1
|
Peluzzo AM, St Paul A, Corbett CB, Kelemen SE, Fossati S, Liu X, Autieri MV. IL-19 Is a Novel Lymphangiocrine Factor Inducing Lymphangiogenesis and Lymphatic Junctional Regulation. Arterioscler Thromb Vasc Biol 2025. [PMID: 40371466 DOI: 10.1161/atvbaha.125.322669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND The lymphatic system functions by removing fluid, macromolecules, and immune cells to maintain tissue homeostasis. The structural organization of junctional protein complexes is vital to lymphatic function where initial lymphatics have permeable button junctions and collecting lymphatics have relatively impermeable zipper junctions. During inflammation, this junctional morphology appears to reverse, contributing to overall lymphatic malfunction. Little is known about the effects of immunomodulatory cytokines on lymphatic vessel formation and function during inflammation. The purpose of this study is to test the hypothesis that IL (interleukin)-19 promotes lymphangiogenesis and proper lymphatic function during inflammation. METHODS We used cultured human dermal lymphatic endothelial cells to determine IL-19 expression and its effects on lymphangiogenesis assays. Immunocytochemistry and electric cell-substrate impedance sensing determined effects on junctional morphology as it relates to permeability in vitro. RNA sequencing determined the effects of IL-19 on gene expression. Il19-/-Ldlr-/- double knockout mice were used to determine IL-19 effects on lymphatic function and lymphatic vessel visualization in vivo. RESULTS Endogenous IL-19 expression is induced by exogenous IL-19 and VEGF (vascular endothelial growth factor) C stimulation. IL-19 is lymphangiogenic, increasing human dermal lymphatic endothelial cell migration, network formation, and proliferation. IL-19 induces expression of transcription factors and permeability-associated genes. IL-19 induces rapid VE-cadherin (vascular endothelial cadherin) phosphorylation, increases permeability of human dermal lymphatic endothelial cell monolayers, and mitigates oxidized low-density lipoprotein-associated decrease in human dermal lymphatic endothelial cell permeability. In vivo, Il19-/-Ldlr-/- double knockout mice on a high-fat diet have impaired lymphatic drainage, decreased lymphatic branch points, and increased percentage of zippered junctions compared with control mice. CONCLUSIONS Taken together, these data show that IL-19 has potent effects on lymphatic vessel formation and function in vitro and that IL-19 regulates lymphatic drainage in vivo. IL-19 may represent an immunomodulatory cytokine with therapeutic potential for improving impaired lymphatic function consequent to inflammation.
Collapse
Affiliation(s)
- Amanda M Peluzzo
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Amanda St Paul
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Cali B Corbett
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Sheri E Kelemen
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine at Temple University, Philadelphia, PA (S.F.)
| | - Xiaolei Liu
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| | - Michael V Autieri
- Lemole Center for Integrated Lymphatics and Vascular Research (A.M.P., A.S.P., C.B.C., S.E.K., X.L., M.V.A.)
| |
Collapse
|
2
|
Vu HH, Moellmer SA, McCarty OJ, Puy C. New mechanisms and therapeutic approaches to regulate vascular permeability in systemic inflammation. Curr Opin Hematol 2025; 32:130-137. [PMID: 40063579 PMCID: PMC11949701 DOI: 10.1097/moh.0000000000000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
PURPOSE OF REVIEW This review summarizes mechanisms that regulate endothelial vascular permeability in health and disease. In systemic inflammation, the endothelial barrier integrity is disrupted, which exacerbates vascular permeability, leading to organ failure and death. Herein we provide an overview of emerging therapeutic targets to reverse barrier dysfunction and preserve vascular permeability in inflammatory diseases like sepsis. RECENT FINDINGS Endothelial barrier function is regulated in part by the endothelial cell-specific protein, Roundabout 4 (ROBO4), and vascular endothelial (VE)-cadherin, a critical adherens junction protein, which act in concert to suppresses vascular permeability by stabilizing endothelial cell-cell interactions. We recently discovered a pathway by which activation of coagulation factor XI (FXI) enhances the cleavage of VE-cadherin by the metalloproteinase ADAM10, contributing to sepsis-related endothelial damage and loss of barrier function. Targeting FXI improved survival and reduced sVE-cadherin levels in a baboon model of sepsis while enhancing Robo4 expression decreased mortality in LPS-treated mice. SUMMARY Endothelial cell barrier dysfunction is a hallmark of excessive immune responses characteristic of systemic inflammatory diseases such as sepsis. Advances in understanding the molecular mechanisms regulating vascular permeability, for instance the newly discovered roles of FXI or ROBO4, may help identify novel therapeutic targets for mitigating vascular hyperpermeability in septic patients.
Collapse
Affiliation(s)
- Helen H. Vu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Portland, OR, USA
| | - Cristina Puy
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
3
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
4
|
Hamad I, Sepic S, Moztarzadeh S, García-Ponce A, Waschke J, Radeva MY. Plakoglobin does not participate in endothelial barrier stabilization mediated by cAMP. Sci Rep 2025; 15:9043. [PMID: 40091082 PMCID: PMC11911453 DOI: 10.1038/s41598-025-93756-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Critical for maintenance of endothelial barrier is the remodeling of the actin cytoskeleton and the precise control of junctional integrity. Plakoglobin (PG) is a structural and signaling protein involved in vascular permeability regulation together with key signaling molecules such as cAMP, Rho GTPases and actin-binding proteins. Here, we investigated the role of PG in cAMP-mediated endothelial barrier stabilization by establishing myocardial endothelial cells derived from wild type (WT) and PG knock-out (PG-KO) mice. Under basal conditions, TEER measurements showed increased barrier function of PG-KO, an effect associated with enhanced protein levels and junctional VE-cadherin and β-catenin accumulation. PG-KO cells also displayed more PECAM-1 and VE-PTP-phosphatase and less phosphorylated VE-cadherin, typically linked with modulation of junctional integrity. PG ablation neither changed the composition of VE-cadherin/β-catenin complex nor activities of Rac1 and RhoA but decreased the basal intracellular cAMP concentration. Remarkably, cAMP augmentation led to enhanced Rac1 activity and TEER in both cell lines, but the effect was less prominent in PG-KO. The tighter barrier in WT was paralleled with more VE-cadherin, β-catenin and cortactin, an actin-binding protein, towards junctions. Surprisingly, PG phosphorylation at Ser665 was not required for cAMP-mediated endothelial barrier integrity, which is different to cardiomyocyte and keratinocyte cell adhesion.
Collapse
Affiliation(s)
- Ibrahim Hamad
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sara Sepic
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Alexander García-Ponce
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University (LMU), Pettenkoferstraße 11, 80336, Munich, Germany.
| |
Collapse
|
5
|
Razavi MS, Munn LL, Padera TP. Mechanics of Lymphatic Pumping and Lymphatic Function. Cold Spring Harb Perspect Med 2025; 15:a041171. [PMID: 38692743 PMCID: PMC11875091 DOI: 10.1101/cshperspect.a041171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
The lymphatic system plays a crucial role in maintaining tissue fluid balance, immune surveillance, and the transport of lipids and macromolecules. Lymph is absorbed by initial lymphatics and then driven through lymph nodes and to the blood circulation by the contraction of collecting lymphatic vessels. Intraluminal valves in collecting lymphatic vessels ensure the unidirectional flow of lymph centrally. The lymphatic muscle cells that invest in collecting lymphatic vessels impart energy to propel lymph against hydrostatic pressure gradients and gravity. A variety of mechanical and biochemical stimuli modulate the contractile activity of lymphatic vessels. This review focuses on the recent advances in our understanding of the mechanisms involved in regulating and collecting lymphatic vessel pumping in normal tissues and the association between lymphatic pumping, infection, inflammatory disease states, and lymphedema.
Collapse
Affiliation(s)
- Mohammad S Razavi
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Lance L Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Timothy P Padera
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
6
|
Sun M, Angelillo J, Hugues S. Lymphatic transport in anti-tumor immunity and metastasis. J Exp Med 2025; 222:e20231954. [PMID: 39969537 PMCID: PMC11837853 DOI: 10.1084/jem.20231954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Although lymphatic vessels (LVs) are present in many tumors, their importance in cancer has long been underestimated. In contrast to the well-studied tumor-associated blood vessels, LVs were previously considered to function as passive conduits for tumor metastasis. However, emerging evidence over the last two decades has shed light on their critical role in locally shaping the tumor microenvironment (TME). Here we review the involvement of LVs in tumor progression, metastasis, and modulation of anti-tumor immune response.
Collapse
Affiliation(s)
- Mengzhu Sun
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Julien Angelillo
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
7
|
Kizhatil K, Clark GM, Sunderland DK, Bhandari A, Horbal LJ, Balasubramanian R, John SWM. FYN regulates aqueous humor outflow and IOP through the phosphorylation of VE-CADHERIN. Nat Commun 2025; 16:51. [PMID: 39746990 PMCID: PMC11696269 DOI: 10.1038/s41467-024-55232-8] [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: 10/18/2023] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Schlemm's canal endothelial cells (SECs) serve as the final barrier to aqueous humor (AQH) drainage from the eye. SECs adjust permeability to AQH outflow to modulate intraocular pressure (IOP). The broad identification of IOP-related genes implicates SECs in glaucoma. However, the molecular mechanisms by which SECs sense and respond to pressure changes to regulate fluid permeability and IOP remain largely undefined. We hypothesize that mechano-responsive phosphorylation of the cell adhesion molecule VE-CADHERIN (CDH5) in SECs, by FYN and possibly other SRC family kinases, regulates adherens junction (AJ) permeability to AQH in response to IOP. On experimentally raising IOP in mouse eyes, AJ permeability, CDH5 phosphorylation, and FYN activation at the AJ all increase. FYN null mutant mice display disrupted IOP regulation and reduced AQH outflow. These findings demonstrate an important role of mechanotransducive signaling within SECs in maintaining IOP homeostasis and implicate FYN as a potential target for developing IOP-lowering treatments.
Collapse
Affiliation(s)
- Krishnakumar Kizhatil
- Department of Ophthalmology and Visual Sciences, The Ohio State Medical Center, Columbus, Ohio, 43210, USA.
| | | | | | - Aakriti Bhandari
- The Jackson Laboratory, Bar Harbor, Maine, 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | - Logan J Horbal
- The Jackson Laboratory, Bar Harbor, Maine, 04609, USA
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Simon W M John
- Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.
- Zuckerman Mind Brain Behavior Institute, New York, NY, 10027, USA.
| |
Collapse
|
8
|
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
|
9
|
Serafin DS, Harris NR, Bálint L, Douglas ES, Caron KM. Proximity interactome of lymphatic VE-cadherin reveals mechanisms of junctional remodeling and reelin secretion. Nat Commun 2024; 15:7734. [PMID: 39232006 PMCID: PMC11374903 DOI: 10.1038/s41467-024-51918-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
The adhesion receptor vascular endothelial (VE)-cadherin transduces an array of signals that modulate crucial lymphatic cell behaviors including permeability and cytoskeletal remodeling. Consequently, VE-cadherin must interact with a multitude of intracellular proteins to exert these functions. Yet, the full protein interactome of VE-cadherin in endothelial cells remains a mystery. Here, we use proximity proteomics to illuminate how the VE-cadherin interactome changes during junctional reorganization from dis-continuous to continuous junctions, triggered by the lymphangiogenic factor adrenomedullin. These analyses identified interactors that reveal roles for ADP ribosylation factor 6 (ARF6) and the exocyst complex in VE-cadherin trafficking and recycling. We also identify a requisite role for VE-cadherin in the in vitro and in vivo control of secretion of reelin-a lymphangiocrine glycoprotein with recently appreciated roles in governing heart development and injury repair. This VE-cadherin protein interactome shines light on mechanisms that control adherens junction remodeling and secretion from lymphatic endothelial cells.
Collapse
Affiliation(s)
- D Stephen Serafin
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, 27599, NC, USA
| | - Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, 27599, NC, USA
| | - László Bálint
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, 27599, NC, USA
| | - Elizabeth S Douglas
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, 27599, NC, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, 27599, NC, USA.
| |
Collapse
|
10
|
Wakasugi R, Suzuki K, Kaneko-Kawano T. Molecular Mechanisms Regulating Vascular Endothelial Permeability. Int J Mol Sci 2024; 25:6415. [PMID: 38928121 PMCID: PMC11203514 DOI: 10.3390/ijms25126415] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Vascular endothelial cells form a monolayer in the vascular lumen and act as a selective barrier to control the permeability between blood and tissues. To maintain homeostasis, the endothelial barrier function must be strictly integrated. During acute inflammation, vascular permeability temporarily increases, allowing intravascular fluid, cells, and other components to permeate tissues. Moreover, it has been suggested that the dysregulation of endothelial cell permeability may cause several diseases, including edema, cancer, and atherosclerosis. Here, we reviewed the molecular mechanisms by which endothelial cells regulate the barrier function and physiological permeability.
Collapse
Affiliation(s)
| | | | - Takako Kaneko-Kawano
- Graduate School of Pharmacy, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu 525-8577, Shiga, Japan; (R.W.); (K.S.)
| |
Collapse
|
11
|
Salehi A, Sprejz S, Ruehl H, Olayioye M, Cattaneo G. An imprint-based approach to replicate nano- to microscale roughness on gelatin hydrogel scaffolds: surface characterization and effect on endothelialization. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1214-1235. [PMID: 38431849 DOI: 10.1080/09205063.2024.2322771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Biologization of biomaterials with endothelial cells (ECs) is an important step in vascular tissue engineering, aiming at improving hemocompatibility and diminishing the thrombo-inflammatory response of implants. Since subcellular topography in the scale of nano to micrometers can influence cellular adhesion, proliferation, and differentiation, we here investigate the effect of surface roughness on the endothelialization of gelatin hydrogel scaffolds. Considering the micron and sub-micron features of the different native tissues underlying the endothelium in the body, we carried out a biomimetic approach to replicate the surface roughness of tissues and analyzed how this impacted the adhesion and proliferation of human umbilical endothelial cells (HUVECs). Using an imprinting technique, nano and micro-roughness ranging from Sa= 402 nm to Sa= 8 μm were replicated on the surface of gelatin hydrogels. Fluorescent imaging of HUVECs on consecutive days after seeding revealed that microscale topographies negatively affect cell spreading and proliferation. By contrast, nanoscale roughnesses of Sa= 402 and Sa= 538 nm promoted endothelialization as evidenced by the formation of confluent cell monolayers with prominent VE-cadherin surface expression. Collectively, we present an affordable and flexible imprinting method to replicate surface characteristics of tissues on hydrogels and demonstrate how nanoscale roughness positively supports their endothelialization.
Collapse
Affiliation(s)
- Ali Salehi
- Institute of Biomedical Engineering, University of Stuttgart, Stuttgart, Germany
| | - Stefanie Sprejz
- Institute of Biomedical Engineering, University of Stuttgart, Stuttgart, Germany
| | - Holger Ruehl
- Institute for Micro Integration, University of Stuttgart, Stuttgart, Germany
| | - Monilola Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Giorgio Cattaneo
- Institute of Biomedical Engineering, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
12
|
Lorente-Herraiz L, Cuesta AM, Granado J, Recio-Poveda L, Botella LM, Albiñana V. Molecular and Cellular Characterization of Primary Endothelial Cells from a Familial Cavernomatosis Patient. Int J Mol Sci 2024; 25:3952. [PMID: 38612762 PMCID: PMC11012380 DOI: 10.3390/ijms25073952] [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: 02/27/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Cerebral cavernous malformation (CCM) or familial cavernomatosis is a rare, autosomal dominant, inherited disease characterized by the presence of vascular malformations consisting of blood vessels with an abnormal structure in the form of clusters. Based on the altered gene (CCM1/Krit1, CCM2, CCM3) and its origin (spontaneous or familial), different types of this disease can be found. In this work we have isolated and cultivated primary endothelial cells (ECs) from peripheral blood of a type 1 CCM patient. Differential functional and gene expression profiles of these cells were analyzed and compared to primary ECs from a healthy donor. The mutation of the familial index case consisted of a heterozygous point mutation in the position +1 splicing consensus between exons 15 and 16, causing failure in RNA processing and in the final protein. Furthermore, gene expression analysis by quantitative PCR revealed a decreased expression of genes involved in intercellular junction formation, angiogenesis, and vascular homeostasis. Cell biology analysis showed that CCM1 ECs were impaired in angiogenesis and cell migration. Taken together, the results obtained suggest that the alterations found in CCM1 ECs are already present in the heterozygous condition, suffering from vascular impairment and somewhat predisposed to vascular damage.
Collapse
Affiliation(s)
- Laura Lorente-Herraiz
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| | - Angel M. Cuesta
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Jaime Granado
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
| | - Lucía Recio-Poveda
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| | - Luisa-María Botella
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| | - Virginia Albiñana
- Departamento de Biomedicina Molecular, Centro de Investigaciones Biológicas Margaritas Salas, 28040 Madrid, Spain; (L.L.-H.); (J.G.); (L.R.-P.); (V.A.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain;
| |
Collapse
|
13
|
Shekhtman O, Sioutas GS, Piavchenko G, Bhalla S, Cooke DL, Winkler E, Burkhardt JK, Srinivasan VM. Endovascular biopsy in neurointerventional surgery: A systematic review. Interv Neuroradiol 2024:15910199241240508. [PMID: 38515364 PMCID: PMC11569772 DOI: 10.1177/15910199241240508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Endothelial cells (ECs) continuously line the cerebrovasculature. Molecular aberrations in the ECs are hallmarks and contributory factors to the development of cerebrovascular diseases, including intracranial aneurysms and arteriovenous malformations (AVMs). Endovascular biopsy has been introduced as a method to harvest ECs and obtain relevant biologic information. We aimed to summarize the literature on endovascular biopsy in neurointerventional surgery. METHODS We conducted a comprehensive literature search in multiple databases, identifying eligible studies focusing on neurosurgical applications of endovascular biopsy. The systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. The relevant information was collected, including study characteristics, biopsy techniques, and key findings. RESULTS Nine studies met the inclusion criteria and were included. The studies involved the collection of ECs using various endovascular devices including coils, guide wires, different stents, and forceps. Endothelial-enrichment techniques, such fluorescence-activated cell sorting (FACS), collected ECs and facilitated downstream applications of bulk or single-cell RNA sequencing (scRNAseq). The studies provided insights into gene expression profiles and identified potential biomarkers associated with intracranial aneurysms. However, challenges were observed in obtaining an adequate number of ECs and identifying consistent biomarkers. CONCLUSION Endovascular biopsy of endothelial cells (ECs) in cerebrovascular pathologies shows promise for gene expression profiling. However, many studies have been limited in sample size and underpowered to identify "signature genes" for aneurysm growth or rupture. Advancements in minimally invasive biopsy methods have potential to facilitate applications of precision medicine in the treatment of cerebrovascular disorders.
Collapse
Affiliation(s)
- Oleg Shekhtman
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Georgios S Sioutas
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Gennadii Piavchenko
- Department of Human Anatomy and Histology, Sechenov University, Moscow, Russia
| | - Shubhang Bhalla
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Daniel L Cooke
- Department of Interventional Radiology, University of California San Francisco, San Francisco, CA, USA
| | - Ethan Winkler
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jan-Karl Burkhardt
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Visish M Srinivasan
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Peters MM, Brister JK, Tang EM, Zhang FW, Lucian VM, Trackey PD, Bone Z, Zimmerman JF, Jin Q, Burpo FJ, Parker KK. Self-organizing behaviors of cardiovascular cells on synthetic nanofiber scaffolds. APL Bioeng 2023; 7:046114. [PMID: 38046543 PMCID: PMC10693444 DOI: 10.1063/5.0172423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023] Open
Abstract
In tissues and organs, the extracellular matrix (ECM) helps maintain inter- and intracellular architectures that sustain the structure-function relationships defining physiological homeostasis. Combining fiber scaffolds and cells to form engineered tissues is a means of replicating these relationships. Engineered tissues' fiber scaffolds are designed to mimic the topology and chemical composition of the ECM network. Here, we asked how cells found in the heart compare in their propensity to align their cytoskeleton and self-organize in response to topological cues in fibrous scaffolds. We studied cardiomyocytes, valvular interstitial cells, and vascular endothelial cells as they adapted their inter- and intracellular architectures to the extracellular space. We used focused rotary jet spinning to manufacture aligned fibrous scaffolds to mimic the length scale and three-dimensional (3D) nature of the native ECM in the muscular, valvular, and vascular tissues of the heart. The representative cardiovascular cell types were seeded onto fiber scaffolds and infiltrated the fibrous network. We measured different cell types' propensity for cytoskeletal alignment in response to fiber scaffolds with differing levels of anisotropy. The results indicated that valvular interstitial cells on moderately anisotropic substrates have a higher propensity for cytoskeletal alignment than cardiomyocytes and vascular endothelial cells. However, all cell types displayed similar levels of alignment on more extreme (isotropic and highly anisotropic) fiber scaffold organizations. These data suggest that in the hierarchy of signals that dictate the spatiotemporal organization of a tissue, geometric cues within the ECM and cellular networks may homogenize behaviors across cell populations and demographics.
Collapse
Affiliation(s)
- Michael M. Peters
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Jackson K. Brister
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Edward M. Tang
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Felita W. Zhang
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Veronica M. Lucian
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Paul D. Trackey
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - Zachary Bone
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | - John F. Zimmerman
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - Qianru Jin
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts 02134, USA
| | - F. John Burpo
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, USA
| | | |
Collapse
|
15
|
Stierschneider A, Wiesner C. Shedding light on the molecular and regulatory mechanisms of TLR4 signaling in endothelial cells under physiological and inflamed conditions. Front Immunol 2023; 14:1264889. [PMID: 38077393 PMCID: PMC10704247 DOI: 10.3389/fimmu.2023.1264889] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/08/2023] [Indexed: 12/18/2023] Open
Abstract
Toll-like receptor 4 (TLR4) are part of the innate immune system. They are capable of recognizing pathogen-associated molecular patterns (PAMPS) of microbes, and damage-associated molecular patterns (DAMPs) of damaged tissues. Activation of TLR4 initiates downstream signaling pathways that trigger the secretion of cytokines, type I interferons, and other pro-inflammatory mediators that are necessary for an immediate immune response. However, the systemic release of pro-inflammatory proteins is a powerful driver of acute and chronic inflammatory responses. Over the past decades, immense progress has been made in clarifying the molecular and regulatory mechanisms of TLR4 signaling in inflammation. However, the most common strategies used to study TLR4 signaling rely on genetic manipulation of the TLR4 or the treatment with agonists such as lipopolysaccharide (LPS) derived from the outer membrane of Gram-negative bacteria, which are often associated with the generation of irreversible phenotypes in the target cells or unintended cytotoxicity and signaling crosstalk due to off-target or pleiotropic effects. Here, optogenetics offers an alternative strategy to control and monitor cellular signaling in an unprecedented spatiotemporally precise, dose-dependent, and non-invasive manner. This review provides an overview of the structure, function and signaling pathways of the TLR4 and its fundamental role in endothelial cells under physiological and inflammatory conditions, as well as the advances in TLR4 modulation strategies.
Collapse
Affiliation(s)
| | - Christoph Wiesner
- Department Science & Technology, Institute Biotechnology, IMC Krems University of Applied Sciences, Krems, Austria
| |
Collapse
|
16
|
Wang B, Yang Y, Wang Y, Yang Y, Li Y, Hu C, Xue C. Matrix stiffness regulates neovascular homeostasis through autophagy in nude mice. J Cell Physiol 2023; 238:2135-2146. [PMID: 37565586 DOI: 10.1002/jcp.31074] [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: 03/21/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 08/12/2023]
Abstract
One of the major obstacles to the effective application of vascularized fruit is an insufficient understanding of the relationship between the microenvironment and neovascular homeostasis. The role of extracellular matrix stiffness in regulating the structural and functional stability of neovascularization has not yet been elucidated. This study explored the effects of matrix stiffness on neovascular homeostasis in nude mice. Dextran hydrogels with three different stiffnesses were separately combined with mouse bone marrow-derived endothelial progenitor cells (EPCs) and subcutaneously implanted into the backs of nude mice. After 14 days, neovascular homeostasis indicators in the different groups were measured. Cell autophagy levels were evaluated, and inhibitor assays were performed to explore the underlying mechanism. New blood vessels were generated in the three stiffnesses of the EPC-loaded dextran hydrogels 14 days after implantation. The newly formed vessels tended to have better structural stability in softer hydrogels. Endothelial function markers, such as endothelial nitric oxide synthase and E-selectin, were downregulated as the matrix stiffness increased. Furthermore, we found that cell autophagy levels decreased in stiffer matrices, and autophagy inhibition attenuated neovascular homeostasis. A soft matrix is conducive to maintaining neovascular homeostasis through autophagy in nude mice.
Collapse
Affiliation(s)
- Bingqing Wang
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yuqing Yang
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yichen Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yanxin Yang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yi Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Chenxi Hu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Changyue Xue
- Department of Oral Implantology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Karlupia N, Schalek RL, Wu Y, Meirovitch Y, Wei D, Charney AW, Kopell BH, Lichtman JW. Immersion Fixation and Staining of Multicubic Millimeter Volumes for Electron Microscopy-Based Connectomics of Human Brain Biopsies. Biol Psychiatry 2023; 94:352-360. [PMID: 36740206 PMCID: PMC10397365 DOI: 10.1016/j.biopsych.2023.01.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Connectomics allows mapping of cells and their circuits at the nanometer scale in volumes of approximately 1 mm3. Given that the human cerebral cortex can be 3 mm in thickness, larger volumes are required. Larger-volume circuit reconstructions of human brain are limited by 1) the availability of fresh biopsies; 2) the need for excellent preservation of ultrastructure, including extracellular space; and 3) the requirement of uniform staining throughout the sample, among other technical challenges. Cerebral cortical samples from neurosurgical patients are available owing to lead placement for deep brain stimulation. Described here is an immersion fixation, heavy metal staining, and tissue processing method that consistently provides excellent ultrastructure throughout human and rodent surgical brain samples of volumes 2 × 2 × 2 mm3 and up to 37 mm3 with one dimension ≤2 mm. This method should allow synapse-level circuit analysis in samples from patients with psychiatric and neurologic disorders.
Collapse
Affiliation(s)
- Neha Karlupia
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, Massachusetts.
| | - Richard L Schalek
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, Massachusetts
| | - Yuelong Wu
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, Massachusetts
| | - Yaron Meirovitch
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, Massachusetts
| | - Donglai Wei
- Department of Computer Science, Boston College, Boston, Massachusetts
| | | | - Brian H Kopell
- Center for Neuromodulation, Department of Neurosurgery, The Icahn School of Medicine, Mount Sinai, New York, New York
| | - Jeff W Lichtman
- Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, Cambridge, Massachusetts.
| |
Collapse
|
18
|
Mulorz J, Ibing W, Cappallo M, Braß SM, Takeuchi K, Raaz U, Schellinger IN, Krott KJ, Schelzig H, Aubin H, Oberhuber A, Elvers M, Wagenhäuser MU. Ethanol Enhances Endothelial Rigidity by Targeting VE-Cadherin-Implications for Acute Aortic Dissection. J Clin Med 2023; 12:4967. [PMID: 37568369 PMCID: PMC10420172 DOI: 10.3390/jcm12154967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
(1) Background: Acute aortic dissection (AAD) is caused by an endothelial entry tear followed by intimomedial delamination of the outer layers of the vessel wall. The established risk factors include hypertension and smoking. Another rising candidate risk factor is excessive alcohol consumption. This experimental study explores the effects of nicotine (Nic), angiotensin II (Ang II), and ethanol (EtOH) on human aortic endothelial cells (hAoEC). (2) Methods: HAoECs were exposed to Nic, Ang II, and EtOH at different dose levels. Cell migration was studied using the scratch assay and live-cell imaging. The metabolic viability and permeability capacity was investigated using the water-soluble tetrazolium (WST)-1 assay and an in vitro vascular permeability assay. Cell adherence was studied by utilizing the hanging drop assay. The transcriptional and protein level changes were analyzed by RT-qPCR, Western blotting and immunohistochemistry for major junctional complexing proteins. (3) Results: We observed reduced metabolic viability following Ang II and EtOH exposure vs. control. Further, cell adherence was enhanced by EtOH exposure prior to trituration and by all risk factors after trituration, which correlated with the increased gene and protein expression of VE-cadherin upon EtOH exposure. The cell migration capacity was reduced upon EtOH exposure vs. controls. (4) Conclusion: Marked functional changes were observed upon exposure to established and potential risk factors for AAD development in hAoECs. Our findings advocate for an enhanced mechanical rigidity in hAoECs in response to the three substances studied, which in turn might increase endothelial rigidity, suggesting a novel mechanism for developing an endothelial entry tear due to reduced deformability in response to increased shear and pulsatile stress.
Collapse
Affiliation(s)
- Joscha Mulorz
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Wiebke Ibing
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Melanie Cappallo
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
- Clinic for Cardiac Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
- CURE3D Lab, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Sönke Maximilian Braß
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Kiku Takeuchi
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Uwe Raaz
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, 37075 Göttingen, Germany
- University Heart Center, 37075 Göttingen, Germany
| | - Isabel Nahal Schellinger
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August-University, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site, 37075 Göttingen, Germany
- University Heart Center, 37075 Göttingen, Germany
| | - Kim Jürgen Krott
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Hubert Schelzig
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Hug Aubin
- Clinic for Cardiac Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
- CURE3D Lab, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Alexander Oberhuber
- Clinic for Vascular and Endovascular Surgery, University Hospital Münster, 48149 Münster, Germany
| | - Margitta Elvers
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Markus Udo Wagenhäuser
- Clinic for Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| |
Collapse
|
19
|
Zhou R, Li J, Chen Z, Wang R, Shen Y, Zhang R, Zhou F, Zhang Y. Pathological hemodynamic changes and leukocyte transmigration disrupt the blood-spinal cord barrier after spinal cord injury. J Neuroinflammation 2023; 20:118. [PMID: 37210532 DOI: 10.1186/s12974-023-02787-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 04/21/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Blood-spinal cord barrier (BSCB) disruption is a key event after spinal cord injury (SCI), which permits unfavorable blood-derived substances to enter the neural tissue and exacerbates secondary injury. However, limited mechanical impact is usually followed by a large-scale BSCB disruption in SCI. How the BSCB disruption is propagated along the spinal cord in the acute period of SCI remains unclear. Thus, strategies for appropriate clinical treatment are lacking. METHODS A SCI contusion mouse model was established in wild-type and LysM-YFP transgenic mice. In vivo two-photon imaging and complementary studies, including immunostaining, capillary western blotting, and whole-tissue clearing, were performed to monitor BSCB disruption and verify relevant injury mechanisms. Clinically applied target temperature management (TTM) to reduce the core body temperature was tested for the efficacy of attenuating BSCB disruption. RESULTS Barrier leakage was detected in the contusion epicenter within several minutes and then gradually spread to more distant regions. Membrane expression of the main tight junction proteins remained unaltered at four hours post-injury. Many junctional gaps emerged in paracellular tight junctions at the small vessels from multiple spinal cord segments at 15 min post-injury. A previously unnoticed pathological hemodynamic change was observed in the venous system, which likely facilitated gap formation and barrier leakage by exerting abnormal physical force on the BSCB. Leukocytes were quickly initiated to transverse through the BSCB within 30 min post-SCI, actively facilitating gap formation and barrier leakage. Inducing leukocyte transmigration generated gap formation and barrier leakage. Furthermore, pharmacological alleviation of pathological hemodynamic changes or leukocyte transmigration reduced gap formation and barrier leakage. TTM had very little protective effects on the BSCB in the early period of SCI other than partially alleviating leukocyte infiltration. CONCLUSIONS Our data show that BSCB disruption in the early period of SCI is a secondary change, which is indicated by widespread gap formation in tight junctions. Pathological hemodynamic changes and leukocyte transmigration contribute to gap formation, which could advance our understanding of BSCB disruption and provide new clues for potential treatment strategies. Ultimately, TTM is inadequate to protect the BSCB in early SCI.
Collapse
Affiliation(s)
- Rubing Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China
| | - Junzhao Li
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Hubei, Wuhan, 430060, People's Republic of China
| | - Rong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, People's Republic of China.
| | - Yong Zhang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Peking University, Beijing, 100191, People's Republic of China.
- Key Laboratory for Neuroscience, Ministry of Education of China and National Health Commission of P.R. China, Beijing, 100191, People's Republic of China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, 100871, People's Republic of China.
| |
Collapse
|
20
|
Peluzzo AM, Bkhache M, Do LNH, Autieri MV, Liu X. Differential regulation of lymphatic junctional morphology and the potential effects on cardiovascular diseases. Front Physiol 2023; 14:1198052. [PMID: 37187962 PMCID: PMC10175597 DOI: 10.3389/fphys.2023.1198052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
The lymphatic vasculature provides an essential route to drain fluid, macromolecules, and immune cells from the interstitium as lymph, returning it to the bloodstream where the thoracic duct meets the subclavian vein. To ensure functional lymphatic drainage, the lymphatic system contains a complex network of vessels which has differential regulation of unique cell-cell junctions. The lymphatic endothelial cells lining initial lymphatic vessels form permeable "button-like" junctions which allow substances to enter the vessel. Collecting lymphatic vessels form less permeable "zipper-like" junctions which retain lymph within the vessel and prevent leakage. Therefore, sections of the lymphatic bed are differentially permeable, regulated in part by its junctional morphology. In this review, we will discuss our current understanding of regulating lymphatic junctional morphology, highlighting how it relates to lymphatic permeability during development and disease. We will also discuss the effect of alterations in lymphatic permeability on efficient lymphatic flux in health and how it may affect cardiovascular diseases, with a focus on atherosclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Xiaolei Liu
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
21
|
Astolfi G, Ciavarella C, Valente S, Coslovi C, Iannetta D, Fontana L, Pasquinelli G, Versura P. Human glial müller and umbilical vein endothelial cell coculture as an in vitro model to investigate retinal oxidative damage. A morphological and molecular assessment. Microsc Res Tech 2023; 86:439-451. [PMID: 36579625 DOI: 10.1002/jemt.24284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/29/2022] [Accepted: 12/17/2022] [Indexed: 12/30/2022]
Abstract
The aim of this study was to optimize a coculture in vitro model established between the human Müller glial cells and human umbilical vein endothelial cells, mimicking the inner blood-retinal barrier, and to explore its resistance to damage induced by oxidative stress. A spontaneously immortalized human Müller cell line MIO-M1 and human umbilical vein endothelial cells (HUVEC) were plated together at a density ratio 1:1 and maintained up to the 8th passage (p8). The MIO-M1/HUVECs p1 through p8 were treated with increasing concentrations (range 200-800 μM) of H2 O2 to evaluate oxidative stress induced damage and comparing data with single cell cultures. The following features were assayed p1 through p8: doubling time maintenance, cell viability using MTS assay, ultrastructure of cell-cell contacts, immunofluorescence for Vimentin and GFAP, molecular biology (q-PCR) for GFAP and CD31 mRNA. MIO-M1/HUVECs cocultures maintained distinct cell cytotype up to p8 as shown by flow cytometry analysis, without evidence of cross activation, displaying cell-cell tight junctions mimicking those found in human retina, only acquiring a slight resistance to oxidative stress induction over the passages. This MIO-M1/HUVECs coculture represents a simple, reproducible and affordable model for in vitro studies on oxidative stress-induced retinal damages.
Collapse
Affiliation(s)
- Gloria Astolfi
- Ophthalmology Unit, DIMES, Alma Mater Studiorum Università di Bologna, Italy
| | - Carmen Ciavarella
- Laboratory of Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Italy
| | - Sabrina Valente
- Laboratory of Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Italy
| | - Chiara Coslovi
- Ophthalmology Unit, DIMES, Alma Mater Studiorum Università di Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Danilo Iannetta
- Ophthalmology Unit, DIMES, Alma Mater Studiorum Università di Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Luigi Fontana
- Ophthalmology Unit, DIMES, Alma Mater Studiorum Università di Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Gianandrea Pasquinelli
- Laboratory of Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| | - Piera Versura
- Ophthalmology Unit, DIMES, Alma Mater Studiorum Università di Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Italy
| |
Collapse
|
22
|
Mamana J, Humber GM, Espinal ER, Seo S, Vollmuth N, Sin J, Kim BJ. Coxsackievirus B3 infects and disrupts human induced-pluripotent stem cell derived brain-like endothelial cells. Front Cell Infect Microbiol 2023; 13:1171275. [PMID: 37139492 PMCID: PMC10149843 DOI: 10.3389/fcimb.2023.1171275] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Coxsackievirus B3 (CVB3) is a significant human pathogen that is commonly found worldwide. CVB3 among other enteroviruses, are the leading causes of aseptic meningo-encephalitis which can be fatal especially in young children. How the virus gains access to the brain is poorly-understood, and the host-virus interactions that occur at the blood-brain barrier (BBB) is even less-characterized. The BBB is a highly specialized biological barrier consisting primarily of brain endothelial cells which possess unique barrier properties and facilitate the passage of nutrients into the brain while restricting access to toxins and pathogens including viruses. To determine the effects of CVB3 infection on the BBB, we utilized a model of human induced-pluripotent stem cell-derived brain-like endothelial cells (iBECs) to ascertain if CVB3 infection may alter barrier cell function and overall survival. In this study, we determined that these iBECs indeed are susceptible to CVB3 infection and release high titers of extracellular virus. We also determined that infected iBECs maintain high transendothelial electrical resistance (TEER) during early infection despite possessing high viral load. TEER progressively declines at later stages of infection. Interestingly, despite the high viral burden and TEER disruptions at later timepoints, infected iBEC monolayers remain intact, indicating a low degree of late-stage virally-mediated cell death, which may contribute to prolonged viral shedding. We had previously reported that CVB3 infections rely on the activation of transient receptor vanilloid potential 1 (TRPV1) and found that inhibiting TRPV1 activity with SB-366791 significantly limited CVB3 infection of HeLa cervical cancer cells. Similarly in this study, we observed that treating iBECs with SB-366791 significantly reduced CVB3 infection, which suggests that not only can this drug potentially limit viral entry into the brain, but also demonstrates that this infection model could be a valuable platform for testing antiviral treatments of neurotropic viruses. In all, our findings elucidate the unique effects of CVB3 infection on the BBB and shed light on potential mechanisms by which the virus can initiate infections in the brain.
Collapse
Affiliation(s)
- Julia Mamana
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Gabrielle M. Humber
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Eric R. Espinal
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Soojung Seo
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Nadine Vollmuth
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Jon Sin
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- *Correspondence: Jon Sin, ; Brandon J. Kim,
| | - Brandon J. Kim
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, AL, United States
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, United States
- *Correspondence: Jon Sin, ; Brandon J. Kim,
| |
Collapse
|
23
|
Sanati M, Afshari AR, Amini J, Mollazadeh H, Jamialahmadi T, Sahebkar A. Targeting angiogenesis in gliomas: Potential role of phytochemicals. J Funct Foods 2022; 96:105192. [DOI: 10.1016/j.jff.2022.105192] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
24
|
Feng C, Liu X, Hu N, Tang Y, Feng M, Zhou Z. Aeromonas hydrophila Ssp1: A secretory serine protease that disrupts tight junction integrity and is essential for host infection. FISH & SHELLFISH IMMUNOLOGY 2022; 127:530-541. [PMID: 35798244 DOI: 10.1016/j.fsi.2022.06.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 06/15/2023]
Abstract
Aeromonas hydrophila is a Gram-negative bacterial pathogen with a broad host range, including fish and humans. In this study, we examined the function of a secretory serine protease (named Ssp1) identified in pathogenic A. hydrophila CCL1. Ssp1 possesses a trypsin-like serine protease domain and contains two conserved PDZ domains. Recombinant Ssp1 protein (rSsp1) treatment increased intestinal permeability by downregulating and redistributing tight junction protein Occludin in intestinal Caco-2 cells in vitro. Western blot demonstrated that rSsp1 treatment in Caco-2 cells resulted in marked increases in the expressions of myosin light chain kinase (MLCK) and phosphorylated myosin light chain (p-MLC). For virulence analysis, an isogenic CCL1 mutant ΔSsp1 was created. ΔSsp1 bears an in-frame deletion of the Ssp1 gene. A live infection study in crucian carps showed that, compared to CCL1, ΔSsp1 infection exhibited increased Occludin expression, reduced intestinal permeability and tissue dissemination capacity, and attenuated overall virulence in vivo. However, ΔSsp1 showed no differences in the biofilm formation, swimming motility, and resistance to environmental stress. These lost virulence capacities of ΔSsp1 were restored by complementation with the Ssp1 gene. Global transcriptome analysis and quantitative real-time RT-PCR showed that compared to CCL1 infection, ΔSsp1 promoted the expressions of antimicrobial molecules (MUC2, LEAP-2, Hepcidin-1, and IL-22). Finally, CCL1 infection caused significant dysbiosis of the gut microbiota, including increased Vibrio and Deefgea compared to ΔSsp1 infected fish. Taken together, these results indicate that Ssp1 is essential for the virulence of A. hydrophila and is required for the perturbation of intestinal tight junction barrier.
Collapse
Affiliation(s)
- Chen Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xiaofeng Liu
- Department of Nutrition, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Niewen Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Yiyang Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Mengzhe Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Zejun Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
25
|
Jeong J, Tanaka M, Iwakiri Y. Hepatic lymphatic vascular system in health and disease. J Hepatol 2022; 77:206-218. [PMID: 35157960 PMCID: PMC9870070 DOI: 10.1016/j.jhep.2022.01.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/13/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023]
Abstract
In recent years, significant advances have been made in the study of lymphatic vessels with the identification of their specific markers and the development of research tools that have accelerated our understanding of their role in tissue homeostasis and disease pathogenesis in many organs. Compared to other organs, the lymphatic system in the liver is understudied despite its obvious importance for hepatic physiology and pathophysiology. In this review, we describe fundamental aspects of the hepatic lymphatic system and its role in a range of liver-related pathological conditions such as portal hypertension, ascites formation, malignant tumours, liver transplantation, congenital liver diseases, non-alcoholic fatty liver disease, and hepatic encephalopathy. The article concludes with a discussion regarding the modulation of lymphangiogenesis as a potential therapeutic strategy for liver diseases.
Collapse
Affiliation(s)
- Jain Jeong
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Masatake Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuko Iwakiri
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
McCright J, Naiknavare R, Yarmovsky J, Maisel K. Targeting Lymphatics for Nanoparticle Drug Delivery. Front Pharmacol 2022; 13:887402. [PMID: 35721179 PMCID: PMC9203826 DOI: 10.3389/fphar.2022.887402] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/25/2022] Open
Abstract
The lymphatics transport material from peripheral tissues to lymph nodes, where immune responses are formed, before being transported into systemic circulation. With key roles in transport and fluid homeostasis, lymphatic dysregulation is linked to diseases, including lymphedema. Fluid within the interstitium passes into initial lymphatic vessels where a valve system prevents fluid backflow. Additionally, lymphatic endothelial cells produce key chemokines, such as CCL21, that direct the migration of dendritic cells and lymphocytes. As a result, lymphatics are an attractive delivery route for transporting immune modulatory treatments to lymph nodes where immunotherapies are potentiated in addition to being an alternative method of reaching systemic circulation. In this review, we discuss the physiology of lymphatic vessels and mechanisms used in the transport of materials from peripheral tissues to lymph nodes. We then summarize nanomaterial-based strategies to take advantage of lymphatic transport functions for delivering therapeutics to lymph nodes or systemic circulation. We also describe opportunities for targeting lymphatic endothelial cells to modulate transport and immune functions.
Collapse
Affiliation(s)
| | | | | | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, MD, United States
| |
Collapse
|
27
|
Puerta-Guardo H, Biering SB, de Sousa FTG, Shu J, Glasner DR, Li J, Blanc SF, Beatty PR, Harris E. Flavivirus NS1 Triggers Tissue-Specific Disassembly of Intercellular Junctions Leading to Barrier Dysfunction and Vascular Leak in a GSK-3β-Dependent Manner. Pathogens 2022; 11:615. [PMID: 35745469 PMCID: PMC9228372 DOI: 10.3390/pathogens11060615] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
The flavivirus nonstructural protein 1 (NS1) is secreted from infected cells and contributes to endothelial barrier dysfunction and vascular leak in a tissue-dependent manner. This phenomenon occurs in part via disruption of the endothelial glycocalyx layer (EGL) lining the endothelium. Additionally, we and others have shown that soluble DENV NS1 induces disassembly of intercellular junctions (IJCs), a group of cellular proteins critical for maintaining endothelial homeostasis and regulating vascular permeability; however, the specific mechanisms by which NS1 mediates IJC disruption remain unclear. Here, we investigated the relative contribution of five flavivirus NS1 proteins, from dengue (DENV), Zika (ZIKV), West Nile (WNV), Japanese encephalitis (JEV), and yellow fever (YFV) viruses, to the expression and localization of the intercellular junction proteins β-catenin and VE-cadherin in endothelial cells from human umbilical vein and brain tissues. We found that flavivirus NS1 induced the mislocalization of β-catenin and VE-cadherin in a tissue-dependent manner, reflecting flavivirus disease tropism. Mechanistically, we observed that NS1 treatment of cells triggered internalization of VE-cadherin, likely via clathrin-mediated endocytosis, and phosphorylation of β-catenin, part of a canonical IJC remodeling pathway during breakdown of endothelial barriers that activates glycogen synthase kinase-3β (GSK-3β). Supporting this model, we found that a chemical inhibitor of GSK-3β reduced both NS1-induced permeability of human umbilical vein and brain microvascular endothelial cell monolayers in vitro and vascular leakage in a mouse dorsal intradermal model. These findings provide insight into the molecular mechanisms regulating NS1-mediated endothelial dysfunction and identify GSK-3β as a potential therapeutic target for treatment of vascular leakage during severe dengue disease.
Collapse
Affiliation(s)
- Henry Puerta-Guardo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
- Laboratorio de Virologia, CIR-Biomedicas y Unidad Colaborativa de Bioensayos Entomologicos (UCBE), Universidad Autonoma de Yucatan, Merida 97000, Mexico
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Francielle Tramontini Gomes de Sousa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Jeffrey Shu
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Dustin R. Glasner
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Jeffrey Li
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Sophie F. Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - P. Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720-3370, USA; (S.B.B.); (F.T.G.d.S.); (J.S.); (D.R.G.); (J.L.); (S.F.B.); (P.R.B.)
| |
Collapse
|
28
|
Churchill MJ, du Bois H, Heim TA, Mudianto T, Steele MM, Nolz JC, Lund AW. Infection-induced lymphatic zippering restricts fluid transport and viral dissemination from skin. J Exp Med 2022; 219:e20211830. [PMID: 35353138 PMCID: PMC8972184 DOI: 10.1084/jem.20211830] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 01/19/2022] [Accepted: 03/14/2022] [Indexed: 01/13/2023] Open
Abstract
Lymphatic vessels are often considered passive conduits that flush antigenic material, pathogens, and cells to draining lymph nodes. Recent evidence, however, suggests that lymphatic vessels actively regulate diverse processes from antigen transport to leukocyte trafficking and dietary lipid absorption. Here we tested the hypothesis that infection-induced changes in lymphatic transport actively contribute to innate host defense. We demonstrate that cutaneous vaccinia virus infection by scarification activates dermal lymphatic capillary junction tightening (zippering) and lymph node lymphangiogenesis, which are associated with reduced fluid transport and cutaneous viral sequestration. Lymphatic-specific deletion of VEGFR2 prevented infection-induced lymphatic capillary zippering, increased fluid flux out of tissue, and allowed lymphatic dissemination of virus. Further, a reduction in dendritic cell migration to lymph nodes in the absence of lymphatic VEGFR2 associated with reduced antiviral CD8+ T cell expansion. These data indicate that VEGFR2-driven lymphatic remodeling is a context-dependent, active mechanism of innate host defense that limits viral dissemination and facilitates protective, antiviral CD8+ T cell responses.
Collapse
Affiliation(s)
- Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Haley du Bois
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Taylor A. Heim
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Tenny Mudianto
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Maria M. Steele
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
| | - Jeffrey C. Nolz
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Amanda W. Lund
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR
- Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
29
|
Li T, Ji X, Liu J, Guo X, Pang R, Zhuang H, Dong L, Duan M, Li A. Ulinastatin Improves Renal Microcirculation by Protecting Endothelial Cells and Inhibiting Autophagy in a Septic Rat Model. Kidney Blood Press Res 2022; 47:256-269. [PMID: 35016182 DOI: 10.1159/000521648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/21/2021] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Increased permeability of the renal capillaries is a common consequence of sepsis-associated acute kidney injury. Vascular endothelial (VE)-cadherin is a strictly endothelial-specific adhesion molecule that can control the permeability of the blood vessel wall. Additionally, autophagy plays an important role in maintaining cell stability. Ulinastatin, a urinary trypsin inhibitor, attenuates the systemic inflammatory response and visceral vasopermeability. However, it is uncertain whether ulinastatin can improve renal microcirculation by acting on the endothelial adhesion junction. METHODS We observed the effect of ulinastatin in a septic rat model using contrast-enhanced ultrasonography (CEUS) to evaluate the perfusion of the renal cortex and medulla. Male adult Sprague Dawley rats were subjected to cecal ligation and puncture and divided into the sham, sepsis, and ulinastatin groups. Ulinastatin (50,000 U/kg) was injected into the tail vein immediately after the operation. The CEUS was performed to evaluate the renal microcirculation perfusion at 3, 6, 12, and 24 h after the operation. Histological staining was used to evaluate kidney injury scores. Western blot was used to quantify the expression of VE-cadherin, LC3II, and inflammatory factors (interleukin-1β, interleukin-6, and tumor necrosis factor-α) in kidney tissue, and enzyme-linked immunosorbent assay detected serum inflammatory factors and kidney function and early kidney injury biomarker levels. RESULTS Compared with the sham group, ulinastatin reduced the inflammatory response, inhibited autophagy, maintained the expression of VE-cadherin, and meliorated cortical and medullary perfusion. CONCLUSION Ulinastatin effectively protects the adhesion junction and helps ameliorate the perfusion of kidney capillaries during sepsis by the inhibition of autophagy and the expression of inflammatory factors.
Collapse
Affiliation(s)
- Tian Li
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Ditan Hospital, Beijing, China
| | - Xiaojun Ji
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Jingfeng Liu
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Xinjie Guo
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Ran Pang
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Haizhou Zhuang
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Lei Dong
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Meili Duan
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Ang Li
- Department of Critical Care Medicine, Capital Medical University Affiliated Beijing Ditan Hospital, Beijing, China
| |
Collapse
|
30
|
Differential Effects of Cytopathic Hypoxia on Human Retinal Endothelial Cellular Behavior: Implication for Ischemic Retinopathies. Int J Mol Sci 2022; 23:ijms23084274. [PMID: 35457092 PMCID: PMC9027301 DOI: 10.3390/ijms23084274] [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: 02/22/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 02/06/2023] Open
Abstract
Loss of barrier integrity of retinal endothelial cells (RECs) is an early feature of ischemic retinopathies (IRs), but the triggering mechanisms remain incompletely understood. Previous studies have reported mitochondrial dysfunction in several forms of IRs, which creates a cytopathic hypoxic environment where cells cannot use oxygen for energy production. Nonetheless, the contribution of cytopathic hypoxia to the REC barrier failure has not been fully explored. In this study, we dissect in-depth the role of cytopathic hypoxia in impairing the barrier function of REC. We employed the electric cell-substrate impedance sensing (ECIS) technology to monitor in real-time the impedance (Z) and hence the barrier functionality of human RECs (HRECs) under cytopathic hypoxia-inducing agent, Cobalt(II) chloride (CoCl2). Furthermore, data were deconvoluted to test the effect of cytopathic hypoxia on the three key components of barrier integrity; Rb (paracellular resistance between HRECs), α (basolateral adhesion between HRECs and the extracellular matrix), and Cm (HREC membrane capacitance). Our results showed that CoCl2 decreased the Z of HRECs dose-dependently. Specifically, the Rb parameter of the HREC barrier was the parameter that declined first and most significantly by the cytopathic hypoxia-inducing agent and in a dose-dependent manner. When Rb began to fall to its minimum, other parameters of the HREC barrier, including α and Cm, were unaffected. Interestingly, the compromised effect of cytopathic hypoxia on Rb was associated with mitochondrial dysfunction but not with cytotoxicity. In conclusion, our results demonstrate distinguishable dielectric properties of HRECs under cytopathic hypoxia in which the paracellular junction between adjacent HRECs is the most vulnerable target. Such selective behavior could be utilized to screen agents or genes that maintain and strengthen the assembly of HRECs tight junction complex.
Collapse
|
31
|
Kolarzyk AM, Wong G, Lee E. Lymphatic Tissue and Organ Engineering for In Vitro Modeling and In Vivo Regeneration. Cold Spring Harb Perspect Med 2022; 12:a041169. [PMID: 35288402 PMCID: PMC9435571 DOI: 10.1101/cshperspect.a041169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The lymphatic system has an important role in maintaining fluid homeostasis and transporting immune cells and biomolecules, such as dietary fat, metabolic products, and antigens in different organs and tissues. Therefore, impaired lymphatic vessel function and/or lymphatic vessel deficiency can lead to numerous human diseases. The discovery of lymphatic endothelial markers and prolymphangiogenic growth factors, along with a growing number of in vitro and in vivo models and technologies has expedited research in lymphatic tissue and organ engineering, advancing therapeutic strategies. In this article, we describe lymphatic tissue and organ engineering in two- and three-dimensional culture systems and recently developed microfluidics and organ-on-a-chip systems in vitro. Next, we discuss advances in lymphatic tissue and organ engineering in vivo, focusing on biomaterial and scaffold engineering and their applications for lymphatic vessels and lymphoid organ regeneration. Last, we provide expert perspective and prospects in the field of lymphatic tissue engineering.
Collapse
Affiliation(s)
- Anna M Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| | - Gigi Wong
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biological Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York 14853, USA
- Biomedical and Biological Sciences PhD Program, Ithaca, New York 14853, USA
| |
Collapse
|
32
|
Kubra KT, Uddin MA, Barabutis N. Tunicamycin Protects against LPS-Induced Lung Injury. Pharmaceuticals (Basel) 2022; 15:ph15020134. [PMID: 35215247 PMCID: PMC8876572 DOI: 10.3390/ph15020134] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 12/29/2022] Open
Abstract
The pulmonary endothelium is a dynamic semipermeable barrier that orchestrates tissue-fluid homeostasis; regulating physiological and immunological responses. Endothelial abnormalities are caused by inflammatory stimuli interacting with intracellular messengers to remodel cytoskeletal junctions and adhesion proteins. Those phenomena are associated with sepsis, acute lung injury, and acute respiratory distress syndrome. The molecular processes beyond those responses are the main interest of our group. Unfolded protein response (UPR) is a highly conserved molecular pathway resolving protein-folding defects to counteract cellular threats. An emerging body of evidence suggests that UPR is a promising target against lung and cardiovascular disease. In the present study, we reveal that Tunicamycin (TM) (UPR inducer) protects against lipopolysaccharide (LPS)-induced injury. The barrier function of the inflamed endothelium was evaluated in vitro (transendothelial and paracellular permeability); as well as in mice exposed to TM after LPS. Our study demonstrates that TM supports vascular barrier function by modulating actomyosin remodeling. Moreover, it reduces the internalization of vascular endothelial cadherin (VE-cadherin), enhancing endothelial integrity. We suggest that UPR activation may deliver novel therapeutic opportunities in diseases related to endothelial dysregulation.
Collapse
|
33
|
Qi S, Wang X, Chang K, Shen W, Yu G, Du J. The bright future of nanotechnology in lymphatic system imaging and imaging-guided surgery. J Nanobiotechnology 2022; 20:24. [PMID: 34991595 PMCID: PMC8740484 DOI: 10.1186/s12951-021-01232-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/28/2021] [Indexed: 12/23/2022] Open
Abstract
Lymphatic system is identified the second vascular system after the blood circulation in mammalian species, however the research on lymphatic system has long been hampered by the lack of comprehensive imaging modality. Nanomaterials have shown the potential to enhance the quality of lymphatic imaging due to the unparalleled advantages such as the specific passive targeting and efficient co-delivery of cocktail to peripheral lymphatic system, ease molecular engineering for precise active targeting and prolonged retention in the lymphatic system of interest. Multimodal lymphatic imaging based on nanotechnology provides a complementary means to understand the kinetics of lymphoid tissues and quantify its function. In this review, we introduce the established approaches of lymphatic imaging used in clinic and summarize their strengths and weaknesses, and list the critical influence factors on lymphatic imaging. Meanwhile, the recent developments in the field of pre-clinical lymphatic imaging are discussed to shed new lights on the design of new imaging agents, the improvement of delivery methods and imaging-guided surgery strategies.
Collapse
Affiliation(s)
- Shaolong Qi
- Key Laboratory & Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, 130031, People's Republic of China.,Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, People's Republic of China
| | - Xinyu Wang
- Key Laboratory & Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, 130031, People's Republic of China
| | - Kun Chang
- Department of Lymphology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, People's Republic of China
| | - Wenbin Shen
- Department of Lymphology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, People's Republic of China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing, 100084, People's Republic of China.
| | - Jianshi Du
- Key Laboratory & Engineering Laboratory of Lymphatic Surgery Jilin Province, China-Japan Union Hospital of Jilin University, Changchun, 130031, People's Republic of China.
| |
Collapse
|
34
|
Xia Q, Dong H, Guo Y, Fang K, Hu M, Xu L, Lu F, Gong J. The role of lacteal integrity and junction transformation in obesity: A promising therapeutic target? Front Endocrinol (Lausanne) 2022; 13:1007856. [PMID: 36506056 PMCID: PMC9729342 DOI: 10.3389/fendo.2022.1007856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/08/2022] [Indexed: 11/26/2022] Open
Abstract
Lacteals are the central lymphatic vessels in the villi of the small intestine and perform nutrient absorption, especially dietary lipids, and the transportation of antigen and antigen-presenting cells. Remodeling, proliferation, and cell-cell junctions of lymphatic endothelial cells (LECs) in lacteals are the basis of the maintenance of lacteal integrity and dietary lipid absorption. Normal lipid absorption in the diet depends on sound lacteal development and proliferation, especially integrity maintenance, namely, maintaining the appropriate proportion of button-like and zipper-like junctions. Maintaining the integrity and transforming button-to-zipper junctions in lacteals are strongly connected with obesity, which could be regulated by intestinal flora and molecular signalings, such as vascular endothelial growth factor C-vascular endothelial growth receptor 3 (VEGFC-VEGFR3) signaling, Hippo signaling, Notch signaling, angiopoietin-TIE signaling, VEGF-A/VEGFR2 signaling, and PROX1. This manuscript reviews the molecular mechanism of development, integrity maintenance, and junction transformation in lacteal related to obesity.
Collapse
Affiliation(s)
- Qingsong Xia
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yujin Guo
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ke Fang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meilin Hu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lijun Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fuer Lu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Jing Gong, ; Fuer Lu,
| | - Jing Gong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- *Correspondence: Jing Gong, ; Fuer Lu,
| |
Collapse
|
35
|
Torres-Terán I, Venczel M, Klein S. Prediction of subcutaneous drug absorption - do we have reliable data to design a simulated interstitial fluid? Int J Pharm 2021; 610:121257. [PMID: 34737015 DOI: 10.1016/j.ijpharm.2021.121257] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023]
Abstract
For many years subcutaneous (SC) administration has represented the main route for delivering biopharmaceuticals. However, little information exists about the milieu in the subcutaneous tissue, especially about the properties/composition of the fluid present in this tissue, the interstitial fluid (ISF), which is one of the key elements for the drug release and absorption. Better knowledge on SC ISF composition, properties and dynamics may provide better insight into in vivo drug performance. In addition, a simulated SC ISF, which allows better prediction of in vivo absorption of drugs after subcutaneous administration based on in vitro release experiments, would help to improve formulation design, and reduce the number of animal studies and clinical trials required to obtain marketing authorization. To date, a universal medium for predicting drug solubility/release in the interstitial space does not exist. This review provides an overview of the currently available information on composition and physicochemical properties of SC ISF and critically discusses different isolation techniques in the context of information that could be gained from the isolated fluid. Moreover, it surveys current in vitro release media aiming to mimic SC ISF composition and highlights information gaps that need to be filled for designing a meaningful artificial SC ISF.
Collapse
Affiliation(s)
- Iria Torres-Terán
- Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt Am Main, Germany; Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, 3 Felix Hausdorff Street, 17489 Greifswald, Germany
| | - Márta Venczel
- Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt Am Main, Germany
| | - Sandra Klein
- Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, 3 Felix Hausdorff Street, 17489 Greifswald, Germany.
| |
Collapse
|
36
|
Rodenburg WS, van Buul JD. Rho GTPase signalling networks in cancer cell transendothelial migration. VASCULAR BIOLOGY 2021; 3:R77-R95. [PMID: 34738075 PMCID: PMC8558887 DOI: 10.1530/vb-21-0008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023]
Abstract
Rho GTPases are small signalling G-proteins that are central regulators of cytoskeleton dynamics, and thereby regulate many cellular processes, including the shape, adhesion and migration of cells. As such, Rho GTPases are also essential for the invasive behaviour of cancer cells, and thus involved in several steps of the metastatic cascade, including the extravasation of cancer cells. Extravasation, the process by which cancer cells leave the circulation by transmigrating through the endothelium that lines capillary walls, is an essential step for metastasis towards distant organs. During extravasation, Rho GTPase signalling networks not only regulate the transmigration of cancer cells but also regulate the interactions between cancer and endothelial cells and are involved in the disruption of the endothelial barrier function, ultimately allowing cancer cells to extravasate into the underlying tissue and potentially form metastases. Thus, targeting Rho GTPase signalling networks in cancer may be an effective approach to inhibit extravasation and metastasis. In this review, the complex process of cancer cell extravasation will be discussed in detail. Additionally, the roles and regulation of Rho GTPase signalling networks during cancer cell extravasation will be discussed, both from a cancer cell and endothelial cell point of view.
Collapse
Affiliation(s)
- Wessel S Rodenburg
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, Section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
37
|
Henderson AR, Ilan IS, Lee E. A bioengineered lymphatic vessel model for studying lymphatic endothelial cell-cell junction and barrier function. Microcirculation 2021; 28:e12730. [PMID: 34569678 DOI: 10.1111/micc.12730] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/09/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Lymphatic vessels (LVs) maintain fluid homeostasis by draining interstitial fluid. A failure in lymphatic drainage triggers lymphatic diseases such as lymphedema. Since lymphatic drainage is regulated by lymphatic barrier function, developing experimental models that assess lymphatic barrier function is critical for better understanding of lymphatic physiology and disease. METHODS We built a lymphatic vessel-on-chip (LV-on-chip) by fabricating a microfluidic device that includes a hollow microchannel embedded in three-dimensional (3D) hydrogel. Employing luminal flow in the microchannel, human lymphatic endothelial cells (LECs) seeded in the microchannel formed an engineered LV exhibiting 3D conduit structure. RESULTS Lymphatic endothelial cells formed relatively permeable junctions in 3D collagen 1. However, adding fibronectin to the collagen 1 apparently tightened LEC junctions. We tested lymphatic barrier function by introducing dextran into LV lumens. While LECs in collagen 1 showed permeable barriers, LECs in fibronectin/collagen 1 showed reduced permeability, which was reversed by integrin α5 inhibition. Mechanistically, LECs expressed inactivated integrin α5 in collagen 1. However, integrin α5 is activated in fibronectin and enhances barrier function. Integrin α5 activation itself also tightened LEC junctions in the absence of fibronectin. CONCLUSIONS Lymphatic vessel-on-chip reveals integrin α5 as a regulator of lymphatic barrier function and provides a platform for studying lymphatic barrier function in various conditions.
Collapse
Affiliation(s)
- Aria R Henderson
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Isabelle S Ilan
- College of Human Ecology, Cornell University, Ithaca, New York, USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
38
|
Dai Y, Liu J, Zhang X, Min X, Wu J, Du S, Li T, Liu L, Ding Z. HSPA12A improves endothelial integrity to attenuate lung injury during endotoxemia through activating ERKs and Akt-dependent signaling. Int Immunopharmacol 2021; 99:107987. [PMID: 34343936 DOI: 10.1016/j.intimp.2021.107987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/27/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
Acute lung injury (ALI) is a critical manifestation of sepsis/septic shock. Disruption of endothelial barrier function is critical for ALI pathogenesis; however, the regulation of endothelial barrier integrity remains largely unclear. Heat shock protein A12A (HSPA12A) is an atypical member of HSP70 family. We have recently demonstrated that hepatocyte HSPA12A attenuated the bacteria endotoxin (lipopolysaccharide, LPS)-induced liver injury. However, the role of HSPA12A in endothelial barrier function and ALI is unknown. Here in this study, HSPA12A showed upregulation in lungs of mice during bacteria endotoxin (lipopolysaccharide, LPS)-induced lung injury in vivo and in primary human umbilical vein endothelial cells (HUVECs) during LPS-induced barrier disruption in vitro. Knockout of HSPA12A in mice exacerbated LPS-induced ALI. Intriguingly, overexpression of HSPA12A in HUVECs attenuated the LPS-induced endothelial hyperpermeability. In line with this, HSPA12A overexpression increased VE-cadherin and decreased VEGF expression following LPS treatment in HUVECs. Also, knockout of HSPA12A enhanced the LPS-evoked pulmonary endothelial cell apoptosis in mice whereas overexpression of HSPA12A inhibited the LPS-induced death of HUVECs. The levels of ERKs and Akt phosphorylation in HUVECs were promoted by HSPA12A overexpression when cells exposed to LPS. Importantly, inhibition of either ERKs or Akt diminished the HSPA12A-induced protection from LPS-induced endothelial hyperpermeability and death. Taken together, these findings indicated that HSPA12A is a novel regulator of endothelial barrier function through both ERKs and Akt-mediated signaling. HSPA12A might represent a viable strategy for the pulmonary protection against endotoxemia challenge.
Collapse
Affiliation(s)
- Yuan Dai
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiali Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinxu Min
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jun Wu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shuya Du
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Tingting Li
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
39
|
Sesorova IS, Dimov ID, Kashin AD, Sesorov VV, Karelina NR, Zdorikova MA, Beznoussenko GV, Mirоnоv AA. Cellular and sub-cellular mechanisms of lipid transport from gut to lymph. Tissue Cell 2021; 72:101529. [PMID: 33915359 DOI: 10.1016/j.tice.2021.101529] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 02/26/2021] [Accepted: 03/11/2021] [Indexed: 12/14/2022]
Abstract
Although the general structure of the barrier between the gut and the blood is well known, many details are still missing. Here, we analyse the literature and our own data related to lipid transcytosis through adult mammalian enterocytes, and their absorption into lymph at the tissue level of the intestine. After starvation, the Golgi complex (GC) of enterocytes is in a resting state. The addition of lipids in the form of chyme leads to the initial appearance of pre-chylomicrons (ChMs) in the tubules of the smooth endoplasmic reticulum, which are attached at the basolateral plasma membrane, immediately below the 'belt' of the adhesive junctions. Then pre-ChMs move into the cisternae of the rough endoplasmic reticulum and then into the expansion of the perforated Golgi cisternae. Next, they pass through the GC, and are concentrated in the distensions of the perforated cisternae on the trans-side of the GC. The arrival of pre-ChMs at the GC leads to the transition of the GC to a state of active transport, with formation of intercisternal connections, attachment of cis-most and trans-most perforated cisternae to the medial Golgi cisternae, and disappearance of COPI vesicles. Post-Golgi carriers then deliver ChMs to the basolateral plasma membrane, fuse with it, and secret ChMs into the intercellular space between enterocytes at the level of their interdigitating contacts. Finally, ChMs are squeezed out into the interstitium through pores in the basal membrane, most likely due to the function of the actin-myosin 'cuff' around the interdigitating contacts. These pores appear to be formed by protrusions of the dendritic cells and the enterocytes per se. ChMs are absorbed from the interstitium into the lymphatic capillaries through the special oblique contacts between endothelial cells, which function as valves through the contraction-relaxation of bundles of smooth muscle cells in the interstitium. Lipid overloading of enterocytes results in accumulation of cytoplasmic lipid droplets, an increase in diameter of ChMs, inhibition of intra-Golgi transport, and fusion of ChMs in the interstitium. Here, we summarise and analyse recent findings, and discuss their functional implications.
Collapse
Affiliation(s)
- Irina S Sesorova
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | - Ivan D Dimov
- Department of Anatomy, Ivanovo State Medical Academy, Ivanovo, Russia
| | - Alexandre D Kashin
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | - Vitaly V Sesorov
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | | | - Maria A Zdorikova
- Department of Anatomy, Saint Petersburg State Paediatric Medical University, S. Petersburg, Russia
| | | | | |
Collapse
|
40
|
Rodent incisor and molar dental follicles show distinct characteristics in tooth eruption. Arch Oral Biol 2021; 126:105117. [PMID: 33845260 DOI: 10.1016/j.archoralbio.2021.105117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Rodent incisors and molars show different eruption patterns. Dental follicles and their interaction with dental epithelia play key roles in tooth eruption. However, little is known about the differences between incisor dental follicle (IF) and molar dental follicle (MF) during tooth eruption of rodents. This study aimed to investigate the differences between IF and MF during tooth eruption under induction with cervical-loop cells (CLC) and Hertwig's epithelial root sheath (HERS) cells of rats. MATERIALS AND METHODS CLC, HERS, IF, MF cells were isolated from 10 postnatal day 7 rats and identified by immunofluorescence staining. CLC or HERS cells-derived conditioned medium (CM) was obtained to induce IF and MF cells. Cell proliferation, mineralization, gene and protein expression related to tooth eruption were detected, and histological analysis was also performed. RESULTS The osteogenic differentiation and mineralization abilities of IF cells were stronger than those of MF cells. Both CLC and HERS cells-derived CM enhanced these abilities of IF cells, whereas they showed the opposite effect on MF cells. At 7, 10, and 15 d after birth, IF cells expressed more OPG and less RANKL than MF cells. CONCLUSIONS IF and MF cells present distinct characteristics in tooth eruption, CLC and HERS cells have significant inductive effects on them.
Collapse
|
41
|
Robertson TF, Chengappa P, Gomez Atria D, Wu CF, Avery L, Roy NH, Maillard I, Petrie RJ, Burkhardt JK. Lymphocyte egress signal sphingosine-1-phosphate promotes ERM-guided, bleb-based migration. J Cell Biol 2021; 220:211919. [PMID: 33764397 PMCID: PMC8006814 DOI: 10.1083/jcb.202007182] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 02/04/2023] Open
Abstract
Ezrin, radixin, and moesin (ERM) family proteins regulate cytoskeletal responses by tethering the plasma membrane to the underlying actin cortex. Mutations in ERM proteins lead to severe combined immunodeficiency, but the function of these proteins in T cells remains poorly defined. Using mice in which T cells lack all ERM proteins, we demonstrate a selective role for these proteins in facilitating S1P-dependent egress from lymphoid organs. ERM-deficient T cells display defective S1P-induced migration in vitro, despite normal responses to standard protein chemokines. Analysis of these defects revealed that S1P promotes a fundamentally different mode of migration than chemokines, characterized by intracellular pressurization and bleb-based motility. ERM proteins facilitate this process, controlling directional migration by limiting blebbing to the leading edge. We propose that the distinct modes of motility induced by S1P and chemokines are specialized to allow T cell migration across lymphatic barriers and through tissue stroma, respectively.
Collapse
Affiliation(s)
- Tanner F Robertson
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Daniela Gomez Atria
- Division of Hematology-Oncology, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Christine F Wu
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lyndsay Avery
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nathan H Roy
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ivan Maillard
- Division of Hematology-Oncology, Department of Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ryan J Petrie
- Department of Biology, Drexel University, Philadelphia, PA
| | - Janis K Burkhardt
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
42
|
Sluiter TJ, van Buul JD, Huveneers S, Quax PHA, de Vries MR. Endothelial Barrier Function and Leukocyte Transmigration in Atherosclerosis. Biomedicines 2021; 9:328. [PMID: 33804952 PMCID: PMC8063931 DOI: 10.3390/biomedicines9040328] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a highly specialized barrier that controls passage of fluids and migration of cells from the lumen into the vessel wall. Endothelial cells assist leukocytes to extravasate and despite the variety in the specific mechanisms utilized by different leukocytes to cross different vascular beds, there is a general principle of capture, rolling, slow rolling, arrest, crawling, and ultimately diapedesis via a paracellular or transcellular route. In atherosclerosis, the barrier function of the endothelium is impaired leading to uncontrolled leukocyte extravasation and vascular leakage. This is also observed in the neovessels that grow into the atherosclerotic plaque leading to intraplaque hemorrhage and plaque destabilization. This review focuses on the vascular endothelial barrier function and the interaction between endothelial cells and leukocytes during transmigration. We will discuss the role of endothelial dysfunction, transendothelial migration of leukocytes and plaque angiogenesis in atherosclerosis.
Collapse
Affiliation(s)
- Thijs J. Sluiter
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaap D. van Buul
- Sanquin Research and Landsteiner Laboratory, Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Paul H. A. Quax
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Margreet R. de Vries
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
43
|
Buchroithner B, Mayr S, Hauser F, Priglinger E, Stangl H, Santa-Maria AR, Deli MA, Der A, Klar TA, Axmann M, Sivun D, Mairhofer M, Jacak J. Dual Channel Microfluidics for Mimicking the Blood-Brain Barrier. ACS NANO 2021; 15:2984-2993. [PMID: 33480670 PMCID: PMC7905877 DOI: 10.1021/acsnano.0c09263] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/19/2021] [Indexed: 05/25/2023]
Abstract
High-resolution imaging is essential for analysis of the steps and way stations of cargo transport in in vitro models of the endothelium. In this study, we demonstrate a microfluidic system consisting of two channels horizontally separated by a cell-growth-promoting membrane. Its design allows for high-resolution (down to single-molecule level) imaging using a high numerical aperture objective with a short working distance. To reduce optical aberrations and enable single-molecule-sensitive imaging, an observation window was constructed in the membrane via laser cutting with subsequent structuring using 3D multiphoton lithography for improved cell growth. The upper channel was loaded with endothelial cells under flow conditions, which showed polarization and junction formation. A coculture of human vascular endothelial cells with pericytes was developed that mimics the blood-brain barrier. Finally, this dual channel microfluidics system enabled 3D localization microscopy of the cytoskeleton and 3D single-molecule-sensitive tracing of lipoprotein particles.
Collapse
Affiliation(s)
- Boris Buchroithner
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Sandra Mayr
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Fabian Hauser
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Eleni Priglinger
- Ludwig
Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
| | - Herbert Stangl
- Institute
of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria
| | - Ana Raquel Santa-Maria
- Institute
of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Maria A. Deli
- Institute
of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Andras Der
- Institute
of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Thomas A. Klar
- Institute
of Applied Physics, Johannes Kepler University
Linz, Altenberger Straße 69, 4040 Linz, Austria
| | - Markus Axmann
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Dmitry Sivun
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Mario Mairhofer
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Jaroslaw Jacak
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| |
Collapse
|
44
|
Pezhman L, Tahrani A, Chimen M. Dysregulation of Leukocyte Trafficking in Type 2 Diabetes: Mechanisms and Potential Therapeutic Avenues. Front Cell Dev Biol 2021; 9:624184. [PMID: 33692997 PMCID: PMC7937619 DOI: 10.3389/fcell.2021.624184] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/04/2021] [Indexed: 12/18/2022] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a chronic inflammatory disorder that is characterized by chronic hyperglycemia and impaired insulin signaling which in addition to be caused by common metabolic dysregulations, have also been associated to changes in various immune cell number, function and activation phenotype. Obesity plays a central role in the development of T2DM. The inflammation originating from obese adipose tissue develops systemically and contributes to insulin resistance, beta cell dysfunction and hyperglycemia. Hyperglycemia can also contribute to chronic, low-grade inflammation resulting in compromised immune function. In this review, we explore how the trafficking of innate and adaptive immune cells under inflammatory condition is dysregulated in T2DM. We particularly highlight the obesity-related accumulation of leukocytes in the adipose tissue leading to insulin resistance and beta-cell dysfunction and resulting in hyperglycemia and consequent changes of adhesion and migratory behavior of leukocytes in different vascular beds. Thus, here we discuss how potential therapeutic targeting of leukocyte trafficking could be an efficient way to control inflammation as well as diabetes and its vascular complications.
Collapse
Affiliation(s)
- Laleh Pezhman
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Abd Tahrani
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom.,University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Myriam Chimen
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
45
|
Abstract
The lymphatic system has received increasing scientific and clinical attention because a wide variety of diseases are linked to lymphatic pathologies and because the lymphatic system serves as an ideal conduit for drug delivery. Lymphatic vessels exert heterogeneous roles in different organs and vascular beds, and consequently, their dysfunction leads to distinct organ-specific outcomes. Although studies in animal model systems have led to the identification of crucial lymphatic genes with potential therapeutic benefit, effective lymphatic-targeted therapeutics are currently lacking for human lymphatic pathological conditions. Here, we focus on the therapeutic roles of lymphatic vessels in diseases and summarize the promising therapeutic targets for modulating lymphangiogenesis or lymphatic function in preclinical or clinical settings. We also discuss considerations for drug delivery or targeting of lymphatic vessels for treatment of lymphatic-related diseases. The lymphatic vasculature is rapidly emerging as a critical system for targeted modulation of its function and as a vehicle for innovative drug delivery.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| | - Natalie R Harris
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA;
| |
Collapse
|
46
|
CD112 Regulates Angiogenesis and T Cell Entry into the Spleen. Cells 2021; 10:cells10010169. [PMID: 33467729 PMCID: PMC7830896 DOI: 10.3390/cells10010169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Junctional adhesion proteins play important roles in controlling angiogenesis, vascular permeability and leukocyte trafficking. CD112 (nectin-2) belongs to the immunoglobulin superfamily and was shown to engage in homophilic and heterophilic interactions with a variety of binding partners expressed on endothelial cells and on leukocytes. Recent in vitro studies suggested that CD112 regulates human endothelial cell migration and proliferation as well as transendothelial migration of leukocytes. However, so far, the role of CD112 in endothelial cell biology and in leukocyte trafficking has not been elucidated in vivo. We found CD112 to be expressed by lymphatic and blood endothelial cells in different murine tissues. In CD112-deficient mice, the blood vessel coverage in the retina and spleen was significantly enhanced. In functional in vitro studies, a blockade of CD112 modulated endothelial cell migration and significantly enhanced endothelial tube formation. An antibody-based blockade of CD112 also significantly reduced T cell transmigration across endothelial monolayers in vitro. Moreover, T cell homing to the spleen was significantly reduced in CD112-deficient mice. Overall, our results identify CD112 as a regulator of angiogenic processes in vivo and demonstrate a novel role for CD112 in T cell entry into the spleen.
Collapse
|
47
|
Norden PR, Kume T. Molecular Mechanisms Controlling Lymphatic Endothelial Junction Integrity. Front Cell Dev Biol 2021; 8:627647. [PMID: 33521001 PMCID: PMC7841202 DOI: 10.3389/fcell.2020.627647] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022] Open
Abstract
The lymphatic system is essential for lipid absorption/transport from the digestive system, maintenance of tissue fluid and protein homeostasis, and immune surveillance. Despite recent progress toward understanding the cellular and molecular mechanisms underlying the formation of the lymphatic vascular system, the nature of lymphatic vessel abnormalities and disease in humans is complex and poorly understood. The mature lymphatic vasculature forms a hierarchical network in which lymphatic endothelial cells (LECs) are joined by functionally specialized cell-cell junctions to maintain the integrity of lymphatic vessels. Blind-ended and highly permeable lymphatic capillaries drain interstitial fluid via discontinuous, button-like LEC junctions, whereas collecting lymphatic vessels, surrounded by intact basement membranes and lymphatic smooth muscle cells, have continuous, zipper-like LEC junctions to transport lymph to the blood circulatory system without leakage. In this review, we discuss the recent advances in our understanding of the mechanisms by which lymphatic button- and zipper-like junctions play critical roles in lymphatic permeability and function in a tissue- and organ-specific manner, including lacteals of the small intestine. We also provide current knowledge related to key pathways and factors such as VEGF and RhoA/ROCK signaling that control lymphatic endothelial cell junctional integrity.
Collapse
Affiliation(s)
- Pieter R Norden
- Department of Medicine, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, United States
| | - Tsutomu Kume
- Department of Medicine, Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, United States
| |
Collapse
|
48
|
Surmounting the endothelial barrier for delivery of drugs and imaging tracers. Atherosclerosis 2020; 315:93-101. [DOI: 10.1016/j.atherosclerosis.2020.04.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/14/2020] [Accepted: 04/29/2020] [Indexed: 12/18/2022]
|
49
|
Ristori E, Cicaloni V, Salvini L, Tinti L, Tinti C, Simons M, Corti F, Donnini S, Ziche M. Amyloid-β Precursor Protein APP Down-Regulation Alters Actin Cytoskeleton-Interacting Proteins in Endothelial Cells. Cells 2020; 9:cells9112506. [PMID: 33228083 PMCID: PMC7699411 DOI: 10.3390/cells9112506] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
The amyloid-β precursor protein (APP) is a ubiquitous membrane protein often associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). Despite its role in the development of the pathogenesis, APP exerts several physiological roles that have been mainly investigated in neuronal tissue. To date, the role of APP in vasculature and endothelial cells has not been fully elucidated. In this study, we used molecular and proteomic approaches to identify and investigate major cellular targets of APP down-regulation in endothelial cells. We found that APP is necessary for endothelial cells proliferation, migration and adhesion. The loss of APP alters focal adhesion stability and cell-cell junctions' expression. Moreover, APP is necessary to mediate endothelial response to the VEGF-A growth factor. Finally, we document that APP propagates exogenous stimuli and mediates cellular response in endothelial cells by modulating the Scr/FAK signaling pathway. Thus, the intact expression and processing of APP is required for normal endothelial function. The identification of molecular mechanisms responsible for vasoprotective properties of endothelial APP may have an impact on clinical efforts to preserve and protect healthy vasculature in patients at risk of the development of cerebrovascular disease and dementia including AD and CAA.
Collapse
Affiliation(s)
- Emma Ristori
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Vittoria Cicaloni
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Salvini
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Cristina Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Michael Simons
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
- Departments of Medicine (Cardiology) and Cell Biology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Federico Corti
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
| | - Sandra Donnini
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| | - Marina Ziche
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| |
Collapse
|
50
|
Abstract
The lymphatic vasculature is a vital component of the vertebrate vascular system that mediates tissue fluid homeostasis, lipid uptake and immune surveillance. The development of the lymphatic vasculature starts in the early vertebrate embryo, when a subset of blood vascular endothelial cells of the cardinal veins acquires lymphatic endothelial cell fate. These cells sprout from the veins, migrate, proliferate and organize to give rise to a highly structured and unique vascular network. Cellular cross-talk, cell-cell communication and the interpretation of signals from surrounding tissues are all essential for coordinating these processes. In this chapter, we highlight new findings and review research progress with a particular focus on LEC migration and guidance, expansion of the LEC lineage, network remodeling and morphogenesis of the lymphatic vasculature.
Collapse
|