each could increase delivery of insulin and nutrients to muscle. The capillary microvasculature possesses the greatest endothelial surface area of the body. Yet, whether insulin acts on the capillary endothelial cell is not known. Here, we review insulin's actions at each of three levels of the arterial vasculature as well as recent data suggesting that insulin can regulate a vesicular transport system within the endothelial cell. This latter action, if it occurs at the capillary level, could enhance insulin delivery to muscle interstitium and thereby complement insulin's actions on arteriolar endothelium to increase insulin delivery. We also review work that suggests that this action of insulin on vesicle transport depends on endothelial cell nitric oxide generation and that insulin's ability to regulate this vesicular transport system is impaired by inflammatory cytokines that provoke insulin resistance.
Collapse
Affiliation(s)
- Eugene J Barrett
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | |
Collapse
|
658
|
Chong DC, Koo Y, Xu K, Fu S, Cleaver O. Stepwise arteriovenous fate acquisition during mammalian vasculogenesis. Dev Dyn 2011; 240:2153-65. [PMID: 21793101 DOI: 10.1002/dvdy.22706] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2011] [Indexed: 11/07/2022] Open
Abstract
Arteriovenous (AV) differentiation is a critical step during blood vessel formation and stabilization. Defects in arterial or venous fate lead to inappropriate fusion of vessels, resulting in damaging arteriovenous shunts. While many studies have unraveled the molecular underpinnings that drive AV fate, surprisingly, the spatiotemporal emergence of arteries and veins in mammalian embryos remains unknown. Here, we examine artery and vein specification and differentiation during vasculogenesis. We show that the first intraembryonic vessels formed are arteries, which differentiate in a stepwise manner. By contrast, veins emerge later, progressively forming after embryonic turning. In addition, we demonstrate that hemodynamic flow is not required for arterial specification, but is required for maintenance of select arterial markers. Together, our results provide a first spatiotemporal analysis of mammalian AV cell fate establishment and anatomy, as well as a delineation of a molecular toolkit for analysis of arteries and veins during early vessel development.
Collapse
Affiliation(s)
- Diana C Chong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | |
Collapse
|
659
|
Bovill EG, van der Vliet A. Venous valvular stasis-associated hypoxia and thrombosis: what is the link? Annu Rev Physiol 2011; 73:527-45. [PMID: 21034220 DOI: 10.1146/annurev-physiol-012110-142305] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This review focuses on the role of the venous valves in the genesis of thrombus formation in venous thromboembolic disease (VTE). Clinical VTE and the evidence for the valvular origin of venous thrombosis are reviewed. Virchow's triad is then used as a framework for discussion to approach the question posed regarding the link between venous valvular stasis-associated hypoxia and thrombosis. Thus, the effects of blood flow stasis, hypercoagulability of blood, and the characteristics of the vessel wall within the venous valvular sinus are assessed in turn.
Collapse
Affiliation(s)
- Edwin G Bovill
- Department of Pathology, University of Vermont College of Medicine, Burlington, 05405, USA.
| | | |
Collapse
|
660
|
Félétou M. The Endothelium, Part I: Multiple Functions of the Endothelial Cells -- Focus on Endothelium-Derived Vasoactive Mediators. ACTA ACUST UNITED AC 2011. [DOI: 10.4199/c00031ed1v01y201105isp019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
661
|
|
662
|
Cerveró J, Montes R, España F, Esmon CT, Hermida J. Limited ability to activate protein C confers left atrial endocardium a thrombogenic phenotype: a role in cardioembolic stroke? Stroke 2011; 42:2622-4. [PMID: 21700937 PMCID: PMC3178262 DOI: 10.1161/strokeaha.111.614420] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Atrial fibrillation is the most important risk factor for cardioembolic stroke. Thrombi form in the left atrial appendage rather than in the right. The causes of this different thrombogenicity are not well-understood. The goal herein was to compare the activation of the anticoagulant protein C and the thrombomodulin and endothelial protein C receptor/activated protein C receptor expression on the endocardium between right and left atria. METHODS We harvested the atria of 6 monkeys (Macaca fascicularis) and quantified their ability to activate protein C ex vivo and we measured the thrombomodulin and endothelial protein C receptor expression by immunofluorescence. RESULTS We found the ability to activate protein C decreased by half (P=0.028) and there was lower expression of thrombomodulin in the left atrial endocardium than the right (52.5±19.9 and 72.1±18.8 arbitrary intensity units, mean±standard deviation; P=0.028). No differences were detected in endothelial protein C receptor expression. CONCLUSIONS Impaired protein C activation on the left atrial endocardium attributable to low thrombomodulin expression may explain its higher thrombogenicity and play a role in cardioembolic stroke.
Collapse
Affiliation(s)
- Jorge Cerveró
- Division of Cardiovascular Sciences, Centre for Applied Medical Research, University of Navarra, Avenida Pío XII, 55, 31008 Pamplona, Spain
| | | | | | | | | |
Collapse
|
663
|
Goldenberg NM, Steinberg BE, Slutsky AS, Lee WL. Broken Barriers: A New Take on Sepsis Pathogenesis. Sci Transl Med 2011; 3:88ps25. [DOI: 10.1126/scitranslmed.3002011] [Citation(s) in RCA: 227] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
664
|
Svensen N, Díaz-Mochón JJ, Dhaliwal K, Planonth S, Dewar M, Armstrong JD, Bradley M. Screening of a Combinatorial Homing Peptide Library for Selective Cellular Delivery. Angew Chem Int Ed Engl 2011; 50:6133-6. [DOI: 10.1002/anie.201101804] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
665
|
Epigenetically coordinated GATA2 binding is necessary for endothelium-specific endomucin expression. EMBO J 2011; 30:2582-95. [PMID: 21666600 DOI: 10.1038/emboj.2011.173] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 05/01/2011] [Indexed: 11/08/2022] Open
Abstract
GATA2 is well recognized as a key transcription factor and regulator of cell-type specificity and differentiation. Here, we carried out comparative chromatin immunoprecipitation with comprehensive sequencing (ChIP-seq) to determine genome-wide occupancy of GATA2 in endothelial cells and erythroids, and compared the occupancy to the respective gene expression profile in each cell type. Although GATA2 was commonly expressed in both cell types, different GATA2 bindings and distinct cell-specific gene expressions were observed. By using the ChIP-seq with epigenetic histone modifications and chromatin conformation capture assays; we elucidated the mechanistic regulation of endothelial-specific GATA2-mediated endomucin gene expression, that was regulated by the endothelial-specific chromatin loop with a GATA2-associated distal enhancer and core promoter. Knockdown of endomucin markedly attenuated endothelial cell growth, migration and tube formation. Moreover, abrogation of GATA2 in endothelium demonstrated not only a reduction of endothelial-specific markers, but also induction of mesenchymal transition promoting gene expression. Our findings provide new insights into the correlation of endothelial-expressed GATA2 binding, epigenetic modification, and the determination of endothelial cell specificity.
Collapse
|
666
|
Deletion of Adam10 in endothelial cells leads to defects in organ-specific vascular structures. Blood 2011; 118:1163-74. [PMID: 21652679 DOI: 10.1182/blood-2011-04-348557] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
During vertebrate angiogenesis, Notch regulates the cell-fate decision between vascular tip cells versus stalk cells. Canonical Notch signaling depends on sequential proteolytic events, whereby interaction of Notch with membrane-anchored ligands triggers proteolytic processing, first by Adam10 and then presenilins. This liberates the Notch intracellular domain, allowing it to enter the nucleus and activate Notch-dependent genes. Here we report that conditional inactivation of Adam10 in endothelial cells (A10ΔEC) recapitulates the increased branching and density of the retinal vasculature that is also caused by interfering with Notch signaling. Moreover, A10ΔEC mice have additional vascular abnormalities, including aberrant subcapsular hepatic veins, enlarged glomeruli, intestinal polyps containing endothelial cell masses, abnormal endochondral ossification, leading to stunted long bone growth and increased pathologic neovascularization following oxygen-induced retinopathy. Our findings support a model in which Adam10 is a crucial regulator of endothelial cell-fate decisions, most likely because of its essential role in canonical Notch signaling.
Collapse
|
667
|
Fliser D, Wiecek A, Suleymanlar G, Ortiz A, Massy Z, Lindholm B, Martinez-Castelao A, Agarwal R, Jager KJ, Dekker FW, Blankestijn PJ, Goldsmith D, Covic A, London G, Zoccali C. The dysfunctional endothelium in CKD and in cardiovascular disease: mapping the origin(s) of cardiovascular problems in CKD and of kidney disease in cardiovascular conditions for a research agenda. Kidney Int Suppl (2011) 2011; 1:6-9. [PMID: 25018895 PMCID: PMC4089605 DOI: 10.1038/kisup.2011.6] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Endothelial dysfunction resulting in disintegration of vascular structure and function is a key element in the progression of chronic kidney disease (CKD). Many risk factors—traditional and non-traditional—are thought to have a role in the progression and development of cardiovascular disease (CVD) in patients with CKD. However, many risk factors await definitive confirmation of their clinical relevance obtained from intervention trials. Moreover, the investigation of the relative contribution of these factors to the twin-risk problem of CVD and progression in patients with CKD is one of the most important future challenges for nephrologists.
Collapse
Affiliation(s)
- Danilo Fliser
- Department of Internal Medicine IV, Saarland University Medical Centre , Homburg/Saar, Germany
| | - Andrzej Wiecek
- Department of Nephrology, Endocrinology and Metabolic Diseases, Medical University of Silesia , Katowice, Poland
| | - Gultekin Suleymanlar
- Nephrology Division, Department of Medicine, Akdeniz University Medical School , Antalya, Turkey
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Fundación Renal Iñigo Alvarez de Toledo , Madrid, Spain
| | - Ziad Massy
- INSERM ERI-12 (EA 4292) , Amiens, France ; Amiens University Hospital and the Jules Verne University of Picardie , Amiens, France
| | - Bengt Lindholm
- Divisions of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet , Stockholm, Sweden
| | | | - Rajiv Agarwal
- Indiana University and VAMC , Indianapolis, Indiana, USA
| | - Kitty J Jager
- ERA-EDTA Registry, Department of Medical Informatics, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands
| | - Friedo W Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center , Leiden, The Netherlands
| | - Peter J Blankestijn
- Department of Nephrology, University Medical Center , Utrecht, The Netherlands
| | - David Goldsmith
- Renal Unit, Guy's and St Thomas' NHS Foundation Hospital, King's Health Partners , London, UK
| | - Adrian Covic
- Clinic of Nephrology, C.I. Parhon University Hospital, Gr. T. Popa University of Medicine and Pharmacy , Iasi, Romania
| | - Gerard London
- INSERM U970, Hôpital Européen Georges Pompidou , Paris, France
| | - Carmine Zoccali
- Nephrology, Dialysis and Transplantation Unit and CNR-IBIM Clinical Epidemiology and Pathophysiology of Renal Diseases and Hypertension , Reggio Calabria, Italy
| | | |
Collapse
|
668
|
Bartkowiak K, Riethdorf S, Pantel K. The interrelating dynamics of hypoxic tumor microenvironments and cancer cell phenotypes in cancer metastasis. CANCER MICROENVIRONMENT 2011; 5:59-72. [PMID: 21626313 DOI: 10.1007/s12307-011-0067-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 05/18/2011] [Indexed: 02/04/2023]
Abstract
The interrelating dynamics of the primary tumor cells and their surrounding microenvironment might determine phenotypic characteristics of disseminated tumor cells and contribute to cancer metastasis. Cytoprotective mechanisms (e.g., energy metabolism control, DNA damage response, global translation control and unfolded protein response) exert selective pressure in the tumor microenvironment. In particular, adaptation to hypoxia is vital for survival of malignant cells in the tumor and at distant sites such as the bone marrow. In addition to the stress response, the ability of tumor cells to undergo certain cellular re-differentiation programmes like the epithelial-mesenchymal transition (EMT), which is linked to cancer stemness, appears to be important for successful cancer cell spread. Here we will discuss the selection pressures that eventually lead to the formation of overt metastases. We will focus the properties of the microenvironment including (i) metabolic and cytoprotective programs that ensure survival of disseminated tumor cells, (ii) blood vessel structure, and (iii) the hypoxia-normoxia switch as well as intrinsic factors affecting the evolvement of novel tumor cell populations.
Collapse
Affiliation(s)
- Kai Bartkowiak
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | | | | |
Collapse
|
669
|
Human liver sinusoidal endothelial cells respond to interaction with Entamoeba histolytica by changes in morphology, integrin signalling and cell death. Cell Microbiol 2011; 13:1091-106. [DOI: 10.1111/j.1462-5822.2011.01604.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
670
|
Metzger W, Sossong D, Bächle A, Pütz N, Wennemuth G, Pohlemann T, Oberringer M. The liquid overlay technique is the key to formation of co-culture spheroids consisting of primary osteoblasts, fibroblasts and endothelial cells. Cytotherapy 2011; 13:1000-12. [PMID: 21619419 DOI: 10.3109/14653249.2011.583233] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND AIMS The 3-dimensional (3-D) culture of various cell types reflects the in vivo situation more precisely than 2-dimensional (2-D) cell culture techniques. Spheroids as 3-D cell constructs have been used in tumor research for a long time. They have also been used to study angiogenic mechanisms, which are essential for the success of many tissue-engineering approaches. Several methods of forming spheroids are known, but there is a lack of systematic studies evaluating the performance of these techniques. METHODS We evaluated the performance of the hanging drop technique, carboxymethyl cellulose technique and liquid overlay technique to form both mono- and co-culture spheroids consisting of primary osteoblasts, fibroblasts and endothelial cells. The performance of the three techniques was evaluated in terms of rate of yield and reproducibility. The size of the generated spheroids was determined systematically. RESULTS The liquid overlay technique was the most suitable for generating spheroids reproducibly. The rate of yield for this technique was between 60% and 100% for monoculture spheroids and 100% for co-culture spheroids. The size of the spheroids could be adjusted easily and precisely by varying the number of seeded cells organized in one spheroid. The formation of co-culture spheroids consisting of three different cell types was possible. CONCLUSIONS Our results show that the most suitable technique for forming spheroids can vary from the chosen cell type, especially if primary cells are used. Co-culture spheroids consisting of three different cell types will be used to study angiogenic phenomena in further studies.
Collapse
Affiliation(s)
- Wolfgang Metzger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, Homburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
671
|
Svensen N, Díaz-Mochón JJ, Dhaliwal K, Planonth S, Dewar M, Armstrong JD, Bradley M. Screening of a Combinatorial Homing Peptide Library for Selective Cellular Delivery. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201101804] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
672
|
Paul DM, Vilas SP, Kumar JM. A flow-cytometry assisted segregation of responding and non-responding population of endothelial cells for enhanced detection of intracellular nitric oxide production. Nitric Oxide 2011; 25:31-40. [PMID: 21550415 DOI: 10.1016/j.niox.2011.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Revised: 03/04/2011] [Accepted: 04/23/2011] [Indexed: 11/26/2022]
Abstract
Nitric oxide (NO) is an important paracrine substance released by the endothelium to regulate vasomotor tone. The constitutive levels of endothelium dependent NO production is low. However, it is induced significantly in response to certain environmental and biological stimuli. An accurate evaluation of such stimulus induced NO release is of pharmacological significance. We observed that the sensitivity of NO detection in endothelial cells is compromised by baseline fluorescence emanated from non-activated cells resulting in ambiguous detection. In order to measure NO levels in activated population independent of non-activated cells, we segregated DAF-FM loaded cells based on their fluorescence intensity using flow-cytometry. Specific agonists like bradykinin, VEGF and insulin enhanced the proportion of activated cells. This effect was partially blocked in presence of NO synthase inhibitor, N(G)-nitro-L-arginine-methyl ester (L-NAME). We demonstrate that the fluorescence yield of activated population serves as a sensitive measure to evaluate agonist induced nitric oxide production in endothelial cells. Such increase in NO production in activated cells was also associated with increased eNOS phosphorylation at Ser-1177. While the endothelial cells showed heterogeneity with respect to NO production, immuno-phenotyping for endothelial cell-surface markers revealed a homogenous population.
Collapse
Affiliation(s)
- Dias M Paul
- Hindustan Unilever Research Centre, # 64 Main Road, Whitefield, Bangalore, Karnataka 560 066, India.
| | | | | |
Collapse
|
673
|
Novosel EC, Kleinhans C, Kluger PJ. Vascularization is the key challenge in tissue engineering. Adv Drug Deliv Rev 2011; 63:300-11. [PMID: 21396416 DOI: 10.1016/j.addr.2011.03.004] [Citation(s) in RCA: 703] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 02/09/2011] [Accepted: 03/02/2011] [Indexed: 12/11/2022]
Abstract
The main limitation in engineering in vitro tissues is the lack of a sufficient blood vessel system - the vascularization. In vivo almost all tissues are supplied by these endothelial cell coated tubular networks. Current strategies to create vascularized tissues are discussed in this review. The first strategy is based on the endothelial cells and their ability to form new vessels known as neoangiogenesis. Herein prevascularization techniques are compared to approaches in which biomolecules, such as growth factors, cytokines, peptides and proteins as well as cells are applied to generate new vessels. The second strategy is focused on scaffold-based techniques. Naturally-derived scaffolds, which contain vessels, are distinguished from synthetically manufactured matrices. Advantages and pitfalls of the approaches to create vascularized tissues in vitro are outlined and feasible future strategies are discussed.
Collapse
|
674
|
Franses JW, Baker AB, Chitalia VC, Edelman ER. Stromal endothelial cells directly influence cancer progression. Sci Transl Med 2011; 3:66ra5. [PMID: 21248315 DOI: 10.1126/scitranslmed.3001542] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer growth and metastasis are regulated in part by stromal cells such as fibroblasts and immune cells within the tumor microenvironment. Endothelial cells (ECs) are also ubiquitous within tumors because tumors are vascular, and yet, the impact of tumor-resident ECs is less well understood. Through paracrine regulation, ECs modulate a diverse spectrum of pathophysiologic processes in normal and hyperplastic tissues. We hypothesized that ECs offer similar paracrine regulatory control of cancer biology. Indeed, secretions from quiescent ECs muted the proliferative and invasive phenotype of lung and breast cancer cells in vitro and reduced cancer cell protumorigenic and proinflammatory signaling. EC perlecan silencing significantly changed this regulatory relationship, eliminating the ability of ECs to inhibit cancer cell invasiveness via increased interleukin-6 secretion. Moreover, implanting ECs embedded within porous matrices slowed adjacent xenograft tumor growth and prevented architectural degeneration, with a concomitant reduction in proliferative and tumorigenic markers. Finally, lung carcinoma cells pretreated with intact EC-conditioned media, but not media conditioned with perlecan-silenced ECs, exhibited reduced micrometastatic burden after tail vein injection. These findings add to an emerging appreciation of EC-regulatory effects that transcend their structural roles and pave the way for improved characterization and control of EC-cancer cross-talk interactions for diagnosis, prognosis, and treatment of cancer.
Collapse
Affiliation(s)
- Joseph W Franses
- Biomedical Engineering Center, Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, E25-438, Cambridge, MA 02139, USA
| | | | | | | |
Collapse
|
675
|
Abstract
Regulation of fluid and material movement between the vascular space of microvessels penetrating functioning organs and the cells therein has been studied extensively. Unanswered questions as to the regulatory mechanisms and routes remain. Significantly less is known about the lymphatic vascular system given the difficulties in seeing, no less isolating, these vessels lying deeper in these same tissues. It has become evident that the exchange microvasculature is not simply a passive biophysical barrier separating the vascular and interstitial compartments but a dynamic, multicellular structure subject to acute regulation and chronic adaptation to stimuli including inflammation, sepsis, diabetes, injury, hypoxia and exercise. Similarly lymphatic vessels range, in their simplest form, from lymphatic endothelium attached to the interstitial matrix, to endothelia and phasic lymphatic smooth muscle that act as Starling resistors. Recent work has demonstrated that among the microvascular lymphatic elements, the collecting lymphatics have barrier properties similar to venules, and thus participate in exchange. As with venules, vasoactive agents can alter both the permeability and contractile properties thereby setting up previously unanticipated gradients in the tissue space and providing potential targets for the pharmacological prevention and/or resolution of oedema.
Collapse
Affiliation(s)
- Virginia H Huxley
- University of Missouri School of Medicine, Department of Physiology, MA 415 Medical Sciences Bldg, Columbia, MO 65212-0001, USA.
| | | |
Collapse
|
676
|
Nabzdyk CS, Chun M, Pradhan L, Logerfo FW. High throughput RNAi assay optimization using adherent cell cytometry. J Transl Med 2011; 9:48. [PMID: 21518450 PMCID: PMC3111359 DOI: 10.1186/1479-5876-9-48] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 04/25/2011] [Indexed: 11/29/2022] Open
Abstract
Background siRNA technology is a promising tool for gene therapy of vascular disease. Due to the multitude of reagents and cell types, RNAi experiment optimization can be time-consuming. In this study adherent cell cytometry was used to rapidly optimize siRNA transfection in human aortic vascular smooth muscle cells (AoSMC). Methods AoSMC were seeded at a density of 3000-8000 cells/well of a 96well plate. 24 hours later AoSMC were transfected with either non-targeting unlabeled siRNA (50 nM), or non-targeting labeled siRNA, siGLO Red (5 or 50 nM) using no transfection reagent, HiPerfect or Lipofectamine RNAiMax. For counting cells, Hoechst nuclei stain or Cell Tracker green were used. For data analysis an adherent cell cytometer, Celigo® was used. Data was normalized to the transfection reagent alone group and expressed as red pixel count/cell. Results After 24 hours, none of the transfection conditions led to cell loss. Red fluorescence counts were normalized to the AoSMC count. RNAiMax was more potent compared to HiPerfect or no transfection reagent at 5 nM siGLO Red (4.12 +/-1.04 vs. 0.70 +/-0.26 vs. 0.15 +/-0.13 red pixel/cell) and 50 nM siGLO Red (6.49 +/-1.81 vs. 2.52 +/-0.67 vs. 0.34 +/-0.19). Fluorescence expression results supported gene knockdown achieved by using MARCKS targeting siRNA in AoSMCs. Conclusion This study underscores that RNAi delivery depends heavily on the choice of delivery method. Adherent cell cytometry can be used as a high throughput-screening tool for the optimization of RNAi assays. This technology can accelerate in vitro cell assays and thus save costs.
Collapse
Affiliation(s)
- Christoph S Nabzdyk
- Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
677
|
Civelek M, Manduchi E, Riley RJ, Stoeckert CJ, Davies PF. Coronary artery endothelial transcriptome in vivo: identification of endoplasmic reticulum stress and enhanced reactive oxygen species by gene connectivity network analysis. ACTA ACUST UNITED AC 2011; 4:243-52. [PMID: 21493819 DOI: 10.1161/circgenetics.110.958926] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Endothelial function is central to the localization of atherosclerosis. The in vivo endothelial phenotypic footprints of arterial bed identity and site-specific atherosusceptibility are addressed. METHODS AND RESULTS Ninety-eight endothelial cell samples from 13 discrete coronary and noncoronary arterial regions of varying susceptibilities to atherosclerosis were isolated from 76 normal swine. Transcript profiles were analyzed to determine the steady-state in vivo endothelial phenotypes. An unsupervised systems biology approach using weighted gene coexpression networks showed highly correlated endothelial genes. Connectivity network analysis identified 19 gene modules, 12 of which showed significant association with circulatory bed classification. Differential expression of 1300 genes between coronary and noncoronary artery endothelium suggested distinct coronary endothelial phenotypes, with highest significance expressed in gene modules enriched for biological functions related to endoplasmic reticulum (ER) stress and unfolded protein binding, regulation of transcription and translation, and redox homeostasis. Furthermore, within coronary arteries, comparison of endothelial transcript profiles of susceptible proximal regions to protected distal regions suggested the presence of ER stress conditions in susceptible sites. Accumulation of reactive oxygen species throughout coronary endothelium was greater than in noncoronary endothelium consistent with coronary artery ER stress and lower endothelial expression of antioxidant genes in coronary arteries. CONCLUSIONS Gene connectivity analyses discriminated between coronary and noncoronary endothelial transcript profiles and identified differential transcript levels associated with increased ER and oxidative stress in coronary arteries consistent with enhanced susceptibility to atherosclerosis.
Collapse
Affiliation(s)
- Mete Civelek
- Department of Bioengineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
678
|
Feng J, Zhang D, Chen B. Endothelial mechanisms of endothelial dysfunction in patients with obstructive sleep apnea. Sleep Breath 2011; 16:283-94. [PMID: 21479903 DOI: 10.1007/s11325-011-0519-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/23/2011] [Accepted: 03/30/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) occurs in 2% of middle-aged women and 4% of middle-aged men in the general population and the prevalence is much higher in specific patient groups. Intermittent hypoxia (IH, oxygen desaturation and re-oxygenation) cycle, a major pathophysiologic character of OSA, and the physiological responses this evokes are thought to be responsible for its association with increased cardiovascular morbidity and mortality. Endothelial dysfunction, resulting from IH and as a key early event in atherosclerosis, was demonstrated repeatedly in patients with OSA and in animal models of IH, providing an important mechanistic link between the acute cyclical IH during sleep and the increased prevalence of chronic vascular diseases. CONCLUSIONS From this work, we conclude that IH from OSA may result in endothelial dysfunction, as a potential promoter of atherosclerosis, through nitric oxide unavailability, oxidative stress and inflammation, cell apoptosis, the crosstalk between endothelial cells and circulating inflammatory cells, microparticles, and damage repairing process. Though effective continuous positive airway pressure (CPAP) may specifically improve endothelial function, more controlled larger interventional trials that will include multiple centers and randomized allocation of CPAP therapy are needed to see if such changes are reversible before cause and effect can be implied finally, while further studies on cellular and animal level are also needed to elucidate molecular biologic/pathologic pathways.
Collapse
Affiliation(s)
- Jing Feng
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, 300052, China
| | | | | |
Collapse
|
679
|
Xu K, Sacharidou A, Fu S, Chong DC, Skaug B, Chen ZJ, Davis GE, Cleaver O. Blood vessel tubulogenesis requires Rasip1 regulation of GTPase signaling. Dev Cell 2011; 20:526-39. [PMID: 21396893 DOI: 10.1016/j.devcel.2011.02.010] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 01/06/2011] [Accepted: 02/23/2011] [Indexed: 01/30/2023]
Abstract
Cardiovascular function depends on patent blood vessel formation by endothelial cells (ECs). However, the mechanisms underlying vascular "tubulogenesis" are only beginning to be unraveled. We show that endothelial tubulogenesis requires the Ras interacting protein 1, Rasip1, and its binding partner, the RhoGAP Arhgap29. Mice lacking Rasip1 fail to form patent lumens in all blood vessels, including the early endocardial tube. Rasipl null angioblasts fail to properly localize the polarity determinant Par3 and display defective cell polarity, resulting in mislocalized junctional complexes and loss of adhesion to extracellular matrix (ECM). Similarly, depletion of either Rasip1 or Arhgap29 in cultured ECs blocks in vitro lumen formation, fundamentally alters the cytoskeleton, and reduces integrin-dependent adhesion to ECM. These defects result from increased RhoA/ROCK/myosin II activity and blockade of Cdc42 and Rac1 signaling. This study identifies Rasip1 as a unique, endothelial-specific regulator of Rho GTPase signaling, which is essential for blood vessel morphogenesis.
Collapse
Affiliation(s)
- Ke Xu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | |
Collapse
|
680
|
Smalberg JH, Kruip MJ, Janssen HL, Rijken DC, Leebeek FW, de Maat MP. Hypercoagulability and Hypofibrinolysis and Risk of Deep Vein Thrombosis and Splanchnic Vein Thrombosis. Arterioscler Thromb Vasc Biol 2011; 31:485-93. [DOI: 10.1161/atvbaha.110.213371] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Jasper H. Smalberg
- From the Departments of Hematology (J.H.S., M.J.H.A.K., D.C.R., F.W.G.L., M.P.M.d.M.) and Hepatology and Gastroenterology (H.L.A.J.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marieke J.H.A. Kruip
- From the Departments of Hematology (J.H.S., M.J.H.A.K., D.C.R., F.W.G.L., M.P.M.d.M.) and Hepatology and Gastroenterology (H.L.A.J.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Harry L.A. Janssen
- From the Departments of Hematology (J.H.S., M.J.H.A.K., D.C.R., F.W.G.L., M.P.M.d.M.) and Hepatology and Gastroenterology (H.L.A.J.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Dingeman C. Rijken
- From the Departments of Hematology (J.H.S., M.J.H.A.K., D.C.R., F.W.G.L., M.P.M.d.M.) and Hepatology and Gastroenterology (H.L.A.J.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Frank W.G. Leebeek
- From the Departments of Hematology (J.H.S., M.J.H.A.K., D.C.R., F.W.G.L., M.P.M.d.M.) and Hepatology and Gastroenterology (H.L.A.J.), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Moniek P.M. de Maat
- From the Departments of Hematology (J.H.S., M.J.H.A.K., D.C.R., F.W.G.L., M.P.M.d.M.) and Hepatology and Gastroenterology (H.L.A.J.), Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
681
|
Endothelial progenitor cells: novel biomarker and promising cell therapy for cardiovascular disease. Clin Sci (Lond) 2011; 120:263-83. [PMID: 21143202 DOI: 10.1042/cs20100429] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bone-marrow-derived EPCs (endothelial progenitor cells) play an integral role in the regulation and protection of the endothelium, as well as new vessel formation. Peripheral circulating EPC number and function are robust biomarkers of vascular risk for a multitude of diseases, particularly CVD (cardiovascular disease). Importantly, using EPCs as a biomarker is independent of both traditional and non-traditional risk factors (e.g. hypertension, hypercholesterolaemia and C-reactive protein), with infused ex vivo-expanded EPCs showing potential for improved endothelial function and either reducing the risk of events or enhancing recovery from ischaemia. However, as the number of existing cardiovascular risk factors is variable between patients, simple EPC counts do not adequately describe vascular disease risk in all clinical conditions and, as such, the risk of CVD remains. It is likely that this limitation is attributable to variation in the definition of EPCs, as well as a difference in the interaction between EPCs and other cells involved in vascular control such as pericytes, smooth muscle cells and macrophages. For EPCs to be used regularly in clinical practice, agreement on definitions of EPC subtypes is needed, and recognition that function of EPCs (rather than number) may be a better marker of vascular risk in certain CVD risk states. The present review focuses on the identification of measures to improve individual risk stratification and, further, to potentially individualize patient care to address specific EPC functional abnormalities. Herein, we describe that future therapeutic use of EPCs will probably rely on a combination of strategies, including optimization of the function of adjunct cell types to prime tissues for the effect of EPCs.
Collapse
|
682
|
Tyurina YY, Tyurin VA, Kapralova VI, Wasserloos K, Mosher M, Epperly MW, Greenberger JS, Pitt BR, Kagan VE. Oxidative lipidomics of γ-radiation-induced lung injury: mass spectrometric characterization of cardiolipin and phosphatidylserine peroxidation. Radiat Res 2011; 175:610-21. [PMID: 21338246 DOI: 10.1667/rr2297.1] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Oxidative damage plays a significant role in the pathogenesis of γ-radiation-induced lung injury. Endothelium is a preferred target for early radiation-induced damage and apoptosis. Given the newly discovered role of oxidized phospholipids in apoptotic signaling, we performed oxidative lipidomics analysis of phospholipids in irradiated mouse lungs and cultured mouse lung endothelial cells. C57BL/6NHsd female mice were subjected to total-body irradiation (10 Gy, 15 Gy) and euthanized 24 h thereafter. Mouse lung endothelial cells were analyzed 48 h after γ irradiation (15 Gy). We found that radiation-induced apoptosis in vivo and in vitro was accompanied by non-random oxidation of phospholipids. Cardiolipin and phosphatidylserine were the major oxidized phospholipids, while more abundant phospholipids (phosphatidylcholine, phosphatidylethanolamine) remained non-oxidized. Electrospray ionization mass spectrometry analysis revealed the formation of cardiolipin and phosphatidylserine oxygenated molecular species in the irradiated lung and cells. Analysis of fatty acids after hydrolysis of cardiolipin and phosphatidylserine by phospholipase A(2) revealed the presence of mono-hydroperoxy and/or mono-hydroxy/mono-epoxy, mono-hydroperoxy/mono-oxo molecular species of linoleic acid. We speculate that cyt c-driven oxidations of cardiolipin and phosphatidylserine associated with the execution of apoptosis in pulmonary endothelial cells are important contributors to endothelium dysfunction in γ-radiation-induced lung injury.
Collapse
Affiliation(s)
- Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh, Bridgeside Point, 100 Technology Drive, Suite 350, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
683
|
Wu X, Zou Y, Liang Y, Zhou Q, Gong H, Sun A, Yuan L, Wang K, Ge J. COUP-TFII switches responses of venous endothelium to atherosclerotic factors through controlling the profile of various inherent genes expression. J Cell Biochem 2011; 112:256-64. [DOI: 10.1002/jcb.22923] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
684
|
The endothelial microenvironment in the venous valvular sinus: thromboresistance trends and inter-individual variation. Histochem Cell Biol 2011; 135:141-52. [PMID: 21298440 DOI: 10.1007/s00418-011-0783-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2011] [Indexed: 10/18/2022]
Abstract
The valve sinuses of the deep venous system are frequent sites of venous thrombus initiation. We previously reported that, in comparison with the non-valvular lumenal endothelium, the valve sinus endothelium had decreased expression of von Willebrand factor (vWF) and increased expression of endothelial protein C receptor (EPCR) and thrombomodulin (TM), suggesting alteration in the procoagulant/anticoagulant balance. We hypothesized that increased stasis in the deeper recesses of the venous valves would be associated with a gradient of increased thromboresistance. Expression of EPCR, TM, and vWF was analyzed via quantitative confocal immunofluorescence in residual saphenous veins collected following coronary artery bypass procedures. In agreement with our hypothesis, endothelial expression of vWF in the valve sinus decreased from the uppermost to the deepest region of the valve sinus. In contrast to our hypothesis, EPCR expression decreased from the uppermost to the deepest region of the valve sinus (p < 0.001) and TM expression remained unchanged throughout the valve sinus. Comparison of the non-valvular lumenal endothelium with the valve sinus endothelium demonstrated significantly decreased vWF expression (p < 0.001) in the valvular sinus consistent with our previous report; however, we did not observe statistically significant differences in EPCR or TM expression in this comparison. In addition, remarkable inter-individual variation in expression of these three proteins was also observed. These findings suggest that the genesis of these observations is more complex than predicted by our initial hypothesis, likely due, at least in part, to the complex rheology of the valvular sinus microenvironment.
Collapse
|
685
|
Sammons V, Davidson A, Tu J, Stoodley MA. Endothelial cells in the context of brain arteriovenous malformations. J Clin Neurosci 2011; 18:165-70. [PMID: 21167719 DOI: 10.1016/j.jocn.2010.04.045] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 04/14/2010] [Indexed: 10/18/2022]
|
686
|
Laurance S, Lansiaux P, Pellay FX, Hauchecorne M, Benecke A, Elion J, Lapoumeroulie C. Differential modulation of adhesion molecule expression by hydroxycarbamide in human endothelial cells from the micro- and macrocirculation: potential implications in sickle cell disease vasoocclusive events. Haematologica 2011; 96:534-42. [PMID: 21228039 DOI: 10.3324/haematol.2010.026740] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND All the cellular partners of the vascular system and especially endothelial cells are involved in the pathophysiology of the vasoocclusive crises associated with sickle cell disease. In sickle cell disease, circulating cells adhere abnormally to endothelial cells in a chronic pro-inflammatory context. Hydroxycarbamide is the only drug with demonstrated efficacy to reduce the frequency of vasoocclusive crises. Here, we investigated the effects of hydroxycarbamide and/or cytokines on the expression of genes related to adhesion events in endothelial cells from three different vascular sites. DESIGN AND METHODS Endothelial cells representative of the macro- (HUVEC) or microcirculation (TrHBMEC and HPMEC) were grown in the presence or absence of hydroxycarbamide and/or cytokines (TNFα and IFNγ). Expression of genes encoding adhesion proteins was analyzed by RQ-PCR, ELISA, flow cytometry, in situ ELISA for extracellular matrix proteins, and Western blot. RESULTS In cells from the microcirculation, expression of TSP-1, vWF, and PECAM-1 genes was decreased by hydroxycarbamide and/or cytokine treatment at the mRNA level. In the macro-circulation their expression was unaffected or increased. Hydroxycarbamide significantly decreased vWF incorporated in the TrHBMEC extracellular matrix. CD36 mRNA was strongly down-regulated by cytokines in HPMEC, the only cell type in which it is expressed. Hydroxycarbamide decreased soluble PECAM-1 in HUVEC supernatants. CONCLUSIONS Our results highlight the heterogeneity of vascular endothelial cell responses to hydroxycarbamide and/or cytokines depending upon their origin. They also suggest that hydroxycarbamide has an anti-adhesogenic effect on endothelial cells, but by mechanisms which could vary according to their macro- or microcirculation and organ origin.
Collapse
Affiliation(s)
- Sandrine Laurance
- INSERM, UMR_S763, Hôpital, Robert Debré, 48 boulevard, Sérurier, 75019 Paris, France
| | | | | | | | | | | | | |
Collapse
|
687
|
Chiu JJ, Chien S. Effects of disturbed flow on vascular endothelium: pathophysiological basis and clinical perspectives. Physiol Rev 2011; 91:327-87. [PMID: 21248169 PMCID: PMC3844671 DOI: 10.1152/physrev.00047.2009] [Citation(s) in RCA: 1547] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Vascular endothelial cells (ECs) are exposed to hemodynamic forces, which modulate EC functions and vascular biology/pathobiology in health and disease. The flow patterns and hemodynamic forces are not uniform in the vascular system. In straight parts of the arterial tree, blood flow is generally laminar and wall shear stress is high and directed; in branches and curvatures, blood flow is disturbed with nonuniform and irregular distribution of low wall shear stress. Sustained laminar flow with high shear stress upregulates expressions of EC genes and proteins that are protective against atherosclerosis, whereas disturbed flow with associated reciprocating, low shear stress generally upregulates the EC genes and proteins that promote atherogenesis. These findings have led to the concept that the disturbed flow pattern in branch points and curvatures causes the preferential localization of atherosclerotic lesions. Disturbed flow also results in postsurgical neointimal hyperplasia and contributes to pathophysiology of clinical conditions such as in-stent restenosis, vein bypass graft failure, and transplant vasculopathy, as well as aortic valve calcification. In the venous system, disturbed flow resulting from reflux, outflow obstruction, and/or stasis leads to venous inflammation and thrombosis, and hence the development of chronic venous diseases. Understanding of the effects of disturbed flow on ECs can provide mechanistic insights into the role of complex flow patterns in pathogenesis of vascular diseases and can help to elucidate the phenotypic and functional differences between quiescent (nonatherogenic/nonthrombogenic) and activated (atherogenic/thrombogenic) ECs. This review summarizes the current knowledge on the role of disturbed flow in EC physiology and pathophysiology, as well as its clinical implications. Such information can contribute to our understanding of the etiology of lesion development in vascular niches with disturbed flow and help to generate new approaches for therapeutic interventions.
Collapse
Affiliation(s)
- Jeng-Jiann Chiu
- Division of Medical Engineering Research, National Health Research Institutes, Taiwan
| | | |
Collapse
|
688
|
|
689
|
Sandoo A, van Zanten JJCSV, Metsios GS, Carroll D, Kitas GD. The endothelium and its role in regulating vascular tone. Open Cardiovasc Med J 2010; 4:302-12. [PMID: 21339899 PMCID: PMC3040999 DOI: 10.2174/1874192401004010302] [Citation(s) in RCA: 533] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 11/22/2010] [Accepted: 11/23/2010] [Indexed: 12/16/2022] Open
Abstract
The endothelium forms an important part of the vasculature and is involved in promoting an atheroprotective environment via the complementary actions of endothelial cell-derived vasoactive factors. Disruption of vascular homeostasis can lead to the development of endothelial dysfunction which in turn contributes to the early and late stages of atherosclerosis. In recent years an increasing number of non-invasive vascular tests have been developed to assess vascular structure and function in different clinical populations. The present review aims to provide an insight into the anatomy of the vasculature as well as the underlying endothelial cell physiology. In addition, an in-depth overview of the current methods used to assess vascular function and structure is provided as well as their link to certain clinical populations.
Collapse
Affiliation(s)
- Aamer Sandoo
- School of Sport and Exercise Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | | | | | | | | |
Collapse
|
690
|
Quantification and cell-to-cell variation of vascular endothelial growth factor receptors. Exp Cell Res 2010; 317:955-65. [PMID: 21185287 DOI: 10.1016/j.yexcr.2010.12.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 12/03/2010] [Accepted: 12/15/2010] [Indexed: 01/13/2023]
Abstract
The vascular endothelial growth factor receptors (VEGFR) play a significant role in angiogenesis, the formation of new blood vessels from existing vasculature. Systems biology offers promising approaches to better understand angiogenesis by computational modeling the key molecular interactions in this process. Such modeling requires quantitative knowledge of cell surface density of pro-angiogenic receptors versus anti-angiogenic receptors, their regulation, and their cell-to-cell variability. Using quantitative fluorescence, we systematically characterized the endothelial surface density of VEGFRs and neuropilin-1 (NRP1). We also determined the role of VEGF in regulating the surface density of these receptors. Applying cell-by-cell analysis revealed heterogeneity in receptor surface density and VEGF tuning of this heterogeneity. Altogether, we determine inherent differences in the surface expression levels of these receptors and the role of VEGF in regulating the balance of anti-angiogenic or modulatory (VEGFR1) and pro-angiogenic (VEGFR2) receptors.
Collapse
|
691
|
Ghelfi E, Karaaslan C, Berkelhamer S, Akar S, Kozakewich H, Cataltepe S. Fatty acid-binding proteins and peribronchial angiogenesis in bronchopulmonary dysplasia. Am J Respir Cell Mol Biol 2010; 45:550-6. [PMID: 21177979 DOI: 10.1165/rcmb.2010-0376oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Inflammation plays a key role in the pathogenesis of bronchopulmonary dysplasia (BPD). Fatty acid-binding proteins (FABPs) 4 and 5 regulate the inflammatory activity of macrophages. Whether FABPs 4 and 5 could play a role in the pathogenesis of BPD via the promotion of macrophage inflammatory activity is unknown. This study sought to examine whether the expression levels of FABP4 and FABP5 were altered in bronchoalveolar lavage fluid and lung tissue in a baboon model of BPD. This study also sought to characterize the cell types that express these proteins. Real-time PCR, immunoblotting, immunohistochemistry, and double immunofluorescence were used to examine the expression of FABPs in samples of BPD. Morphometric analysis was used to quantify FABP4-positive peribronchial blood vessels in lung sections. FABP4 was primarily expressed in macrophages in samples of BPD. In addition, FABP4 was expressed in the endothelial cells of blood vessels in peribronchial areas and the vasa vasorum, but not in the alveolar vasculature in samples of BPD. FABP4 concentrations were significantly increased in lungs and bronchoalveolar lavage fluid samples with BPD. An increased density of FABP4-positive peribronchial blood vessels was evident in both baboon and human BPD sections. FABP5 was expressed in several cell types, including alveolar epithelial cells and macrophages. FABP5 concentrations did not show any significant alterations in BPD. In conclusion, FABP4 but not FABP5 levels are increased in BPD. FABP4 is differentially expressed in endothelial cells of the bronchial microvasculature, which demonstrates a previously unrecognized expansion in BPD.
Collapse
Affiliation(s)
- Elisa Ghelfi
- Division of Neonatology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
692
|
Heterogeneous distribution of angiotensin I-converting enzyme (CD143) in the human and rat vascular systems: vessel, organ and species specificity. Microvasc Res 2010; 81:206-15. [PMID: 21167844 DOI: 10.1016/j.mvr.2010.12.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/06/2010] [Accepted: 12/09/2010] [Indexed: 11/20/2022]
Abstract
Angiotensin I-converting enzyme (kininase II, ACE, CD143) availability is a determinant of local angiotensin and kinin concentrations and physiological actions. Limited information is available on ACE synthesis in peripheral vascular beds. We studied the distribution of ACE along the human and rat vascular tree, and determined whether the enzyme was uniformly distributed in all endothelial cells (EC) or if differences occurred among vessels and organs. The distribution of ACE was assessed by using a panel of anti-human ACE monoclonal antibodies and serial sections of the entire vascular tree of humans. Comparison was made with other EC markers. EC of small muscular arteries and arterioles displayed high ACE immunoreactivity in all organs studied except the kidney, while EC of large arteries and of veins were poorly reactive or completely negative. Only 20% on average of capillary EC in each organ, including the heart, stained for ACE, with the remarkable exception of the lung and kidney. In the lung all capillary EC were labeled intensively for ACE, whereas in the kidney the entire vasculature was devoid of detectable enzyme. In contrast to the man, the rat showed homogeneous endothelial expression of ACE in all large and middle-sized arteries, and in veins, but in renal vessels ACE expression was reduced. This study documents a vessel, organ and species specific pattern of distribution of endothelial ACE. The markedly reduced ACE content of the renal vasculature may protect the renal circulation against excess angiotensin II formation and kinin depletion, and maintain high renal blood flow.
Collapse
|
693
|
Valsami S, Ruf W, Leikauf MS, Madon J, Kaech A, Asmis LM. Immunomodulatory drugs increase endothelial tissue factor expression in vitro. Thromb Res 2010; 127:264-71. [PMID: 21159364 DOI: 10.1016/j.thromres.2010.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 11/17/2010] [Accepted: 11/17/2010] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Immunomodulatory compounds such as thalidomide (THL) and lenalidomide (LEN) represent treatment options for multiple myeloma. Venous thromboembolism is a potential complication of immunomodulatory treatment in myeloma patients. The optimal thromboprophylactic strategy to prevent this drug-induced hypercoagulable state is debated. It is the aim of this study to investigate i) the effect of immunomodulatory treatment on endothelial cell tissue factor (TF) expression and function, ii) the regulation of the observed TF activity, and iii) the modulating effect of low molecular weight heparin and aspirin on TF activity in vitro. MATERIALS AND METHODS These aims were addressed in an in vitro culture model, human umbilical vein endothelial cells, using TF activity and antigen assays as well flow cytometry, real time PCR and electron microscopy. RESULTS At THL and LEN concentrations resembling those observed in myeloma patients in vivo and in the presence of tumor necrosis factor-α (TNFα) we observed significantly increased TF activity in human umbilical vein endothelial cells in vitro. Concordant changes were detected for tissue factor mRNA and TF whole cell antigen. Dalteparin and a mixture of monoclonal anti-TF antibodies inhibited TF activity by 100% and more than 80% respectively, while aspirin's inhibitory effect was only approximately 30%. In the presence of TNFα we detected the generation of endothelial cell-derived microparticles which expressed TF activity. CONCLUSIONS Our in vitro data support the hypothesis that THL and LEN induce a hypercoagulable state through increased endothelial TF expression.
Collapse
Affiliation(s)
- Serena Valsami
- Division of Hematology, University Hospital of Zurich, CH 8091 Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
694
|
Experimental models for assaying microvascular endothelial cell pathophysiology in stroke. Molecules 2010; 15:9104-34. [PMID: 21150829 PMCID: PMC6259215 DOI: 10.3390/molecules15129104] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 11/29/2010] [Accepted: 12/08/2010] [Indexed: 02/06/2023] Open
Abstract
It is important to understand the molecular mechanisms underlying neuron death following stroke in order to develop effective neuroprotective strategies. Since studies on human stroke are extremely limited due to the difficulty in collecting post-mortem tissue at different time points after the onset of stroke, brain ischaemia research focuses on information derived from in-vitro models of neuronal death through ischaemic injury [1]. This review aims to provide an update on the different in-vitro stroke models with brain microvascular endothelial cells that are currently being used. These models provide a physiologically relevant tool to screen potential neuroprotective drugs in stroke and to study the molecular mechanisms involved in brain ischaemia.
Collapse
|
695
|
Kuhnert F, Mancuso MR, Shamloo A, Wang HT, Choksi V, Florek M, Su H, Fruttiger M, Young WL, Heilshorn SC, Kuo CJ. Essential regulation of CNS angiogenesis by the orphan G protein-coupled receptor GPR124. Science 2010; 330:985-9. [PMID: 21071672 PMCID: PMC3099479 DOI: 10.1126/science.1196554] [Citation(s) in RCA: 245] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The orphan G protein-coupled receptor (GPCR) GPR124/tumor endothelial marker 5 is highly expressed in central nervous system (CNS) endothelium. Here, we show that complete null or endothelial-specific GPR124 deletion resulted in embryonic lethality from CNS-specific angiogenesis arrest in forebrain and neural tube. Conversely, GPR124 overexpression throughout all adult vascular beds produced CNS-specific hyperproliferative vascular malformations. In vivo, GPR124 functioned cell-autonomously in endothelium to regulate sprouting, migration, and developmental expression of the blood-brain barrier marker Glut1, whereas in vitro, GPR124 mediated Cdc42-dependent directional migration to forebrain-derived, vascular endothelial growth factor-independent cues. Our results demonstrate CNS-specific angiogenesis regulation by an endothelial receptor and illuminate functions of the poorly understood adhesion GPCR subfamily. Further, the functional tropism of GPR124 marks this receptor as a therapeutic target for CNS-related vascular pathologies.
Collapse
Affiliation(s)
- Frank Kuhnert
- Department of Medicine, Hematology Division, Stanford University, Stanford, CA 94305, USA
| | - Michael R. Mancuso
- Department of Medicine, Hematology Division, Stanford University, Stanford, CA 94305, USA
| | - Amir Shamloo
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Hsiao-Ting Wang
- Department of Medicine, Hematology Division, Stanford University, Stanford, CA 94305, USA
| | - Vir Choksi
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Mareike Florek
- Division of Blood and Marrow Transplantation, Stanford University, Stanford, CA 94305, USA
| | - Hua Su
- UCSF Center for Cerebrovascular Research, Departments of Anesthesia and Perioperative Care, Neurological Surgery and Neurology, San Francisco, CA 94110
| | | | - William L. Young
- UCSF Center for Cerebrovascular Research, Departments of Anesthesia and Perioperative Care, Neurological Surgery and Neurology, San Francisco, CA 94110
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Calvin J. Kuo
- Department of Medicine, Hematology Division, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
696
|
|
697
|
Relevance of disease- and organ-specific endothelial cells forin vitroresearch. Cell Biol Int 2010; 34:1231-8. [DOI: 10.1042/cbi20100531] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
698
|
Hsu J, Serrano D, Bhowmick T, Kumar K, Shen Y, Kuo YC, Garnacho C, Muro S. Enhanced endothelial delivery and biochemical effects of α-galactosidase by ICAM-1-targeted nanocarriers for Fabry disease. J Control Release 2010; 149:323-31. [PMID: 21047542 DOI: 10.1016/j.jconrel.2010.10.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 08/19/2010] [Accepted: 10/21/2010] [Indexed: 01/31/2023]
Abstract
Fabry disease, due to the deficiency of α-galactosidase A (α-Gal), causes lysosomal accumulation of globotriaosylceramide (Gb3) in multiple tissues and prominently in the vascular endothelium. Although enzyme replacement therapy (ERT) by injection of recombinant α-Gal improves the disease outcome, the effects on the vasculopathy associated with life-threatening cerebrovascular, cardiac and renal complications are still limited. We designed a strategy to enhance the delivery of α-Gal to organs and endothelial cells (ECs). We targeted α-Gal to intercellular adhesion molecule 1 (ICAM-1), a protein expressed on ECs throughout the vasculature, by loading this enzyme on nanocarriers coated with anti-ICAM (anti-ICAM/α-Gal NCs). In vitro radioisotope tracing showed efficient loading of α-Gal on anti-ICAM NCs, stability of this formulation under storage and in model physiological fluids, and enzyme release in response to lysosome environmental conditions. In mice, the delivery of (125)I-α-Gal was markedly enhanced by anti-ICAM/(125)I-α-Gal NCs in brain, kidney, heart, liver, lung, and spleen, and transmission electron microscopy showed anti-ICAM/α-Gal NCs attached to and internalized into the vascular endothelium. Fluorescence microscopy proved targeting, endocytosis and lysosomal transport of anti-ICAM/α-Gal NCs in macro- and micro-vascular ECs and a marked enhancement of Gb3 degradation. Therefore, this ICAM-1-targeting strategy may help improve the efficacy of therapeutic enzymes for Fabry disease.
Collapse
Affiliation(s)
- Janet Hsu
- Fischell Department of Bioengineering, School of Engineering, University of Maryland College Park, College Park, MD 20742, USA
| | | | | | | | | | | | | | | |
Collapse
|
699
|
Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA. The mouse blood-brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS One 2010; 5:e13741. [PMID: 21060791 PMCID: PMC2966423 DOI: 10.1371/journal.pone.0013741] [Citation(s) in RCA: 439] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 08/24/2010] [Indexed: 01/21/2023] Open
Abstract
The blood-brain barrier (BBB) maintains brain homeostasis and limits the entry of toxins and pathogens into the brain. Despite its importance, little is known about the molecular mechanisms regulating the development and function of this crucial barrier. In this study we have developed methods to highly purify and gene profile endothelial cells from different tissues, and by comparing the transcriptional profile of brain endothelial cells with those purified from the liver and lung, we have generated a comprehensive resource of transcripts that are enriched in the BBB forming endothelial cells of the brain. Through this comparison we have identified novel tight junction proteins, transporters, metabolic enzymes, signaling components, and unknown transcripts whose expression is enriched in central nervous system (CNS) endothelial cells. This analysis has identified that RXRalpha signaling cascade is specifically enriched at the BBB, implicating this pathway in regulating this vital barrier. This dataset provides a resource for understanding CNS endothelial cells and their interaction with neural and hematogenous cells.
Collapse
Affiliation(s)
- Richard Daneman
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America.
| | | | | | | | | | | |
Collapse
|
700
|
Danilov SM, Balyasnikova IV, Danilova AS, Naperova IA, Arablinskaya NE, Borisov SE, Metzger R, Franke FE, Schwartz DE, Gachok IV, Trakht IN, Kost OA, Garcia JGN. Conformational fingerprinting of the angiotensin I-converting enzyme (ACE). 1. Application in sarcoidosis. J Proteome Res 2010; 9:5782-93. [PMID: 20873814 DOI: 10.1021/pr100564r] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fine epitope mapping of monoclonal antibodies (mAbs) to 16 epitopes on human angiotensin I-converting enzyme (ACE) revealed that the epitopes of all mAbs contained putative glycosylation sites. ACE glycosylation is both cell- and tissue-specific and, therefore, the local conformation of ACE produced by different cells could be also unique. The pattern of ACE binding by a set of mAbs to 16 epitopes of human ACE - "conformational fingerprint of ACE" - is the most sensitive marker of ACE conformation and could be cell- and tissue-specific. The recognition of ACEs by mAbs to ACE was estimated using an immune-capture enzymatic plate precipitation assay. Precipitation patterns of soluble recombinant ACE released from Chinese hamster ovary (CHO)-ACE cells was influenced by conditions that alter ACE glycosylation. This pattern was also strongly cell type specific. Patients with sarcoidosis exhibited conformational fingerprints of tissue ACE (lungs and lymph nodes), as well as blood ACE, which were distinct from controls. Conformational fingerprinting of ACE may detect ACE originated from the cells other than endothelial cells in the blood and when combined with elevated blood ACE levels in patients with sarcoidosis may potentially reflect extrapulmonary sarcoidosis involvement (bone marrow, spleen, liver). If proven true, this would serve as a biomarker of enormous potential clinical significance.
Collapse
Affiliation(s)
- Sergei M Danilov
- Department of Anesthesiology, University of Illinois, Chicago, IL, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|