451
|
Olver TD, Laughlin MH. Endurance, interval sprint, and resistance exercise training: impact on microvascular dysfunction in type 2 diabetes. Am J Physiol Heart Circ Physiol 2016; 310:H337-50. [PMID: 26408541 PMCID: PMC4796622 DOI: 10.1152/ajpheart.00440.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/16/2015] [Indexed: 01/02/2023]
Abstract
Type 2 diabetes (T2D) alters capillary hemodynamics, causes capillary rarefaction in skeletal muscle, and alters endothelial and vascular smooth muscle cell phenotype, resulting in impaired vasodilatory responses. These changes contribute to altered blood flow responses to physiological stimuli, such as exercise and insulin secretion. T2D-induced microvascular dysfunction impairs glucose and insulin delivery to skeletal muscle (and other tissues such as skin and nervous), thereby reducing glucose uptake and perpetuating hyperglycemia and hyperinsulinemia. In patients with T2D, exercise training (EX) improves microvascular vasodilator and insulin signaling and attenuates capillary rarefaction in skeletal muscle. EX-induced changes subsequently augment glucose and insulin delivery as well as glucose uptake. If these adaptions occur in a sufficient amount of tissue, and skeletal muscle in particular, chronic exposure to hyperglycemia and hyperinsulinemia and the risk of microvascular complications in all vascular beds will decrease. We postulate that EX programs that engage as much skeletal muscle mass as possible and recruit as many muscle fibers within each muscle as possible will generate the greatest improvements in microvascular function, providing that the duration of the stimulus is sufficient. Primary improvements in microvascular function occur in tissues (skeletal muscle primarily) engaged during exercise, and secondary improvements in microvascular function throughout the body may result from improved blood glucose control. We propose that the added benefit of combined resistance and aerobic EX programs and of vigorous intensity EX programs is not simply "more is better." Rather, we believe the additional benefit is the result of EX-induced adaptations in and around more muscle fibers, resulting in more muscle mass and the associated microvasculature being changed. Thus, to acquire primary and secondary improvements in microvascular function and improved blood glucose control, EX programs should involve upper and lower body exercise and modulate intensity to augment skeletal muscle fiber recruitment. Under conditions of limited mobility, it may be necessary to train skeletal muscle groups separately to maximize whole body skeletal muscle fiber recruitment.
Collapse
Affiliation(s)
- T Dylan Olver
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri;
| | - M Harold Laughlin
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri; Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
452
|
Velasco V, Gruenthal M, Zusstone E, Thomas JMD, Berson RE, Keynton RS, Williams SJ. An orbital shear platform for real-time, in vitro endothelium characterization. Biotechnol Bioeng 2016; 113:1336-44. [PMID: 26615057 DOI: 10.1002/bit.25893] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 10/29/2015] [Accepted: 11/24/2015] [Indexed: 01/13/2023]
Abstract
Electrical impedance techniques have been used to characterize endothelium morphology, permeability, and motility in vitro. However, these impedance platforms have been limited to either static endothelium studies and/or induced laminar fluid flow at a constant, single shear stress value. In this work, we present a microfabricated impedance sensor for real-time, in vitro characterization of human umbilical vein endothelial cells (HUVECs) undergoing oscillatory hydrodynamic shear. Oscillatory shear was applied with an orbital shaker and the electrical impedance was measured by a microfabricated impedance chip with discrete electrodes positioned at radial locations of 0, 2.5, 5.0, 7.5, 10.0, and 12.5 mm from the center of the chip. Depending on their radial position within the circular orbital platform, HUVECs were exposed to shear values ranging between 0.6 and 6.71 dyne/cm(2) (according to numerical simulations) for 22 h. Impedance spectra were fit to an equivalent circuit model and the trans-endothelial resistance and monolayer's capacitance were extracted. Results demonstrated that, compared to measurements acquired before the onset of shear, cells at the center of the platform that experienced low steady shear stress (∼2.2 dyne/cm(2) ) had an average change in trans-endothelial resistance of 6.99 ± 4.06% and 1.78 ± 2.40% change in cell capacitance after 22 hours of shear exposure; cells near the periphery of the well (r = 12.5 mm) experienced transient shears (2.5-6.7 dyne/cm(2) ) and exhibited a greater change in trans-endothelial resistance (24.2 ± 10.8%) and cell capacitance (4.57 ± 5.39%). This study, demonstrates that the orbital shear platform provides a simple system that can capture and quantify the real-time cellular morphology as a result of induced shear stress. The orbital shear platform presented in this work, compared to traditional laminar platforms, subjects cells to more physiologically relevant oscillatory shear as well as exposes the sample to several shear values simultaneously. Biotechnol. Bioeng. 2016;113: 1336-1344. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vanessa Velasco
- Department of Mechanical Engineering, University of Louisville, Louisville, 40292, Kentucky
| | - Mark Gruenthal
- Department of Mechanical Engineering, University of Louisville, Louisville, 40292, Kentucky
| | - Esther Zusstone
- School of Nursing, University of Louisville, Louisville, Kentucky
| | - Jonathan M D Thomas
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky
| | - R Eric Berson
- Department of Chemical Engineering, University of Louisville, Louisville, Kentucky
| | - Robert S Keynton
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Stuart J Williams
- Department of Mechanical Engineering, University of Louisville, Louisville, 40292, Kentucky.
| |
Collapse
|
453
|
Lewis PL, Shah RN. 3D Printing for Liver Tissue Engineering: Current Approaches and Future Challenges. CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0084-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
454
|
Yuan L, Chan GC, Beeler D, Janes L, Spokes KC, Dharaneeswaran H, Mojiri A, Adams WJ, Sciuto T, Garcia-Cardeña G, Molema G, Kang PM, Jahroudi N, Marsden PA, Dvorak A, Regan ER, Aird WC. A role of stochastic phenotype switching in generating mosaic endothelial cell heterogeneity. Nat Commun 2016; 7:10160. [PMID: 26744078 PMCID: PMC5154372 DOI: 10.1038/ncomms10160] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/10/2015] [Indexed: 01/20/2023] Open
Abstract
Previous studies have shown that biological noise may drive dynamic phenotypic mosaicism in isogenic unicellular organisms. However, there is no evidence for a similar mechanism operating in metazoans. Here we show that the endothelial-restricted gene, von Willebrand factor (VWF), is expressed in a mosaic pattern in the capillaries of many vascular beds and in the aorta. In capillaries, the mosaicism is dynamically regulated, with VWF switching between ON and OFF states during the lifetime of the animal. Clonal analysis of cultured endothelial cells reveals that dynamic mosaic heterogeneity is controlled by a low-barrier, noise-sensitive bistable switch that involves random transitions in the DNA methylation status of the VWF promoter. Finally, the hearts of VWF-null mice demonstrate an abnormal endothelial phenotype as well as cardiac dysfunction. Together, these findings suggest a novel stochastic phenotype switching strategy for adaptive homoeostasis in the adult vasculature.
Collapse
Affiliation(s)
- Lei Yuan
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Gary C Chan
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - David Beeler
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Lauren Janes
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Katherine C Spokes
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Harita Dharaneeswaran
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Anahita Mojiri
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - William J Adams
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Tracey Sciuto
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Guillermo Garcia-Cardeña
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02215, USA
| | - Grietje Molema
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, 9700 AB Groningen, The Netherlands
| | - Peter M Kang
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Cardiovascular Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Nadia Jahroudi
- Department of Medicine, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Philip A Marsden
- Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada.,St. Michaels's Hospital, Toronto, Ontario M5B 1W8, Canada
| | - Ann Dvorak
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Erzsébet Ravasz Regan
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - William C Aird
- Center for Vascular Biology Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| |
Collapse
|
455
|
Greathouse KM, Palladino SP, Dong C, Helton ES, Ubogu EE. Modeling leukocyte trafficking at the human blood-nerve barrier in vitro and in vivo geared towards targeted molecular therapies for peripheral neuroinflammation. J Neuroinflammation 2016; 13:3. [PMID: 26732309 PMCID: PMC4702318 DOI: 10.1186/s12974-015-0469-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 12/24/2015] [Indexed: 12/19/2022] Open
Abstract
Peripheral neuroinflammation is characterized by hematogenous mononuclear leukocyte infiltration into peripheral nerves. Despite significant clinical knowledge, advancements in molecular biology and progress in developing specific drugs for inflammatory disorders such as rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis, there are currently no specific therapies that modulate pathogenic peripheral nerve inflammation. Modeling leukocyte trafficking at the blood-nerve barrier using a reliable human in vitro model and potential intravital microscopy techniques in representative animal models guided by human observational data should facilitate the targeted modulation of the complex inflammatory cascade needed to develop safe and efficacious therapeutics for immune-mediated neuropathies and chronic neuropathic pain.
Collapse
Affiliation(s)
- Kelsey M Greathouse
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Steven P Palladino
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Chaoling Dong
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Eric S Helton
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| | - Eroboghene E Ubogu
- Department of Neurology, Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, The University of Alabama at Birmingham, 1825 University Boulevard, Room 1131, Birmingham, AL, 35294-0017, USA.
| |
Collapse
|
456
|
Goldenberg NM, Kuebler WM. Endothelial cell regulation of pulmonary vascular tone, inflammation, and coagulation. Compr Physiol 2016; 5:531-59. [PMID: 25880504 DOI: 10.1002/cphy.c140024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pulmonary endothelium represents a heterogeneous cell monolayer covering the luminal surface of the entire lung vasculature. As such, this cell layer lies at a critical interface between the blood, airways, and lung parenchyma, and must act as a selective barrier between these diverse compartments. Lung endothelial cells are able to produce and secrete mediators, display surface receptor, and cellular adhesion molecules, and metabolize circulating hormones to influence vasomotor tone, both local and systemic inflammation, and coagulation functions. In this review, we will explore the role of the pulmonary endothelium in each of these systems, highlighting key regulatory functions of the pulmonary endothelial cell, as well as novel aspects of the pulmonary endothelium in contrast to the systemic cell type. The interactions between pulmonary endothelial cells and both leukocytes and platelets will be discussed in detail, and wherever possible, elements of endothelial control over physiological and pathophysiological processes will be examined.
Collapse
Affiliation(s)
- Neil M Goldenberg
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; Department of Anesthesia, University of Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- The Keenan Research Centre for Biomedical Science of St. Michael's, Toronto, Ontario, Canada; German Heart Institute Berlin, Germany; Institute of Physiology, Charité-Universitätsmedizin Berlin, Germany; Department of Surgery, University of Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Ontario,Canada
| |
Collapse
|
457
|
Kuo MM, Kim DH, Jandu S, Bergman Y, Tan S, Wang H, Pandey DR, Abraham TP, Shoukas AA, Berkowitz DE, Santhanam L. MPST but not CSE is the primary regulator of hydrogen sulfide production and function in the coronary artery. Am J Physiol Heart Circ Physiol 2016; 310:H71-9. [PMID: 26519030 PMCID: PMC4796461 DOI: 10.1152/ajpheart.00574.2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/08/2015] [Indexed: 01/09/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as an important gasotransmitter in the vasculature. In this study, we tested the hypothesis that H2S contributes to coronary vasoregulation and evaluated the physiological relevance of two sources of H2S, namely, cystathionine-γ-lyase (CSE) and 3-mercaptypyruvate sulfertransferase (MPST). MPST was detected in human coronary artery endothelial cells as well as rat and mouse coronary artery; CSE was not detected in the coronary vasculature. Rat coronary artery homogenates produced H2S through the MPST pathway but not the CSE pathway in vitro. In vivo coronary vasorelaxation response was similar in CSE knockout mice, wild-type mice (WT), and WT mice treated with the CSE inhibitor propargylglycine, suggesting that CSE-produced H2S does not have a significant role in coronary vasoregulation in vivo. Ex vivo, the MPST substrate 3-mercaptopyruvate (3-MP) and H2S donor sodium hydrosulfide (NaHS) elicited similar coronary vasoreactivity responses. Pyruvate did not have any effects on vasoreactivity. The vasoactive effect of H2S appeared to be nitric oxide (NO) dependent: H2S induced coronary vasoconstriction in the presence of NO and vasorelaxation in its absence. Maximal endothelial-dependent relaxation was intact after 3-MP and NaHS induced an increase in preconstriction tone, suggesting that endothelial NO synthase activity was not significantly inhibited. In vitro, H2S reacted with NO, which may, in part explain the vasoconstrictive effects of 3-MP and NaHS. Taken together, these data show that MPST rather than CSE generates H2S in coronary artery, mediating its effects through direct modulation of NO. This has important implications for H2S-based therapy in healthy and diseased coronary arteries.
Collapse
Affiliation(s)
- Maggie M Kuo
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Dae Hee Kim
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland; Department of Anethesiology and Pain Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Sandeep Jandu
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Yehudit Bergman
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Siqi Tan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Huilei Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Deepesh R Pandey
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Theodore P Abraham
- Department of Medicine (Cardiology), Johns Hopkins University, Baltimore, Maryland; and
| | - Artin A Shoukas
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Dan E Berkowitz
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Lakshmi Santhanam
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland;
| |
Collapse
|
458
|
Sriram G, Tan JY, Islam I, Rufaihah AJ, Cao T. Efficient differentiation of human embryonic stem cells to arterial and venous endothelial cells under feeder- and serum-free conditions. Stem Cell Res Ther 2015; 6:261. [PMID: 26718617 PMCID: PMC4697311 DOI: 10.1186/s13287-015-0260-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/25/2015] [Accepted: 08/26/2015] [Indexed: 12/30/2022] Open
Abstract
Background Heterogeneity of endothelial cells (ECs) is a hallmark of the vascular system which may impact the development and management of vascular disorders. Despite the tremendous progress in differentiation of human embryonic stem cells (hESCs) towards endothelial lineage, differentiation into arterial and venous endothelial phenotypes remains elusive. Additionally, current differentiation strategies are hampered by inefficiency, lack of reproducibility, and use of animal-derived products. Methods To direct the differentiation of hESCs to endothelial subtypes, H1- and H9-hESCs were seeded on human plasma fibronectin and differentiated under chemically defined conditions by sequential modulation of glycogen synthase kinase-3 (GSK-3), basic fibroblast growth factor (bFGF), bone morphogenetic protein 4 (BMP4) and vascular endothelial growth factor (VEGF) signaling pathways for 5 days. Following the initial differentiation, the endothelial progenitor cells (CD34+CD31+ cells) were sorted and terminally differentiated under serum-free conditions to arterial and venous ECs. The transcriptome and secretome profiles of the two distinct populations of hESC-derived arterial and venous ECs were characterized. Furthermore, the safety and functionality of these cells upon in vivo transplantation were characterized. Results Sequential modulation of hESCs with GSK-3 inhibitor, bFGF, BMP4 and VEGF resulted in stages reminiscent of primitive streak, early mesoderm/lateral plate mesoderm, and endothelial progenitors under feeder- and serum-free conditions. Furthermore, these endothelial progenitors demonstrated differentiation potential to almost pure populations of arterial and venous endothelial phenotypes under serum-free conditions. Specifically, the endothelial progenitors differentiated to venous ECs in the absence of VEGF, and to arterial phenotype under low concentrations of VEGF. Additionally, these hESC-derived arterial and venous ECs showed distinct molecular and functional profiles in vitro. Furthermore, these hESC-derived arterial and venous ECs were nontumorigenic and were functional in terms of forming perfused microvascular channels upon subcutaneous implantation in the mouse. Conclusions We report a simple, rapid, and efficient protocol for directed differentiation of hESCs into endothelial progenitor cells capable of differentiation to arterial and venous ECs under feeder-free and serum-free conditions. This could offer a human platform to study arterial–venous specification for various applications related to drug discovery, disease modeling and regenerative medicine in the future. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0260-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gopu Sriram
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore, 119083, Singapore. .,Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Groove, #06-06 Immunos, Singapore, 138648, Singapore.
| | - Jia Yong Tan
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore, 119083, Singapore.
| | - Intekhab Islam
- Oral and Maxillofacial Surgery Disciplines, Faculty of Dentistry, National University of Singapore, Singapore, 119083, Singapore.
| | - Abdul Jalil Rufaihah
- Cardiac, Thoracic and Vascular Surgery (CTVS) Laboratory, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117510, Singapore. .,Singapore-Technion Alliance For Research and Technology (START) Regenerative Medicine Laboratory, Campus for Research Excellence And Technological Enterprise (CREATE), Singapore, 138602, Singapore.
| | - Tong Cao
- Oral Sciences Disciplines, Faculty of Dentistry, National University of Singapore, Singapore, 119083, Singapore. .,NUS Graduate School for Integrative Science and Engineering, Singapore, 117456, Singapore. .,Tissue Engineering Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
459
|
Lokhov PG, Balashova EE. Design of universal cancer vaccines using natural tumor vessel-specific antigens (SANTAVAC). Hum Vaccin Immunother 2015; 11:689-98. [PMID: 25714389 PMCID: PMC4514425 DOI: 10.1080/21645515.2015.1011022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Vaccination against endothelial cells (ECs) lining the tumor vasculature represents one of the most attractive potential cancer immunotherapy options due to its ability to prevent solid tumor growth. Using this approach, target antigens can be derived from ECs and used to develop a universal cancer vaccine. Unfortunately, direct immunization with EC preparations can elicit autoimmune vasculitis in normal tissues. Recently, tumor-induced changes to the human EC surface were described that provided a basis for designing efficient EC-based vaccines capable of eliciting immune responses that targeted the tumor endothelium directly. This review examines these data from the perspective of designing EC-based cancer vaccines for the treatment of all solid tumors, including the antigen composition of vaccine formulations, the selection ECs for antigen derivation, the production and control of antigens, and the method for estimating vaccine efficacy and safety. As the vaccine preparation requires a specifically derived set of natural cell surface antigens, a new vaccine preparation concept was formulated. Antigen compositions prepared according to this concept were named SANTAVAC (Set of All Natural Target Antigens for Vaccination Against Cancer).
Collapse
Affiliation(s)
- Petr G Lokhov
- a Institute of Biomedical Chemistry ; Moscow , Russia
| | | |
Collapse
|
460
|
Garciarena CD, McHale TM, Watkin RL, Kerrigan SW. Coordinated Molecular Cross-Talk between Staphylococcus aureus, Endothelial Cells and Platelets in Bloodstream Infection. Pathogens 2015; 4:869-82. [PMID: 26690226 PMCID: PMC4693168 DOI: 10.3390/pathogens4040869] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/02/2015] [Indexed: 01/06/2023] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen often carried asymptomatically on the human body. Upon entry to the otherwise sterile environment of the cardiovascular system, S. aureus can lead to serious complications resulting in organ failure and death. The success of S. aureus as a pathogen in the bloodstream is due to its ability to express a wide array of cell wall proteins on its surface that recognise host receptors, extracellular matrix proteins and plasma proteins. Endothelial cells and platelets are important cells in the cardiovascular system and are a major target of bloodstream infection. Endothelial cells form the inner lining of a blood vessel and provide an antithrombotic barrier between the vessel wall and blood. Platelets on the other hand travel throughout the cardiovascular system and respond by aggregating around the site of injury and initiating clot formation. Activation of either of these cells leads to functional dysregulation in the cardiovascular system. In this review, we will illustrate how S. aureus establish intimate interactions with both endothelial cells and platelets leading to cardiovascular dysregulation.
Collapse
Affiliation(s)
- Carolina D Garciarena
- Cardiovascular Infection Research Group, School of Pharmacy & Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | - Tony M McHale
- Cardiovascular Infection Research Group, School of Pharmacy & Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | - Rebecca L Watkin
- Cardiovascular Infection Research Group, School of Pharmacy & Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | - Steven W Kerrigan
- Cardiovascular Infection Research Group, School of Pharmacy & Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
461
|
Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins: A need for ADME characterization in drug discovery and development. MAbs 2015; 8:229-45. [PMID: 26636901 PMCID: PMC4966629 DOI: 10.1080/19420862.2015.1115937] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein therapeutics represent a diverse array of biologics including antibodies, fusion proteins, and therapeutic replacement enzymes. Since their inception, they have revolutionized the treatment of a wide range of diseases including respiratory, vascular, autoimmune, inflammatory, infectious, and neurodegenerative diseases, as well as cancer. While in vivo pharmacokinetic, pharmacodynamic, and efficacy studies are routinely carried out for protein therapeutics, studies that identify key factors governing their absorption, distribution, metabolism, and excretion (ADME) properties have not been fully investigated. Thorough characterization and in-depth study of their ADME properties are critical in order to support drug discovery and development processes for the production of safer and more effective biotherapeutics. In this review, we discuss the main factors affecting the ADME characteristics of these large macromolecular therapies. We also give an overview of the current tools, technologies, and approaches available to investigate key factors that influence the ADME of recombinant biotherapeutic drugs, and demonstrate how ADME studies will facilitate their future development.
Collapse
|
462
|
Zhou Y, Williams J, Smallwood PM, Nathans J. Sox7, Sox17, and Sox18 Cooperatively Regulate Vascular Development in the Mouse Retina. PLoS One 2015; 10:e0143650. [PMID: 26630461 PMCID: PMC4667919 DOI: 10.1371/journal.pone.0143650] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 11/06/2015] [Indexed: 02/06/2023] Open
Abstract
Vascular development and maintenance are controlled by a complex transcriptional program, which integrates both extracellular and intracellular signals in endothelial cells. Here we study the roles of three closely related SoxF family transcription factors–Sox7, Sox17, and Sox18 –in the developing and mature mouse vasculature using targeted gene deletion on a mixed C57/129/CD1 genetic background. In the retinal vasculature, each SoxF gene exhibits a distinctive pattern of expression in different classes of blood vessels. On a mixed genetic background, vascular endothelial-specific deletion of individual SoxF genes has little or no effect on vascular architecture or differentiation, a result that can be explained by overlapping function and by reciprocal regulation of gene expression between Sox7 and Sox17. By contrast, combined deletion of Sox7, Sox17, and Sox18 at the onset of retinal angiogenesis leads to a dense capillary plexus with a nearly complete loss of radial arteries and veins, whereas the presence of a single Sox17 allele largely restores arterial identity, as determined by vascular smooth muscle cell coverage. In the developing retina, expression of all three SoxF genes is reduced in the absence of Norrin/Frizzled4-mediated canonical Wnt signaling, but SoxF gene expression is unaffected by reduced VEGF signaling in response to deletion of Neuropilin1 (Npn1). In adulthood, Sox7, Sox17, and Sox18 act in a largely redundant manner to maintain blood vessel function, as adult onset vascular endothelial-specific deletion of all three SoxF genes leads to massive edema despite nearly normal vascular architecture. These data reveal critical and partially redundant roles for Sox7, Sox17 and Sox18 in vascular growth, differentiation, and maintenance.
Collapse
Affiliation(s)
- Yulian Zhou
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - John Williams
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Philip M. Smallwood
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
| | - Jeremy Nathans
- Departments of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, United States of America
- * E-mail:
| |
Collapse
|
463
|
Leclercq A, Veillat V, Loriot S, Spuul P, Madonna F, Roques X, Génot E. A Methodology for Concomitant Isolation of Intimal and Adventitial Endothelial Cells from the Human Thoracic Aorta. PLoS One 2015; 10:e0143144. [PMID: 26599408 PMCID: PMC4658207 DOI: 10.1371/journal.pone.0143144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/30/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Aortic diseases are diverse and involve a multiplicity of biological systems in the vascular wall. Aortic dissection, which is usually preceded by aortic aneurysm, is a leading cause of morbidity and mortality in modern societies. Although the endothelium is now known to play an important role in vascular diseases, its contribution to aneurysmal aortic lesions remains largely unknown. The aim of this study was to define a reliable methodology for the isolation of aortic intimal and adventitial endothelial cells in order to throw light on issues relevant to endothelial cell biology in aneurysmal diseases. METHODOLOGY/PRINCIPAL FINDINGS We set up protocols to isolate endothelial cells from both the intima and the adventitia of human aneurysmal aortic vessel segments. Throughout the procedure, analysis of cell morphology and endothelial markers allowed us to select an endothelial fraction which after two rounds of expansion yielded a population of >90% pure endothelial cells. These cells have the features and functionalities of freshly isolated cells and can be used for biochemical studies. The technique was successfully used for aortic vessel segments of 20 patients and 3 healthy donors. CONCLUSIONS/SIGNIFICANCE This simple and highly reproducible method allows the simultaneous preparation of reasonably pure primary cultures of intimal and adventitial human endothelial cells, thus providing a reliable source for investigating their biology and involvement in both thoracic aneurysms and other aortic diseases.
Collapse
Affiliation(s)
- Anne Leclercq
- Université de Bordeaux, Bordeaux, France
- INSERM, U1045, Bordeaux, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
- * E-mail: (AL); (EG)
| | - Véronique Veillat
- Université de Bordeaux, Bordeaux, France
- INSERM, U1045, Bordeaux, France
| | - Sandrine Loriot
- Université de Bordeaux, Bordeaux, France
- SFR TransBioMed, Bordeaux, France
| | - Pirjo Spuul
- Université de Bordeaux, Bordeaux, France
- INSERM, U1045, Bordeaux, France
| | - Francesco Madonna
- Service de chirurgie cardiaque et vasculaire, Hôpital Haut-L’Evêque, Pessac, France
| | - Xavier Roques
- Service de chirurgie cardiaque et vasculaire, Hôpital Haut-L’Evêque, Pessac, France
| | - Elisabeth Génot
- Université de Bordeaux, Bordeaux, France
- INSERM, U1045, Bordeaux, France
- * E-mail: (AL); (EG)
| |
Collapse
|
464
|
Carman CV, Martinelli R. T Lymphocyte-Endothelial Interactions: Emerging Understanding of Trafficking and Antigen-Specific Immunity. Front Immunol 2015; 6:603. [PMID: 26635815 PMCID: PMC4657048 DOI: 10.3389/fimmu.2015.00603] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/10/2015] [Indexed: 12/26/2022] Open
Abstract
Antigen-specific immunity requires regulated trafficking of T cells in and out of diverse tissues in order to orchestrate lymphocyte development, immune surveillance, responses, and memory. The endothelium serves as a unique barrier, as well as a sentinel, between the blood and the tissues, and as such it plays an essential locally tuned role in regulating T cell migration and information exchange. While it is well established that chemoattractants and adhesion molecules are major determinants of T cell trafficking, emerging studies have now enumerated a large number of molecular players as well as a range of discrete cellular remodeling activities (e.g., transmigratory cups and invadosome-like protrusions) that participate in directed migration and pathfinding by T cells. In addition to providing trafficking cues, intimate cell-cell interaction between lymphocytes and endothelial cells provide instruction to T cells that influence their activation and differentiation states. Perhaps the most intriguing and underappreciated of these "sentinel" roles is the ability of the endothelium to act as a non-hematopoietic "semiprofessional" antigen-presenting cell. Close contacts between circulating T cells and antigen-presenting endothelium may play unique non-redundant roles in shaping adaptive immune responses within the periphery. A better understanding of the mechanisms directing T cell trafficking and the antigen-presenting role of the endothelium may not only increase our knowledge of the adaptive immune response but also empower the utility of emerging immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Christopher V Carman
- Center for Vascular Biology Research, Department of Medicine and Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Roberta Martinelli
- Center for Vascular Biology Research, Department of Medicine and Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
465
|
Bogorad MI, DeStefano J, Karlsson J, Wong AD, Gerecht S, Searson PC. Review: in vitro microvessel models. LAB ON A CHIP 2015; 15:4242-55. [PMID: 26364747 PMCID: PMC9397147 DOI: 10.1039/c5lc00832h] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
A wide range of perfusable microvessel models have been developed, exploiting advances in microfabrication, microfluidics, biomaterials, stem cell technology, and tissue engineering. These models vary in complexity and physiological relevance, but provide a diverse tool kit for the study of vascular phenomena and methods to vascularize artificial organs. Here we review the state-of-the-art in perfusable microvessel models, summarizing the different fabrication methods and highlighting advantages and limitations.
Collapse
Affiliation(s)
- Max I Bogorad
- Institute for Nanobiotechnology (INBT), 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, Maryland 21218, USA.
| | - Jackson DeStefano
- Institute for Nanobiotechnology (INBT), 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, Maryland 21218, USA.
| | - Johan Karlsson
- Institute for Nanobiotechnology (INBT), 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, Maryland 21218, USA.
| | - Andrew D Wong
- Institute for Nanobiotechnology (INBT), 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, Maryland 21218, USA.
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology (INBT), 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, Maryland 21218, USA. and Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| |
Collapse
|
466
|
Pfaltzgraff ER, Bader DM. Heterogeneity in vascular smooth muscle cell embryonic origin in relation to adult structure, physiology, and disease. Dev Dyn 2015; 244:410-6. [PMID: 25546231 DOI: 10.1002/dvdy.24247] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/18/2014] [Accepted: 12/19/2014] [Indexed: 12/22/2022] Open
Abstract
Regional differences in vascular physiology and disease response exist throughout the vascular tree. While these differences in physiology and disease correspond to regional vascular environmental conditions, there is also compelling evidence that the embryonic origins of the smooth muscle inherent to the vessels may play a role. Here, we review what is known regarding the role of embryonic origin of vascular smooth muscle cells during vascular development. The focus of this review is to highlight the heterogeneity in the origins of vascular smooth muscle cells and the resulting regional physiologies of the vessels. Our goal is to stimulate future investigation into this area and provide a better understanding of vascular organogenesis and disease. .
Collapse
Affiliation(s)
- Elise R Pfaltzgraff
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
467
|
Koudelkova P, Weber G, Mikulits W. Liver Sinusoidal Endothelial Cells Escape Senescence by Loss of p19ARF. PLoS One 2015; 10:e0142134. [PMID: 26528722 PMCID: PMC4631446 DOI: 10.1371/journal.pone.0142134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/16/2015] [Indexed: 11/18/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) represent a highly differentiated cell type that lines hepatic sinusoids. LSECs form a discontinuous endothelium due to fenestrations under physiological conditions, which are reduced upon chronic liver injury. Cultivation of rodent LSECs associates with a rapid onset of stress-induced senescence a few days post isolation, which limits genetic and biochemical studies ex vivo. Here we show the establishment of LSECs isolated from p19ARF-/- mice which undergo more than 50 cell doublings in the absence of senescence. Isolated p19ARF-/- LSECs display a cobblestone-like morphology and show the ability of tube formation. Analysis of DNA content revealed a stable diploid phenotype after long-term passaging without a gain of aneuploidy. Notably, p19ARF-/- LSECs express the endothelial markers CD31, vascular endothelial growth factor receptor (VEGFR)-2, VE-cadherin, von Willebrand factor, stabilin-2 and CD146 suggesting that these cells harbor and maintain an endothelial phenotype. In line, treatment with small molecule inhibitors against VEGFR-2 caused cell death, demonstrating the sustained ability of p19ARF-/- LSECs to respond to anti-angiogenic therapeutics. From these data we conclude that loss of p19ARF overcomes senescence of LSECs, allowing immortalization of cells without losing endothelial characteristics. Thus, p19ARF-/- LSECs provide a novel cellular model to study endothelial cell biology.
Collapse
Affiliation(s)
- Petra Koudelkova
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Gerhard Weber
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
468
|
Mueller KB, Bender SB, Hong K, Yang Y, Aronovitz M, Jaisser F, Hill MA, Jaffe IZ. Endothelial Mineralocorticoid Receptors Differentially Contribute to Coronary and Mesenteric Vascular Function Without Modulating Blood Pressure. Hypertension 2015; 66:988-97. [PMID: 26351033 PMCID: PMC4600033 DOI: 10.1161/hypertensionaha.115.06172] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 08/17/2015] [Indexed: 12/20/2022]
Abstract
Arteriolar vasoreactivity tightly regulates tissue-specific blood flow and contributes to systemic blood pressure (BP) but becomes dysfunctional in the setting of cardiovascular disease. The mineralocorticoid receptor (MR) is known to regulate BP via the kidney and by vasoconstriction in smooth muscle cells. Although endothelial cells (EC) express MR, the contribution of EC-MR to BP and resistance vessel function remains unclear. To address this, we created a mouse with MR specifically deleted from EC (EC-MR knockout [EC-MR-KO]) but with intact leukocyte MR expression and normal renal MR function. Telemetric BP studies reveal no difference between male EC-MR-KO mice and MR-intact littermates in systolic, diastolic, circadian, or salt-sensitive BP or in the hypertensive responses to aldosterone±salt or angiotensin II±l-nitroarginine methyl ester. Vessel myography demonstrated normal vasorelaxation in mesenteric and coronary arterioles from EC-MR-KO mice. After exposure to angiotensin II-induced hypertension, impaired endothelial-dependent relaxation was prevented in EC-MR-KO mice in mesenteric vessels but not in coronary vessels. Mesenteric vessels from angiotensin II-exposed EC-MR-KO mice showed increased maximum responsiveness to acetylcholine when compared with MR-intact vessels, a difference that is lost with indomethacin+l-nitroarginine methyl ester pretreatment. These data support that EC-MR plays a role in regulating endothelial function in hypertension. Although there was no effect of EC-MR deletion on mesenteric vasoconstriction, coronary arterioles from EC-MR-KO mice showed decreased constriction to endothelin-1 and thromboxane agonist at baseline and also after exposure to hypertension. These data support that EC-MR participates in regulation of vasomotor function in a vascular bed-specific manner that is also modulated by risk factors, such as hypertension.
Collapse
Affiliation(s)
- Katelee Barrett Mueller
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Shawn B Bender
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Kwangseok Hong
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Yan Yang
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Mark Aronovitz
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Frederic Jaisser
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Michael A Hill
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.)
| | - Iris Z Jaffe
- From the Molecular Cardiology Research Institute, Tufts Medical Center, and Sackler School of Biomedical Graduate Studies, Tufts University School of Medicine, Boston, MA (K.B.M., M.A., I.Z.J.); Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO (S.B.B.); Department of Biomedical Sciences (S.B.B.), Dalton Cardiovascular Research Center (S.B.B., K.H., Y.Y., M.A.H.), and Department of Medical Pharmacology and Physiology, School of Medicine (K.H., M.A.H.), University of Missouri, Columbia; and INSERM, UMR 1138, Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris, France (F.J.).
| |
Collapse
|
469
|
Vascularization mediated by mesenchymal stem cells from bone marrow and adipose tissue: a comparison. CELL REGENERATION 2015; 4:8. [PMID: 26500761 PMCID: PMC4619361 DOI: 10.1186/s13619-015-0025-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/01/2015] [Indexed: 02/08/2023]
Abstract
Tissue-engineered constructs are promising to overcome shortage of organ donors and to reconstruct at least parts of injured or diseased tissues or organs. However, oxygen and nutrient supply are limiting factors in many tissues, especially after implantation into the host. Therefore, the development of a vascular system prior to implantation appears crucial. To develop a functional vascular system, different cell types that interact with each other need to be co-cultured to simulate a physiological environment in vitro. This review provides an overview and a comparison of the current knowledge of co-cultures of human endothelial cells (ECs) with human adipose tissue-derived stem/stromal cells (ASCs) or bone marrow-mesenchymal stem cells (BMSCs) in three dimensional (3D) hydrogel matrices. Mesenchymal stem cells (MSCs), BMSCs or ASCs, have been shown to enhance vascular tube formation of ECs and to provide a stabilizing function in addition to growth factor delivery and permeability control for ECs. Although phenotypically similar, MSCs from different tissues promote tubulogenesis through distinct mechanisms. In this report, we describe differences and similarities regarding molecular interactions in order to investigate which of these two cell types displays more favorable characteristics to be used in clinical applications. Our comparative study shows that ASCs as well as BMSCs are both promising cell types to induce vascularization with ECs in vitro and consequently are promising candidates to support in vivo vascularization.
Collapse
|
470
|
Yau JW, Teoh H, Verma S. Endothelial cell control of thrombosis. BMC Cardiovasc Disord 2015; 15:130. [PMID: 26481314 PMCID: PMC4617895 DOI: 10.1186/s12872-015-0124-z] [Citation(s) in RCA: 459] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/09/2015] [Indexed: 02/07/2023] Open
Abstract
Hemostasis encompasses a set of tightly regulated processes that govern blood clotting, platelet activation, and vascular repair. Upon vascular injury, the hemostatic system initiates a series of vascular events and activates extravascular receptors that act in concert to seal off the damage. Blood clotting is subsequently attenuated by a plethora of inhibitors that prevent excessive clot formation and eventual thrombosis. The endothelium which resides at the interface between the blood and surrounding tissues, serves an integral role in the hemostatic system. Depending on specific tissue needs and local stresses, endothelial cells are capable of evoking either antithrombotic or prothrombotic events. Healthy endothelial cells express antiplatelet and anticoagulant agents that prevent platelet aggregation and fibrin formation, respectively. In the face of endothelial dysfunction, endothelial cells trigger fibrin formation, as well as platelet adhesion and aggregation. Finally, endothelial cells release pro-fibrinolytic agents that initiate fibrinolysis to degrade the clot. Taken together, a functional endothelium is essential to maintain hemostasis and prevent thrombosis. Thus, a greater understanding into the role of the endothelium can provide new avenues for exploration and novel therapies for the management of thromboembolisms.
Collapse
Affiliation(s)
- Jonathan W Yau
- Division of Cardiac Surgery, St. Michael's Hospital, Suite 8-003, Bond Wing, 30 Bond St., Toronto, ON, M5B 1W8, Canada.
| | - Hwee Teoh
- Division of Cardiac Surgery, St. Michael's Hospital, Suite 8-003, Bond Wing, 30 Bond St., Toronto, ON, M5B 1W8, Canada. .,Divisions of Endocrinology & Metabolism, Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, ON, Canada.
| | - Subodh Verma
- Division of Cardiac Surgery, St. Michael's Hospital, Suite 8-003, Bond Wing, 30 Bond St., Toronto, ON, M5B 1W8, Canada. .,Department of Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
471
|
Su EJ. Role of the fetoplacental endothelium in fetal growth restriction with abnormal umbilical artery Doppler velocimetry. Am J Obstet Gynecol 2015; 213:S123-30. [PMID: 26428491 DOI: 10.1016/j.ajog.2015.06.038] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 01/30/2023]
Abstract
Growth-restricted fetuses with absent or reversed end-diastolic velocities in the umbilical artery are at substantially increased risk for adverse perinatal and long-term outcome, even in comparison to growth-restricted fetuses with preserved end-diastolic velocities. Translational studies show that this Doppler velocimetry correlates with fetoplacental blood flow, with absent or reversed end-diastolic velocities signifying abnormally elevated resistance within the placental vasculature. The fetoplacental vasculature is unique in that it is not subject to autonomic regulation, unlike other vascular beds. Instead, humoral mediators, many of which are synthesized by local endothelial cells, regulate placental vascular resistance. Existing data demonstrate that in growth-restricted pregnancies complicated by absent or reversed umbilical artery end-diastolic velocities, an imbalance in production of these vasoactive substances occurs, favoring vasoconstriction. Morphologically, placentas from these pregnancies also demonstrate impaired angiogenesis, whereby vessels within the terminal villi are sparsely branched, abnormally thin, and elongated. This structural deviation from normal placental angiogenesis restricts blood flow and further contributes to elevated fetoplacental vascular resistance. Although considerable work has been done in the field of fetoplacental vascular development and function, much remains unknown about the mechanisms underlying impaired development and function of the human fetoplacental vasculature, especially in the context of severe fetal growth restriction with absent or reversed umbilical artery end-diastolic velocities. Fetoplacental endothelial cells are key regulators of angiogenesis and vasomotor tone. A thorough understanding of their role in placental vascular biology carries the significant potential of discovering clinically relevant and innovative approaches to prevention and treatment of fetal growth restriction with compromised umbilical artery end-diastolic velocities.
Collapse
|
472
|
Inflammatory neuropathies: pathology, molecular markers and targets for specific therapeutic intervention. Acta Neuropathol 2015; 130:445-68. [PMID: 26264608 DOI: 10.1007/s00401-015-1466-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 08/01/2015] [Accepted: 08/02/2015] [Indexed: 12/21/2022]
Abstract
Inflammatory neuropathies encompass groups of heterogeneous disorders characterized by pathogenic immune-mediated hematogenous leukocyte infiltration of peripheral nerves, nerve roots or both, with resultant demyelination or axonal degeneration or both. Inflammatory neuropathies may be divided into three major disease categories: Guillain-Barré syndrome (particularly the acute inflammatory demyelinating polyradiculoneuropathy variant), chronic inflammatory demyelinating polyradiculoneuropathy and nonsystemic vasculitic neuropathy (or peripheral nerve vasculitis). Despite major advances in molecular biology, pathology and genetics, the pathogenesis of these disorders remains elusive. There is insufficient knowledge on the mechanisms of hematogenous leukocyte trafficking into the peripheral nervous system to guide the development of specific molecular therapies for immune-mediated inflammatory neuropathies compared to disorders such as psoriasis, inflammatory bowel disease, rheumatoid arthritis or multiple sclerosis. The recent isolation and characterization of human endoneurial endothelial cells that form the blood-nerve barrier provides an opportunity to elucidate leukocyte-endothelial cell interactions critical to the pathogenesis of inflammatory neuropathies at the interface between the systemic circulation and peripheral nerve endoneurium. This review discusses our current knowledge of the classic pathological features of inflammatory neuropathies, attempts at molecular classification and genetic determinants, the utilization of in vitro and in vivo animal models to determine pathogenic mechanisms at the interface between the systemic circulation and the peripheral nervous system relevant to these disorders and prospects for future potential molecular pathology biomarkers and targets for specific therapeutic intervention.
Collapse
|
473
|
Stabler CT, Lecht S, Mondrinos MJ, Goulart E, Lazarovici P, Lelkes PI. Revascularization of decellularized lung scaffolds: principles and progress. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1273-85. [PMID: 26408553 DOI: 10.1152/ajplung.00237.2015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/23/2015] [Indexed: 02/07/2023] Open
Abstract
There is a clear unmet clinical need for novel biotechnology-based therapeutic approaches to lung repair and/or replacement, such as tissue engineering of whole bioengineered lungs. Recent studies have demonstrated the feasibility of decellularizing the whole organ by removal of all its cellular components, thus leaving behind the extracellular matrix as a complex three-dimensional (3D) biomimetic scaffold. Implantation of decellularized lung scaffolds (DLS), which were recellularized with patient-specific lung (progenitor) cells, is deemed the ultimate alternative to lung transplantation. Preclinical studies demonstrated that, upon implantation in rodent models, bioengineered lungs that were recellularized with airway and vascular cells were capable of gas exchange for up to 14 days. However, the long-term applicability of this concept is thwarted in part by the failure of current approaches to reconstruct a physiologically functional, quiescent endothelium lining the entire vascular tree of reseeded lung scaffolds, as inferred from the occurrence of hemorrhage into the airway compartment and thrombosis in the vasculature in vivo. In this review, we explore the idea that successful whole lung bioengineering will critically depend on 1) preserving and/or reestablishing the integrity of the subendothelial basement membrane, especially of the ultrathin respiratory membrane separating airways and capillaries, during and following decellularization and 2) restoring vascular physiological functionality including the barrier function and quiescence of the endothelial lining following reseeding of the vascular compartment. We posit that physiological reconstitution of the pulmonary vascular tree in its entirety will significantly promote the clinical translation of the next generation of bioengineered whole lungs.
Collapse
Affiliation(s)
- Collin T Stabler
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Shimon Lecht
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania
| | - Mark J Mondrinos
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ernesto Goulart
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, Brazil; and
| | - Philip Lazarovici
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, Pennsylvania;
| |
Collapse
|
474
|
Vandersmissen I, Craps S, Depypere M, Coppiello G, van Gastel N, Maes F, Carmeliet G, Schrooten J, Jones EAV, Umans L, Devlieger R, Koole M, Gheysens O, Zwijsen A, Aranguren XL, Luttun A. Endothelial Msx1 transduces hemodynamic changes into an arteriogenic remodeling response. J Cell Biol 2015; 210:1239-56. [PMID: 26391659 PMCID: PMC4586738 DOI: 10.1083/jcb.201502003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 08/18/2015] [Indexed: 12/25/2022] Open
Abstract
During peripheral arterial disease, MSX1 acts downstream of BMP–SMAD signaling to transduce the arterial shear stimulus into an arteriogenic remodeling response. MSX1 activates collateral endothelium into a proinflammatory state through ICAM1/VCAM1 up-regulation, resulting in increased leukocyte infiltration and collateral remodeling. Collateral remodeling is critical for blood flow restoration in peripheral arterial disease and is triggered by increasing fluid shear stress in preexisting collateral arteries. So far, no arterial-specific mediators of this mechanotransduction response have been identified. We show that muscle segment homeobox 1 (MSX1) acts exclusively in collateral arterial endothelium to transduce the extrinsic shear stimulus into an arteriogenic remodeling response. MSX1 was specifically up-regulated in remodeling collateral arteries. MSX1 induction in collateral endothelial cells (ECs) was shear stress driven and downstream of canonical bone morphogenetic protein–SMAD signaling. Flow recovery and collateral remodeling were significantly blunted in EC-specific Msx1/2 knockout mice. Mechanistically, MSX1 linked the arterial shear stimulus to arteriogenic remodeling by activating the endothelial but not medial layer to a proinflammatory state because EC but not smooth muscle cellMsx1/2 knockout mice had reduced leukocyte recruitment to remodeling collateral arteries. This reduced leukocyte infiltration in EC Msx1/2 knockout mice originated from decreased levels of intercellular adhesion molecule 1 (ICAM1)/vascular cell adhesion molecule 1 (VCAM1), whose expression was also in vitro driven by promoter binding of MSX1.
Collapse
Affiliation(s)
- Ine Vandersmissen
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Sander Craps
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Maarten Depypere
- Department of Electrical Engineering/Processing Speech and Images, Medical Image Computing, KU Leuven, 3000 Leuven, Belgium
| | - Giulia Coppiello
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
| | - Nick van Gastel
- Laboratory of Clinical and Experimental Endocrinology, Division of Skeletal Tissue Engineering, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Frederik Maes
- Department of Electrical Engineering/Processing Speech and Images, Medical Image Computing, KU Leuven, 3000 Leuven, Belgium
| | - Geert Carmeliet
- Laboratory of Clinical and Experimental Endocrinology, Division of Skeletal Tissue Engineering, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Jan Schrooten
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium Department of Materials Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Umans
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Roland Devlieger
- Department of Gynecology and Obstetrics, University Hospital Leuven, 3000 Leuven, Belgium Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Olivier Gheysens
- Department of Nuclear Medicine University Hospital Leuven, 3000 Leuven, Belgium Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - An Zwijsen
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, KU Leuven, 3000 Leuven, Belgium Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Xabier L Aranguren
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium Hematology and Cell Therapy Area, Clínica Universidad de Navarra and Division of Oncology, Center for Applied Medical Research, University of Navarra, 31008 Pamplona, Spain
| | - Aernout Luttun
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
475
|
Alba MM, Citarelli AN, Menni F, Agricola M, Braicovich A, De Horta E, De Rosa F, Filanino G, Gaggiotti R, Junqueras N, Martinelli S, Milan A, Morales ME, Setti S, Villalba DO. Tobacco and end stage renal disease: a multicenter, cross-sectional study in Argentinian Northern Patagonia. Tob Induc Dis 2015; 13:28. [PMID: 26327820 PMCID: PMC4553923 DOI: 10.1186/s12971-015-0051-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 08/20/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Smoking and chronic kidney disease are major public health problems with common features -high prevalence and mortality, high cardiovascular risk, gender differences and high prevalence in low income people-, but the link between them is poorly recognized. Our objectives were to investigate the exposure of dialysis patients to tobacco and to know their smoking behavior. METHODS We performed a multicenter, cross-sectional study in nine dialysis units in the Argentinian Northern Patagonia. We investigated smoker status, lifetime tobacco consumption, current tobacco use, breath carbon monoxide and % carboxyhaemoglobin. Fagerström and Richmond tests were performed for active smokers. STATISTICAL ANALYSIS one way ANOVA and Tukey's test for post hoc test. For exploratory analysis, frequency tables through chi-square distribution and single correspondence analysis were performed. RESULTS Six hundred thirty six patients (60.9 % males, 39.1 % females) were interviewed. Almost 70 % of them had had tobacco exposure. Excluding light smokers, the lifetime consumption was significantly different (p = 0.0052) between sexes (33.1 ± 2.4 pack/years in males and 18.2 ± 2.1 pack/years in females) The distribution of etiologies changed significantly (χ (2) p < 0.0001) with smoker status and the dose of tobacco smoking, with an increase in the diagnosis of nephrosclerosis in patients with high and very high lifetime consumption (from 16.1 % in non-smokers to 28.2 and 27 % respectively), and in passive smokers (from 16.1 to 27.3 %). The male preponderance of end-stage renal disease disappeared when only non-smokers were considered and grew with the increase in the lifetime consumption. Active smokers have small consumption, both low CO level and % COHb, low dependence and a good motivation to quit, but a high lifetime consumption. CONCLUSIONS Exposure of dialysis patients to tobacco is high and could be related to the progression to the final stage of the renal disease. It seems that tobacco renal damage is mostly hidden in the diagnosis of nephrosclerosis. The gender difference observed in these patients could also have a nexus with the men's higher tobacco exposure. Active smokers have a low current consumption but a high lifetime tobacco dose.
Collapse
Affiliation(s)
- Maria M Alba
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina ; Unidad Renal Cipolletti, España 885, Cipolletti 8332 Río Negro, Argentina
| | - Alicia N Citarelli
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Fernanda Menni
- Department of Economy and Statistics of INTA (National Institute of Agrological Technology) Estación Experimental Alto Valle del Río Negro, Ruta Nacional 22 Km 1190, 8332 Allen Río Negro, Argentina
| | - Maria Agricola
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Alejandra Braicovich
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Eduardo De Horta
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Fernando De Rosa
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Graciela Filanino
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Raul Gaggiotti
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Nelson Junqueras
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Sandra Martinelli
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Adriana Milan
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Mabel E Morales
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Silvia Setti
- Northern Patagonia Association of Nephrology, Entre Ríos 651, Neuquén, 8300 Argentina
| | - Daniel O Villalba
- Unidad Renal Cipolletti, España 885, Cipolletti 8332 Río Negro, Argentina
| |
Collapse
|
476
|
Yu JA, Castranova D, Pham VN, Weinstein BM. Single-cell analysis of endothelial morphogenesis in vivo. Development 2015; 142:2951-61. [PMID: 26253401 PMCID: PMC4582182 DOI: 10.1242/dev.123174] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 07/27/2015] [Indexed: 12/19/2022]
Abstract
Vessel formation has been extensively studied at the tissue level, but the difficulty in imaging the endothelium with cellular resolution has hampered study of the morphogenesis and behavior of endothelial cells (ECs) in vivo. We are using endothelial-specific transgenes and high-resolution imaging to examine single ECs in zebrafish. By generating mosaics with transgenes that simultaneously mark endothelial nuclei and membranes we are able to definitively identify and study the morphology and behavior of individual ECs during vessel sprouting and lumen formation. Using these methods, we show that developing trunk vessels are composed of ECs of varying morphology, and that single-cell analysis can be used to quantitate alterations in morphology and dynamics in ECs that are defective in proper guidance and patterning. Finally, we use single-cell analysis of intersegmental vessels undergoing lumen formation to demonstrate the coexistence of seamless transcellular lumens and single or multicellular enclosed lumens with autocellular or intercellular junctions, suggesting that heterogeneous mechanisms contribute to vascular lumen formation in vivo. The tools that we have developed for single EC analysis should facilitate further rigorous qualitative and quantitative analysis of EC morphology and behavior in vivo.
Collapse
Affiliation(s)
- Jianxin A Yu
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Castranova
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
477
|
Promising approaches to circumvent the blood-brain barrier: progress, pitfalls and clinical prospects in brain cancer. Ther Deliv 2015; 6:989-1016. [PMID: 26488496 DOI: 10.4155/tde.15.48] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain drug delivery is a major challenge for therapy of central nervous system (CNS) diseases. Biochemical modifications of drugs or drug nanocarriers, methods of local delivery, and blood-brain barrier (BBB) disruption with focused ultrasound and microbubbles are promising approaches which enhance transport or bypass the BBB. These approaches are discussed in the context of brain cancer as an example in CNS drug development. Targeting to receptors enabling transport across the BBB offers noninvasive delivery of small molecule and biological cancer therapeutics. Local delivery methods enable high dose delivery while avoiding systemic exposure. BBB disruption with focused ultrasound and microbubbles offers local and noninvasive treatment. Clinical trials show the prospects of these technologies and point to challenges for the future.
Collapse
|
478
|
Setyawati MI, Tay CY, Docter D, Stauber RH, Leong DT. Understanding and exploiting nanoparticles' intimacy with the blood vessel and blood. Chem Soc Rev 2015; 44:8174-99. [PMID: 26239875 DOI: 10.1039/c5cs00499c] [Citation(s) in RCA: 222] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While the blood vessel is seldom the target tissue, almost all nanomedicine will interact with blood vessels and blood at some point of time along its life cycle in the human body regardless of their intended destination. Despite its importance, many bionanotechnologists do not feature endothelial cells (ECs), the blood vessel cells, or consider blood effects in their studies. Including blood vessel cells in the study can greatly increase our understanding of the behavior of any given nanomedicine at the tissue of interest or to understand side effects that may occur in vivo. In this review, we will first describe the diversity of EC types found in the human body and their unique behaviors and possibly how these important differences can implicate nanomedicine behavior. Subsequently, we will discuss about the protein corona derived from blood with foci on the physiochemical aspects of nanoparticles (NPs) that dictate the protein corona characteristics. We would also discuss about how NPs characteristics can affect uptake by the endothelium. Subsequently, mechanisms of how NPs could cross the endothelium to access the tissue of interest. Throughout the paper, we will share some novel nanomedicine related ideas and insights that were derived from the understanding of the NPs' interaction with the ECs. This review will inspire more exciting nanotechnologies that had accounted for the complexities of the real human body.
Collapse
Affiliation(s)
- Magdiel Inggrid Setyawati
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | | | | | | | | |
Collapse
|
479
|
Laughlin MH, Padilla J, Jenkins NT, Thorne PK, Martin JS, Rector RS, Akter S, Davis JW. Exercise training causes differential changes in gene expression in diaphragm arteries and 2A arterioles of obese rats. J Appl Physiol (1985) 2015; 119:604-16. [PMID: 26183478 DOI: 10.1152/japplphysiol.00317.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/10/2015] [Indexed: 12/20/2022] Open
Abstract
We employed next-generation, transcriptome-wide RNA sequencing (RNA-Seq) technology to assess the effects of two different exercise training protocols on transcriptional profiles in diaphragm second-order arterioles (D2a) and in the diaphragm feed artery (DFA) from Otsuka Long Evans Tokushima Fatty (OLETF) rats. Arterioles were isolated from the diaphragm of OLETF rats that underwent an endurance exercise training program (EX; n = 13), interval sprint training program (SPRINT; n = 14), or remained sedentary (Sed; n = 12). Our hypothesis was that exercise training would have similar effects on gene expression in the diaphragm and soleus muscle arterioles because diaphragm blood flow increases during exercise to a similar extent as in soleus. Results reveal that several canonical pathways that were significantly altered by exercise in limb skeletal muscles were not among the pathways significantly changed in the diaphragm arterioles including actin cytoskeleton signaling, role of NFAT in regulation of immune response, protein kinase A signaling, and protein ubiquitination pathway. EX training altered the expression of a smaller number of genes than did SPRINT in the DFA but induced a larger number of genes with altered expression in the D2a than did SPRINT. In fact, FDR differential expression analysis (FDR, 10%) indicated that only two genes exhibited altered expression in D2a of SPRINT rats. Very few of the genes that exhibited altered expression in the DFA or D2a were also altered in limb muscle arterioles. Finally, results indicate that the 2a arterioles of soleus muscle (S2a) from endurance-trained animals and the DFA of SPRINT animals exhibited the largest number of genes with altered expression.
Collapse
Affiliation(s)
- M Harold Laughlin
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri; Biomedical Sciences, University of Missouri, Columbia, Missouri; Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Jaume Padilla
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; Child Health, University of Missouri, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | | | - Pamela K Thorne
- Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Jeffrey S Martin
- Cell Biology and Physiology, Edward Via College of Osteopathic Medicine-Auburn Campus, Auburn, Alabama; Kinesiology, Auburn University, Auburn, Alabama
| | - R Scott Rector
- Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri; Research Service-Harry S Truman Memorial VA Medical Center, Columbia, Missouri; Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri
| | - Sadia Akter
- MU Informatics Institute, University of Missouri, Columbia, Missouri; and
| | - J Wade Davis
- Health Management and Informatics, University of Missouri, Columbia, Missouri; Statistics, University of Missouri, Columbia, Missouri; MU Informatics Institute, University of Missouri, Columbia, Missouri; and
| |
Collapse
|
480
|
Sarker M, Chen X, Schreyer D. Experimental approaches to vascularisation within tissue engineering constructs. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2015; 26:683-734. [DOI: 10.1080/09205063.2015.1059018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
481
|
Xiao X, Zhang C, Ma X, Miao H, Wang J, Liu L, Chen S, Zeng R, Chen Y, Bihl JC. Angiotensin-(1-7) counteracts angiotensin II-induced dysfunction in cerebral endothelial cells via modulating Nox2/ROS and PI3K/NO pathways. Exp Cell Res 2015; 336:58-65. [PMID: 26101159 DOI: 10.1016/j.yexcr.2015.06.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/29/2022]
Abstract
Angiotensin (Ang) II, the main effector of the renin-angiotensin system, has been implicated in the pathogenesis of vascular diseases. Ang-(1-7) binds to the G protein-coupled Mas receptor (MasR) and can exert vasoprotective effects. We investigated the effects and underlying mechanisms of Ang-(1-7) on Ang II-induced dysfunction and oxidative stress in human brain microvascular endothelial cells (HbmECs). The pro-apoptotic activity, reactive oxygen species (ROS) and nitric oxide (NO) productions in HbmECs were measured. The protein expressions of nicotinamide adenine dinucleotide phosphate oxidase 2 (Nox2), serine/threonine kinase (Akt), endothelial nitric oxide synthase (eNOS) and their phosphorylated forms (p-Akt and p-eNOS) were examined by western blot. MasR antagonist and phosphatidylinositol-3-kinase (PI3K) inhibitor were used for receptor/pathway verification. We found that Ang-(1-7) suppressed Ang II-induced pro-apoptotic activity, ROS over-production and NO reduction in HbmECs, which were abolished by MasR antagonist. In addition, Ang-(1-7) down-regulated the expression of Nox2, and up-regulated the ratios of p-Akt/Akt and its downstream p-eNOS/eNOS in HbmECs. Exposure to PI3K inhibitor partially abrogated Ang-(1-7)-mediated protective effects in HbmECs. Our data suggests that Ang-(1-7)/MasR axis protects HbmECs from Ang II-induced dysfunction and oxidative stress via inhibition of Nox2/ROS and activation of PI3K/NO pathways.
Collapse
Affiliation(s)
- Xiang Xiao
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Cheng Zhang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Xiaotang Ma
- Institute of Neurology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Huilai Miao
- Department of Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Jinju Wang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Langni Liu
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Shuzhen Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Rong Zeng
- Department of Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Yanfang Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA.
| | - Ji C Bihl
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; Institute of Neurology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China.
| |
Collapse
|
482
|
Obesity-induced adipokine imbalance impairs mouse pulmonary vascular endothelial function and primes the lung for injury. Sci Rep 2015; 5:11362. [PMID: 26068229 PMCID: PMC4464323 DOI: 10.1038/srep11362] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/22/2015] [Indexed: 12/13/2022] Open
Abstract
Obesity is a risk factor for the development of acute respiratory distress syndrome (ARDS) but mechanisms mediating this association are unknown. While obesity is known to impair systemic blood vessel function, and predisposes to systemic vascular diseases, its effects on the pulmonary circulation are largely unknown. We hypothesized that the chronic low grade inflammation of obesity impairs pulmonary vascular homeostasis and primes the lung for acute injury. The lung endothelium from obese mice expressed higher levels of leukocyte adhesion markers and lower levels of cell-cell junctional proteins when compared to lean mice. We tested whether systemic factors are responsible for these alterations in the pulmonary endothelium; treatment of primary lung endothelial cells with obese serum enhanced the expression of adhesion proteins and reduced the expression of endothelial junctional proteins when compared to lean serum. Alterations in pulmonary endothelial cells observed in obese mice were associated with enhanced susceptibility to LPS-induced lung injury. Restoring serum adiponectin levels reversed the effects of obesity on the lung endothelium and attenuated susceptibility to acute injury. Our work indicates that obesity impairs pulmonary vascular homeostasis and enhances susceptibility to acute injury and provides mechanistic insight into the increased prevalence of ARDS in obese humans.
Collapse
|
483
|
Sacerdoti D, Pesce P, Di Pascoli M, Brocco S, Cecchetto L, Bolognesi M. Arachidonic acid metabolites and endothelial dysfunction of portal hypertension. Prostaglandins Other Lipid Mediat 2015; 120:80-90. [PMID: 26072731 DOI: 10.1016/j.prostaglandins.2015.05.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/20/2015] [Accepted: 05/25/2015] [Indexed: 12/12/2022]
Abstract
Increased resistance to portal flow and increased portal inflow due to mesenteric vasodilatation represent the main factors causing portal hypertension in cirrhosis. Endothelial cell dysfunction, defined as an imbalance between the synthesis, release, and effect of endothelial mediators of vascular tone, inflammation, thrombosis, and angiogenesis, plays a major role in the increase of resistance in portal circulation, in the decrease in the mesenteric one, in the development of collateral circulation. Reduced response to vasodilators in liver sinusoids and increased response in the mesenteric arterioles, and, viceversa, increased response to vasoconstrictors in the portal-sinusoidal circulation and decreased response in the mesenteric arterioles are also relevant to the pathophysiology of portal hypertension. Arachidonic acid (AA) metabolites through the three pathways, cyclooxygenase (COX), lipoxygenase, and cytochrome P450 monooxygenase and epoxygenase, are involved in endothelial dysfunction of portal hypertension. Increased thromboxane-A2 production by liver sinusoidal endothelial cells (LSECs) via increased COX-1 activity/expression, increased leukotriens, increased epoxyeicosatrienoic acids (EETs) (dilators of the peripheral arterial circulation, but vasoconstrictors of the portal-sinusoidal circulation), represent a major component in the increased portal resistance, in the decreased portal response to vasodilators and in the hyper-response to vasoconstrictors. Increased prostacyclin (PGI2) via COX-1 and COX-2 overexpression, and increased EETs/heme-oxygenase-1/K channels/gap junctions (endothelial derived hyperpolarizing factor system) play a major role in mesenteric vasodilatation, hyporeactivity to vasoconstrictors, and hyper-response to vasodilators. EETs, mediators of liver regeneration after hepatectomy and of angiogenesis, may play a role in the development of regenerative nodules and collateral circulation, through stimulation of vascular endothelial growth factor (VEGF) inside the liver and in the portal circulation. Pharmacological manipulation of AA metabolites may be beneficial for cirrhotic portal hypertension.
Collapse
Affiliation(s)
- David Sacerdoti
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy.
| | - Paola Pesce
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Marco Di Pascoli
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Silvia Brocco
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Lara Cecchetto
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| | - Massimo Bolognesi
- Department of Medicine, University of Padova Via Giustiniani, 2, 35100 Padova, Italy
| |
Collapse
|
484
|
Kolka CM, Richey JM, Castro AVB, Broussard JL, Ionut V, Bergman RN. Lipid-induced insulin resistance does not impair insulin access to skeletal muscle. Am J Physiol Endocrinol Metab 2015; 308:E1001-9. [PMID: 25852002 PMCID: PMC4451289 DOI: 10.1152/ajpendo.00015.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/03/2015] [Indexed: 11/22/2022]
Abstract
Elevated plasma free fatty acids (FFA) induce insulin resistance in skeletal muscle. Previously, we have shown that experimental insulin resistance induced by lipid infusion prevents the dispersion of insulin through the muscle, and we hypothesized that this would lead to an impairment of insulin moving from the plasma to the muscle interstitium. Thus, we infused lipid into our anesthetized canine model and measured the appearance of insulin in the lymph as a means to sample muscle interstitium under hyperinsulinemic euglycemic clamp conditions. Although lipid infusion lowered the glucose infusion rate and induced both peripheral and hepatic insulin resistance, we were unable to detect an impairment of insulin access to the lymph. Interestingly, despite a significant, 10-fold increase in plasma FFA, we detected little to no increase in free fatty acids or triglycerides in the lymph after lipid infusion. Thus, we conclude that experimental insulin resistance induced by lipid infusion does not reduce insulin access to skeletal muscle under clamp conditions. This would suggest that the peripheral insulin resistance is likely due to reduced cellular sensitivity to insulin in this model, and yet we did not detect a change in the tissue microenvironment that could contribute to cellular insulin resistance.
Collapse
Affiliation(s)
- Cathryn M Kolka
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Joyce M Richey
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ana Valeria B Castro
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Josiane L Broussard
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Viorica Ionut
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Richard N Bergman
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
485
|
Wang J, Xiao J, Wen D, Wu X, Mao Z, Zhang J, Ma D. Endothelial cell-anchored tissue factor pathway inhibitor regulates tumor metastasis to the lung in mice. Mol Carcinog 2015; 55:882-96. [DOI: 10.1002/mc.22329] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 02/27/2015] [Accepted: 03/26/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Jiping Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology; Institute of Biomedical Sciences, School of Basic Medical Sciences, Collaborative Innovation Center of Genetics and Development, Fudan University; Shanghai China
| | - Jiajun Xiao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology; Institute of Biomedical Sciences, School of Basic Medical Sciences, Collaborative Innovation Center of Genetics and Development, Fudan University; Shanghai China
| | - Danping Wen
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology; Institute of Biomedical Sciences, School of Basic Medical Sciences, Collaborative Innovation Center of Genetics and Development, Fudan University; Shanghai China
| | - Xie Wu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology; Institute of Biomedical Sciences, School of Basic Medical Sciences, Collaborative Innovation Center of Genetics and Development, Fudan University; Shanghai China
| | - Zuohua Mao
- Department of Parasitology and Microbiology; Shanghai Medical College, Fudan University; Shanghai China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology; Institute of Biomedical Sciences, School of Basic Medical Sciences, Collaborative Innovation Center of Genetics and Development, Fudan University; Shanghai China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology; Institute of Biomedical Sciences, School of Basic Medical Sciences, Collaborative Innovation Center of Genetics and Development, Fudan University; Shanghai China
- Children's Hospital; Fudan University; Shanghai China
| |
Collapse
|
486
|
Lan KC, Chiu CY, Kao CW, Huang KH, Wang CC, Huang KT, Tsai KS, Sheu ML, Liu SH. Advanced glycation end-products induce apoptosis in pancreatic islet endothelial cells via NF-κB-activated cyclooxygenase-2/prostaglandin E2 up-regulation. PLoS One 2015; 10:e0124418. [PMID: 25898207 PMCID: PMC4405342 DOI: 10.1371/journal.pone.0124418] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/13/2015] [Indexed: 12/23/2022] Open
Abstract
Microvascular complications eventually affect nearly all patients with diabetes. Advanced glycation end-products (AGEs) resulting from hyperglycemia are a complex and heterogeneous group of compounds that accumulate in the plasma and tissues in diabetic patients. They are responsible for both endothelial dysfunction and diabetic vasculopathy. The aim of this study was to investigate the cytotoxicity of AGEs on pancreatic islet microvascular endothelial cells. The mechanism underlying the apoptotic effect of AGEs in pancreatic islet endothelial cell line MS1 was explored. The results showed that AGEs significantly decreased MS1 cell viability and induced MS1 cell apoptosis in a dose-dependent manner. AGEs dose-dependently increased the expressions of cleaved caspase-3, and cleaved poly (ADP-ribose) polymerase in MS1 cells. Treatment of MS1 cells with AGEs also resulted in increased nuclear factor (NF)-κB-p65 phosphorylation and cyclooxygenase (COX)-2 expression. However, AGEs did not affect the expressions of endoplasmic reticulum (ER) stress-related molecules in MS1 cells. Pretreatment with NS398 (a COX-2 inhibitor) to inhibit prostaglandin E2 (PGE2) production reversed the induction of cleaved caspase-3, cleaved PARP, and MS1 cell viability. Moreover, AGEs significantly increased the receptor for AGEs (RAGE) protein expression in MS1 cells, which could be reversed by RAGE neutralizing antibody. RAGE Neutralizing antibody could also reverse the induction of cleaved caspase-3 and cleaved PARP and decreased cell viability induced by AGEs. These results implicate the involvement of NF-κB-activated COX-2/PGE2 up-regulation in AGEs/RAGE-induced islet endothelial cell apoptosis and cytotoxicity. These findings may provide insight into the pathological processes within the pancreatic islet microvasculature induced by AGEs accumulation.
Collapse
Affiliation(s)
- Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chen-Yuan Chiu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Wei Kao
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-How Huang
- Department of Urology, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Ching-Chia Wang
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Kuo-Tong Huang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keh-Sung Tsai
- Departments of Laboratory Medicine, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shing Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pediatrics, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
487
|
The combination of N-acetyl cysteine, alpha-lipoic acid, and bromelain shows high anti-inflammatory properties in novel in vivo and in vitro models of endometriosis. Mediators Inflamm 2015; 2015:918089. [PMID: 25960622 PMCID: PMC4415658 DOI: 10.1155/2015/918089] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/03/2014] [Indexed: 02/03/2023] Open
Abstract
To evaluate the efficacy of an association of N-acetyl cystein, alpha-lipoic acid, and bromelain (NAC/LA/Br) in the treatment of endometriosis we set up a new in vivo murine model. We explored the anti-inflammatory and proapoptotic effect of this combination on human endometriotic endothelial cells (EECs) and on endothelial cells isolated from normal uterus (UtMECs). We implanted fragments of human endometriotic cysts intraperitoneally into SCID mice to evaluate the efficacy of NAC/LA/Br treatment. UtMECs and EECs, untreated or treated with NAC/LA/Br, were activated with the proinflammatory stimulus TNF-α and their response in terms of VCAM1 expression was evaluated. The proapoptotic effect of higher doses of NAC/LA/Br on UtMECs and EECs was measured with a fluorogenic substrate for activated caspases 3 and 7. The preincubation of EECs with NAC/LA/Br prior to cell stimulation with TNF-α prevents the upregulation of the expression of the inflammatory “marker” VCAM1. Furthermore NAC/LA/Br were able to induce EEC, but not UtMEC, apoptosis. Finally, the novel mouse model allowed us to demonstrate that mice treated with NAC/LA/Br presented a lower number of cysts, smaller in size, compared to untreated mice. Our findings suggest that these dietary supplements may have potential therapeutic uses in the treatment of chronic inflammatory diseases like endometriosis.
Collapse
|
488
|
Béziau DM, Toussaint F, Blanchette A, Dayeh NR, Charbel C, Tardif JC, Dupuis J, Ledoux J. Expression of phosphoinositide-specific phospholipase C isoforms in native endothelial cells. PLoS One 2015; 10:e0123769. [PMID: 25875657 PMCID: PMC4395365 DOI: 10.1371/journal.pone.0123769] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/25/2015] [Indexed: 11/18/2022] Open
Abstract
Phospholipase C (PLC) comprises a superfamily of enzymes that play a key role in a wide array of intracellular signalling pathways, including protein kinase C and intracellular calcium. Thirteen different mammalian PLC isoforms have been identified and classified into 6 families (PLC-β, γ, δ, ε, ζ and η) based on their biochemical properties. Although the expression of PLC isoforms is tissue-specific, concomitant expression of different PLC has been reported, suggesting that PLC family is involved in multiple cellular functions. Despite their critical role, the PLC isoforms expressed in native endothelial cells (ECs) remains undetermined. A conventional PCR approach was initially used to elucidate the mRNA expression pattern of PLC isoforms in 3 distinct murine vascular beds: mesenteric (MA), pulmonary (PA) and middle cerebral arteries (MCA). mRNA encoding for most PLC isoforms was detected in MA, MCA and PA with the exception of η2 and β2 (only expressed in PA), δ4 (only expressed in MCA), η1 (expressed in all but MA) and ζ (not detected in any vascular beds tested). The endothelial-specific PLC expression was then sought in freshly isolated ECs. Interestingly, the PLC expression profile appears to differ across the investigated arterial beds. While mRNA for 8 of the 13 PLC isoforms was detected in ECs from MA, two additional PLC isoforms were detected in ECs from PA and MCA. Co-expression of multiple PLC isoforms in ECs suggests an elaborate network of signalling pathways: PLC isoforms may contribute to the complexity or diversity of signalling by their selective localization in cellular microdomains. However in situ immunofluorescence revealed a homogeneous distribution for all PLC isoforms probed (β3, γ2 and δ1) in intact endothelium. Although PLC isoforms play a crucial role in endothelial signal transduction, subcellular localization alone does not appear to be sufficient to determine the role of PLC in the signalling microdomains found in the native endothelium.
Collapse
Affiliation(s)
- Delphine M. Béziau
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Molecular and Integrative Physiology, Université de Montréal, Montreal, Qc, Canada
| | - Fanny Toussaint
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Molecular and Integrative Physiology, Université de Montréal, Montreal, Qc, Canada
| | | | - Nour R. Dayeh
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Medicine, Université de Montréal, Montreal, Qc, Canada
| | - Chimène Charbel
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Qc, Canada
| | - Jean-Claude Tardif
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Medicine, Université de Montréal, Montreal, Qc, Canada
| | - Jocelyn Dupuis
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Medicine, Université de Montréal, Montreal, Qc, Canada
| | - Jonathan Ledoux
- Research Center, Montreal Heart Institute, Montreal, Qc, Canada
- Department of Molecular and Integrative Physiology, Université de Montréal, Montreal, Qc, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Qc, Canada
- Department of Medicine, Université de Montréal, Montreal, Qc, Canada
- * E-mail:
| |
Collapse
|
489
|
Lilly B. We have contact: endothelial cell-smooth muscle cell interactions. Physiology (Bethesda) 2015; 29:234-41. [PMID: 24985327 DOI: 10.1152/physiol.00047.2013] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Blood vessels are composed of two primary cell types, endothelial cells and smooth muscle cells, each providing a unique contribution to vessel function. Signaling between these two cell types is essential for maintaining tone in mature vessels, and their communication is critical during development, and for repair and remodeling associated with blood vessel growth. This review will highlight the pathways that endothelial cells and smooth muscle cells utilize to communicate during vessel formation and discuss how disruptions in these pathways contribute to disease.
Collapse
Affiliation(s)
- Brenda Lilly
- Department of Pediatrics, Nationwide Children's Hospital, The Heart Center, The Ohio State University, Columbus, Ohio
| |
Collapse
|
490
|
Williams RM, Shah J, Ng BD, Minton DR, Gudas LJ, Park CY, Heller DA. Mesoscale nanoparticles selectively target the renal proximal tubule epithelium. NANO LETTERS 2015; 15:2358-64. [PMID: 25811353 PMCID: PMC4518714 DOI: 10.1021/nl504610d] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
We synthesized "mesoscale" nanoparticles, approximately 400 nm in diameter, which unexpectedly localized selectively in renal proximal tubules and up to 7 times more efficiently in the kidney than other organs. Although nanoparticles typically localize in the liver and spleen, modulating their size and opsonization potential allowed for stable targeting of the kidneys through a new proposed uptake mechanism. Applying this kidney targeting strategy, we anticipate use in the treatment of renal disease and the study of renal physiology.
Collapse
Affiliation(s)
- Ryan M. Williams
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Janki Shah
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Brandon D. Ng
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Denise R. Minton
- Weill Cornell Graduate School of Medical Sciences, New York, New York 10065, United States
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Christopher Y. Park
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, New York 10065
| | - Daniel A. Heller
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Pharmacology, Weill Cornell Medical College, New York, New York 10065, United States
| |
Collapse
|
491
|
Cheng H, Harris RC. Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Hematol Disord Drug Targets 2015; 14:22-33. [PMID: 24720460 DOI: 10.2174/1871529x14666140401110841] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 03/21/2014] [Accepted: 03/26/2014] [Indexed: 12/24/2022]
Abstract
Endothelial dysfunction has been posited to play an important role in the pathogenesis of diabetic nephropathy (DN). Due to the heterogeneity of endothelial cells (ECs), it is difficult to generalize about endothelial responses to diabetic stimuli. At present, there are limited techniques fordirectly measuring EC function in vivo, so diagnosis of endothelial disorders still largely depends on indirect assessment of mediators arising from EC injury. In the kidney microcirculation, both afferent and efferent arteries, arterioles and glomerular endothelial cells (GEnC) have all been implicated as targets of diabetic injury. Both hyperglycemia per se, as well as the metabolic consequences of glucose dysregulation, are thought to lead to endothelial cell dysfunction. In this regard, endothelial nitric oxide synthase (eNOS) plays a central role in EC dysfunction. Impaired eNOS activity can occur at numerous levels, including enzyme uncoupling, post-translational modifications, internalization and decreased expression. Reduced nitric oxide (NO) bioavailability exacerbates oxidative stress, further promoting endothelial dysfunction and injury. The injured ECs may then function as active signal transducers of metabolic, hemodynamic and inflammatory factors that modify the function and morphology of the vessel wall and interact with adjacent cells, which may activate a cascade of inflammatory and proliferative and profibrotic responses in progressive DN. Both pharmacological approaches and potential regenerative therapies hold promise for restoration of impaired endothelial cells in diabetic nephropathy.
Collapse
Affiliation(s)
| | - Raymond C Harris
- Division of Nephrology, S3223 MCN, Vanderbilt University School of Medicine, and Nashville Veterans Affairs Hospital, Nashville, TN 37232, USA.
| |
Collapse
|
492
|
|
493
|
van Bussel BCT, Henry RMA, Ferreira I, van Greevenbroek MMJ, van der Kallen CJH, Twisk JWR, Feskens EJM, Schalkwijk CG, Stehouwer CDA. A healthy diet is associated with less endothelial dysfunction and less low-grade inflammation over a 7-year period in adults at risk of cardiovascular disease. J Nutr 2015; 145:532-40. [PMID: 25733469 DOI: 10.3945/jn.114.201236] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND A healthy diet rich in fish, fruit, and vegetables, but moderate in alcohol and low in dairy products and meat, has been associated with a lower rate of incident cardiovascular disease (CVD). The underlying mechanisms, however, remain unclear. Endothelial dysfunction and low-grade inflammation play important roles in CVD. A healthy diet might modify these phenomena. OBJECTIVE We investigated the associations between the above food groups and overall biomarker scores of endothelial dysfunction and low-grade inflammation in a 7-y longitudinal study. METHODS Using longitudinal data from 557 participants at increased CVD risk from the CODAM (Cohort on Diabetes and Atherosclerosis Maastricht) Study, we assessed diet intake by food-frequency questionnaire and measured plasma biomarkers of endothelial dysfunction [von Willebrand factor, soluble vascular cell adhesion molecule 1, soluble endothelial selectin, soluble thrombomodulin, soluble intercellular adhesion molecule 1 (sICAM-1)] and low-grade inflammation [C-reactive protein, serum amyloid A, interleukin (IL)-6, IL-8, tumor necrosis factor α, and sICAM-1]. At baseline, participants were aged 59.6 ± 6.9 y. Measurements were performed then and after 7 y. Biomarkers were combined into overall scores (sum of z scores; higher scores indicating worse function). Longitudinal data were analyzed with generalized estimating equations and adjusted for sex, age, glucose metabolism, energy intake, body mass index, physical activity, alcohol consumption, and smoking. RESULTS Higher consumption of fish (per 100 g/wk), but not total consumption of vegetables, fruit, alcohol-containing beverages, dairy products, or meat, was associated with a lower overall endothelial dysfunction score over 7 y (β: -0.027; 95% CI: -0.051, -0.004). No associations were observed with the overall low-grade inflammation score. Further food component analyses indicated that consumption of more lean fish (per 100 g/wk) and raw vegetables (per 100 g/d), and fewer high-fat dairy products (per 100 g/d) was associated with less endothelial dysfunction [(β: -0.038; 95% CI: -0.072, -0.005), (β: -0.095; 95% CI: -0.191, 0.000), and (β: -0.070; 95% CI: -0.131, -0.009), respectively]. Consumption of more fresh fruit (per 100 g/d), wine (per 100 mL/wk), and poultry (per 100 g/d), and fewer high-fat dairy products (per 100 g/d) was associated with less low-grade inflammation [(β: -0.074; 95% CI: -0.133, -0.015), (β:-0.006; 95% CI: -0.013, 0.001), (β:-0.247; 95% CI: -0.479, -0.014), and (β:-0.100; 95% CI: -0.182, -0.019), respectively]. CONCLUSION These data suggest that the dietary modification of endothelial dysfunction and low-grade inflammation, processes that are important in atherothrombosis, is possible.
Collapse
Affiliation(s)
- Bas C T van Bussel
- Department of Medicine School for Nutrition, Toxicology, and Metabolism Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Ronald M A Henry
- Department of Medicine Cardiovascular Research Institute Maastricht Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Isabel Ferreira
- Department of Medicine Cardiovascular Research Institute Maastricht School for Public Health and Primary Care, and Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | - Jos W R Twisk
- The Institute for Research in Extramural Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Edith J M Feskens
- Top Institute Food and Nutrition, Wageningen, The Netherlands Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Casper G Schalkwijk
- Department of Medicine Cardiovascular Research Institute Maastricht Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Coen D A Stehouwer
- Department of Medicine Cardiovascular Research Institute Maastricht Top Institute Food and Nutrition, Wageningen, The Netherlands
| |
Collapse
|
494
|
Mobilization of endothelial progenitor cells in acute cardiovascular events in the PROCELL study: Time-course after acute myocardial infarction and stroke. J Mol Cell Cardiol 2015; 80:146-55. [DOI: 10.1016/j.yjmcc.2015.01.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 01/08/2015] [Accepted: 01/13/2015] [Indexed: 11/19/2022]
|
495
|
Stanczuk L, Martinez-Corral I, Ulvmar MH, Zhang Y, Laviña B, Fruttiger M, Adams R, Saur D, Betsholtz C, Ortega S, Alitalo K, Graupera M, Mäkinen T. cKit Lineage Hemogenic Endothelium-Derived Cells Contribute to Mesenteric Lymphatic Vessels. Cell Rep 2015; 10:1708-1721. [DOI: 10.1016/j.celrep.2015.02.026] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 12/29/2014] [Accepted: 02/05/2015] [Indexed: 10/23/2022] Open
|
496
|
Chen HL, Lu CH, Lin HC, Chen PC, Chou KH, Lin WM, Tsai NW, Su YJ, Friedman M, Lin CP, Lin WC. White matter damage and systemic inflammation in obstructive sleep apnea. Sleep 2015; 38:361-70. [PMID: 25325459 DOI: 10.5665/sleep.4490] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 07/17/2014] [Indexed: 12/22/2022] Open
Abstract
STUDY OBJECTIVES To evaluate white matter integrity in patients with obstructive sleep apnea (OSA) using diffusion tensor imaging (DTI) and to assess its relationship with systemic inflammation. DESIGN Cross-sectional study. SETTING One tertiary medical center research institute. PATIENTS OR PARTICIPANTS Twenty patients with severe OSA (apnea-hypopnea index [AHI] > 30, 18 men and 2 women) and 14 healthy volunteers (AHI < 5, 11 men and 3 women). INTERVENTIONS N/A. MEASUREMENTS AND RESULTS Patients with severe OSA and healthy volunteers underwent polysomnography to determine the severity of sleep apnea, and DTI scanning to determine fiber integrity. Early or late phase changes in leukocyte apoptosis and its subsets were determined by flow cytometry. DTI-related indices (including fractional anisotropy [FA], axial diffusivity [AD], radial diffusivity [RD], and mean diffusivity [MD]) were derived from DTI. The FA maps were compared using voxel-based statistics to determine differences between the severe OSA and control groups. The differences in DTI indices, clinical severity, and leukocyte apoptosis were correlated after adjusting for age, sex, body mass index, and systolic blood pressure. Exploratory group-wise comparison between the two groups revealed that patients with OSA exhibited low FA accomplished by high RD in several brain locations, without any differences in AD and MD. The FA values were negatively correlated with clinical disease severity and leukocyte early apoptosis. CONCLUSIONS Obstructive sleep apnea impairs white matter integrity in vulnerable regions, and this impairment is associated with increased disease severity. The possible interactions between systemic inflammation and central nervous system microstructural damage may represent variant hypoxic patterns and their consequent processes in OSA.
Collapse
Affiliation(s)
- Hsiu-Ling Chen
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Hsien Lu
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Hsin-Ching Lin
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Sleep Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pei-Chin Chen
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kun-Hsien Chou
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Ming Lin
- Department of Diagnostic Radiology, Chiayi Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Chiayi, Taiwan
| | - Nai-Wen Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Jih Su
- Department of Biological Science, National Sun Yat-Sen University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Michael Friedman
- Department of Otolaryngology-Head and Neck Surgery, Division of Sleep Surgery, Rush University Medical Center, Chicago, IL.,Department of Otolaryngology, Advanced Center for Specialty Care, Advocate Illinois Masonic Medical Center, Chicago, IL
| | - Ching-Po Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Che Lin
- Department of Diagnostic Radiology, Kaohsiung Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
497
|
Reinitz A, DeStefano J, Ye M, Wong AD, Searson PC. Human brain microvascular endothelial cells resist elongation due to shear stress. Microvasc Res 2015; 99:8-18. [PMID: 25725258 DOI: 10.1016/j.mvr.2015.02.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 02/02/2023]
Abstract
Endothelial cells in straight sections of vessels are known to elongate and align in the direction of flow. This phenotype has been replicated in confluent monolayers of bovine aortic endothelial cells and human umbilical vein endothelial cells (HUVECs) in cell culture under physiological shear stress. Here we report on the morphological response of human brain microvascular endothelial cells (HBMECs) in confluent monolayers in response to shear stress. Using a microfluidic platform we image confluent monolayers of HBMECs and HUVECs under shear stresses up to 16 dyne cm(-2). From live-cell imaging we quantitatively analyze the cell morphology and cell speed as a function of time. We show that HBMECs do not undergo a classical transition from cobblestone to spindle-like morphology in response to shear stress. We further show that under shear stress, actin fibers are randomly oriented in the cells indicating that there is no cytoskeletal remodeling. These results suggest that HBMECs are programmed to resist elongation and alignment under shear stress, a phenotype that may be associated with the unique properties of the blood-brain barrier.
Collapse
Affiliation(s)
- Adam Reinitz
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Jackson DeStefano
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Mao Ye
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Andrew D Wong
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Peter C Searson
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
498
|
Dane MJC, van den Berg BM, Lee DH, Boels MGS, Tiemeier GL, Avramut MC, van Zonneveld AJ, van der Vlag J, Vink H, Rabelink TJ. A microscopic view on the renal endothelial glycocalyx. Am J Physiol Renal Physiol 2015; 308:F956-66. [PMID: 25673809 DOI: 10.1152/ajprenal.00532.2014] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 02/04/2015] [Indexed: 11/22/2022] Open
Abstract
Endothelial cells perform key homeostatic functions such as regulating blood flow, permeability, and aiding immune surveillance for pathogens. While endothelial activation serves normal physiological adaptation, maladaptation of these endothelial functions has been identified as an important effector mechanism in the progression of renal disease as well as the associated development of cardiovascular disease. The primary interface between blood and the endothelium is the glycocalyx. This carbohydrate-rich gel-like structure with its associated proteins mediates most of the regulatory functions of the endothelium. Because the endothelial glycocalyx is a highly dynamic and fragile structure ex vivo, and traditional tissue processing for staining and perfusion-fixation usually results in a partial or complete loss of the glycocalyx, studying its dimensions and function has proven to be challenging. In this review, we will outline the core functions of the glycocalyx and focus on different techniques to study structure-function relationships in kidney and vasculature.
Collapse
Affiliation(s)
- Martijn J C Dane
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernard M van den Berg
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Dae Hyun Lee
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Margien G S Boels
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Gesa L Tiemeier
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - M Cristina Avramut
- Department of Molecular Cell Biology, Section Electron Microscopy LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Anton Jan van Zonneveld
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; and
| | - Hans Vink
- Department of Physiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ton J Rabelink
- Department of Nephrology, Einthoven laboratory for Vascular Medicine, LUMC, Leiden University Medical Center, Leiden, The Netherlands;
| |
Collapse
|
499
|
Sandoo A, Kitas GD. A methodological approach to non-invasive assessments of vascular function and morphology. J Vis Exp 2015. [PMID: 25741637 PMCID: PMC4354622 DOI: 10.3791/52339] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The endothelium is the innermost lining of the vasculature and is involved in the maintenance of vascular homeostasis. Damage to the endothelium may predispose the vessel to atherosclerosis and increase the risk for cardiovascular disease. Assessments of peripheral endothelial function are good indicators of early abnormalities in the vascular wall and correlate well with assessments of coronary endothelial function. The present manuscript details the important methodological steps necessary for the assessment of microvascular endothelial function using laser Doppler imaging with iontophoresis, large vessel endothelial function using flow-mediated dilatation, and carotid atherosclerosis using carotid artery ultrasound. A discussion on the methodological considerations for each of the techniques is also presented, and recommendations are made for future research.
Collapse
Affiliation(s)
- Aamer Sandoo
- School of Sport, Health and Exercise Sciences, Bangor University; Department of Rheumatology, Dudley Group of Hospitals NHS Trust, Russells Hall Hospital;
| | - George D Kitas
- Department of Rheumatology, Dudley Group of Hospitals NHS Trust, Russells Hall Hospital; Arthritis Research UK Epidemiology Unit, University of Manchester
| |
Collapse
|
500
|
Development and characterization of an endothelial cell line from the bulbus arteriosus of walleye, Sander vitreus. Comp Biochem Physiol A Mol Integr Physiol 2015; 180:57-67. [DOI: 10.1016/j.cbpa.2014.10.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 09/29/2014] [Accepted: 10/10/2014] [Indexed: 11/15/2022]
|