1
|
Song J, Lee Y, Kim MS, Ha G, Jang W, Batjargal U, Kim Y, Kim HJ, Lee J. High throughput drug screening platform utilizing capillary and artery cell layered models based on tumor-vascular cell interactions. LAB ON A CHIP 2025; 25:2349-2363. [PMID: 40177711 DOI: 10.1039/d4lc00950a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Interactions between tumors and adjacent blood vessels are critical in the tumor microenvironment (TME) for influencing angiogenesis and hematogenous metastasis. Understanding these interactions within the native TME is vital for targeting various tumors, including brain tumors, due to the complexities of the blood-brain barrier. Developing an accurate tumor model that includes cell-cell and cell-matrix interactions, as well as blood flow-induced shear stress, is essential for high-throughput screening (HTS) of anti-cancer drugs. Here, we developed a glioblastoma (GBM) model surrounded by vascular cells. The arterial model was constructed by encapsulating GBM spheroids with layers of human smooth muscle cells (SMCs) and human umbilical vein endothelial cells (HUVECs), while the capillary cell layered model used only HUVECs. Comparative analysis with tumors from different organs revealed the significant role for platelet endothelial cell adhesion molecule (PECAM) in GBM-blood vascular cell interactions. Cytokine secretion analysis demonstrated PECAM's impact on tumor-specific angiogenic potential. Testing with anti-cancer drugs revealed increased expression of PECAM-associated proteins, drug resistance cytokines, and genes associated with tumor progression and metastasis. Additionally, we developed a HTS platform by encapsulating these tumor models in hydrogels and subjecting them to media circulation, effectively mimicking the dynamic TME, suitable for cancer treatment research and drug development.
Collapse
Affiliation(s)
- Jihyeon Song
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| | - Yeji Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| | - Min-Seok Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
| | - Giheon Ha
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| | - WonJun Jang
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Ulziituya Batjargal
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Younggyun Kim
- Department of Bioengineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Han-Jun Kim
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
- Interdisciplinary Major Program in Innovative Pharmaceutical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| |
Collapse
|
2
|
Zablotskii V, Polyakova T, Dejneka A. Exploring Ion Channel Magnetic Pharmacology: Are Magnetic Cues a Viable Alternative to Ion Channel Drugs? Bioessays 2025; 47:e202400200. [PMID: 39651810 PMCID: PMC11848120 DOI: 10.1002/bies.202400200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 12/11/2024]
Abstract
We explore the potential of using magnetic cues as a novel approach to modulating ion channel expression, which could provide an alternative to traditional pharmacological interventions. Ion channels are crucial targets for pharmacological therapies, and ongoing research in this field continues to introduce new methods for treating various diseases. However, the efficacy of ion channel drugs is often compromised by issues such as target selectivity, leading to side effects, toxicity, and complex drug interactions. These challenges, along with problems like drug resistance and difficulties in crossing biological barriers, highlight the need for innovative strategies. In this context, the proposed use of magnetic cues to modulate ion channel expression may offer a promising solution to address these limitations, potentially improving the safety and effectiveness of treatments, particularly for long-term use. Key developments in this area are reviewed, the relationships between changes in ion channel expression and magnetic fields are summarized, knowledge gaps are identified, and central issues relevant to future research are discussed.
Collapse
Affiliation(s)
- Vitalii Zablotskii
- Institute of Physics of the Czech Academy of SciencesPragueCzech Republic
| | - Tatyana Polyakova
- Institute of Physics of the Czech Academy of SciencesPragueCzech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
3
|
Brandon KD, Frank WE, Stroka KM. Junctions at the crossroads: the impact of mechanical cues on endothelial cell-cell junction conformations and vascular permeability. Am J Physiol Cell Physiol 2024; 327:C1073-C1086. [PMID: 39129490 PMCID: PMC11481987 DOI: 10.1152/ajpcell.00605.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
Cells depend on precisely regulating barrier function within the vasculature to maintain physiological stability and facilitate essential substance transport. Endothelial cells achieve this through specialized adherens and tight junction protein complexes, which govern paracellular permeability across vascular beds. Adherens junctions, anchored by vascular endothelial (VE)-cadherin and associated catenins to the actin cytoskeleton, mediate homophilic adhesion crucial for barrier integrity. In contrast, tight junctions composed of occludin, claudin, and junctional adhesion molecule A interact with Zonula Occludens proteins, reinforcing intercellular connections essential for barrier selectivity. Endothelial cell-cell junctions exhibit dynamic conformations during development, maturation, and remodeling, regulated by local biochemical and mechanical cues. These structural adaptations play pivotal roles in disease contexts such as chronic inflammation, where junctional remodeling contributes to increased vascular permeability observed in conditions from cancer to cardiovascular diseases. Conversely, the brain microvasculature's specialized junctional arrangements pose challenges for therapeutic drug delivery due to their unique molecular compositions and tight organization. This commentary explores the molecular mechanisms underlying endothelial cell-cell junction conformations and their implications for vascular permeability. By highlighting recent advances in quantifying junctional changes and understanding mechanotransduction pathways, we elucidate how physical forces from cellular contacts and hemodynamic flow influence junctional dynamics.
Collapse
Affiliation(s)
- Ken D Brandon
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
| | - William E Frank
- Department of Biology, University of Puerto Rico in Ponce, Ponce, Puerto Rico
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, United States
- Biophysics Program, University of Maryland, College Park, Maryland, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, Maryland, United States
| |
Collapse
|
4
|
Ting KK, Coleman P, Kim HJ, Zhao Y, Mulangala J, Cheng NC, Li W, Gunatilake D, Johnstone DM, Loo L, Neely GG, Yang P, Götz J, Vadas MA, Gamble JR. Vascular senescence and leak are features of the early breakdown of the blood-brain barrier in Alzheimer's disease models. GeroScience 2023; 45:3307-3331. [PMID: 37782439 PMCID: PMC10643714 DOI: 10.1007/s11357-023-00927-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 08/27/2023] [Indexed: 10/03/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related disease, with loss of integrity of the blood-brain barrier (BBB) being an early feature. Cellular senescence is one of the reported nine hallmarks of aging. Here, we show for the first time the presence of senescent cells in the vasculature in AD patients and mouse models of AD. Senescent endothelial cells and pericytes are present in APP/PS1 transgenic mice but not in wild-type littermates at the time of amyloid deposition. In vitro, senescent endothelial cells display altered VE-cadherin expression and loss of cell junction formation and increased permeability. Consistent with this, senescent endothelial cells in APP/PS1 mice are present at areas of vascular leak that have decreased claudin-5 and VE-cadherin expression confirming BBB breakdown. Furthermore, single cell sequencing of endothelial cells from APP/PS1 transgenic mice confirms that adhesion molecule pathways are among the most highly altered pathways in these cells. At the pre-plaque stage, the vasculature shows significant signs of breakdown, with a general loss of VE-cadherin, leakage within the microcirculation, and obvious pericyte perturbation. Although senescent vascular cells were not directly observed at sites of vascular leak, senescent cells were close to the leak area. Thus, we would suggest in AD that there is a progressive induction of senescence in constituents of the neurovascular unit contributing to an increasing loss of vascular integrity. Targeting the vasculature early in AD, either with senolytics or with drugs that improve the integrity of the BBB may be valid therapeutic strategies.
Collapse
Affiliation(s)
- Ka Ka Ting
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia.
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.
| | - Paul Coleman
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Hani Jieun Kim
- Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Yang Zhao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jocelyne Mulangala
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia
| | - Ngan Ching Cheng
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia
| | - Wan Li
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Dilini Gunatilake
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia
| | - Daniel M Johnstone
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Lipin Loo
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, Centenary Institute, & School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - G Gregory Neely
- Charles Perkins Centre, Dr. John and Anne Chong Lab for Functional Genomics, Centenary Institute, & School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Pengyi Yang
- Computational Systems Biology Group, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Mathew A Vadas
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia
- Heart Research Institute, Sydney, NSW, Australia
| | - Jennifer R Gamble
- Vascular Biology Program, Centenary Institute, Camperdown, NSW, Australia.
- School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
5
|
Shi H, Koronyo Y, Fuchs DT, Sheyn J, Jallow O, Mandalia K, Graham SL, Gupta VK, Mirzaei M, Kramerov AA, Ljubimov AV, Hawes D, Miller CA, Black KL, Carare RO, Koronyo-Hamaoui M. Retinal arterial Aβ 40 deposition is linked with tight junction loss and cerebral amyloid angiopathy in MCI and AD patients. Alzheimers Dement 2023; 19:5185-5197. [PMID: 37166032 PMCID: PMC10638467 DOI: 10.1002/alz.13086] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 05/12/2023]
Abstract
INTRODUCTION Vascular amyloid beta (Aβ) protein deposits were detected in retinas of mild cognitively impaired (MCI) and Alzheimer's disease (AD) patients. We tested the hypothesis that the retinal vascular tight junctions (TJs) were compromised and linked to disease status. METHODS TJ components and Aβ expression in capillaries and larger blood vessels were determined in post mortem retinas from 34 MCI or AD patients and 27 cognitively normal controls and correlated with neuropathology. RESULTS Severe decreases in retinal vascular zonula occludens-1 (ZO-1) and claudin-5 correlating with abundant arteriolar Aβ40 deposition were identified in MCI and AD patients. Retinal claudin-5 deficiency was closely associated with cerebral amyloid angiopathy, whereas ZO-1 defects correlated with cerebral pathology and cognitive deficits. DISCUSSION We uncovered deficiencies in blood-retinal barrier markers for potential retinal imaging targets of AD screening and monitoring. Intense retinal arteriolar Aβ40 deposition suggests a common pathogenic mechanism of failed Aβ clearance via intramural periarterial drainage.
Collapse
Affiliation(s)
- Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Krishna Mandalia
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stuart L. Graham
- Macquarie Medical school, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Vivek K. Gupta
- Macquarie Medical school, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Mehdi Mirzaei
- Macquarie Medical school, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Andrei A. Kramerov
- Department of Biomedical Sciences and Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alexander V. Ljubimov
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences and Eye Program, Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Debra Hawes
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90048, USA
| | - Carol A. Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90048, USA
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Roxana O. Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton SO16 6YD, UK
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
6
|
Gorobets O, Gorobets S, Sharai I, Polyakova T, Zablotskii V. Interaction of magnetic fields with biogenic magnetic nanoparticles on cell membranes: Physiological consequences for organisms in health and disease. Bioelectrochemistry 2023; 151:108390. [PMID: 36746089 DOI: 10.1016/j.bioelechem.2023.108390] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
The interaction mechanisms between magnetic fields (MFs) and living systems, which remained hidden for more than a hundred years, continue to attract the attention of researchers from various disciplines: physics, biology, medicine, and life sciences. Revealing these mechanisms at the cellular level would allow to understand complex cell systems and could help to explain and predict cell responses to MFs, intervene in organisms' reactions to MFs of different strengths, directions, and spatial distributions. We suggest several new physical mechanisms of the MF impacts on endothelial and cancer cells by the MF interaction with chains of biogenic and non-biogenic magnetic nanoparticles on cell membranes. The revealed mechanisms can play a hitherto unexpected role in creating physiological responses of organisms to externally applied MFs. We have also a set of theoretical models that can predict how cells will individually and collectively respond to a MF exposure. The physiological sequences of the MF - cell interactions for organisms in health and disease are discussed. The described effects and their underlying mechanisms are general and should take place in a large family of biological effects of MFs. The results are of great importance for further developing novel approaches in cell biology, cell therapy and medicine.
Collapse
Affiliation(s)
- Oksana Gorobets
- National Technical University of Ukraine, "Igor Sikorsky Kyiv Polytechnic Institute", Ukraine; Institute of Magnetism of NAS and MES of Ukraine, Ukraine.
| | - Svitlana Gorobets
- National Technical University of Ukraine, "Igor Sikorsky Kyiv Polytechnic Institute", Ukraine
| | - Iryna Sharai
- National Technical University of Ukraine, "Igor Sikorsky Kyiv Polytechnic Institute", Ukraine; Institute of Magnetism of NAS and MES of Ukraine, Ukraine
| | - Tatyana Polyakova
- Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vitalii Zablotskii
- Institute of Physics of the Czech Academy of Sciences, Prague, Czech Republic; International Magnetobiology Frontier Research Center (iMFRC), Science Island, China
| |
Collapse
|
7
|
Sung B, Baek YY, Kim YM, You JC. Topical Administration of a Novel Acetylated Tetrapeptide Suppresses Vascular Permeability and Immune Responses and Alleviates Atopic Dermatitis in a Murine Model. Int J Mol Sci 2022; 23:ijms232113498. [PMID: 36362286 PMCID: PMC9658216 DOI: 10.3390/ijms232113498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/25/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Although the pathogenesis of atopic dermatitis (AD) remains to be fully deciphered, skin barrier abnormality and immune dysregulation are known to be involved. Recently, the vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR) system has also been implicated in the pathogenesis of this multifactorial chronic inflammatory skin disorder. Previously, we showed that a novel tetrapeptide, N-acetyl-Arg-Leu-Tyr-Glu (Ac-RLYE), inhibits angiogenesis and vascular permeability effectively by selectively antagonizing VEGFR-2. The current study aimed to investigate the pharmacological effect of Ac-RLYE on AD in vitro and in vivo. The in vitro experiments demonstrated that Ac-RLYE inhibited VEGF-induced vascular permeability in endothelial cells. Moreover, in an in vivo animal model of AD, Ac-RLYE relieved AD-like symptoms such as ear thickness and dermatitis severity scores and infiltration of immune cells, including mast cells and eosinophils. Ac-RLYE inhibited IgE secretion, restored the skin barrier protein filaggrin level, and markedly downregulated gene expression of AD-related Th1, Th2, and Th17 cytokines. Collectively, these findings suggest that Ac-RLYE would be useful for the treatment of AD and associated inflammatory skin disorders.
Collapse
Affiliation(s)
- Bokyung Sung
- Avixgen Inc., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, Korea
| | - Yi-Yong Baek
- Avixgen Inc., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Ji Chang You
- Avixgen Inc., 2477, Nambusunhwan-ro, Seocho-gu, Seoul 06725, Korea
- National Research Laboratory for Molecular Virology, Department of Pathology, School of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
8
|
Man K, Liu J, Phan KM, Wang K, Lee JY, Sun X, Story M, Saha D, Liao J, Sadat H, Yang Y. Dimensionality-Dependent Mechanical Stretch Regulation of Cell Behavior. ACS APPLIED MATERIALS & INTERFACES 2022; 14:17081-17092. [PMID: 35380801 DOI: 10.1021/acsami.2c01266] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A variety of cells are subject to mechanical stretch in vivo, which plays a critical role in the function and homeostasis of cells, tissues, and organs. Deviations from the physiologically relevant mechanical stretch are often associated with organ dysfunction and various diseases. Although mechanical stretch is provided in some in vitro cell culture models, the effects of stretch dimensionality on cells are often overlooked and it remains unclear whether and how stretch dimensionality affects cell behavior. Here we develop cell culture platforms that provide 1-D uniaxial, 2-D circumferential, or 3-D radial mechanical stretches, which recapitulate the three major types of mechanical stretches that cells experience in vivo. We investigate the behavior of human microvascular endothelial cells and human alveolar epithelial cells cultured on these platforms, showing that the mechanical stretch influences cell morphology and cell-cell and cell-substrate interactions in a stretch dimensionality-dependent manner. Furthermore, the endothelial and epithelial cells are sensitive to the physiologically relevant 2-D and 3-D stretches, respectively, which could promote the formation of endothelium and epithelium. This study underscores the importance of recreating the physiologically relevant mechanical stretch in the development of in vitro tissue/organ models.
Collapse
Affiliation(s)
- Kun Man
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Khang Minh Phan
- Department of Mechanical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Kai Wang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Jung Yeon Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Michael Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76010, United States
| | - Hamid Sadat
- Department of Mechanical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| |
Collapse
|
9
|
Jones JH, Minshall RD. Endothelial Transcytosis in Acute Lung Injury: Emerging Mechanisms and Therapeutic Approaches. Front Physiol 2022; 13:828093. [PMID: 35431977 PMCID: PMC9008570 DOI: 10.3389/fphys.2022.828093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury (ALI) is characterized by widespread inflammation which in its severe form, Acute Respiratory Distress Syndrome (ARDS), leads to compromise in respiration causing hypoxemia and death in a substantial number of affected individuals. Loss of endothelial barrier integrity, pneumocyte necrosis, and circulating leukocyte recruitment into the injured lung are recognized mechanisms that contribute to the progression of ALI/ARDS. Additionally, damage to the pulmonary microvasculature by Gram-negative and positive bacteria or viruses (e.g., Escherichia coli, SARS-Cov-2) leads to increased protein and fluid permeability and interstitial edema, further impairing lung function. While most of the vascular leakage is attributed to loss of inter-endothelial junctional integrity, studies in animal models suggest that transendothelial transport of protein through caveolar vesicles, known as transcytosis, occurs in the early phase of ALI/ARDS. Here, we discuss the role of transcytosis in healthy and injured endothelium and highlight recent studies that have contributed to our understanding of the process during ALI/ARDS. We also cover potential approaches that utilize caveolar transport to deliver therapeutics to the lungs which may prevent further injury or improve recovery.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,*Correspondence: Richard D. Minshall,
| |
Collapse
|
10
|
Choi J, Mathew S, Oerter S, Appelt-Menzel A, Hansmann J, Schmitz T. Online Measurement System for Dynamic Flow Bioreactors to Study Barrier Integrity of hiPSC-Based Blood-Brain Barrier In Vitro Models. Bioengineering (Basel) 2022; 9:bioengineering9010039. [PMID: 35049748 PMCID: PMC8773345 DOI: 10.3390/bioengineering9010039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/31/2022] Open
Abstract
Electrochemical impedance spectroscopy (EIS) is a noninvasive, reliable, and efficient method to analyze the barrier integrity of in vitro tissue models. This well-established tool is used most widely to quantify the transendothelial/epithelial resistance (TEER) of Transwell-based models cultured under static conditions. However, dynamic culture in bioreactors can achieve advanced cell culture conditions that mimic a more tissue-specific environment and stimulation. This requires the development of culture systems that also allow for the assessment of barrier integrity under dynamic conditions. Here, we present a bioreactor system that is capable of the automated, continuous, and non-invasive online monitoring of cellular barrier integrity during dynamic culture. Polydimethylsiloxane (PDMS) casting and 3D printing were used for the fabrication of the bioreactors. Additionally, attachable electrodes based on titanium nitride (TiN)-coated steel tubes were developed to perform EIS measurements. In order to test the monitored bioreactor system, blood–brain barrier (BBB) in vitro models derived from human-induced pluripotent stem cells (hiPSC) were cultured for up to 7 days. We applied equivalent electrical circuit fitting to quantify the electrical parameters of the cell layer and observed that TEER gradually decreased over time from 2513 Ω·cm2 to 285 Ω·cm2, as also specified in the static control culture. Our versatile system offers the possibility to be used for various dynamic tissue cultures that require a non-invasive monitoring system for barrier integrity.
Collapse
Affiliation(s)
- Jihyoung Choi
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
- Correspondence: (J.C.); (A.A.-M.)
| | - Sanjana Mathew
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
| | - Sabrina Oerter
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research, Röntgenring 11, 97070 Würzburg, Germany;
| | - Antje Appelt-Menzel
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research, Röntgenring 11, 97070 Würzburg, Germany;
- Correspondence: (J.C.); (A.A.-M.)
| | - Jan Hansmann
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
- Faculty of Electronics, University of Applied Science Würzburg-Schweinfurt, Ignaz-Schön-Straße 11, 97421 Schweinfurt, Germany
| | - Tobias Schmitz
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Röntgenring 11, 97070 Würzburg, Germany; (S.M.); (J.H.); (T.S.)
| |
Collapse
|
11
|
Wang L, Astone M, Alam SK, Zhu Z, Pei W, Frank DA, Burgess SM, Hoeppner LH. Suppressing STAT3 activity protects the endothelial barrier from VEGF-mediated vascular permeability. Dis Model Mech 2021; 14:272222. [PMID: 34542605 PMCID: PMC8592016 DOI: 10.1242/dmm.049029] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 09/10/2021] [Indexed: 12/27/2022] Open
Abstract
Vascular permeability triggered by inflammation or ischemia promotes edema, exacerbates disease progression and impairs tissue recovery. Vascular endothelial growth factor (VEGF) is a potent inducer of vascular permeability. VEGF plays an integral role in regulating vascular barrier function physiologically and in pathologies, including cancer, stroke, cardiovascular disease, retinal conditions and COVID-19-associated pulmonary edema, sepsis and acute lung injury. Understanding temporal molecular regulation of VEGF-induced vascular permeability will facilitate developing therapeutics to inhibit vascular permeability, while preserving tissue-restorative angiogenesis. Here, we demonstrate that VEGF signals through signal transducer and activator of transcription 3 (STAT3) to promote vascular permeability. We show that genetic STAT3 ablation reduces vascular permeability in STAT3-deficient endothelium of mice and VEGF-inducible zebrafish crossed with CRISPR/Cas9-generated Stat3 knockout zebrafish. Intercellular adhesion molecule 1 (ICAM-1) expression is transcriptionally regulated by STAT3, and VEGF-dependent STAT3 activation is regulated by JAK2. Pyrimethamine, an FDA-approved antimicrobial agent that inhibits STAT3-dependent transcription, substantially reduces VEGF-induced vascular permeability in zebrafish, mouse and human endothelium. Collectively, our findings suggest that VEGF/VEGFR-2/JAK2/STAT3 signaling regulates vascular barrier integrity, and inhibition of STAT3-dependent activity reduces VEGF-induced vascular permeability. This article has an associated First Person interview with the first author of the paper. Summary: Genetic STAT3 ablation in mice and VEGF-inducible zebrafish reveals that VEGF signals through STAT3 to promote vascular permeability. Pyrimethamine reduces VEGF-induced permeability in animal models.
Collapse
Affiliation(s)
- Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Zhu Zhu
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
12
|
Kotlyarov S. Diversity of Lipid Function in Atherogenesis: A Focus on Endothelial Mechanobiology. Int J Mol Sci 2021; 22:11545. [PMID: 34768974 PMCID: PMC8584259 DOI: 10.3390/ijms222111545] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the most important problems in modern medicine. Its high prevalence and social significance determine the need for a better understanding of the mechanisms of the disease's development and progression. Lipid metabolism and its disorders are one of the key links in the pathogenesis of atherosclerosis. Lipids are involved in many processes, including those related to the mechanoreception of endothelial cells. The multifaceted role of lipids in endothelial mechanobiology and mechanisms of atherogenesis are discussed in this review. Endothelium is involved in ensuring adequate vascular hemodynamics, and changes in blood flow characteristics are detected by endothelial cells and affect their structure and function.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
13
|
Gorzelak-Pabis P, Broncel M, Wojdan K, Gajewski A, Chalubinski M, Gawrysiak M, Wozniak E. Rivaroxaban protects from the oxysterol-induced damage and inflammatory activation of the vascular endothelium. Tissue Barriers 2021; 9:1956284. [PMID: 34323663 PMCID: PMC8794498 DOI: 10.1080/21688370.2021.1956284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Rivaroxaban is one of the direct factor Xa inhibitors. Its function in the inactivated coagulation cascade is unclear. The aim of the study was to assess the effect of rivaroxaban on the endothelial integrity and inflammatory properties of endothelial cells stimulated by 25-hydroxycholesterol (25-OHC). METHODS HUVECs were stimulated with 25-OHC, rivaroxaban and 25-OHC+ rivaroxaban. HUVEC integrity and permeability were measured using the xCELLigence system and paracellular flux assay. The mRNA expression of tissue factor, ICAM-1, VEGF, IL-33, MCP-1, TNF-α was analyzed in the real-time PCR. Apoptosis and viability were measured by flow cytometry. The VEGF protein concentration was assessed by ELISA. The confocal microscope was used to evaluate the expression of VE-cadherin in endothelial cells. RESULTS 25-OHC decreased endothelial cell integrity and increased the mRNA expression of IL-33, tissue factor, ICAM-1, MCP-1, VEGF, TNF-α as compared to unstimulated controls. Following the stimulation with rivaroxaban, HUVEC restored integrity disrupted by 25-OHC (p < .01). In HUVECs pre-stimulated with oxysterol, rivaroxaban decreased mRNA expression of IL-33, TNF-α, chemokines MCP-1, ICAM-1, VEGF and tissue factor (p < .01). Rivaroxaban 100 mg/ml+25-OHC increased the VE-cadherin expression in endothelium as compared to 25-OHC (p < .05). CONCLUSION Our finding suggests that rivaroxaban may restore the endothelial barrier and inhibit the inflammatory activation caused by oxysterol in vitro.
Collapse
Affiliation(s)
- Paulina Gorzelak-Pabis
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Marlena Broncel
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Katarzyna Wojdan
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| | - Adrian Gajewski
- Department of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Maciej Chalubinski
- Department of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Mateusz Gawrysiak
- Department of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Ewelina Wozniak
- Dept. of Internal Diseases and Clinical Pharmacology, The Laboratory of Tissue Immunopharmacology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
14
|
Li J, Zheng M, Shimoni O, Banks WA, Bush AI, Gamble JR, Shi B. Development of Novel Therapeutics Targeting the Blood-Brain Barrier: From Barrier to Carrier. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101090. [PMID: 34085418 PMCID: PMC8373165 DOI: 10.1002/advs.202101090] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/11/2021] [Indexed: 05/05/2023]
Abstract
The blood-brain barrier (BBB) is a highly specialized neurovascular unit, initially described as an intact barrier to prevent toxins, pathogens, and potentially harmful substances from entering the brain. An intact BBB is also critical for the maintenance of normal neuronal function. In cerebral vascular diseases and neurological disorders, the BBB can be disrupted, contributing to disease progression. While restoration of BBB integrity serves as a robust biomarker of better clinical outcomes, the restrictive nature of the intact BBB presents a major hurdle for delivery of therapeutics into the brain. Recent studies show that the BBB is actively engaged in crosstalk between neuronal and the circulatory systems, which defines another important role of the BBB: as an interfacing conduit that mediates communication between two sides of the BBB. This role has been subject to extensive investigation for brain-targeted drug delivery and shows promising results. The dual roles of the BBB make it a unique target for drug development. Here, recent developments and novel strategies to target the BBB for therapeutic purposes are reviewed, from both barrier and carrier perspectives.
Collapse
Affiliation(s)
- Jia Li
- School of PharmacyHenan UniversityKaifeng475001China
- Centre for Motor Neuron DiseaseDepartment of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNew South Wales2109Australia
| | - Meng Zheng
- Henan‐Macquarie University Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenan475004China
| | - Olga Shimoni
- Institute for Biomedical Materials and DevicesSchool of Mathematical and Physical SciencesFaculty of ScienceUniversity of Technology SydneySydneyNew South Wales2007Australia
| | - William A. Banks
- Geriatric Research Education and Clinical CenterVeterans Affairs Puget Sound Health Care System and Division of Gerontology and Geriatric MedicineDepartment of MedicineUniversity of Washington School of MedicineSeattleWA98108USA
| | - Ashley I. Bush
- Melbourne Dementia Research CenterThe Florey Institute for Neuroscience and Mental HealthThe University of MelbourneParkvilleVictoria3052Australia
| | - Jennifer R. Gamble
- Center for the EndotheliumVascular Biology ProgramCentenary InstituteThe University of SydneySydneyNew South Wales2042Australia
| | - Bingyang Shi
- School of PharmacyHenan UniversityKaifeng475001China
- Centre for Motor Neuron DiseaseDepartment of Biomedical SciencesFaculty of Medicine & Health SciencesMacquarie UniversitySydneyNew South Wales2109Australia
- Henan‐Macquarie University Joint Center for Biomedical InnovationSchool of Life SciencesHenan UniversityKaifengHenan475004China
| |
Collapse
|
15
|
Medina-Leyte DJ, Zepeda-García O, Domínguez-Pérez M, González-Garrido A, Villarreal-Molina T, Jacobo-Albavera L. Endothelial Dysfunction, Inflammation and Coronary Artery Disease: Potential Biomarkers and Promising Therapeutical Approaches. Int J Mol Sci 2021; 22:3850. [PMID: 33917744 PMCID: PMC8068178 DOI: 10.3390/ijms22083850] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Coronary artery disease (CAD) and its complications are the leading cause of death worldwide. Inflammatory activation and dysfunction of the endothelium are key events in the development and pathophysiology of atherosclerosis and are associated with an elevated risk of cardiovascular events. There is great interest to further understand the pathophysiologic mechanisms underlying endothelial dysfunction and atherosclerosis progression, and to identify novel biomarkers and therapeutic strategies to prevent endothelial dysfunction, atherosclerosis and to reduce the risk of developing CAD and its complications. The use of liquid biopsies and new molecular biology techniques have allowed the identification of a growing list of molecular and cellular markers of endothelial dysfunction, which have provided insight on the molecular basis of atherosclerosis and are potential biomarkers and therapeutic targets for the prevention and or treatment of atherosclerosis and CAD. This review describes recent information on normal vascular endothelium function, as well as traditional and novel potential biomarkers of endothelial dysfunction and inflammation, and pharmacological and non-pharmacological therapeutic strategies aimed to protect the endothelium or reverse endothelial damage, as a preventive treatment for CAD and related complications.
Collapse
Affiliation(s)
- Diana Jhoseline Medina-Leyte
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City 04510, Mexico
| | - Oscar Zepeda-García
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Coyoacán, Mexico City 04510, Mexico
| | - Mayra Domínguez-Pérez
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| | - Antonia González-Garrido
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| | - Teresa Villarreal-Molina
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| | - Leonor Jacobo-Albavera
- Genomics of Cardiovascular Diseases Laboratory, National Institute of Genomic Medicine (INMEGEN), Mexico City 14610, Mexico; (D.J.M.-L.); (O.Z.-G.); (M.D.-P.); (A.G.-G.); (T.V.-M.)
| |
Collapse
|
16
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
17
|
Kolářová H, Víteček J, Černá A, Černík M, Přibyl J, Skládal P, Potěšil D, Ihnatová I, Zdráhal Z, Hampl A, Klinke A, Kubala L. Myeloperoxidase mediated alteration of endothelial function is dependent on its cationic charge. Free Radic Biol Med 2021; 162:14-26. [PMID: 33271281 DOI: 10.1016/j.freeradbiomed.2020.11.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/02/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022]
Abstract
Endothelial cell (EC) glycocalyx (GLX) comprise a multicomponent layer of proteoglycans and glycoproteins. Alteration of its integrity contributes to chronic vascular inflammation and leads to the development of cardiovascular diseases. Myeloperoxidase (MPO), a highly abundant enzyme released by polymorphonuclear neutrophils, binds to the GLX and deleteriously affects vascular EC functions. The focus of this study was to elucidate the mechanisms of MPO-mediated alteration of GLX molecules, and to unravel subsequent changes in endothelial integrity and function. MPO binding to GLX of human ECs and subsequent internalization was mediated by cell surface heparan sulfate chains. Moreover, interaction of MPO, which is carrying a cationic charge, with anionic glycosaminoglycans (GAGs) resulted in reduction of their relative charge. By means of micro-viscometry and atomic force microscopy, we disclosed that MPO can crosslink GAG chains. MPO-dependent modulation of GLX structure was further supported by alteration of wheat germ agglutinin staining. Increased expression of ICAM-1 documented endothelial cell activation by both catalytically active and also inactive MPO. Furthermore, MPO increased vascular permeability connected with reorganization of intracellular junctions, however, this was dependent on MPO's catalytic activity. Novel proteins interacting with MPO during transcytosis were identified by proteomic analysis. Altogether, these findings provide evidence that MPO through interaction with GAGs modulates overall charge of the GLX, causing modification of its structure and thus affecting EC function. Importantly, our results also suggest a number of proteins interacting with MPO that possess a variety of cellular localizations and functions.
Collapse
Affiliation(s)
- Hana Kolářová
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Jan Víteček
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Anna Černá
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Marek Černík
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic
| | - Jan Přibyl
- Central European Institute for Technology, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - Petr Skládal
- Central European Institute for Technology, Masaryk University, Kamenice 5, Brno, Czech Republic
| | - David Potěšil
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Ivana Ihnatová
- Institute of Biostatistics and Analyses, Masaryk University, Kamenice 3, Brno, Czech Republic
| | - Zbyněk Zdráhal
- Central European Institute for Technology, Masaryk University, Kamenice 5, Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 62500, Brno, Czech Republic
| | - Aleš Hampl
- Faculty of Medicine, Department of Histology and Embryology, Masaryk University, Kamenice 3, 625 00, Brno, Czech Republic
| | - Anna Klinke
- Clinic of General and Interventional Cardiology/Angiology, Agnes Wittenborg Institute of Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Lukáš Kubala
- Department of Biophysics of Immune System, Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno, Czech Republic.
| |
Collapse
|
18
|
Daum R, Mrsic I, Hutterer J, Junginger A, Hinderer S, Meixner AJ, Gauglitz G, Chassé T, Schenke-Layland K. Fibronectin adsorption on oxygen plasma-treated polyurethane surfaces modulates endothelial cell response. J Mater Chem B 2021; 9:1647-1660. [DOI: 10.1039/d0tb02757j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Fibronectin coating increases implant biocompatibility by enhancing surface endothelialization via integrin-mediated binding.
Collapse
Affiliation(s)
- Ruben Daum
- NMI Natural and Medical Sciences
- Institute at the University of Tübingen
- 72770 Reutlingen
- Germany
- Department of Women's Health
| | - Ivana Mrsic
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Johanna Hutterer
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Achim Junginger
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Svenja Hinderer
- NMI Natural and Medical Sciences
- Institute at the University of Tübingen
- 72770 Reutlingen
- Germany
- Department of Women's Health
| | - Alfred J. Meixner
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
- Center for Light–Matter Interaction
| | - Günter Gauglitz
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
| | - Thomas Chassé
- Institute of Physical and Theoretical Chemistry
- Eberhard Karls University Tübingen
- 72076 Tübingen
- Germany
- Center for Light–Matter Interaction
| | - Katja Schenke-Layland
- NMI Natural and Medical Sciences
- Institute at the University of Tübingen
- 72770 Reutlingen
- Germany
- Department of Women's Health
| |
Collapse
|
19
|
Adil MS, Narayanan SP, Somanath PR. Cell-cell junctions: structure and regulation in physiology and pathology. Tissue Barriers 2020; 9:1848212. [PMID: 33300427 DOI: 10.1080/21688370.2020.1848212] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Epithelial and endothelial cell-cell contacts are established and maintained by several intercellular junctional complexes. These structurally and biochemically differentiated regions on the plasma membrane primarily include tight junctions (TJs), and anchoring junctions. While the adherens junctions (AJs) provide essential adhesive and mechanical properties, TJs hold the cells together and form a near leak-proof intercellular seal by the fusion of adjacent cell membranes. AJs and TJs play essential roles in vascular permeability. Considering their involvement in several key cellular functions such as barrier formation, proliferation, migration, survival, and differentiation, further research is warranted on the composition and signaling pathways regulating cell-cell junctions to develop novel therapeutics for diseases such as organ injuries. The current review article presents our current state of knowledge on various cell-cell junctions, their molecular composition, and mechanisms regulating their expression and function in endothelial and epithelial cells.
Collapse
Affiliation(s)
- Mir S Adil
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center , Augusta, GA, USA
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center , Augusta, GA, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, University of Georgia and Charlie Norwood VA Medical Center , Augusta, GA, USA
| |
Collapse
|
20
|
Potential role of the gut microbiota in neuromyelitis optica spectrum disorder: Implication for intervention. J Clin Neurosci 2020; 82:193-199. [PMID: 33257156 DOI: 10.1016/j.jocn.2020.11.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/22/2020] [Accepted: 11/01/2020] [Indexed: 12/19/2022]
Abstract
The gut microbiota plays an important role in the occurrence and development of neuroimmunological diseases. Neuromyelitis optica spectrum disorder (NMOSD) is an autoimmune disease of the central nervous system that is characterized by the peripheral production of the disease-specific serum autoantibody aquaporin-4 (AQP4)-IgG. Recently, accumulating evidence has provided insights into the associations of gut microbiota dysbiosis and intestinal mucosal barrier destruction with NMOSD, but the underlying pathogenesis remains unclear. Thus, a microbiota intervention might be a potential therapeutic strategy for NMOSD by regulating the gut microbiota, repairing the intestinal mucosal barrier, and modulating intestinal immunity and peripheral immunity.
Collapse
|
21
|
Retinal capillary degeneration and blood-retinal barrier disruption in murine models of Alzheimer's disease. Acta Neuropathol Commun 2020; 8:202. [PMID: 33228786 PMCID: PMC7686701 DOI: 10.1186/s40478-020-01076-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023] Open
Abstract
Extensive effort has been made studying retinal pathology in Alzheimer’s disease (AD) to improve early noninvasive diagnosis and treatment. Particularly relevant are vascular changes, which appear prominent in early brain pathogenesis and could predict cognitive decline. Recently, we identified platelet-derived growth factor receptor beta (PDGFRβ) deficiency and pericyte loss associated with vascular Aβ deposition in the neurosensory retina of mild cognitively impaired (MCI) and AD patients. However, the pathological mechanisms of retinal vascular changes and their possible relationships with vascular amyloidosis, pericyte loss, and blood-retinal barrier (BRB) integrity remain unknown. Here, we evaluated the retinas of transgenic APPSWE/PS1ΔE9 mouse models of AD (ADtg mice) and wild-type mice at different ages for capillary degeneration, PDGFRβ expression, vascular amyloidosis, permeability and inner BRB tight-junction molecules. Using a retinal vascular isolation technique followed by periodic acid-Schiff or immunofluorescent staining, we discovered significant retinal capillary degeneration in ADtg mice compared to age- and sex-matched wild-type mice (P < 0.0001). This small vessel degeneration reached significance in 8-month-old mice (P = 0.0035), with males more susceptible than females. Degeneration of retinal capillaries also progressively increased with age in healthy mice (P = 0.0145); however, the phenomenon was significantly worse during AD-like progression (P = 0.0001). A substantial vascular PDGFRβ deficiency (~ 50% reduction, P = 0.0017) along with prominent vascular Aβ deposition was further detected in the retina of ADtg mice, which inversely correlated with the extent of degenerated capillaries (Pearson’s r = − 0.8, P = 0.0016). Importantly, tight-junction alterations such as claudin-1 downregulation and increased BRB permeability, demonstrated in vivo by retinal fluorescein imaging and ex vivo following injection of FITC-dextran (2000 kD) and Texas Red-dextran (3 kD), were found in ADtg mice. Overall, the identification of age- and Alzheimer’s-dependent retinal capillary degeneration and compromised BRB integrity starting at early disease stages in ADtg mice could contribute to the development of novel targets for AD diagnosis and therapy.
Collapse
|
22
|
Wang L, Astone M, Alam SK, Zhu Z, Pei W, Frank DA, Burgess SM, Hoeppner LH. Suppressing STAT3 activity protects the endothelial barrier from VEGF-mediated vascular permeability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33140053 PMCID: PMC7605565 DOI: 10.1101/2020.10.27.358374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Vascular permeability triggered by inflammation or ischemia promotes edema, exacerbates disease progression, and impairs tissue recovery. Vascular endothelial growth factor (VEGF) is a potent inducer of vascular permeability. VEGF plays an integral role in regulating vascular barrier function physiologically and in pathologies, such as cancer, ischemic stroke, cardiovascular disease, retinal conditions, and COVID-19-associated pulmonary edema and sepsis, which often leads to acute lung injury, including acute respiratory distress syndrome. However, after initially stimulating permeability, VEGF subsequently mediates angiogenesis to repair damaged tissue. Consequently, understanding temporal molecular regulation of VEG-Finduced vascular permeability will facilitate developing therapeutics that achieve the delicate balance of inhibiting vascular permeability while preserving tissue repair. Here, we demonstrate that VEGF signals through signal transducer and activator of transcription 3 (STAT3) to promote vascular permeability. Specifically, we show that genetic STAT3 ablation reduces vascular permeability in STAT3-deficient endothelium of mice and VEGF-inducible zebrafish crossed with CRISPR/Cas9 generated genomic STAT3 knockout zebrafish. Importantly, STAT3 deficiency does not impair vascular development and function in vivo. We identify intercellular adhesion molecule 1 (ICAM-1) as a STAT3-dependent transcriptional regulator and show VEGF-dependent STAT3 activation is regulated by JAK2. Pyrimethamine, an FDA-approved antimicrobial agent that inhibits STAT3-dependent transcription, substantially reduces VEGF-induced vascular permeability in zebrafish, mouse, and human endothelium. Indeed, pharmacologically targeting STAT3 increases vascular barrier integrity using two additional compounds, atovaquone and C188-9. Collectively, our findings suggest that the VEGF, VEGFR-2, JAK2, and STAT3 signaling cascade regulates vascular barrier integrity, and inhibition of STAT3-dependent activity reduces VEGF-induced vascular permeability in vertebrate models.
Collapse
Affiliation(s)
- Li Wang
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Matteo Astone
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sk Kayum Alam
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Zhu Zhu
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luke H Hoeppner
- The Hormel Institute, University of Minnesota, Austin, MN, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
23
|
Hiepen C, Mendez PL, Knaus P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020; 9:E1965. [PMID: 32858894 PMCID: PMC7564048 DOI: 10.3390/cells9091965] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGFβ) superfamily of cytokines. While some ligand members are potent inducers of angiogenesis, others promote vascular homeostasis. However, the precise understanding of the molecular mechanisms underlying these functions is still a growing research field. In bone, the tissue in which BMPs were first discovered, crosstalk of TGFβ/BMP signaling with mechanobiology is well understood. Likewise, the endothelium represents a tissue that is constantly exposed to multiple mechanical triggers, such as wall shear stress, elicited by blood flow or strain, and tension from the surrounding cells and to the extracellular matrix. To integrate mechanical stimuli, the cytoskeleton plays a pivotal role in the transduction of these forces in endothelial cells. Importantly, mechanical forces integrate on several levels of the TGFβ/BMP pathway, such as receptors and SMADs, but also global cell-architecture and nuclear chromatin re-organization. Here, we summarize the current literature on crosstalk mechanisms between biochemical cues elicited by TGFβ/BMP growth factors and mechanical cues, as shear stress or matrix stiffness that collectively orchestrate endothelial function. We focus on the different subcellular compartments in which the forces are sensed and integrated into the TGFβ/BMP growth factor signaling.
Collapse
Affiliation(s)
| | | | - Petra Knaus
- Knaus-Lab/Signal Transduction, Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (C.H.); (P.-L.M.)
| |
Collapse
|
24
|
Kakogiannos N, Ferrari L, Giampietro C, Scalise AA, Maderna C, Ravà M, Taddei A, Lampugnani MG, Pisati F, Malinverno M, Martini E, Costa I, Lupia M, Cavallaro U, Beznoussenko GV, Mironov AA, Fernandes B, Rudini N, Dejana E, Giannotta M. JAM-A Acts via C/EBP-α to Promote Claudin-5 Expression and Enhance Endothelial Barrier Function. Circ Res 2020; 127:1056-1073. [PMID: 32673519 PMCID: PMC7508279 DOI: 10.1161/circresaha.120.316742] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
RATIONALE Intercellular tight junctions are crucial for correct regulation of the endothelial barrier. Their composition and integrity are affected in pathological contexts, such as inflammation and tumor growth. JAM-A (junctional adhesion molecule A) is a transmembrane component of tight junctions with a role in maintenance of endothelial barrier function, although how this is accomplished remains elusive. OBJECTIVE We aimed to understand the molecular mechanisms through which JAM-A expression regulates tight junction organization to control endothelial permeability, with potential implications under pathological conditions. METHODS AND RESULTS Genetic deletion of JAM-A in mice significantly increased vascular permeability. This was associated with significantly decreased expression of claudin-5 in the vasculature of various tissues, including brain and lung. We observed that C/EBP-α (CCAAT/enhancer-binding protein-α) can act as a transcription factor to trigger the expression of claudin-5 downstream of JAM-A, to thus enhance vascular barrier function. Accordingly, gain-of-function for C/EBP-α increased claudin-5 expression and decreased endothelial permeability, as measured by the passage of fluorescein isothiocyanate (FITC)-dextran through endothelial monolayers. Conversely, C/EBP-α loss-of-function showed the opposite effects of decreased claudin-5 levels and increased endothelial permeability. Mechanistically, JAM-A promoted C/EBP-α expression through suppression of β-catenin transcriptional activity, and also through activation of EPAC (exchange protein directly activated by cAMP). C/EBP-α then directly binds the promoter of claudin-5 to thereby promote its transcription. Finally, JAM-A-C/EBP-α-mediated regulation of claudin-5 was lost in blood vessels from tissue biopsies from patients with glioblastoma and ovarian cancer. CONCLUSIONS We describe here a novel role for the transcription factor C/EBP-α that is positively modulated by JAM-A, a component of tight junctions that acts through EPAC to up-regulate the expression of claudin-5, to thus decrease endothelial permeability. Overall, these data unravel a regulatory molecular pathway through which tight junctions limit vascular permeability. This will help in the identification of further therapeutic targets for diseases associated with endothelial barrier dysfunction. Graphic Abstract: An graphic abstract is available for this article.
Collapse
Affiliation(s)
- Nikolaos Kakogiannos
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Laura Ferrari
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Costanza Giampietro
- EMPA, Swiss Federal Laboratories for Material Science and Technologies, Experimental Continuum Mechanics, Dübendorf, Switzerland (C.G.)
| | - Anna Agata Scalise
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Claudio Maderna
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Micol Ravà
- Experimental Oncology (M.R.), European Institute of Oncology IRCSS, Milan
| | | | - Maria Grazia Lampugnani
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.).,Mario Negri Institute for Pharmacological Research, Milan (M.G.L.)
| | | | - Matteo Malinverno
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Emanuele Martini
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Ilaria Costa
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Michela Lupia
- Unit of Gynaecological Oncology Research (M.L., U.C.), European Institute of Oncology IRCSS, Milan
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research (M.L., U.C.), European Institute of Oncology IRCSS, Milan
| | - Galina V Beznoussenko
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Alexander A Mironov
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| | - Bethania Fernandes
- Pathology Unit, Humanitas Clinical and Research Centre, Rozzano, Milan (B.F., N.R.)
| | - Noemi Rudini
- Pathology Unit, Humanitas Clinical and Research Centre, Rozzano, Milan (B.F., N.R.)
| | - Elisabetta Dejana
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.).,Oncology and Haemato-Oncology, School of Medicine, University of Milan (E.D.).,Immunology, Genetics and Pathology, Uppsala University, Sweden (E.D.)
| | - Monica Giannotta
- From the FIRC Institute of Molecular Oncology, Milan, Italy (N.K., L.F., A.A.S., C.M., M.G.L., M.M., E.M., I.C., G.V.B., A.A.M., E.D., M.G.)
| |
Collapse
|
25
|
Koskimäki J, Polster SP, Li Y, Romanos S, Srinath A, Zhang D, Carrión-Penagos J, Lightle R, Moore T, Lyne SB, Stadnik A, Piedad K, Cao Y, Shenkar R, Dimov AV, Hobson N, Christoforidis GA, Carroll T, Girard R, Awad IA. Common transcriptome, plasma molecules, and imaging signatures in the aging brain and a Mendelian neurovascular disease, cerebral cavernous malformation. GeroScience 2020; 42:1351-1363. [PMID: 32556941 DOI: 10.1007/s11357-020-00201-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022] Open
Abstract
Brain senescence is associated with impaired endothelial barrier function, angiogenic and inflammatory activity, and propensity to brain hemorrhage. The same pathological changes occur in cerebral cavernous malformations (CCM), a genetic neurovascular anomaly. We hypothesized common transcriptomic and plasma cytokine signatures in the aging brain and CCM. We identified 320 genes [fold change ≥1.5; p < 0.05; false discovery rate (FDR) corrected] commonly dysregulated in the aging brain and CCM. Ontology and pathway analyses of the common differentially expressed genes were related to inflammation and extracellular matrix organization. Plasma levels of C-reactive protein and angiopoietin-2 were significantly greater in older compared to younger healthy non-CCM subjects and were also greater in CCM (Sporadic and Familial) subjects regardless of age (all: p < 0.05; FDR corrected). Plasma levels of vascular endothelial growth factor were significantly greater in older compared to younger subjects, in both healthy non-CCM and Sporadic-CCM groups (all: padj < 0.05). Plasma levels of vascular endothelial growth factor were also significantly greater in Familial-CCM cases with germ line mutations regardless of age (all: padj < 0.05) compared to both healthy non-CCM and Sporadic-CCM subjects. Brain white matter vascular permeability assessed by MRI followed the same pattern as vascular endothelial growth factor across all groups. In addition, quantitative susceptibility mapping of brain white matter, a measure of iron deposition, was increased in older compared to younger healthy non-CCM subjects. Genetic aberrations, plasma molecules, and imaging biomarkers in a well characterized Mendelian neurovascular disease may also be applicable in the aging brain. Graphical abstract.
Collapse
Affiliation(s)
- Janne Koskimäki
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Sean P Polster
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Yan Li
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA.,Center for Research Informatics, The University of Chicago, Chicago, IL, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Dongdong Zhang
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Julián Carrión-Penagos
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Rhonda Lightle
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Thomas Moore
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Seán B Lyne
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Agnieszka Stadnik
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Kristina Piedad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Ying Cao
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Robert Shenkar
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Alexey V Dimov
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Nick Hobson
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Gregory A Christoforidis
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Timothy Carroll
- Department of Diagnostic Radiology, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Romuald Girard
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA
| | - Issam A Awad
- Neurovascular Surgery Program, Section of Neurosurgery, The University of Chicago Medicine and Biological Sciences, Chicago, IL, USA.
| |
Collapse
|
26
|
EMT and EndMT: Emerging Roles in Age-Related Macular Degeneration. Int J Mol Sci 2020; 21:ijms21124271. [PMID: 32560057 PMCID: PMC7349630 DOI: 10.3390/ijms21124271] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 02/06/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) and endothelial–mesenchymal transition (EndMT) are physiological processes required for normal embryogenesis. However, these processes can be hijacked in pathological conditions to facilitate tissue fibrosis and cancer metastasis. In the eye, EMT and EndMT play key roles in the pathogenesis of subretinal fibrosis, the end-stage of age-related macular degeneration (AMD) that leads to profound and permanent vision loss. Predominant in subretinal fibrotic lesions are matrix-producing mesenchymal cells believed to originate from the retinal pigment epithelium (RPE) and/or choroidal endothelial cells (CECs) through EMT and EndMT, respectively. Recent evidence suggests that EMT of RPE may also be implicated during the early stages of AMD. Transforming growth factor-beta (TGFβ) is a key cytokine orchestrating both EMT and EndMT. Investigations in the molecular mechanisms underpinning EMT and EndMT in AMD have implicated a myriad of contributing factors including signaling pathways, extracellular matrix remodelling, oxidative stress, inflammation, autophagy, metabolism and mitochondrial dysfunction. Questions arise as to differences in the mesenchymal cells derived from these two processes and their distinct mechanistic contributions to the pathogenesis of AMD. Detailed discussion on the AMD microenvironment highlights the synergistic interactions between RPE and CECs that may augment the EMT and EndMT processes in vivo. Understanding the differential regulatory networks of EMT and EndMT and their contributions to both the dry and wet forms of AMD can aid the development of therapeutic strategies targeting both RPE and CECs to potentially reverse the aberrant cellular transdifferentiation processes, regenerate the retina and thus restore vision.
Collapse
|
27
|
Kwak BS, Jin SP, Kim SJ, Kim EJ, Chung JH, Sung JH. Microfluidic skin chip with vasculature for recapitulating the immune response of the skin tissue. Biotechnol Bioeng 2020; 117:1853-1863. [PMID: 32100875 DOI: 10.1002/bit.27320] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/16/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022]
Abstract
There is a considerable need for cell-based in vitro skin models for studying dermatological diseases and testing cosmetic products, but current in vitro skin models lack physiological relevance compared to human skin tissue. For example, many dermatological disorders involve complex immune responses, but current skin models are not capable of recapitulating the phenomena. Previously, we reported development of a microfluidic skin chip with a vessel structure and vascular endothelial cells. In this study, we cocultured dermal fibroblasts and keratinocytes with vascular endothelial cells, human umbilical vascular endothelial cells. We verified the formation of a vascular endothelium in the presence of the dermis and epidermis layers by examining the expression of tissue-specific markers. As the vascular endothelium plays a critical role in the migration of leukocytes to inflammation sites, we incorporated leukocytes in the circulating media and attempted to mimic the migration of neutrophils in response to external stimuli. Increased secretion of cytokines and migration of neutrophils was observed when the skin chip was exposed to ultraviolet irradiation, showing that the microfluidic skin chip may be useful for studying the immune response of the human tissue.
Collapse
Affiliation(s)
- Bong Shin Kwak
- Department of Chemical Engineering, Hongik University, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University Hospital, Republic of Korea.,Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University College of Medicine, Republic of Korea
| | - Su Jung Kim
- DYNEBIO INC., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eun Joo Kim
- DYNEBIO INC., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University Hospital, Republic of Korea.,Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University College of Medicine, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Republic of Korea
| |
Collapse
|
28
|
Internalization Characterization of Si Nanorod with Camouflaged Cell Membrane Proteins Reveals ATXN2 as a Negative Regulator. Cells 2019; 8:cells8080931. [PMID: 31430912 PMCID: PMC6721741 DOI: 10.3390/cells8080931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
The fabrication of shape-controlled nanocarriers is critical for efficient delivery of biomolecules across the cell membrane. Surface coating of the nanocarrier can improve internalization efficiency. Here, we developed a facile method of silicon nanorod fabrication leading to a controlled size and shape. We then systematically evaluated five surface modifications with membrane proteins from different cancer cell lines including MCF7, MD231, Hela, Panc-PDX, and Panc-1. We demonstrated that silicon nanorods coated with either a homolytic or heterolytic membrane protein coating have significantly improved internalization efficiency as compared with uncoated Si nanorods. To elucidate the molecular mechanism of the improved efficiency associated with a modified coating, we analyzed the coating membrane proteins derived from five cell lines with proteomics and identified 601 proteins shared by different cell sources. These proteins may function as cell-substrate adhesion molecules that contribute to the enhanced internalization. We also tested the internalization efficiency of nanorods with different coatings in each of the five cell lines to determine the influencing factors from target cells. We found that the internalization efficiency varied among different target cells, and the ranking of the average efficiency was as follows: Hela > Panc-PDX > MD231 > MCF7 > Panc-1. The bioinformatics analysis suggested that the low internalization efficiency in Panc-1 cells might be associated with the upregulation of ATXN2, which is a negative regulator of endocytosis. We further demonstrated that ATXN2 knockdown with specific siRNA significantly improved nanorod internalization efficiency in Panc-1 cells suggesting that ATXN2 can be a reference for efficiency prediction of nanoparticle delivery to tumor cells. Thus, we studied the effect of different cancer cell membrane proteins on nanorod uptake efficiencies. These results can improve nanorod internalization to cancer cells, including a fundamental understanding of the internalization efficiency of cancer cells.
Collapse
|
29
|
Yun JH, Han MH, Jeong HS, Lee DH, Cho CH. Angiopoietin 1 attenuates interleukin-6-induced endothelial cell permeability through SHP-1. Biochem Biophys Res Commun 2019; 518:286-293. [PMID: 31427082 DOI: 10.1016/j.bbrc.2019.08.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/08/2019] [Indexed: 02/06/2023]
Abstract
The regulation of endothelial cell (EC) permeability is critical for the physiological homeostasis of blood vessels and tissues. The elevation of pro-inflammatory cytokines is highly associated with lesions, such as the increased vascular permeability of diabetic retinas. We have previously reported that interleukin-6 (IL-6) increases EC permeability through the downregulation of tight junction protein expression. Angiopoietin 1 (Ang1) has an anti-permeability function, but the effect of Ang1 on vascular permeability induced by inflammatory cytokines is unclear. In the present study, we investigated the effect of Ang1 on IL-6-induced EC permeability and its underlying molecular mechanisms. We demonstrated that Ang1 inhibited the IL-6-induced increase in EC permeability by inhibiting the reductions in the levels of tight junction protein ZO-1 and occludin, which was related to the decrease in vascular endothelial growth factor (VEGF) secretion through the inhibition of STAT3 activation by Ang1. Mechanistically, Ang1 induced the dissociation of the tyrosine phosphatase SHP-1 from the Tie2 receptor and increased the binding of SHP-1 to JAK1, JAK2, and STAT3, which are IL-6 downstream signaling proteins. We conclude that SHP-1 plays an important role in the Ang1-induced inhibition of JAK/STAT3 signaling. These results provide evidence for a potential beneficial role of Ang1 in suppressing the vascular permeability induced by the pro-inflammatory cytokine IL-6 in diabetic retinopathy.
Collapse
Affiliation(s)
- Jang-Hyuk Yun
- Vascular Microenvironment Laboratory, Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Man Hyup Han
- Vascular Microenvironment Laboratory, Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Han-Seok Jeong
- Vascular Microenvironment Laboratory, Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Da-Hye Lee
- Vascular Microenvironment Laboratory, Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chung-Hyun Cho
- Vascular Microenvironment Laboratory, Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Ischemic/Hypoxic Disease Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Mundi S, Massaro M, Scoditti E, Carluccio MA, van Hinsbergh VWM, Iruela-Arispe ML, De Caterina R. Endothelial permeability, LDL deposition, and cardiovascular risk factors-a review. Cardiovasc Res 2019; 114:35-52. [PMID: 29228169 DOI: 10.1093/cvr/cvx226] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022] Open
Abstract
Early atherosclerosis features functional and structural changes in the endothelial barrier function that affect the traffic of molecules and solutes between the vessel lumen and the vascular wall. Such changes are mechanistically related to the development of atherosclerosis. Proatherogenic stimuli and cardiovascular risk factors, such as dyslipidaemias, diabetes, obesity, and smoking, all increase endothelial permeability sharing a common signalling denominator: an imbalance in the production/disposal of reactive oxygen species (ROS), broadly termed oxidative stress. Mostly as a consequence of the activation of enzymatic systems leading to ROS overproduction, proatherogenic factors lead to a pro-inflammatory status that translates in changes in gene expression and functional rearrangements, including changes in the transendothelial transport of molecules, leading to the deposition of low-density lipoproteins (LDL) and the subsequent infiltration of circulating leucocytes in the intima. In this review, we focus on such early changes in atherogenesis and on the concept that proatherogenic stimuli and risk factors for cardiovascular disease, by altering the endothelial barrier properties, co-ordinately trigger the accumulation of LDL in the intima and ultimately plaque formation.
Collapse
Affiliation(s)
- Santa Mundi
- Department of Biological and Environmental Science and Technology (DISTEBA), University of Salento, via Monteroni, 73100, Lecce, Italy
| | - Marika Massaro
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Maria Annunziata Carluccio
- National Research Council (CNR), Department of Biomedical sciences, Institute of Clinical Physiology, Via Monteroni, 73100, Lecce, Italy
| | - Victor W M van Hinsbergh
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat, NL-1081 BT, Amsterdam, The Netherlands
| | - Marial Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California, 610 Charles E Young Dr S, 90095, Los Angeles, USA; and
| | - Raffaele De Caterina
- Department of Neuroscience, Imaging and Clinical Science and Institute of Advanced Biomedical Technologies, University G. D'Annunzio, via dei Vestini, 66100 Chieti, Italy
| |
Collapse
|
31
|
Angiopoietin-1 Promotes the Integrity of Neovascularization in the Subcutaneous Matrigel of Type 1 Diabetic Rats. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2016972. [PMID: 30729120 PMCID: PMC6343146 DOI: 10.1155/2019/2016972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 12/30/2018] [Indexed: 12/21/2022]
Abstract
Objective This study aimed to investigate the effects of Ang-1 on neovascularization of diabetic organs by subcutaneous Matrigel angiogenesis model, established in type 1 diabetic rats. Methods Ang-1 adenoviral vector was constructed. The rat model was established by STZ and divided into four group. The Matrigel was inserted subcutaneously into the abdominal cavity of rats at 8 weeks, the treatment group was injected with Ang-1 adenovirus vector via tail vein, and the rats were sacrificed at 10 weeks. Neovascularization of Matrigel was observed with transmission electron microscopy. The marker of vascular endothelial cell and pericyte were detected by immunofluorescence. Immunohistochemical detection of the neovascular endothelial junction protein was performed. RT-PCR was used to determine protein expression of neovascular in Matrigel. Results Vascular cavity-like structure could be seen in subcutaneous Matrigel of diabetic rats, and the cavity was filled with a lot of red blood cells. Transmission electron microscopy showed that neovascular endothelial structure of the Matrigel was incomplete, while the Ang-1 treatment group had more vascular cavity-like structures, intact vascular endothelial structure, and reduced inflammatory cell infiltration in Matrigel. Additionally, the integrity of vascularization improved, and the marker of pericyte and the cell tight junctions protein was upregulated in Ang-1 treatment group. Conclusion Hyperglycemia could induce pathological angiogenesis in subcutaneous Matrigel of diabetic rats, and Ang-1 could upregulate the expression of intercellular junction protein in subcutaneous Matrigel of diabetic rats and promote the integrity of neovascularization in the subcutaneous Matrigel of diabetic rats.
Collapse
|
32
|
Santos C, Turiel S, Sousa Gomes P, Costa E, Santos-Silva A, Quadros P, Duarte J, Battistuzzo S, Fernandes MH. Vascular biosafety of commercial hydroxyapatite particles: discrepancy between blood compatibility assays and endothelial cell behavior. J Nanobiotechnology 2018; 16:27. [PMID: 29566760 PMCID: PMC5863823 DOI: 10.1186/s12951-018-0357-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/19/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Vascular homeostasis is ensured by a dynamic interplay involving the endothelium, the platelets and the coagulation system. Thus, the vascular safety of particulate materials must address this integrated system, an approach that has been largely neglected. This work analysed the effects of commercial hydroxyapatite (HA) particles in blood compatibility and in endothelial cell behavior, due to their clinical relevance and scarcity of data on their vascular biosafety. RESULTS Particles with similar chemical composition and distinct size and morphology were tested, i.e. rod-like, nano dimensions and low aspect ratio (HAp1) and needle-shape with wider size and aspect ratio (HAp2). HAp1 and HAp2, at 1 to 10 mg/mL, did not affect haemolysis, platelet adhesion, aggregation and activation, or the coagulation system (intrinsic and extrinsic pathways), although HAp2 exhibited a slight thrombogenic potential at 10 mg/mL. Notwithstanding, significantly lower levels presented dose-dependent toxicity on endothelial cells' behavior. HAp1 and HAp2 decreased cell viability at levels ≥ 250 and ≥ 50 μg/mL, respectively. At 10 and 50 μg/mL, HAp1 did not interfere with the F-actin cytoskeleton, apoptotic index, cell cycle progression, expression of vWF, VECad and CD31, and the ability to form a network of tubular-like structures. Comparatively, HAp2 caused dose-dependent toxic effects in these parameters in the same concentration range. CONCLUSION The most relevant observation is the great discrepancy of HA particles' levels that interfere with the routine blood compatibility assays and the endothelial cell behavior. Further, this difference was also found to be dependent on the particles' size, morphology and aspect ratio, emphasizing the need of a complementary biological characterization, taking into consideration the endothelial cells' functionality, to establish the vascular safety of particulate HA.
Collapse
Affiliation(s)
- Catarina Santos
- EST Setúbal, DEM, Instituto Politécnico de Setúbal, Campus IPS, 2914-508, Setúbal, Portugal.,CQE, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal
| | - Suzy Turiel
- Faculdade de Medicina Dentária, U. Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal
| | - Pedro Sousa Gomes
- Faculdade de Medicina Dentária, U. Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal.,REQUIMTE/LAQV - U. Porto, Porto, Portugal
| | - Elísio Costa
- UCIBIO/REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, U. Porto (FFUP), Porto, Portugal
| | - Alice Santos-Silva
- UCIBIO/REQUIMTE, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia, U. Porto (FFUP), Porto, Portugal
| | | | - José Duarte
- CIAFEL, Faculdade de Desporto, Universidade do Porto, Porto, Portugal
| | - Sílvia Battistuzzo
- Laboratório de Biologia Molecular e Genômica, Centro de Biociências, Universidade Federal do Rio Grande do Norte (UFRN), Campus Universitário s/n, Lagoa Nova, Natal, RN, 59072-970, Brazil
| | - Maria Helena Fernandes
- Faculdade de Medicina Dentária, U. Porto, Rua Dr. Manuel Pereira da Silva, 4200-393, Porto, Portugal. .,REQUIMTE/LAQV - U. Porto, Porto, Portugal.
| |
Collapse
|
33
|
Yuksel H, Turkeli A. Airway epithelial barrier dysfunction in the pathogenesis and prognosis of respiratory tract diseases in childhood and adulthood. Tissue Barriers 2017; 5:e1367458. [PMID: 28886270 DOI: 10.1080/21688370.2017.1367458] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The lungs are in direct contact with the environment through the tubular structure that constitutes the airway. Starting from the nasal orifice, the airway is exposed to foreign particles including infectious agents, allergens, and other substances that can damage the airways. Therefore, the airway must have a functional epithelial barrier both in the upper and lower airways to protect against these threats. As with the skin, it is likely that the pathogenesis of respiratory diseases is a consequence of epithelial barrier defects in these airways. The characteristics of this system, starting from the beginning of life and extending into maturing and aging, determine the prognosis of respiratory diseases. In this article, we discuss the pathogenesis, clinical phenotype, and prognosis of respiratory diseases from newborns to adulthood in the context of epithelial barrier function and dysfunction.
Collapse
Affiliation(s)
- Hasan Yuksel
- a Department of Pediatric Allergy and Pulmonology , Celal Bayar University Medical Faculty , Manisa , Turkey
| | - Ahmet Turkeli
- a Department of Pediatric Allergy and Pulmonology , Celal Bayar University Medical Faculty , Manisa , Turkey
| |
Collapse
|
34
|
Chang F, Flavahan S, Flavahan NA. Impaired activity of adherens junctions contributes to endothelial dilator dysfunction in ageing rat arteries. J Physiol 2017; 595:5143-5158. [PMID: 28561330 DOI: 10.1113/jp274189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Ageing-induced endothelial dysfunction contributes to organ dysfunction and progression of cardiovascular disease. VE-cadherin clustering at adherens junctions promotes protective endothelial functions, including endothelium-dependent dilatation. Ageing increased internalization and degradation of VE-cadherin, resulting in impaired activity of adherens junctions. Inhibition of VE-cadherin clustering at adherens junctions (function-blocking antibody; FBA) reduced endothelial dilatation in young arteries but did not affect the already impaired dilatation in old arteries. After junctional disruption with the FBA, dilatation was similar in young and old arteries. Src tyrosine kinase activity and tyrosine phosphorylation of VE-cadherin were increased in old arteries. Src inhibition increased VE-cadherin at adherens junctions and increased endothelial dilatation in old, but not young, arteries. Src inhibition did not increase dilatation in old arteries treated with the VE-cadherin FBA. Ageing impairs the activity of adherens junctions, which contributes to endothelial dilator dysfunction. Restoring the activity of adherens junctions could be of therapeutic benefit in vascular ageing. ABSTRACT Endothelial dilator dysfunction contributes to pathological vascular ageing. Experiments assessed whether altered activity of endothelial adherens junctions (AJs) might contribute to this dysfunction. Aortas and tail arteries were isolated from young (3-4 months) and old (22-24 months) F344 rats. VE-cadherin immunofluorescent staining at endothelial AJs and AJ width were reduced in old compared to young arteries. A 140 kDa VE-cadherin species was present on the cell surface and in TTX-insoluble fractions, consistent with junctional localization. Levels of the 140 kDa VE-cadherin were decreased, whereas levels of a TTX-soluble 115 kDa VE-cadherin species were increased in old compared to young arteries. Acetylcholine caused endothelium-dependent dilatation that was decreased in old compared to young arteries. Disruption of VE-cadherin clustering at AJs (function-blocking antibody, FBA) inhibited dilatation to acetylcholine in young, but not old, arteries. After the FBA, there was no longer any difference in dilatation between old and young arteries. Src activity and tyrosine phosphorylation of VE-cadherin were increased in old compared to young arteries. In old arteries, Src inhibition (saracatinib) increased: (i) 140 kDa VE-cadherin in the TTX-insoluble fraction, (ii) VE-cadherin intensity at AJs, (iii) AJ width, and (iv) acetylcholine dilatation. In old arteries treated with the FBA, saracatinib no longer increased acetylcholine dilatation. Saracatinib did not affect dilatation in young arteries. Therefore, ageing impairs AJ activity, which appears to reflect Src-induced phosphorylation, internalization and degradation of VE-cadherin. Moreover, impaired AJ activity can account for the endothelial dilator dysfunction in old arteries. Restoring endothelial AJ activity may be a novel therapeutic approach to vascular ageing.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
35
|
Tongxinluo Regulates Expression of Tight Junction Proteins and Alleviates Endothelial Cell Monolayer Hyperpermeability via ERK-1/2 Signaling Pathway in Oxidized Low-Density Lipoprotein-Induced Human Umbilical Vein Endothelial Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:4198486. [PMID: 28400842 PMCID: PMC5376437 DOI: 10.1155/2017/4198486] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 02/13/2017] [Accepted: 03/02/2017] [Indexed: 12/26/2022]
Abstract
Vascular hyperpermeability resulting from distortion of endothelial junctions is associated with a number of cardiovascular diseases. Endothelial tight junction regulates the paracellular permeability of macromolecules, a function of Human Umbilical Vein Endothelial Cells (HUVEC) monolayers that can be regulated by oxidized Low-density Lipoprotein (ox-LDL). However, the understanding of drug regulation of vascular hyperpermeability is so far limited. This study thus aimed to investigate the role of Tongxinluo (TXL) in the maintenance of the vascular endothelial paracellular permeability. Here, changes in permeability were determined by measuring the paracellular flux of FITC-dextran 40000 (FD40), while protein expression and intercellular distribution were examined by western blot and immunofluorescence assay, respectively. We found that TXL alleviated the ox-LDL-induced increase in flux of FD40 and then reduced the hyperpermeability. Moreover, ox-LDL-induced disruptions of ZO-1, occludin, and claudin1 were also restored. This is via the activation of ERK1/2 in the vascular endothelial cells. Our results provide insights into the molecular mechanism by which TXL alleviates ox-LDL-induced hyperpermeability and provide the basis for further investigations of TXL as regulators of vascular barrier function.
Collapse
|
36
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Gupta A, Bhatnagar S. Vasoregression: A Shared Vascular Pathology Underlying Macrovascular And Microvascular Pathologies? OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 19:733-53. [PMID: 26669709 DOI: 10.1089/omi.2015.0128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vasoregression is a common phenomenon underlying physiological vessel development as well as pathological microvascular diseases leading to peripheral neuropathy, nephropathy, and vascular oculopathies. In this review, we describe the hallmarks and pathways of vasoregression. We argue here that there is a parallel between characteristic features of vasoregression in the ocular microvessels and atherosclerosis in the larger vessels. Shared molecular pathways and molecular effectors in the two conditions are outlined, thus highlighting the possible systemic causes of local vascular diseases. Our review gives us a system-wide insight into factors leading to multiple synchronous vascular diseases. Because shared molecular pathways might usefully address the diagnostic and therapeutic needs of multiple common complex diseases, the literature analysis presented here is of broad interest to readership in integrative biology, rational drug development and systems medicine.
Collapse
Affiliation(s)
- Akanksha Gupta
- 1 Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology , Dwarka, New Delhi, India .,2 Department of Biotechnology, IMS Engineering College , Ghaziabad, India
| | - Sonika Bhatnagar
- 1 Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology , Dwarka, New Delhi, India
| |
Collapse
|
38
|
Yun JH, Park SW, Kim KJ, Bae JS, Lee EH, Paek SH, Kim SU, Ye S, Kim JH, Cho CH. Endothelial STAT3 Activation Increases Vascular Leakage Through Downregulating Tight Junction Proteins: Implications for Diabetic Retinopathy. J Cell Physiol 2016; 232:1123-1134. [PMID: 27580405 DOI: 10.1002/jcp.25575] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 08/29/2016] [Indexed: 02/06/2023]
Abstract
Vascular inflammation is characteristic feature of diabetic retinopathy. In diabetic retina, a variety of the pro-inflammatory cytokines are elevated and involved in endothelial dysfunction. STAT3 transcription factor has been implicated in mediating cytokine signaling during vascular inflammation. However, whether and how STAT3 is involved in the direct regulation of the endothelial permeability is currently undefined. Our studies revealed that IL-6-induced STAT3 activation increases retinal endothelial permeability and vascular leakage in retinas of mice through the reduced expression of the tight junction proteins ZO-1 and occludin. In a co-culture model with microglia and endothelial cells under a high glucose condition, the microglia-derived IL-6 induced STAT3 activation in the retinal endothelial cells, leading to increasing endothelial permeability. In addition, IL-6-induced STAT3 activation was independent of ROS generation in the retinal endothelial cells. Moreover, we demonstrated that STAT3 activation downregulates the ZO-1 and occludin levels and increases the endothelial permeability through the induction of VEGF production in retinal endothelial cells. These results suggest the potential importance of IL-6/STAT3 signaling in regulating endothelial permeability and provide a therapeutic target to prevent the pathology of diabetic retinopathy. J. Cell. Physiol. 232: 1123-1134, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jang-Hyuk Yun
- Department of Pharmacology and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Fight Against Angiogenesis-Related Blindness Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyung-Jin Kim
- Department of Pharmacology and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung U Kim
- Medical Research Institute, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Sangkyu Ye
- Department of Pharmacology and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeong-Hun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Fight Against Angiogenesis-Related Blindness Laboratory, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Ophthalmology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chung-Hyun Cho
- Department of Pharmacology and Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
39
|
|
40
|
Scotti L, Abramovich D, Pascuali N, Durand LH, Irusta G, de Zúñiga I, Tesone M, Parborell F. Inhibition of angiopoietin-1 (ANGPT1) affects vascular integrity in ovarian hyperstimulation syndrome (OHSS). Reprod Fertil Dev 2016; 28:690-9. [DOI: 10.1071/rd13356] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 09/11/2014] [Indexed: 01/29/2023] Open
Abstract
Ovarian hyperstimulation syndrome (OHSS) is a complication of ovarian stimulation with gonadotrophins following human chorionic gonadotrophin (hCG) administration. The relationship between hCG and OHSS is partly mediated via the production of angiogenic factors, such as vascular endothelial growth factor A (VEGFA) and angiopoietins (ANGPTs). Here, we investigated the effect of ANGPT1 inhibition on ovarian angiogenesis in follicular fluid (FF) from women at risk of OHSS, using the chorioallantoic membrane (CAM) of quail embryos as an experimental model. We also analysed cytoskeletal changes and endothelial junction protein expression induced by this FF in the presence or absence of an ANGPT1-neutralising antibody in endothelial cell cultures. The presence of this antibody restored the number of vascular branch points and integrin αvβ3 levels in the CAMs to control values. ANGPT1 inhibition in FF from OHSS patients also restored the levels of claudin-5, vascular endothelial cadherin and phosphorylated β-catenin and partially reversed actin redistribution in endothelial cells. Our findings suggest that ANGPT1 increases pathophysiological angiogenesis in patients at risk of OHSS by acting on tight and adherens junction proteins. Elucidating the mechanisms by which ANGPT1 regulates vascular development and cell–cell junctions in OHSS will contribute to identifying new therapeutic targets for the treatment of human diseases with aberrant vascular leakage.
Collapse
|
41
|
Cheng X, Wang X, Wan Y, Zhou Q, Zhu H, Wang Y. Myosin light chain kinase inhibitor ML7 improves vascular endothelial dysfunction via tight junction regulation in a rabbit model of atherosclerosis. Mol Med Rep 2015; 12:4109-4116. [PMID: 26096176 PMCID: PMC4526030 DOI: 10.3892/mmr.2015.3973] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 05/08/2015] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial dysfunction (VED) is an important factor in the initiation and development of atherosclerosis (AS). Previous studies have demonstrated that endothelial permeability is increased in diet‑induced AS. However, the precise underlying mechanisms remain poorly understood. The present study aimed to analyze whether the myosin light chain kinase (MLCK) inhibitor ML7 is able to improve VED and AS by regulating the expression of the tight junction (TJ) proteins zona occludens (ZO)‑1 and occludin via mechanisms involving MLCK and MLC phosphorylation in high‑fat diet‑fed rabbits. New Zealand white rabbits were randomly divided into three groups: Control group, AS group and ML7 group. The rabbits were fed a standard diet (control group), a high‑fat diet (AS group) or a high‑fat diet supplemented with 1 mg/kg/day ML7 (ML7 group). After 12 weeks, endothelium‑dependent relaxation and endothelium‑independent relaxation were measured using high-frequency ultrasound. Administration of a high‑fat diet significantly increased the levels of serum lipids and inflammatory markers in the rabbits in the AS group, as compared with those in the rabbits in the control group. Furthermore, a high‑fat diet contributed to the formation of a typical atherosclerotic plaque, as well as an increase in endothelial permeability and VED. These symptoms of AS were significantly improved following treatment with ML7, as demonstrated in the ML7 group. Hematoxylin & eosin and immunohistochemical staining indicated that ML7 was able to decrease the expression of MLCK and MLC phosphorylation in the arterial wall of rabbits fed a high‑fat diet. A similar change was observed for the TJ proteins ZO‑1 and occludin. In addition, western blot analysis demonstrated that ML7 increased the expression levels of occludin in the precipitate, but reduced its expression in the supernatant of lysed aortas. These results indicated that occludin, which is a dynamic protein at the TJ, is associated with remodeling from cell membrane to cytoplasm. The present study was the first, to the best of our knowledge, to indicate that ML7 may ameliorate VED and AS by regulating the TJ proteins ZO‑1 and occludin via mechanisms involving MLCK and MLC phosphorylation.
Collapse
Affiliation(s)
- Xiaowen Cheng
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xiaobian Wang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yufeng Wan
- Department of Otolaryngology, The Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Qing Zhou
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Huaqing Zhu
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Wang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
42
|
Tornavaca O, Chia M, Dufton N, Almagro LO, Conway DE, Randi AM, Schwartz MA, Matter K, Balda MS. ZO-1 controls endothelial adherens junctions, cell-cell tension, angiogenesis, and barrier formation. ACTA ACUST UNITED AC 2015; 208:821-38. [PMID: 25753039 PMCID: PMC4362456 DOI: 10.1083/jcb.201404140] [Citation(s) in RCA: 413] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intercellular junctions are crucial for mechanotransduction, but whether tight junctions contribute to the regulation of cell-cell tension and adherens junctions is unknown. Here, we demonstrate that the tight junction protein ZO-1 regulates tension acting on VE-cadherin-based adherens junctions, cell migration, and barrier formation of primary endothelial cells, as well as angiogenesis in vitro and in vivo. ZO-1 depletion led to tight junction disruption, redistribution of active myosin II from junctions to stress fibers, reduced tension on VE-cadherin and loss of junctional mechanotransducers such as vinculin and PAK2, and induced vinculin dissociation from the α-catenin-VE-cadherin complex. Claudin-5 depletion only mimicked ZO-1 effects on barrier formation, whereas the effects on mechanotransducers were rescued by inhibition of ROCK and phenocopied by JAM-A, JACOP, or p114RhoGEF down-regulation. ZO-1 was required for junctional recruitment of JACOP, which, in turn, recruited p114RhoGEF. ZO-1 is thus a central regulator of VE-cadherin-dependent endothelial junctions that orchestrates the spatial actomyosin organization, tuning cell-cell tension, migration, angiogenesis, and barrier formation.
Collapse
Affiliation(s)
- Olga Tornavaca
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Minghao Chia
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Neil Dufton
- National Heart and Lung Institute (NHLI) Vascular Sciences Unit, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, Imperial College London, London W12 0NN, England, UK
| | - Lourdes Osuna Almagro
- National Heart and Lung Institute (NHLI) Vascular Sciences Unit, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, Imperial College London, London W12 0NN, England, UK
| | - Daniel E Conway
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284
| | - Anna M Randi
- National Heart and Lung Institute (NHLI) Vascular Sciences Unit, Imperial Centre for Translational and Experimental Medicine (ICTEM), Hammersmith Hospital, Imperial College London, London W12 0NN, England, UK
| | - Martin A Schwartz
- Department of Medicine and Department of Cell Biology, Yale University, New Haven, CT 06520 Department of Medicine and Department of Cell Biology, Yale University, New Haven, CT 06520
| | - Karl Matter
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Maria S Balda
- Department of Cell Biology, UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| |
Collapse
|
43
|
Scotti L, Abramovich D, Pascuali N, Irusta G, Meresman G, Tesone M, Parborell F. Local VEGF inhibition prevents ovarian alterations associated with ovarian hyperstimulation syndrome. J Steroid Biochem Mol Biol 2014; 144 Pt B:392-401. [PMID: 25151950 DOI: 10.1016/j.jsbmb.2014.08.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/13/2014] [Accepted: 08/14/2014] [Indexed: 01/09/2023]
Abstract
The relationship between human chorionic gonadotropin and ovarian hyperstimulation syndrome (OHSS) is partially mediated by vascular endothelial growth factor A (VEGF). The aim of this study was to investigate the effects of VEGF inhibition on the development of corpora lutea (CL) and cystic structures, steroidogenesis, apoptosis, cell proliferation, endothelial cell area, VEGF receptors (KDR and Flt-1), claudin-5 and occludin levels in ovaries from an OHSS rat model. The VEGF inhibitor used (VEGF receptor-1 (FLT-1)/Fc chimera, TRAP) decreased the concentrations of progesterone and estradiol as well as the percentage of CL and cystic structures in OHSS rats, and increased apoptosis in CL. Endothelial cell area in CL and KDR expression and its phosphorylation were increased, whereas claudin-5 and occludin levels were decreased in the OHSS compared to the control TRAP reversed these parameters. Our findings indicate that VEGF inhibition prevents the early onset of OHSS and decreases its severity in rats.
Collapse
Affiliation(s)
- Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Griselda Irusta
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Gabriela Meresman
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Marta Tesone
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina; Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.
| |
Collapse
|
44
|
Skachkov I, Luan Y, van der Steen AFW, de Jong N, Kooiman K. Targeted microbubble mediated sonoporation of endothelial cells in vivo. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2014; 61:1661-1667. [PMID: 25265175 DOI: 10.1109/tuffc.2014.006440] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Ultrasound contrast agents as drug-delivery systems are an emerging field. Recently, we reported that targeted microbubbles are able to sonoporate endothelial cells in vitro. In this study, we investigated whether targeted microbubbles can also induce sonoporation of endothelial cells in vivo, thereby making it possible to combine molecular imaging and drug delivery. Live chicken embryos were chosen as the in vivo model. αvß3-targeted microbubbles attached to the vessel wall of the chicken embryo were insonified at 1 MHz at 150 kPa (1 × 10,000 cycles) and at 200 kPa (1 × 1000 cycles) peak negative acoustic pressure. Sonoporation was studied by intravital microscopy using the model drug propidium iodide (PI). Endothelial cell PI uptake was observed in 48% of microbubble-vessel-wall complexes at 150 kPa (n = 140) and in 33% at 200 kPa (n = 140). Efficiency of PI uptake depended on the local targeted microbubble concentration and increased up to 80% for clusters of 10 to 16 targeted microbubbles. Ultrasound or targeted microbubbles alone did not induce PI uptake. This intravital microscopy study reveals that sonoporation can be visualized and induced in vivo using targeted microbubbles.
Collapse
|
45
|
Newey SE, Tsaknakis G, Khoo CP, Athanassopoulos T, Camicia R, Zhang Y, Grabowska R, Harris AL, Roubelakis MG, Watt SM. The hematopoietic chemokine CXCL12 promotes integration of human endothelial colony forming cell-derived cells into immature vessel networks. Stem Cells Dev 2014; 23:2730-43. [PMID: 24940843 DOI: 10.1089/scd.2014.0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Proangiogenic factors, vascular endothelial growth factor (VEGF), and fibroblast growth factor-2 (FGF-2) prime endothelial cells to respond to "hematopoietic" chemokines and cytokines by inducing/upregulating expression of the respective chemokine/cytokine receptors. Coculture of human endothelial colony forming cell (ECFC)-derived cells with human stromal cells in the presence of VEGF and FGF-2 for 14 days resulted in upregulation of the "hematopoietic" chemokine CXCL12 and its CXCR4 receptor by day 3 of coculture. Chronic exposure to the CXCR4 antagonist AMD3100 in this vasculo/angiogenesis assay significantly reduced vascular tubule formation, an observation recapitulated by delayed AMD3100 addition. While AMD3100 did not affect ECFC-derived cell proliferation, it did demonstrate a dual action. First, over the later stages of the 14-day cocultures, AMD3100 delayed tubule organization into maturing vessel networks, resulting in enhanced endothelial cell retraction and loss of complexity as defined by live cell imaging. Second, at earlier stages of cocultures, we observed that AMD3100 significantly inhibited the integration of exogenous ECFC-derived cells into established, but immature, vascular networks. Comparative proteome profiler array analyses of ECFC-derived cells treated with AMD3100 identified changes in expression of potential candidate molecules involved in adhesion and/or migration. Blocking antibodies to CD31, but not CD146 or CD166, reduced the ECFC-derived cell integration into these extant vascular networks. Thus, CXCL12 plays a key role not only in endothelial cell sensing and guidance, but also in promoting the integration of ECFC-derived cells into developing vascular networks.
Collapse
Affiliation(s)
- Sarah E Newey
- 1 Stem Cell Research Laboratory , Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, and NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
E-cadherin expression in Barrett’s esophagus and esophageal carcinoma. Esophagus 2014. [DOI: 10.1007/s10388-014-0424-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
47
|
Bravi L, Dejana E, Lampugnani MG. VE-cadherin at a glance. Cell Tissue Res 2014; 355:515-22. [PMID: 24643676 DOI: 10.1007/s00441-014-1843-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/04/2014] [Indexed: 10/25/2022]
Abstract
Although being a monolayer the vascular endothelium controls fundamental vessel functions such as permeability, leukocyte extravasation and angiogenesis. The endothelial selective transmembrane constituent of adherens junctions, Vascular Endothelial- (VE-) cadherin plays a crucial role in the regulation of such activities. The signaling pathways controlled by VE-cadherin as well as the ones that regulate VE-cadherin activity start to be elucidated. This delineates a complex network of molecular and functional interactions that can be altered in pathologies.
Collapse
Affiliation(s)
- Luca Bravi
- FIRC Institute of Molecular Oncology (IFOM) Fondazione, Via Adamello 16, 20139, Milan, Italy
| | | | | |
Collapse
|
48
|
Charpentier MS, Conlon FL. Cellular and molecular mechanisms underlying blood vessel lumen formation. Bioessays 2013; 36:251-9. [PMID: 24323945 DOI: 10.1002/bies.201300133] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The establishment of a functional vascular system requires multiple complex steps throughout embryogenesis, from endothelial cell (EC) specification to vascular patterning into venous and arterial hierarchies. Following the initial assembly of ECs into a network of cord-like structures, vascular expansion and remodeling occur rapidly through morphogenetic events including vessel sprouting, fusion, and pruning. In addition, vascular morphogenesis encompasses the process of lumen formation, critical for the transformation of cords into perfusable vascular tubes. Studies in mouse, zebrafish, frog, and human endothelial cells have begun to outline the cellular and molecular requirements underlying lumen formation. Although the lumen can be generated through diverse mechanisms, the coordinated participation of multiple conserved molecules including transcription factors, small GTPases, and adhesion and polarity proteins remains a fundamental principle, leading us closer to a more thorough understanding of this complex event.
Collapse
Affiliation(s)
- Marta S Charpentier
- McAllister Heart Institute, Departments of Biology and Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
49
|
Ding L, Lu Z, Lu Q, Chen YH. The claudin family of proteins in human malignancy: a clinical perspective. Cancer Manag Res 2013; 5:367-375. [PMID: 24232410 PMCID: PMC3825674 DOI: 10.2147/cmar.s38294] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Tight junctions, or zonula occludens, are the most apical component of the junctional complex and provide one form of cell-cell adhesion in epithelial and endothelial cells. Nearly 90% of malignant tumors are derived from the epithelium. Loss of cell-cell adhesion is one of the steps in the progression of cancer to metastasis. At least three main tight junction family proteins have been discovered: occludin, claudin, and junctional adhesion molecule (JAM). Claudins are the most important structural and functional components of tight junction integral membrane proteins, with at least 24 members in mammals. They are crucial for the paracellular flux of ions and small molecules. Overexpression or downregulation of claudins is frequently observed in epithelial-derived cancers. However, molecular mechanisms by which claudins affect tumorigenesis remain largely unknown. As the pivotal proteins in epithelial cells, altered expression and distribution of different claudins have been reported in a wide variety of human malignancies, including pancreatic, colonic, lung, ovarian, thyroid, prostate, esophageal, and breast cancers. In this review, we will give the readers an overall picture of the changes in claudin expression observed in various cancers and their mechanisms of regulation. Downregulation of claudins contributes to epithelial transformation by increasing the paracellular permeability of nutrients and growth factors to cancerous cells. In the cases of upregulation of claudin expression, the barrier function of the cancerous epithelia changes, as they often display a disorganized arrangement of tight junction strands with increased permeability to paracellular markers. Finally, we will summarize the literature suggesting that claudins may become useful biomarkers for cancer detection and diagnosis as well as possible therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Lei Ding
- Department of Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, People’s Republic of China
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC, USA
| | - Zhe Lu
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC, USA
- Department of Basic Medicine, Hangzhou Normal University, Hangzhou, People’s Republic of China
| | - Qun Lu
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC, USA
- Leo W. Jenkins Cancer Center, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Yan-Hua Chen
- Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC, USA
- Leo W. Jenkins Cancer Center, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| |
Collapse
|
50
|
Aamelfot M, Weli SC, Dale OB, Koppang EO, Falk K. Characterisation of a monoclonal antibody detecting Atlantic salmon endothelial and red blood cells, and its association with the infectious salmon anaemia virus cell receptor. J Anat 2013; 222:547-57. [PMID: 23439106 PMCID: PMC3633344 DOI: 10.1111/joa.12033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2013] [Indexed: 01/29/2023] Open
Abstract
Endothelial cells (ECs) line the luminal surfaces of the cardiovascular system and play an important role in cardiovascular functions such as regulation of haemostasis and vasomotor tone. A number of fish and mammalian viruses target these cells in the course of their infection. Infectious salmon anaemia virus (ISAV) attacks ECs and red blood cells (RBCs) of farmed Atlantic salmon (Salmo salar L.), producing the severe disease of infectious salmon anaemia (ISA). The investigation of ISA has up to now been hampered by the lack of a functional marker for ECs in Atlantic salmon in situ. In this study, we report the characterisation and use of a novel monoclonal antibody (MAb) detecting Atlantic salmon ECs (e.g. vessel endothelium, endocardial cells and scavenger ECs) and RBCs. The antibody can be used with immunohistochemistry, IFAT and on Western blots. It appears that the epitope recognised by the antibody is associated with the ISAV cellular receptor. Besides being a tool to identify ECs in situ, it could be useful in further studies of the pathogenicity of ISA. Finally, the detection of an epitope shared by ECs and RBCs agrees with recent findings that these cells share a common origin, thus the MAb can potentially be used to study the ontogeny of these cells in Atlantic salmon.
Collapse
Affiliation(s)
| | | | - Ole B Dale
- Norwegian Veterinary InstituteOslo, Norway
| | | | - Knut Falk
- Norwegian Veterinary InstituteOslo, Norway
| |
Collapse
|