501
|
Abstract
Blood vessels are critical to deliver oxygen and nutrients to all of the tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier, which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and also protects the neural tissue from toxins and pathogens, and alterations of these barrier properties are an important component of pathology and progression of different neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the endothelial cells (ECs) that form the walls of the blood vessels, and these properties are regulated by interactions with different vascular, immune, and neural cells. Understanding how these different cell populations interact to regulate the barrier properties is essential for understanding how the brain functions during health and disease.
Collapse
Affiliation(s)
- Richard Daneman
- Departments of Neuroscience and Pharmacology, University of California, San Diego, San Diego, California 92093
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| |
Collapse
|
502
|
Teo GSL, Yang Z, Carman CV, Karp JM, Lin CP. Intravital imaging of mesenchymal stem cell trafficking and association with platelets and neutrophils. Stem Cells 2015; 33:265-77. [PMID: 25263183 PMCID: PMC4270897 DOI: 10.1002/stem.1848] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 08/18/2014] [Indexed: 12/13/2022]
Abstract
Early events of mesenchymal stem/stromal cell (MSC) adhesion to and transmigration through the vascular wall following systemic infusion are important for MSC trafficking to inflamed sites, yet are poorly characterized in vivo. Here, we used intravital confocal imaging to determine the acute extravasation kinetics and distribution of culture-expanded MSC (2-6 hours postinfusion) in a murine model of dermal inflammation. By 2 hours postinfusion, among the MSC that arrested within the inflamed ear dermis, 47.8% ± 8.2% of MSC had either initiated or completed transmigration into the extravascular space. Arrested and transmigrating MSCs were equally distributed within both small capillaries and larger venules. This suggested existence of an active adhesion mechanism, since venule diameters were greater than those of the MSC. Heterotypic intravascular interactions between distinct blood cell types have been reported to facilitate the arrest and extravasation of leukocytes and circulating tumor cells. We found that 42.8% ± 24.8% of intravascular MSC were in contact with neutrophil-platelet clusters. A role for platelets in MSC trafficking was confirmed by platelet depletion, which significantly reduced the preferential homing of MSC to the inflamed ear, although the total percentage of MSC in contact with neutrophils was maintained. Interestingly, although platelet depletion increased vascular permeability in the inflamed ear, there was decreased MSC accumulation. This suggests that increased vascular permeability is unnecessary for MSC trafficking to inflamed sites. These findings represent the first glimpse into MSC extravasation kinetics and microvascular distribution in vivo, and further clarify the roles of active adhesion, the intravascular cellular environment, and vascular permeability in MSC trafficking.
Collapse
Affiliation(s)
- Grace Sock Leng Teo
- Harvard-MIT Health Sciences and Technology
- Wellman Center for Photomedicine, Massachusetts General Hospital
| | - Zijiang Yang
- Harvard-MIT Health Sciences and Technology
- Harvard Medical School
- Brigham and Women’s Hospital, Harvard Stem Cell Institute
- Wellman Center for Photomedicine, Massachusetts General Hospital
| | - Christopher V. Carman
- Harvard Medical School
- Center for Vascular Biology, Beth Israel Deaconess Medical Center
| | - Jeffrey M. Karp
- Harvard-MIT Health Sciences and Technology
- Harvard Medical School
- Brigham and Women’s Hospital, Harvard Stem Cell Institute
| | - Charles P. Lin
- Harvard Medical School
- Wellman Center for Photomedicine, Massachusetts General Hospital
| |
Collapse
|
503
|
|
504
|
van de Stolpe A, Kauffmann RH. Innovative human-specific investigational approaches to autoimmune disease. RSC Adv 2015. [DOI: 10.1039/c4ra15794j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
An organ-on-chip disease model approach, including “pre-clinical trial-on-chip” is introduced for understanding of human autoimmune disease pathophysiology and drug development.
Collapse
Affiliation(s)
- Anja van de Stolpe
- Precision & Decentralized Diagnostics
- Philips Research
- Eindhoven
- The Netherlands
| | | |
Collapse
|
505
|
Herrnberger L, Hennig R, Kremer W, Hellerbrand C, Goepferich A, Kalbitzer HR, Tamm ER. Formation of fenestrae in murine liver sinusoids depends on plasmalemma vesicle-associated protein and is required for lipoprotein passage. PLoS One 2014; 9:e115005. [PMID: 25541982 PMCID: PMC4277272 DOI: 10.1371/journal.pone.0115005] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 11/17/2014] [Indexed: 12/11/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) are characterized by the presence of fenestrations that are not bridged by a diaphragm. The molecular mechanisms that control the formation of the fenestrations are largely unclear. Here we report that mice, which are deficient in plasmalemma vesicle-associated protein (PLVAP), develop a distinct phenotype that is caused by the lack of sinusoidal fenestrations. Fenestrations with a diaphragm were not observed in mouse LSEC at three weeks of age, but were present during embryonic life starting from embryonic day 12.5. PLVAP was expressed in LSEC of wild-type mice, but not in that of Plvap-deficient littermates. Plvap-/- LSEC showed a pronounced and highly significant reduction in the number of fenestrations, a finding, which was seen both by transmission and scanning electron microscopy. The lack of fenestrations was associated with an impaired passage of macromolecules such as FITC-dextran and quantum dot nanoparticles from the sinusoidal lumen into Disse's space. Plvap-deficient mice suffered from a pronounced hyperlipoproteinemia as evidenced by milky plasma and the presence of lipid granules that occluded kidney and liver capillaries. By NMR spectroscopy of plasma, the nature of hyperlipoproteinemia was identified as massive accumulation of chylomicron remnants. Plasma levels of low density lipoproteins (LDL) were also significantly increased as were those of cholesterol and triglycerides. In contrast, plasma levels of high density lipoproteins (HDL), albumin and total protein were reduced. At around three weeks of life, Plvap-deficient livers developed extensive multivesicular steatosis, steatohepatitis, and fibrosis. PLVAP is critically required for the formation of fenestrations in LSEC. Lack of fenestrations caused by PLVAP deficiency substantially impairs the passage of chylomicron remnants between liver sinusoids and hepatocytes, and finally leads to liver damage.
Collapse
Affiliation(s)
- Leonie Herrnberger
- Department of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Robert Hennig
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Werner Kremer
- Department of Biophysics and Physical Biochemistry, and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Regensburg, Germany
| | - Claus Hellerbrand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Hans Robert Kalbitzer
- Department of Biophysics and Physical Biochemistry, and Centre of Magnetic Resonance in Chemistry and Biomedicine, University of Regensburg, Regensburg, Germany
| | - Ernst R. Tamm
- Department of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
- * E-mail:
| |
Collapse
|
506
|
Azizi PM, Zyla RE, Guan S, Wang C, Liu J, Bolz SS, Heit B, Klip A, Lee WL. Clathrin-dependent entry and vesicle-mediated exocytosis define insulin transcytosis across microvascular endothelial cells. Mol Biol Cell 2014; 26:740-50. [PMID: 25540431 PMCID: PMC4325843 DOI: 10.1091/mbc.e14-08-1307] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
How insulin traverses the continuous endothelium of the microvasculature has been poorly studied. Development of a novel assay to measure insulin transcytosis reveals an unexpected role for clathrin in insulin transendothelial transport. Insulin transcytosis is dynamin and clathrin dependent but does not require cholesterol or caveolin-1. Transport of insulin across the microvasculature is necessary to reach its target organs (e.g., adipose and muscle tissues) and is rate limiting in insulin action. Morphological evidence suggests that insulin enters endothelial cells of the microvasculature, and studies with large vessel–derived endothelial cells show insulin uptake; however, little is known about the actual transcytosis of insulin and how this occurs in the relevant microvascular endothelial cells. We report an approach to study insulin transcytosis across individual, primary human adipose microvascular endothelial cells (HAMECs), involving insulin uptake followed by vesicle-mediated exocytosis visualized by total internal reflection fluorescence microscopy. In this setting, fluorophore-conjugated insulin exocytosis depended on its initial binding and uptake, which was saturable and much greater than in muscle cells. Unlike its degradation within muscle cells, insulin was stable within HAMECs and escaped lysosomal colocalization. Insulin transcytosis required dynamin but was unaffected by caveolin-1 knockdown or cholesterol depletion. Instead, insulin transcytosis was significantly inhibited by the clathrin-mediated endocytosis inhibitor Pitstop 2 or siRNA-mediated clathrin depletion. Accordingly, insulin internalized for 1 min in HAMECs colocalized with clathrin far more than with caveolin-1. This study constitutes the first evidence of vesicle-mediated insulin transcytosis and highlights that its initial uptake is clathrin dependent and caveolae independent.
Collapse
Affiliation(s)
- Paymon M Azizi
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada Programme in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Roman E Zyla
- Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Sha Guan
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada Programme in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Changsen Wang
- Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada
| | - Jun Liu
- Programme in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | | | - Bryan Heit
- Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Amira Klip
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada Programme in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Warren L Lee
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada Keenan Research Centre, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada Department of Microbiology and Immunology, University of Western Ontario, London, ON N6A 5C1, Canada Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
507
|
Zhou X, Chen M, Zeng X, Yang J, Deng H, Yi L, Mi MT. Resveratrol regulates mitochondrial reactive oxygen species homeostasis through Sirt3 signaling pathway in human vascular endothelial cells. Cell Death Dis 2014; 5:e1576. [PMID: 25522270 PMCID: PMC4454164 DOI: 10.1038/cddis.2014.530] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 10/06/2014] [Accepted: 11/04/2014] [Indexed: 12/21/2022]
Abstract
Mitochondrial reactive oxygen species (mtROS) homeostasis plays an essential role in preventing oxidative injury in endothelial cells, an initial step in atherogenesis. Resveratrol (RSV) possesses a variety of cardioprotective activities, however, little is known regarding the effects of RSV on mtROS homeostasis in endothelial cells. Sirt3 is a mitochondrial deacetylase, which plays a key role in mitochondrial bioenergetics and is closely associated with oxidative stress. The goal of the study is to investigate whether RSV could attenuate oxidative injury in endothelial cells via mtROS homeostasis regulation through Sirt3 signaling pathway. We found that pretreatment with RSV suppressed tert-butyl hydroperoxide (t-BHP)-induced oxidative damage in human umbilical vein endothelial cells (HUVECs) by increasing cell viability, inhibiting cell apoptosis, repressing collapse of mitochondrial membrane potential and decreasing mtROS generation. Moreover, the enzymatic activities of isocitrate dehydrogenase 2 (IDH2), glutathione peroxidase (GSH-Px) and manganese superoxide dismutase (SOD2) as well as deacetylation of SOD2 were increased by RSV pretreatment, suggesting RSV notably enhanced mtROS scavenging in t-BHP-induced endothelial cells. Meanwhile, RSV remarkably reduced mtROS generation by promoting Sirt3 enrichment within the mitochondria and subsequent upregulation of forkhead box O3A (FoxO3A)-mediated mitochondria-encoded gene expression of ATP6, CO1, Cytb, ND2 and ND5, thereby leading to increased complex I activity and ATP synthesis. Furthermore, RSV activated the expressions of phosphorylated adenosine monophosphate-activated protein kinase (p-AMPK), peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and Sirt3, as well as estrogen-related receptor-α (ERRα)-dependent Sirt3 mRNA transcription, which were abolished in the presence of AMPK inhibitor and AMPK, PGC-1α or Sirt3 siRNA transfection, indicating the effects of RSV on mtROS homeostasis regulation were dependent on AMPK-PGC-1α-ERRα-Sirt3 signaling pathway. Our findings indicated a novel mechanism that RSV-attenuated oxidative injury in endothelial cells through the regulation of mtROS homeostasis, which, in part, was mediated through the activation of the Sirt3 signaling pathway.
Collapse
Affiliation(s)
- X Zhou
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| | - M Chen
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| | - X Zeng
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| | - J Yang
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| | - H Deng
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| | - L Yi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| | - M T Mi
- Research Center for Nutrition and Food Safety, Chongqing Key Laboratory of Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Medical Nutrition Research Center, Chongqing 400038, PR China
| |
Collapse
|
508
|
Alinovi R, Goldoni M, Pinelli S, Campanini M, Aliatis I, Bersani D, Lottici PP, Iavicoli S, Petyx M, Mozzoni P, Mutti A. Oxidative and pro-inflammatory effects of cobalt and titanium oxide nanoparticles on aortic and venous endothelial cells. Toxicol In Vitro 2014; 29:426-37. [PMID: 25526690 DOI: 10.1016/j.tiv.2014.12.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 11/17/2022]
Abstract
Ultra-fine particles have recently been included among the risk factors for the development of endothelium inflammation and atherosclerosis, and cobalt (CoNPs) and titanium oxide nanoparticles (TiNPs) have attracted attention because of their wide range of applications. We investigated their toxicity profiles in two primary endothelial cell lines derived from human aorta (HAECs) and human umbilical vein (HUVECs) by comparing cell viability, oxidative stress, the expression of adhesion molecules and the release of chemokines during NP exposure. Both NPs were very rapidly internalised, and significantly increased adhesion molecule (ICAM-1, VCAM-1, E-selectin) mRNA and protein levels and the release of monocyte chemoattractant protein-1 (MCP-1) and interleukin 8 (IL-8). However, unlike the TiNPs, the CoNPs also induced time- and concentration-dependent metabolic impairment and oxidative stress without any evident signs of cell death or the induction of apoptosis. There were differences between the HAECs and HUVECs in terms of the extent of oxidative stress-related enzyme and vascular adhesion molecule expression, ROS production, and pro-inflammatory cytokine release despite the similar rate of NP internalisation, thus indicating endothelium heterogeneity in response to exogenous stimuli. Our data indicate that NPs can induce endothelial inflammatory responses via various pathways not involving only oxidative stress.
Collapse
Affiliation(s)
- Rossella Alinovi
- Department of Clinical and Experimental Medicine, University of Parma, Italy
| | - Matteo Goldoni
- Department of Clinical and Experimental Medicine, University of Parma, Italy.
| | - Silvana Pinelli
- Department of Clinical and Experimental Medicine, University of Parma, Italy
| | | | - Irene Aliatis
- Department of Physics and Earth Sciences, University of Parma, Italy
| | - Danilo Bersani
- Department of Physics and Earth Sciences, University of Parma, Italy
| | | | - Sergio Iavicoli
- Italian Workers' Compensation Authority (INAIL), Research Area, Department of Occupational Hygiene, Rome, Italy
| | - Marta Petyx
- Italian Workers' Compensation Authority (INAIL), Research Area, Department of Occupational Hygiene, Rome, Italy
| | - Paola Mozzoni
- Department of Clinical and Experimental Medicine, University of Parma, Italy; Italian Workers' Compensation Authority (INAIL), Research Center at the University of Parma, Italy
| | - Antonio Mutti
- Department of Clinical and Experimental Medicine, University of Parma, Italy
| |
Collapse
|
509
|
Xu N, Cioffi DL, Alexeyev M, Rich TC, Stevens T. Sodium entry through endothelial store-operated calcium entry channels: regulation by Orai1. Am J Physiol Cell Physiol 2014; 308:C277-88. [PMID: 25428882 DOI: 10.1152/ajpcell.00063.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Orai1 interacts with transient receptor potential protein of the canonical subfamily (TRPC4) and contributes to calcium selectivity of the endothelial cell store-operated calcium entry current (ISOC). Orai1 silencing increases sodium permeability and decreases membrane-associated calcium, although it is not known whether Orai1 is an important determinant of cytosolic sodium transitions. We test the hypothesis that, upon activation of store-operated calcium entry channels, Orai1 is a critical determinant of cytosolic sodium transitions. Activation of store-operated calcium entry channels transiently increased cytosolic calcium and sodium, characteristic of release from an intracellular store. The sodium response occurred more abruptly and returned to baseline more rapidly than did the transient calcium rise. Extracellular choline substitution for sodium did not inhibit the response, although 2-aminoethoxydiphenyl borate and YM-58483 reduced it by ∼50%. After this transient response, cytosolic sodium continued to increase due to influx through activated store-operated calcium entry channels. The magnitude of this sustained increase in cytosolic sodium was greater when experiments were conducted in low extracellular calcium and when Orai1 expression was silenced; these two interventions were not additive, suggesting a common mechanism. 2-Aminoethoxydiphenyl borate and YM-58483 inhibited the sustained increase in cytosolic sodium, only in the presence of Orai1. These studies demonstrate that sodium permeates activated store-operated calcium entry channels, resulting in an increase in cytosolic sodium; the magnitude of this response is determined by Orai1.
Collapse
Affiliation(s)
- Ningyong Xu
- Department of Pharmacology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Mikhail Alexeyev
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Thomas C Rich
- Department of Pharmacology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Pharmacology, University of South Alabama, Mobile, Alabama; Department of Medicine, University of South Alabama, Mobile, Alabama; and Center for Lung Biology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
510
|
Ngo JP, Kar S, Kett MM, Gardiner BS, Pearson JT, Smith DW, Ludbrook J, Bertram JF, Evans RG. Vascular geometry and oxygen diffusion in the vicinity of artery-vein pairs in the kidney. Am J Physiol Renal Physiol 2014; 307:F1111-22. [DOI: 10.1152/ajprenal.00382.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal arterial-to-venous (AV) oxygen shunting limits oxygen delivery to renal tissue. To better understand how oxygen in arterial blood can bypass renal tissue, we quantified the radial geometry of AV pairs and how it differs according to arterial diameter and anatomic location. We then estimated diffusion of oxygen in the vicinity of arteries of typical geometry using a computational model. The kidneys of six rats were perfusion fixed, and the vasculature was filled with silicone rubber (Microfil). A single section was chosen from each kidney, and all arteries ( n = 1,628) were identified. Intrarenal arteries were largely divisible into two “types,” characterized by the presence or absence of a close physical relationship with a paired vein. Arteries with a close physical relationship with a paired vein were more likely to have a larger rather than smaller diameter, and more likely to be in the inner-cortex than the mid- or outer cortex. Computational simulations indicated that direct diffusion of oxygen from an artery to a paired vein can only occur when the two vessels have a close physical relationship. However, even in the absence of this close relationship oxygen can diffuse from an artery to periarteriolar capillaries and venules. Thus AV oxygen shunting in the proximal preglomerular circulation is dominated by direct diffusion of oxygen to a paired vein. In the distal preglomerular circulation, it may be sustained by diffusion of oxygen from arteries to capillaries and venules close to the artery wall, which is subsequently transported to renal veins by convection.
Collapse
Affiliation(s)
- Jennifer P. Ngo
- Department of Physiology, Monash University, Melbourne, Australia
| | - Saptarshi Kar
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | - Michelle M. Kett
- Department of Physiology, Monash University, Melbourne, Australia
| | - Bruce S. Gardiner
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | - James T. Pearson
- Department of Physiology, Monash University, Melbourne, Australia
- Monash Biomedical Imaging Facility, Monash University, Melbourne, Australia
| | - David W. Smith
- School of Computer Science and Software Engineering, The University of Western Australia, Perth, Australia; and
| | | | - John F. Bertram
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Roger G. Evans
- Department of Physiology, Monash University, Melbourne, Australia
| |
Collapse
|
511
|
Branco-Price C, Evans CE, Johnson RS. Endothelial hypoxic metabolism in carcinogenesis and dissemination: HIF-A isoforms are a NO metastatic phenomenon. Oncotarget 2014; 4:2567-76. [PMID: 24318195 PMCID: PMC3926849 DOI: 10.18632/oncotarget.1461] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tumor biology is a broad and encompassing field of research, particularly given recent demonstrations of the multicellular nature of solid tumors, which have led to studies of molecular and metabolic intercellular interactions that regulate cancer progression. Hypoxia is a broad stimulus that results in activation of hypoxia inducible factors (HIFs). Downstream HIF targets include angiogenic factors (e.g. vascular endothelial growth factor, VEGF) and highly reactive molecules (e.g. nitric oxide, NO) that act as cell-specific switches with unique spatial and temporal effects on cancer progression. The effect of cell-specific responses to hypoxia on tumour progression and spread, as well as potential therapeutic strategies to target metastatic disease, are currently under active investigation. Vascular endothelial remodelling events at tumour and metastatic sites are responsive to hypoxia, HIF activation, and NO signalling. Here, we describe the interactions between endothelial HIF and NO during tumor growth and spread, and outline the effects of endothelial HIF/NO signalling on cancer progression. In doing so, we attempt to identify areas of metastasis research that require attention, in order to ultimately facilitate the development of novel treatments that reduce or prevent tumour dissemination.
Collapse
Affiliation(s)
- Cristina Branco-Price
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
512
|
Pober JS, Sessa WC. Inflammation and the blood microvascular system. Cold Spring Harb Perspect Biol 2014; 7:a016345. [PMID: 25384307 DOI: 10.1101/cshperspect.a016345] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Acute and chronic inflammation is associated with changes in microvascular form and function. At rest, endothelial cells maintain a nonthrombogenic, nonreactive surface at the interface between blood and tissue. However, on activation by proinflammatory mediators, the endothelium becomes a major participant in the generation of the inflammatory response. These functions of endothelium are modified by the other cell populations of the microvessel wall, namely pericytes, and smooth muscle cells. This article reviews recent advances in understanding the roles played by microvessels in inflammation.
Collapse
Affiliation(s)
- Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520-8089
| | - William C Sessa
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520-8089
| |
Collapse
|
513
|
Li F, Yuan Y, Guo Y, Liu N, Jing D, Wang H, Guo W. Pulsed magnetic field accelerate proliferation and migration of cardiac microvascular endothelial cells. Bioelectromagnetics 2014; 36:1-9. [PMID: 25338938 DOI: 10.1002/bem.21875] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 07/17/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Fei Li
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Yuan Yuan
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Ying Guo
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Nan Liu
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Da Jing
- Faculty of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Haichang Wang
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Wenyi Guo
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an China
| |
Collapse
|
514
|
Sumagin R, Sarelius IH. Emerging understanding of roles for arterioles in inflammation. Microcirculation 2014; 20:679-92. [PMID: 23701383 DOI: 10.1111/micc.12068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 05/20/2013] [Indexed: 12/20/2022]
Abstract
Arterioles, capillaries, and venules all actively change their cellular functions and phenotypes during inflammation in ways that are essential for maintenance of homeostasis and self-defense, and are also associated with many inflammatory disorders. ECs, together with pericytes and ECM proteins, can regulate blood flow, the coagulation cascade, fluid and solute exchange, and leukocyte trafficking. While capillary and venular functions in inflammation are well characterized, the arteriolar contribution to inflammation has only recently come into focus. Arterioles differ from venules in structure, EC morphology, shear environment, expression, and distribution of surface ligands; hence, regulation and function of arteriolar wall cells during inflammation may also be distinct from venules. Recent work indicates that in response to proinflammatory stimuli, arterioles alter barrier function, and support leukocyte and platelet interactions through upregulation of adhesion molecules. This suggests that in addition to their role in blood flow regulation, arterioles may also participate in inflammatory responses. In this review, we will discuss mechanisms that characterize arteriolar responses to proinflammatory stimuli. We will detail how distinct arteriolar features contribute to regulation of barrier function and leukocyte-EC interactions in inflammation, and further highlight the potential priming effects of arteriolar responses on venular function and progression of inflammatory responses.
Collapse
Affiliation(s)
- Ronen Sumagin
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | | |
Collapse
|
515
|
Tran-Dinh A, Diallo D, Delbosc S, Varela-Perez LM, Dang QB, Lapergue B, Burillo E, Michel JB, Levoye A, Martin-Ventura JL, Meilhac O. HDL and endothelial protection. Br J Pharmacol 2014; 169:493-511. [PMID: 23488589 DOI: 10.1111/bph.12174] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/07/2013] [Accepted: 02/24/2013] [Indexed: 12/23/2022] Open
Abstract
High-density lipoproteins (HDLs) represent a family of particles characterized by the presence of apolipoprotein A-I (apoA-I) and by their ability to transport cholesterol from peripheral tissues back to the liver. In addition to this function, HDLs display pleiotropic effects including antioxidant, anti-apoptotic, anti-inflammatory, anti-thrombotic or anti-proteolytic properties that account for their protective action on endothelial cells. Vasodilatation via production of nitric oxide is also a hallmark of HDL action on endothelial cells. Endothelial cells express receptors for apoA-I and HDLs that mediate intracellular signalling and potentially participate in the internalization of these particles. In this review, we will detail the different effects of HDLs on the endothelium in normal and pathological conditions with a particular focus on the potential use of HDL therapy to restore endothelial function and integrity.
Collapse
|
516
|
Cruz-Chu ER, Malafeev A, Pajarskas T, Pivkin IV, Koumoutsakos P. Structure and response to flow of the glycocalyx layer. Biophys J 2014; 106:232-43. [PMID: 24411255 DOI: 10.1016/j.bpj.2013.09.060] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 09/03/2013] [Accepted: 09/30/2013] [Indexed: 12/31/2022] Open
Abstract
The glycocalyx is a sugar-rich layer located at the luminal part of the endothelial cells. It is involved in key metabolic processes and its malfunction is related to several diseases. To understand the function of the glycocalyx, a molecular level characterization is necessary. In this article, we present large-scale molecular-dynamics simulations that provide a comprehensive description of the structure and dynamics of the glycocalyx. We introduce the most detailed, to-date, all-atom glycocalyx model, composed of lipid bilayer, proteoglycan dimers, and heparan sulfate chains with realistic sequences. Our results reveal the folding of proteoglycan ectodomain and the extended conformation of heparan sulfate chains. Furthermore, we study the glycocalyx response under shear flow and its role as a flypaper for binding fibroblast growth factors (FGFs), which are involved in diverse functions related to cellular differentiation, including angiogenesis, morphogenesis, and wound healing. The simulations show that the glycocalyx increases the effective concentration of FGFs, leading to FGF oligomerization, and acts as a lever to transfer mechanical stimulus into the cytoplasmic side of endothelial cells.
Collapse
Affiliation(s)
- Eduardo R Cruz-Chu
- Computational Science and Engineering Laboratory, ETH, Zurich, Switzerland
| | - Alexander Malafeev
- Scientific Computing Group, Institute of Computational Science, University of Lugano, Lugano, Switzerland
| | | | - Igor V Pivkin
- Scientific Computing Group, Institute of Computational Science, University of Lugano, Lugano, Switzerland
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, ETH, Zurich, Switzerland; Scientific Computing Group, Institute of Computational Science, University of Lugano, Lugano, Switzerland.
| |
Collapse
|
517
|
Fadini GP, Ferraro F, Quaini F, Asahara T, Madeddu P. Concise review: diabetes, the bone marrow niche, and impaired vascular regeneration. Stem Cells Transl Med 2014; 3:949-57. [PMID: 24944206 PMCID: PMC4116251 DOI: 10.5966/sctm.2014-0052] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/15/2014] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus is a global health problem that results in multiorgan complications leading to high morbidity and mortality. Until recently, the effects of diabetes and hyperglycemia on the bone marrow microenvironment-a site where multiple organ systems converge and communicate-have been underappreciated. However, several new studies in mice, rats, and humans reveal that diabetes leads to multiple bone marrow microenvironmental defects, such as small vessel disease (microangiopathy), nerve terminal pauperization (neuropathy), and impaired stem cell mobilization (mobilopathy). The discovery that diabetes involves bone marrow-derived progenitors implicated in maintaining cardiovascular homeostasis has been proposed as a bridging mechanism between micro- and macroangiopathy in distant organs. Herein, we review the physiological and molecular bone marrow abnormalities associated with diabetes and discuss how bone marrow dysfunction represents a potential root for the development of the multiorgan failure characteristic of advanced diabetes. The notion of diabetes as a bone marrow and stem cell disease opens new avenues for therapeutic interventions ultimately aimed at improving the outcome of diabetic patients.
Collapse
Affiliation(s)
- Gian Paolo Fadini
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Francesca Ferraro
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Federico Quaini
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Takayuki Asahara
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Department of Medicine, University of Padova, Padova, Italy; Venetian Institute of Molecular Medicine, Padova, Italy; Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania, USA; Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy; Division of Regenerative Medicine, Department of Basic Clinical Science, Tokai University, Tokyo, Japan; Regenerative Medicine Section, Bristol Heart Institute, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
518
|
Zhou Y, Wang Y, Tischfield M, Williams J, Smallwood PM, Rattner A, Taketo MM, Nathans J. Canonical WNT signaling components in vascular development and barrier formation. J Clin Invest 2014; 124:3825-46. [PMID: 25083995 DOI: 10.1172/jci76431] [Citation(s) in RCA: 252] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 06/12/2014] [Indexed: 01/10/2023] Open
Abstract
Canonical WNT signaling is required for proper vascularization of the CNS during embryonic development. Here, we used mice with targeted mutations in genes encoding canonical WNT pathway members to evaluate the exact contribution of these components in CNS vascular development and in specification of the blood-brain barrier (BBB) and blood-retina barrier (BRB). We determined that vasculature in various CNS regions is differentially sensitive to perturbations in canonical WNT signaling. The closely related WNT signaling coreceptors LDL receptor-related protein 5 (LRP5) and LRP6 had redundant functions in brain vascular development and barrier maintenance; however, loss of LRP5 alone dramatically altered development of the retinal vasculature. The BBB in the cerebellum and pons/interpeduncular nuclei was highly sensitive to decrements in canonical WNT signaling, and WNT signaling was required to maintain plasticity of barrier properties in mature CNS vasculature. Brain and retinal vascular defects resulting from ablation of Norrin/Frizzled4 signaling were ameliorated by stabilizing β-catenin, while inhibition of β-catenin-dependent transcription recapitulated the vascular development and barrier defects associated with loss of receptor, coreceptor, or ligand, indicating that Norrin/Frizzled4 signaling acts predominantly through β-catenin-dependent transcriptional regulation. Together, these data strongly support a model in which identical or nearly identical canonical WNT signaling mechanisms mediate neural tube and retinal vascularization and maintain the BBB and BRB.
Collapse
|
519
|
Chen YE, Xie C, Yang B. Stem cells for vascular engineering. BIOMATERIALS AND REGENERATIVE MEDICINE 2014:621-639. [DOI: 10.1017/cbo9780511997839.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
520
|
Wu S, Zhou C, King JAC, Stevens T. A unique pulmonary microvascular endothelial cell niche revealed by Weibel-Palade bodies and Griffonia simplicifolia. Pulm Circ 2014; 4:110-5. [PMID: 25006426 DOI: 10.1086/674879] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 11/13/2013] [Indexed: 12/15/2022] Open
Abstract
Pulmonary endothelium displays considerable heterogeneity along the vascular axis, from arteries to capillaries to veins. Griffonia simplicifolia is a lectin that recognizes pulmonary microvascular endothelium with preference over extra-alveolar endothelium in both arteries and veins, yet the precise vascular location where this phenotypic shift occurs is poorly resolved. We gelatin-filled the circulation and agarose-loaded the airways and then labeled the lung with Griffonia lectin to enable visualization of the endothelial transition zone. Endothelium in vessels with internal diameters less than 38 μm were uniformly Griffonia positive, whereas vessels with internal diameters greater than 60 μm were always Griffonia negative. Two populations of endothelium were identified in vessels ranging from 38 to 60 μm in diameter, including some that were positive and others that were negative for binding to G. simplicifolia. To better resolve this endothelial transition zone, we performed morphology studies to measure the distribution of Weibel-Palade bodies (WPbs), since WPbs are present in conduit vessel endothelium and absent in capillary endothelium. WPbs were found in endothelium with vascular dimensions as small as 18 μm in diameter but were not found in capillaries. Thus, we identify with precision that the endothelial phenotype transition from a cell that does not interact with Griffonia lectin to one that does occurs in blood vessels with internal diameters of approximately 38 μm, and we reveal an unappreciated vascular zone, between 18 and 38 μm in diameter, where endothelium both is Griffonia positive and possesses WPbs.
Collapse
Affiliation(s)
- Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, Georgia, USA
| | - Chun Zhou
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Judy A C King
- Department of Pathology, West Virginia University, Morgantown, West Virginia, USA
| | - Troy Stevens
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA ; Departments of Pharmacology and Internal Medicine, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
521
|
Faust DM, Marquay Markiewicz J, Santi-Rocca J, Guillen N. New insights into host-pathogen interactions during Entamoeba histolytica liver infection. Eur J Microbiol Immunol (Bp) 2014; 1:10-8. [PMID: 24466432 DOI: 10.1556/eujmi.1.2011.1.4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Amoebiasis is the third worldwide disease due to a parasite. The causative agent of this disease, the unicellular eukaryote Entamoeba histolytica, causes dysentery and liver abscesses associated with inflammation and human cell death. During liver invasion, before entering the parenchyma, E. histolytica trophozoites are in contact with liver sinusoidal endothelial cells (LSEC). We present data characterizing human LSEC responses to interaction with E. histolytica and identifying amoebic factors involved in the process of cell death in this cell culture model potentially relevant for early steps of hepatic amoebiasis. E. histolytica interferes with host cell adhesion signalling and leads to diminished adhesion and target cell death. Contact with parasites induces disruption of actin stress fibers and focal adhesion complexes. We conclude that interference with LSEC signalling may result from amoeba-triggered changes in the mechanical forces in the vicinity of cells in contact with parasites, sensed and transmitted by focal adhesion complexes. The study highlights for the first time the potential role in the onset of hepatic amoebiasis of the loss of liver endothelium integrity by disturbance of focal adhesion function and adhesion signalling. Among the amoebic factors required for changed LSEC adherence properties we identified the Gal/GalNAC lectin, cysteine proteases and KERP1.
Collapse
Affiliation(s)
- D M Faust
- Institut Pasteur, Cell Biology of Parasitism Unit Inserm U786, Paris France
| | | | - J Santi-Rocca
- Institut Pasteur, Cell Biology of Parasitism Unit Inserm U786, Paris France
| | - N Guillen
- Institut Pasteur, Cell Biology of Parasitism Unit Inserm U786, Paris France
| |
Collapse
|
522
|
Galectin-3 expressed on different lung compartments promotes organ specific metastasis by facilitating arrest, extravasation and organ colonization via high affinity ligands on melanoma cells. Clin Exp Metastasis 2014; 31:661-73. [PMID: 24952269 DOI: 10.1007/s10585-014-9657-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 05/14/2014] [Indexed: 12/28/2022]
Abstract
Interactions between molecules on the surface of tumor cells and those on the target organ endothelium play an important role in their arrest in an organ. Galectin-3 on the lung endothelium and high affinity ligands poly-N-acetyllactosamine (polyLacNAc) on N-oligosaccharides on melanoma cells facilitate such interactions. However, to extravasate and colonize an organ the cells must stabilize these interactions by spreading to retract endothelium, degrade exposed basement membrane (BM) and move into parenchyma and proliferate. Here, we show that galectin-3 is expressed on all the major compartments of the lungs and participates in not just promoting adhesion but also in spreading. We for the first time demonstrate that both soluble and immobilized galectin-3 induce secretion of MMP-9 required to breach vascular BM. Further, we show that immobilized galectin-3 is used as traction for the movement of cells. Downregulation of galactosyltransferases-I and -V resulted in significant loss in expression of polyLacNAc and thus reduced binding of galectin-3. This was accompanied with a loss in adhesion, spreading, MMP-9 secretion and motility of the cells on galectin-3 and thus their metastasis to lungs. Metastasis could also be inhibited by blocking surface polyLacNAc by pre-incubating cells with truncated galectin-3 (which lacked oligomerization domain) or by feeding mice with modified citrus pectin in drinking water. Overall, these results unequivocally show that polyLacNAc on melanoma cells and galectin-3 on the lungs play a critical role in arrest and extravasation of cells in the lungs and strategies that target these interactions inhibit lung metastasis.
Collapse
|
523
|
Abstract
Endothelial cells (ECs) exhibit dramatic plasticity of form at the single- and collective-cell level during new vessel growth, adult vascular homeostasis, and pathology. Understanding how, when, and why individual ECs coordinate decisions to change shape, in relation to the myriad of dynamic environmental signals, is key to understanding normal and pathological blood vessel behavior. However, this is a complex spatial and temporal problem. In this review we show that the multidisciplinary field of Adaptive Systems offers a refreshing perspective, common biological language, and straightforward toolkit that cell biologists can use to untangle the complexity of dynamic, morphogenetic systems.
Collapse
Affiliation(s)
- Katie Bentley
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Andrew Philippides
- Centre for Computational Neuroscience and Robotics, Department of Informatics, University of Sussex, Brighton BN1 9QJ, UK
| | - Erzsébet Ravasz Regan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
524
|
Kratzer A, Giral H, Landmesser U. High-density lipoproteins as modulators of endothelial cell functions: alterations in patients with coronary artery disease. Cardiovasc Res 2014; 103:350-61. [DOI: 10.1093/cvr/cvu139] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
525
|
O'Toole TE, Abplanalp W, Li X, Cooper N, Conklin DJ, Haberzettl P, Bhatnagar A. Acrolein decreases endothelial cell migration and insulin sensitivity through induction of let-7a. Toxicol Sci 2014; 140:271-82. [PMID: 24812010 DOI: 10.1093/toxsci/kfu087] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Acrolein is a major reactive component of vehicle exhaust, and cigarette and wood smoke. It is also present in several food substances and is generated endogenously during inflammation and lipid peroxidation. Although previous studies have shown that dietary or inhalation exposure to acrolein results in endothelial activation, platelet activation, and accelerated atherogenesis, the basis for these effects is unknown. Moreover, the effects of acrolein on microRNA (miRNA) have not been studied. Using AGILENT miRNA microarray high-throughput technology, we found that treatment of cultured human umbilical vein endothelial cells with acrolein led to a significant (>1.5-fold) upregulation of 12, and downregulation of 15, miRNAs. Among the miRNAs upregulated were members of the let-7 family and this upregulation was associated with decreased expression of their protein targets, β3 integrin, Cdc34, and K-Ras. Exposure to acrolein attenuated β3 integrin-dependent migration and reduced Akt phosphorylation in response to insulin. These effects of acrolein on endothelial cell migration and insulin signaling were reversed by expression of a let-7a inhibitor. Also, inhalation exposure of mice to acrolein (1 ppm x 6 h/day x 4 days) upregulated let-7a and led to a decrease in insulin-stimulated Akt phosphorylation in the aorta. These results suggest that acrolein exposure has broad effects on endothelial miRNA repertoire and that attenuation of endothelial cell migration and insulin signaling by acrolein is mediated in part by the upregulation of let-7a. This mechanism may be a significant feature of vascular injury caused by inflammation, oxidized lipids, and exposure to environmental pollutants.
Collapse
Affiliation(s)
| | | | - Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky 40202
| | - Nigel Cooper
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky 40202
| | | | | | | |
Collapse
|
526
|
Functional and morphological characteristics of the retinal and choroidal vasculature. Prog Retin Eye Res 2014; 40:53-93. [DOI: 10.1016/j.preteyeres.2014.02.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 11/24/2022]
|
527
|
Brugat T, Cunningham D, Sodenkamp J, Coomes S, Wilson M, Spence PJ, Jarra W, Thompson J, Scudamore C, Langhorne J. Sequestration and histopathology in Plasmodium chabaudi malaria are influenced by the immune response in an organ-specific manner. Cell Microbiol 2014; 16:687-700. [PMID: 24003897 PMCID: PMC4234010 DOI: 10.1111/cmi.12212] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/26/2013] [Accepted: 08/26/2013] [Indexed: 11/28/2022]
Abstract
Infection with the malaria parasite, Plasmodium, is associated with a strong inflammatory response and parasite cytoadhesion (sequestration) in several organs. Here, we have carried out a systematic study of sequestration and histopathology during infection of C57Bl/6 mice with Plasmodium chabaudi AS and determined the influence of the immune response. This parasite sequesters predominantly in liver and lung, but not in the brain, kidney or gut. Histopathological changes occur in multiple organs during the acute infection, but are not restricted to the organs where sequestration takes place. Adaptive immunity, and signalling through the IFNγ receptor increased sequestration and histopathology in the liver, but not in the lung, suggesting that there are differences in the adhesion molecules and/or parasite ligands utilized and mechanisms of pathogenesis in these two organs. Exacerbation of pro-inflammatory responses during infection by deletion of the il10 gene resultsin the aggravation of damage to lung and kidney irrespective of the degree of sequestration. The immune response therefore affected both sequestration and histopathology in an organ-specific manner. P. chabaudi AS provides a good model to investigate the influence of the host response on the sequestration and specific organ pathology, which is applicable to human malaria.
Collapse
Affiliation(s)
- Thibaut Brugat
- Division of Parasitology, MRC National Institute for Medical Research, London, NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
528
|
van Sloten TT, Schram MT, Adriaanse MC, Dekker JM, Nijpels G, Teerlink T, Scheffer PG, Pouwer F, Schalkwijk CG, Stehouwer CDA, Henry RMA. Endothelial dysfunction is associated with a greater depressive symptom score in a general elderly population: the Hoorn Study. Psychol Med 2014; 44:1403-1416. [PMID: 23942242 DOI: 10.1017/s0033291713002043] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Endothelial dysfunction (ED), low-grade inflammation (LGI) and oxidative stress (OxS) may be involved in the pathobiology of depression. Previous studies on the association of these processes in depression have yielded contradictory results. We therefore investigated comprehensively, in a population-based cohort study, the association between ED, LGI and OxS on the one hand and depressive symptoms on the other. METHOD We used data from the Hoorn Study and determined biomarkers of ED [flow-mediated dilatation (FMD), von Willebrand factor, soluble intercellular adhesion molecule 1 (sICAM-1), soluble vascular cell adhesion molecule 1, soluble thrombomodulin and soluble endothelial selectin], LGI [C-reactive protein, tumour necrosis factor-α, interleukin 6, interleukin 8, serum amyloid A, myeloperoxidase (MPO) and sICAM-1] and OxS (oxidized low density lipoprotein and MPO). Depressive symptoms were quantified by the Center for Epidemiologic Studies Depression Scale (CES-D) questionnaire (n = 493; age 68 years; 49.9% female). Regression analyses were performed with the use of biomarker Z scores. Adjustments were made for age, sex and glucose metabolism status (cohort stratification variables) and prior cardiovascular disease, hypertension, waist-to-hip ratio, cholesterol levels, education level, physical activity, dietary habits, and the use of antihypertensive and/or lipid-lowering medication and/or metformin (potential confounders). RESULTS After adjustment for age, sex and glucose metabolism status, one standard deviation increase in the ED Z score was associated with a 1.9 [95% confidence interval (CI) 0.7-3.1] higher CES-D score. Additional adjustments did not materially change this result. LGI and OxS were not associated with the CES-D score. CONCLUSIONS ED, as quantified by an array of circulating biomarkers and FMD, was independently associated with depressive symptoms. This study supports the hypothesis that ED plays an important role in the pathobiology of depression.
Collapse
Affiliation(s)
- T T van Sloten
- Department of Medicine, Maastricht University Medical Centre, The Netherlands
| | - M T Schram
- Department of Medicine, Maastricht University Medical Centre, The Netherlands
| | - M C Adriaanse
- EMGO Institute for Health and Care Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - J M Dekker
- EMGO Institute for Health and Care Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - G Nijpels
- EMGO Institute for Health and Care Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - T Teerlink
- Metabolic Laboratory, Department of Clinical Chemistry, VU University Medical Centre, Amsterdam, The Netherlands
| | - P G Scheffer
- Metabolic Laboratory, Department of Clinical Chemistry, VU University Medical Centre, Amsterdam, The Netherlands
| | - F Pouwer
- Centre of Research on Psychology in Somatic Diseases, Department of Medical Psychology and Neuropsychology, Tilburg University, The Netherlands
| | - C G Schalkwijk
- Department of Medicine, Maastricht University Medical Centre, The Netherlands
| | - C D A Stehouwer
- Department of Medicine, Maastricht University Medical Centre, The Netherlands
| | - R M A Henry
- Department of Medicine, Maastricht University Medical Centre, The Netherlands
| |
Collapse
|
529
|
Abstract
The significant role of platelets and P-selectin in assisting tumor cell metastasis to the lungs has been frequently reported and reviewed. However, evidence recently has come to light on other pro-metastatic mechanisms of platelets beyond that of tumor cell protection from immune cell attack and aiding extravasation, such as promoting epithelial to mesenchymal transition in tumor cells and conveying signals from the primary tumor to distant tissues that optimize conditions for metastasis. Moreover, the role of platelets and selectins in hematogenous metastasis to frequently targeted organs other than the lungs has been less well examined. This review aims to summarize the literature on the roles of platelets in all stages of the metastatic process and to examine the participation of platelets and selectins in hematogenous metastasis to the lungs, liver, bone, and brain. In the light of the available evidence, potential therapeutic avenues for the control of metastasis are also discussed.
Collapse
Affiliation(s)
- Lucy A Coupland
- Cancer & Vascular Biology Group, Department of Immuology, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia; Clinical Haematology Unit, The Canberra Hospital, Garran, ACT, Australia
| | - Christopher R Parish
- Cancer & Vascular Biology Group, Department of Immuology, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
530
|
Brain microvascular endothelial cells resist elongation due to curvature and shear stress. Sci Rep 2014; 4:4681. [PMID: 24732421 PMCID: PMC3986701 DOI: 10.1038/srep04681] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/14/2014] [Indexed: 11/08/2022] Open
Abstract
The highly specialized endothelial cells in brain capillaries are a key component of the blood-brain barrier, forming a network of tight junctions that almost completely block paracellular transport. In contrast to vascular endothelial cells in other organs, we show that brain microvascular endothelial cells resist elongation in response to curvature and shear stress. Since the tight junction network is defined by endothelial cell morphology, these results suggest that there may be an evolutionary advantage to resisting elongation by minimizing the total length of cell-cell junctions per unit length of vessel.
Collapse
|
531
|
Ren X, Ott HC. On the road to bioartificial organs. Pflugers Arch 2014; 466:1847-57. [PMID: 24691559 DOI: 10.1007/s00424-014-1504-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 03/18/2014] [Indexed: 01/08/2023]
Abstract
Biological organs are highly orchestrated systems with well-coordinated positioning, grouping, and interaction of different cell types within their specialized extracellular environment. Bioartificial organs are intended to be functional replacements of native organs generated through bioengineering techniques and hold the potential to alleviate donor organ shortage for transplantation. The development, production, and evaluation of such bioartificial organs require synergistic efforts of biology, material science, engineering, and medicine. In this review, we highlight the emerging platforms enabling structured assembly of multiple cell types into functional grafts and discuss recent advances and challenges in the development of bioartificial organs, including cell sources, in vitro organ culture, in vivo evaluation, and clinical considerations.
Collapse
Affiliation(s)
- X Ren
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
532
|
Yoon N, Dang TQ, Chasiotis H, Kelly SP, Sweeney G. Altered transendothelial transport of hormones as a contributor to diabetes. Diabetes Metab J 2014; 38:92-9. [PMID: 24851202 PMCID: PMC4021306 DOI: 10.4093/dmj.2014.38.2.92] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The vascular endothelium is a dynamic structure responsible for the separation and regulated movement of biological material between circulation and interstitial fluid. Hormones and nutrients can move across the endothelium either via a transcellular or paracellular route. Transcellular endothelial transport is well understood and broadly acknowledged to play an important role in the normal and abnormal physiology of endothelial function. However, less is known about the role of the paracellular route. Although the concept of endothelial dysfunction in diabetes is now widely accepted, we suggest that alterations in paracellular transport should be studied in greater detail and incorporated into this model. In this review we provide an overview of endothelial paracellular permeability and discuss its potential importance in contributing to the development of diabetes and associated complications. Accordingly, we also contend that if better understood, altered endothelial paracellular permeability could be considered as a potential therapeutic target for diabetes.
Collapse
Affiliation(s)
- Nanyoung Yoon
- Department of Biology, York University, Toronto, ON, Canada
| | - Thanh Q. Dang
- Department of Biology, York University, Toronto, ON, Canada
| | | | - Scott P. Kelly
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
533
|
Human fetal liver cells for regulated ex vivo erythropoietin gene therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2014; 1:14003. [PMID: 26015950 PMCID: PMC4362349 DOI: 10.1038/mtm.2014.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/11/2014] [Indexed: 11/09/2022]
Abstract
Possible risks and lack of donor livers limit application of liver transplantation. Liver cell transplantation is, at this moment, not a feasible alternative because engraftment in the liver is poor. Furthermore, there is also shortage of cells suitable for transplantation. Fetal liver cells are able to proliferate in cell culture and could therefore present an alternative source of cells for transplantation. In this study, we investigated the utility of human fetal liver cells for therapeutic protein delivery. We transplanted human fetal liver cells in immunodeficient mice but were not able to detect engraftment of human hepatocytes. In contrast, transplantation of human adult hepatocytes led to detectable engraftment of hepatocytes in murine liver. Transplantation of fetal liver cells did lead to abundant reconstitution of murine liver with human endothelium, indicating that endothelial cells are the most promising cell type for ex vivo liver cell gene therapy. Human liver endothelial cells were subsequently transduced with a lentiviral autoregulatory erythropoietin expression vector. After transplantation in immunodeficient mice, these cells mediated long-term regulation of murine hematocrits. Our study shows the potential of human liver endothelial cells for long-term regulated gene therapy.
Collapse
|
534
|
Tognarelli S, Gayet J, Lambert M, Dupuy S, Karras A, Cohen P, Guillevin L, de Menthon M, Caillat-Zucman S. Tissue-specific microvascular endothelial cells show distinct capacity to activate NK cells: implications for the pathophysiology of granulomatosis with polyangiitis. THE JOURNAL OF IMMUNOLOGY 2014; 192:3399-408. [PMID: 24600034 DOI: 10.4049/jimmunol.1301508] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The relevance of tissue specificity of microvascular endothelial cells (MECs) in the response to inflammatory stimuli and sensitivity to immune cell-mediated injury is not well defined. We hypothesized that such MEC characteristics might shape their interaction with NK cells through the use of different adhesion molecules and NK cell receptor ligands or the release of different soluble factors and render them more or less vulnerable to NK cell injury during autoimmune vasculitis, such as granulomatosis with polyangiitis (GPA). To generate a comprehensive expression profile of human MECs of renal, lung, and dermal tissue origin, we characterized, in detail, their response to inflammatory cytokines and to proteinase 3, a major autoantigen in GPA, and analyzed the effects on NK cell activation. In this study, we show that renal MECs were more susceptible than lung and dermal MECs to the effect of inflammatory signals, showing upregulation of ICAM-1 and VCAM-1 on their surface, as well as release of CCL2, soluble fractalkine, and soluble VCAM-1. Proteinase 3-stimulated renal and lung MECs triggered CD107a degranulation in control NK cell. Notably, NK cells from GPA patients expressed markers of recent in vivo activation (CD69, CD107a), degranulated more efficiently than did control NK cells in the presence of renal MECs, and induced direct killing of renal MECs in vitro. These results suggest that, upon inflammatory conditions in GPA, renal MECs may contribute to the recruitment and activation of NK cells in the target vessel wall, which may participate in the necrotizing vasculitis of the kidney during this disease.
Collapse
Affiliation(s)
- Sara Tognarelli
- INSERM, U1016 Hôpital Saint-Vincent de Paul, 75014 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
535
|
Experimental tools to monitor the dynamics of endothelial barrier function: a survey of in vitro approaches. Cell Tissue Res 2014; 355:485-514. [DOI: 10.1007/s00441-014-1810-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/13/2014] [Indexed: 02/05/2023]
|
536
|
Higher central fat mass and lower peripheral lean mass are independent determinants of endothelial dysfunction in the elderly: The Hoorn study. Atherosclerosis 2014; 233:310-8. [DOI: 10.1016/j.atherosclerosis.2013.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 11/14/2013] [Accepted: 12/13/2013] [Indexed: 02/04/2023]
|
537
|
Favero G, Paganelli C, Buffoli B, Rodella LF, Rezzani R. Endothelium and its alterations in cardiovascular diseases: life style intervention. BIOMED RESEARCH INTERNATIONAL 2014; 2014:801896. [PMID: 24719887 PMCID: PMC3955677 DOI: 10.1155/2014/801896] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/11/2014] [Indexed: 01/07/2023]
Abstract
The endothelium, which forms the inner cellular lining of blood vessels and lymphatics, is a highly metabolically active organ that is involved in many physiopathological processes, including the control of vasomotor tone, barrier function, leukocyte adhesion, and trafficking and inflammation. In this review, we summarized and described the following: (i) endothelial cell function in physiological conditions and (ii) endothelial cell activation and dysfunction in the main cardiovascular diseases (such as atherosclerosis, and hypertension) and to diabetes, cigarette smoking, and aging physiological process. Finally, we presented the currently available evidence that supports the beneficial effects of physical activity and various dietary compounds on endothelial functions.
Collapse
Affiliation(s)
- Gaia Favero
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Corrado Paganelli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Barbara Buffoli
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi Fabrizio Rodella
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Rita Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
538
|
Rutkowski JM, Halberg N, Wang QA, Holland WL, Xia JY, Scherer PE. Differential transendothelial transport of adiponectin complexes. Cardiovasc Diabetol 2014; 13:47. [PMID: 24552349 PMCID: PMC3932731 DOI: 10.1186/1475-2840-13-47] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 02/12/2014] [Indexed: 12/15/2022] Open
Abstract
Background Adiponectin’s effects on systemic physiology and cell-specific responses are well-defined, but little is known about how this insulin-sensitizing and anti-inflammatory adipokine reaches its target cells. All molecules face active and passive transport limitations, but adiponectin is particularly noteworthy due to the diverse size range and high molecular weights of its oligomers. Additionally, its metabolic target organs possess a range of endothelial permeability. Methods Full-length recombinant murine adiponectin was produced and oligomer fractions isolated by gel filtration. Adiponectin complex sizes were measured by dynamic light scattering to determine Stokes radii. Transendothelial transport of purified oligomers was quantitatively assessed under a number of different conditions in vitro using murine endothelial cells and in vivo using several mouse models of altered endothelial function. Results Adiponectin oligomers exhibit large transport radii that limit transendothelial transport. Oligomerization is a significant determinant of flux across endothelial monolayers in vitro; low molecular weight adiponectin is preferentially transported. In vivo sampled sera from the heart, liver, and tail vein demonstrated significantly different complex distribution of lower molecular weight oligomers. Pharmacological interventions, such as PPARγ agonist treatment, differentially affect adiponectin plasma clearance and tissue uptake. Exercise induces enhanced adiponectin uptake to oxidative skeletal muscles, wherein adiponectin potently lowers ceramide levels. In total, endothelial barriers control adiponectin transport in a cell- and tissue-specific manner. Conclusions Adiponectin oligomer efficacy in a given tissue may therefore be endothelial transport mediated. Targeting endothelial dysfunction in the metabolic syndrome through exercise and pharmaceuticals may afford an effective approach to increasing adiponectin’s beneficial effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
539
|
Gui L, Niklason LE. Vascular Tissue Engineering: Building Perfusable Vasculature for Implantation. Curr Opin Chem Eng 2014; 3:68-74. [PMID: 24533306 DOI: 10.1016/j.coche.2013.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tissue and organ replacement is required when there are no alternative therapies available. Although vascular tissue engineering was originally developed to meet the clinical demands of small-diameter vascular conduits as bypass grafts, it has evolved into a highly advanced field where perfusable vasculatures are generated for implantation. Herein, we review several cutting-edge techniques that have led to implantable human blood vessels in clinical trials, the novel approaches that build complex perfusable microvascular networks in functional tissues, the use of stem cells to generate endothelial cells for vascularization, as well as the challenges in bringing vascular tissue engineering technologies into the clinics.
Collapse
Affiliation(s)
- Liqiong Gui
- Department of Anesthesiology, Yale University, New Haven, CT ; The Vascular Biology and Therapeutics Program, Yale University, New Haven, CT
| | - Laura E Niklason
- Department of Anesthesiology, Yale University, New Haven, CT ; The Vascular Biology and Therapeutics Program, Yale University, New Haven, CT ; Department of Biomedical Engineering, Yale University, New Haven, CT
| |
Collapse
|
540
|
Möbius-Winkler S, Linke A, Adams V, Schuler G, Erbs S. How to improve endothelial repair mechanisms: the lifestyle approach. Expert Rev Cardiovasc Ther 2014; 8:573-80. [DOI: 10.1586/erc.10.7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
541
|
Abstract
Current advances in nanotechnology have paved the way for the early detection, prevention and treatment of various diseases such as vascular disorders and cancer. These advances have provided novel approaches or modalities of incorporating or adsorbing therapeutic, biosensor and targeting agents into/on nanoparticles. With significant progress, nanomedicine for vascular therapy has shown significant advantages over traditional medicine because of its ability to selectively target the disease site and reduce adverse side effects. Targeted delivery of nanoparticles to vascular endothelial cells or the vascular wall provides an effective and more efficient way for early detection and/or treatment of vascular diseases such as atherosclerosis, thrombosis and Cerebrovascular Amyloid Angiopathy (CAA). Clinical applications of biocompatible and biodegradable polymers in areas such as vascular graft, implantable drug delivery, stent devices and tissue engineering scaffolds have advanced the candidature of polymers as potential nano-carriers for vascular-targeted delivery of diagnostic agents and drugs. This review focuses on the basic aspects of the vasculature and its associated diseases and relates them to polymeric nanoparticle-based strategies for targeting therapeutic agents to diseased vascular site.
Collapse
Affiliation(s)
- Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL ; Division of Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Karunyna Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
542
|
Hupe M, Li MX, Gertow Gillner K, Adams RH, Stenman JM. Evaluation of TRAP-sequencing technology with a versatile conditional mouse model. Nucleic Acids Res 2014; 42:e14. [PMID: 24165879 PMCID: PMC3902954 DOI: 10.1093/nar/gkt995] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/27/2013] [Accepted: 10/03/2013] [Indexed: 11/25/2022] Open
Abstract
Gene expression profiling of various cell lineages has provided invaluable insights into the molecular mechanisms regulating cellular development and differentiation. However, in vivo molecular profiling of rare and interspersed cell populations, such as endothelial cells, has remained challenging. We have generated a versatile floxed translating ribosome affinity purification (TRAP) mouse model, mCherryTRAP, for Cre-dependent translational profiling of distinct cell lineages from intact tissues. To identify cell type-specific transcripts using TRAP, the data have to be filtered to remove both background transcripts not expressed in the profiled cell type and transcripts expressed in all cell populations of the tissue/organ. Filtering has previously been achieved using transcribed RNA from the tissue/organ. Using the mCherryTRAP model, we demonstrate extensive differential expression of RNAs between the translatome and transcriptome of embryonic brains and kidneys. We evaluate the implications of these data for TRAP studies of abundant and rare cell populations. Finally, we demonstrate the applicability of the technology to study organ-specific endothelial cell differentiation.
Collapse
Affiliation(s)
- Mike Hupe
- Ludwig Institute for Cancer Research Ltd, Box 240, Stockholm SE-171 77, Sweden, Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden and Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Muenster, Germany
| | - Minerva Xueting Li
- Ludwig Institute for Cancer Research Ltd, Box 240, Stockholm SE-171 77, Sweden, Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden and Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Muenster, Germany
| | - Karin Gertow Gillner
- Ludwig Institute for Cancer Research Ltd, Box 240, Stockholm SE-171 77, Sweden, Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden and Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Muenster, Germany
| | - Ralf H. Adams
- Ludwig Institute for Cancer Research Ltd, Box 240, Stockholm SE-171 77, Sweden, Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden and Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Muenster, Germany
| | - Jan M. Stenman
- Ludwig Institute for Cancer Research Ltd, Box 240, Stockholm SE-171 77, Sweden, Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden and Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, and University of Münster, Faculty of Medicine, D-48149 Muenster, Germany
| |
Collapse
|
543
|
Dahlman JE, Kauffman KJ, Langer R, Anderson DG. Nanotechnology for in vivo targeted siRNA delivery. ADVANCES IN GENETICS 2014; 88:37-69. [PMID: 25409603 DOI: 10.1016/b978-0-12-800148-6.00003-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small interfering RNAs (siRNAs) can specifically inhibit gene expression. As a result, they have tremendous scientific and clinical potential. However, the use of these molecules in patients and animal models has been limited by challenges with delivery. Intracellular RNA delivery is difficult; it requires a system that protects the siRNA from degradative nucleases in the bloodstream, minimizes clearance by the reticuloendothelial system, maximizes delivery to the target tissue, and promotes entry into, and out of, an endocytic vesicle. Despite these barriers, recent data suggest that RNA may be targeted to cells of interest in vivo. Herein we outline strategies for targeted siRNA delivery, and describe how these strategies may be improved.
Collapse
Affiliation(s)
- James E Dahlman
- The Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kevin J Kauffman
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- The Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- The Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
544
|
Deletion of cavin genes reveals tissue-specific mechanisms for morphogenesis of endothelial caveolae. Nat Commun 2013; 4:1831. [PMID: 23652019 PMCID: PMC3674239 DOI: 10.1038/ncomms2808] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 03/26/2013] [Indexed: 12/23/2022] Open
Abstract
Caveolae are abundant in endothelial cells and are thought to have important roles in endothelial cell biology. The cavin proteins are key components of caveolae, and are expressed at varied amounts in different tissues. Here we use knockout mice to determine the roles of cavins 2 and 3 in caveolar morphogenesis in vivo. Deletion of cavin 2 causes loss of endothelial caveolae in lung and adipose tissue, but has no effect on the abundance of endothelial caveolae in heart and other tissues. Changes in the morphology of endothelium in cavin 2 null mice correlate with changes in caveolar abundance. Cavin 3 is not required for making caveolae in the tissues examined. Cavin 2 determines the size of cavin complexes, and acts to shape caveolae. Cavin 1, however, is essential for normal oligomerization of caveolin 1. Our data reveal that endothelial caveolae are heterogeneous, and identify cavin 2 as a determinant of this heterogeneity. Cavin proteins are key components of mammalian caveolae and are expressed from four genes in a tissue-specific manner. Gram Hansen et al. demonstrate that caveolae in the endothelia of different tissues are remarkably heterogeneous, and reveal a role for cavin 2 in determining the apparent size of cavin complexes.
Collapse
|
545
|
Sullivan DP, Muller WA. Neutrophil and monocyte recruitment by PECAM, CD99, and other molecules via the LBRC. Semin Immunopathol 2013; 36:193-209. [PMID: 24337626 DOI: 10.1007/s00281-013-0412-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 11/28/2013] [Indexed: 12/14/2022]
Abstract
The recruitment of specific leukocyte subtypes to the site of tissue injury is the cornerstone of inflammation and disease progression. This process has become an intense area of research because it presents several possible steps against which disease-specific therapies could be targeted. Leukocytes are recruited out of the blood stream by a series of events that include their capture, rolling, activation, and migration along the endothelium. In the last step, the leukocytes squeeze between adjacent endothelial cells to gain access to the inflamed tissue through a process referred to as transendothelial migration (TEM). Although many of the molecules, such as PECAM and CD99, that regulate these sequential steps have been identified, much less is understood regarding how they work together to coordinate the complex intercellular communications and dramatic shape changes that take place between the endothelial cells and leukocytes. Several of the endothelial cell proteins that function in TEM are localized to the lateral border recycling compartment (LBRC), an interconnected reticulum of membrane that recycles selectively to the endothelial borders. The recruitment of the LBRC to surround the migrating leukocyte is required for efficient TEM. This review will focus on the proteins and mechanisms that mediate TEM and specifically how the LBRC functions in the context of these molecular interactions and membrane movements.
Collapse
Affiliation(s)
- David P Sullivan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Ward Building, Rm 3-140, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | | |
Collapse
|
546
|
Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood-brain barrier. Nat Med 2013; 19:1584-96. [PMID: 24309662 DOI: 10.1038/nm.3407] [Citation(s) in RCA: 1709] [Impact Index Per Article: 142.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/22/2013] [Indexed: 01/01/2023]
Abstract
The interface between the blood circulation and the neural tissue features unique characteristics that are encompassed by the term 'blood-brain barrier' (BBB). The main functions of this barrier, namely maintenance of brain homeostasis, regulation of influx and efflux transport, and protection from harm, are determined by its specialized multicellular structure. Every constituent cell type makes an indispensable contribution to the BBB's integrity. But if one member of the BBB fails, and as a result the barrier breaks down, there can be dramatic consequences and neuroinflammation and neurodegeneration can occur. In this Review, we highlight recently gained mechanistic insights into the development and maintenance of the BBB. We then discuss how BBB disruption can cause or contribute to neurological disease. Finally, we examine how this knowledge can be used to explore new possibilities for BBB repair.
Collapse
Affiliation(s)
- Birgit Obermeier
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | |
Collapse
|
547
|
Torres C, Fonseca AM, Leander M, Matos R, Morais S, Campos M, Lima M. Circulating endothelial cells in patients with venous thromboembolism and myeloproliferative neoplasms. PLoS One 2013; 8:e81574. [PMID: 24339944 PMCID: PMC3855326 DOI: 10.1371/journal.pone.0081574] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 10/15/2013] [Indexed: 11/25/2022] Open
Abstract
Background Circulating endothelial cells (CEC) may be a biomarker of vascular injury and pro-thrombotic tendency, while circulating endothelial progenitor cells (CEP) may be an indicator for angiogenesis and vascular remodelling. However, there is not a universally accepted standardized protocol to identify and quantify these cells and its clinical relevancy remains to be established. Objectives To quantify CEC and CEP in patients with venous thromboembolism (VTE) and with myeloproliferative neoplasms (MPN), to characterize the CEC for the expression of activation (CD54, CD62E) and procoagulant (CD142) markers and to investigate whether they correlate with other clinical and laboratory data. Patients and Methods Sixteen patients with VTE, 17 patients with MPN and 20 healthy individuals were studied. The CEC and CEP were quantified and characterized in the blood using flow cytometry, and the demographic, clinical and laboratory data were obtained from hospital records. Results We found the CEC counts were higher in both patient groups as compared to controls, whereas increased numbers of CEP were found only in patients with MPN. In addition, all disease groups had higher numbers of CD62E+ CEC as compared to controls, whereas only patients with VTE had increased numbers of CD142+ and CD54+ CEC. Moreover, the numbers of total and CD62+ CEC correlated positively with the white blood cells (WBC) counts in both groups of patients, while the numbers of CEP correlated positively with the WBC counts only in patients with MPN. In addition, in patients with VTE a positive correlation was found between the numbers of CD54+ CEC and the antithrombin levels, as well as between the CD142+ CEC counts and the number of thrombotic events. Conclusions Our study suggests that CEC counts may reveal endothelial injury in patients with VTE and MPN and that CEC may express different activation-related phenotypes depending on the disease status.
Collapse
Affiliation(s)
- Cláudia Torres
- Laboratório de Citometria, Serviço de Hematologia Clínica, Hospital de Santo António (HSA), Centro Hospitalar do Porto (CHP), Porto, Portugal
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS/UP), Porto, Portugal
- * E-mail: (ML); (CT)
| | - Ana Mafalda Fonseca
- CICS-UBI-Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Magdalena Leander
- Laboratório de Citometria, Serviço de Hematologia Clínica, Hospital de Santo António (HSA), Centro Hospitalar do Porto (CHP), Porto, Portugal
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS/UP), Porto, Portugal
| | - Rui Matos
- Secção de Trombose e Hemostase, Serviço de Hematologia Clínica, Hospital de Santo António (HSA), Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - Sara Morais
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS/UP), Porto, Portugal
- Secção de Trombose e Hemostase, Serviço de Hematologia Clínica, Hospital de Santo António (HSA), Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - Manuel Campos
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS/UP), Porto, Portugal
- Secção de Trombose e Hemostase, Serviço de Hematologia Clínica, Hospital de Santo António (HSA), Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - Margarida Lima
- Laboratório de Citometria, Serviço de Hematologia Clínica, Hospital de Santo António (HSA), Centro Hospitalar do Porto (CHP), Porto, Portugal
- Unidade Multidisciplinar de Investigação Biomédica (UMIB), Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS/UP), Porto, Portugal
- * E-mail: (ML); (CT)
| |
Collapse
|
548
|
Behfar A, Latere JP, Bartunek J, Homsy C, Daro D, Crespo-Diaz RJ, Stalboerger PG, Steenwinckel V, Seron A, Redfield MM, Terzic A. Optimized delivery system achieves enhanced endomyocardial stem cell retention. Circ Cardiovasc Interv 2013; 6:710-8. [PMID: 24326777 PMCID: PMC4273747 DOI: 10.1161/circinterventions.112.000422] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Regenerative cell-based therapies are associated with limited myocardial retention of delivered stem cells. The objective of this study is to develop an endocardial delivery system for enhanced cell retention. METHODS AND RESULTS Stem cell retention was simulated in silico using 1- and 3-dimensional models of tissue distortion and compliance associated with delivery. Needle designs, predicted to be optimal, were accordingly engineered using nitinol, a nickel and titanium alloy displaying shape memory and superelasticity. Biocompatibility was tested with human mesenchymal stem cells. Experimental validation was performed with species-matched cells directly delivered into Langendorff-perfused porcine hearts or administered percutaneously into the endocardium of infarcted pigs. Cell retention was quantified by flow cytometry and real-time quantitative polymerase chain reaction methodology. Models, computing optimal distribution of distortion calibrated to favor tissue compliance, predicted that a 75°-curved needle featuring small-to-large graded side holes would ensure the highest cell retention profile. In isolated hearts, the nitinol curved needle catheter (C-Cath) design ensured 3-fold superior stem cell retention compared with a standard needle. In the setting of chronic infarction, percutaneous delivery of stem cells with C-Cath yielded a 37.7±7.1% versus 10.0±2.8% retention achieved with a traditional needle without effect on biocompatibility or safety. CONCLUSIONS Modeling-guided development of a nitinol-based curved needle delivery system with incremental side holes achieved enhanced myocardial stem cell retention.
Collapse
Affiliation(s)
- Atta Behfar
- From Division of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN (A.B., R.J.C.-D., P.G.S., M.M.R., A.T.); Cardio3 BioSciences, Mont-Saint-Guibert, Belgium (J.-P.L., C.H., D.D., V.S., A.S.); and Cardiovascular Center, OLV Ziekenhuis, Aalst, Belgium (J.B.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
549
|
Geenen IL, Verbruggen S, Molin DG, Spronk HM, Maessen JG, Meesters B, Schurink GW, Post MJ. Phenotypic fitness of primary endothelial cells cultured from patients with high cardiovascular risk or chronic kidney disease for vascular tissue engineering. Tissue Eng Part A 2013; 20:1049-59. [PMID: 24279825 DOI: 10.1089/ten.tea.2013.0163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vascular tissue engineering relies on the combination of patient-derived cells and biomaterials to create new vessels. For clinical application, data regarding the function and behavior of patient-derived cells are needed. We investigated cell growth and functional characteristics of human venous endothelial cells (HVECs) from coronary arterial bypass graft (CABG), chronic kidney disease (CKD), and control patients. HVECs were isolated from venous specimens that were obtained during elective surgical procedures by means of collagenase digestion. Gene expression, proliferation, migration, secretory functions, and thrombogenic characteristics were evaluated using high-throughput assays. A total of 48 cell batches (14 control, 19 CABG, and 15 CKD subjects) were assessed. Proliferation, population doubling times, and migration of HVECs derived from CABG and CKD patients did not differ from controls. Thrombomodulin expression was higher in CABG-HVECs compared with controls. HVEC-induced thrombin formation in plasma did not differ between groups, and the contact activation pathway was the major contributor to coagulation. Patient-derived HVECs were able to attach and survive on polycaprolactone scaffolds that were coated with fibrin. HVECs from cardiovascular-diseased and CKD patients showed comparable functional characteristics with HVECs derived from uncompromised patients. We, therefore, conclude that endothelial cells from aged patients with comorbidities can be safely used for isolation and in vitro expansion for vascular tissue engineering.
Collapse
Affiliation(s)
- Irma L Geenen
- 1 Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center , Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
550
|
Ni NC, Ballantyne LL, Mewburn JD, Funk CD. Multiple-site activation of the cysteinyl leukotriene receptor 2 is required for exacerbation of ischemia/reperfusion injury. Arterioscler Thromb Vasc Biol 2013; 34:321-30. [PMID: 24285579 DOI: 10.1161/atvbaha.113.302536] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Transgenic overexpression of the human cysteinyl leukotriene receptor 2 (CysLT2R) in murine endothelium exacerbates vascular permeability and ischemia/reperfusion injury. Here, we explore the underlying mechanisms of CysLT2R activation-mediated inflammation and delineate the relative contributions of endogenous murine CysLT2R and the transgene-derived receptor. APPROACH AND RESULTS We created a novel mouse with only endothelial-expressed CysLT2R (endothelium-targeted overexpression mice [EC]/CysLT2R-knockout mice [KO]) by crossing EC with KO to dissect the role of endothelial CysLT2R in tissue injury. Surprisingly, we discovered that damage in EC/KO mice was not elevated (24% versus 47% EC) after ischemia/reperfusion. We examined vascular permeability and leukocyte recruitment/rolling responses in the cremaster vasculature after cysteinyl leukotriene (cysLT) stimulation. Mice possessing transgenic endothelial CysLT2R overexpression, whether EC or EC/KO, when stimulated with cysLTs, exhibited vascular hyperpermeability, declining leukocyte flux, and a transient increase in slow-rolling leukocyte fraction. Mice lacking endogenous CysLT2R (both KO [20 ± 3 cells/min] EC/KO [24 ± 3]) showed lower-rolling leukocyte flux versus wild-type (38 ± 6) and EC (35 ± 6) mice under unstimulated conditions. EC/KO mice differed from EC counterparts in that vascular hyperpermeability was not present in the absence of exogenous cysLTs. CONCLUSIONS These results indicate that endothelial and nonendothelial CysLT2R niches have separate roles in mediating inflammatory responses. Endothelial receptor activation results in increased vascular permeability and leukocyte slow-rolling, facilitating leukocyte transmigration. Nonendothelial receptors, likely located on resident/circulating leukocytes, facilitate endothelial receptor activation and leukocyte transit. Activation of both receptor populations is required for injury exacerbation.
Collapse
Affiliation(s)
- Nathan C Ni
- From the Department of Biomedical and Molecular Sciences (N.C.N., L.L.B., C.D.F.) and Cancer Research Institute (J.D.M.), Queen's University, Kingston, ON, Canada
| | | | | | | |
Collapse
|