51
|
Deppermann C, Kratofil RM, Peiseler M, David BA, Zindel J, Castanheira FVES, van der Wal F, Carestia A, Jenne CN, Marth JD, Kubes P. Macrophage galactose lectin is critical for Kupffer cells to clear aged platelets. J Exp Med 2020; 217:133651. [PMID: 31978220 PMCID: PMC7144524 DOI: 10.1084/jem.20190723] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/01/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Every day, megakaryocytes produce billions of platelets that circulate for several days and eventually are cleared by the liver. The exact removal mechanism, however, remains unclear. Loss of sialic acid residues is thought to feature in the aging and clearance of platelets. Using state-of-the-art spinning disk intravital microscopy to delineate the different compartments and cells of the mouse liver, we observed rapid accumulation of desialylated platelets predominantly on Kupffer cells, with only a few on endothelial cells and none on hepatocytes. Kupffer cell depletion prevented the removal of aged platelets from circulation. Ashwell-Morell receptor (AMR) deficiency alone had little effect on platelet uptake. Macrophage galactose lectin (MGL) together with AMR mediated clearance of desialylated or cold-stored platelets by Kupffer cells. Effective clearance is critical, as mice with an aged platelet population displayed a bleeding phenotype. Our data provide evidence that the MGL of Kupffer cells plays a significant role in the removal of desialylated platelets through a collaboration with the AMR, thereby maintaining a healthy and functional platelet compartment.
Collapse
Affiliation(s)
- Carsten Deppermann
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Rachel M Kratofil
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Moritz Peiseler
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Bruna A David
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Joel Zindel
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Fernanda Vargas E Silva Castanheira
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Fardau van der Wal
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Agostina Carestia
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Craig N Jenne
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jamey D Marth
- Center for Nanomedicine, SBP Medical Discovery Institute, and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA
| | - Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
52
|
Mak KM, Shin DW. Hepatic sinusoids versus central veins: Structures, markers, angiocrines, and roles in liver regeneration and homeostasis. Anat Rec (Hoboken) 2020; 304:1661-1691. [PMID: 33135318 DOI: 10.1002/ar.24560] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/14/2020] [Accepted: 10/22/2020] [Indexed: 01/20/2023]
Abstract
The blood circulates through the hepatic sinusoids delivering nutrients and oxygen to the liver parenchyma and drains into the hepatic central vein, yet the structures and phenotypes of these vessels are distinctively different. Sinusoidal endothelial cells are uniquely fenestrated, lack basal lamina and possess organelles involved in endocytosis, pinocytosis, degradation, synthesis and secretion. Hepatic central veins are nonfenestrated but are also active in synthesis and secretion. Endothelial cells of sinusoids and central veins secrete angiocrines that play respective roles in hepatic regeneration and metabolic homeostasis. The list of markers for identifying sinusoidal endothelial cells is long and their terminologies are complex. Further, their uses vary in different investigations and, in some instances, could be confusing. Central vein markers are fewer but more distinctive. Here we analyze and categorize the molecular pathways/modules associated with the sinusoid-mediated liver regeneration in response to partial hepatectomy and chemical-induced acute or chronic injury. Similarly, we highlight the findings that central vein-derived angiocrines interact with Wnt/β-catenin in perivenous hepatocytes to direct gene expression and maintain pericentral metabolic zonation. The proposal that perivenous hepatocytes behave as stem/progenitor cells to provoke hepatic homeostatic cell renewal is reevaluated and newer concepts of broad zonal distribution of hepatocyte proliferation in liver homeostasis and regeneration are updated. Thus, this review integrates the structures, biology and physiology of liver sinusoids and central veins in mediating hepatic regeneration and metabolic homeostasis.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Da Wi Shin
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
53
|
Verhaegh P, Wisse E, de Munck T, Greve JW, Verheij J, Riedl R, Duimel H, Masclee A, Jonkers D, Koek G. Electron microscopic observations in perfusion-fixed human non-alcoholic fatty liver disease biopsies. Pathology 2020; 53:220-228. [PMID: 33143903 DOI: 10.1016/j.pathol.2020.07.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/23/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a widespread liver disease in Western society, but its multifactorial pathogenesis is not yet fully understood. Ultrastructural analysis of liver sinusoidal endothelial cells (LSECs) in animal models and in vitro studies shows defenestration early in the course of NAFLD, promoting steatosis. LSECs and fenestrae are important in the transport of lipids across the sinusoids. However, human ultrastructural data, especially on LSECs and fenestrae, are scarce. This study aimed to explore the ultrastructural changes in perfusion type fixed liver biopsies of NAFLD patients with and without non-alcoholic steatohepatitis (NASH), with a special focus on LSECs and their fenestration. Liver biopsies from patients with NAFLD were fixed using two perfusion techniques, jet and injection fixation, for needle and wedge biopsies, respectively. Ultrastructural changes were studied using transmission electron microscopy. NASH was diagnosed by bright-field microscopy using the SAF score (steatosis, activity, fibrosis). Thirty-seven patients were included, of which 12 (32.4%) had NASH. Significantly less defenestration was found in NASH compared to noNASH samples (p=0.002). Other features, i.e., giant mitochondria and fenestrae size did not differ between groups. Furthermore, we described new structures, i.e., single cell steatonecrosis and inflammatory fat follicles (IFF) that were observed in both groups. Concluding, defenestration was more common in noNASH compared to NASH in human liver samples. Defenestration was not related to the degree of steatosis or fibrosis. We speculate that defenestration can be a protective mechanism in simple steatosis which is lacking in NASH.
Collapse
Affiliation(s)
- Pauline Verhaegh
- Department of Internal Medicine, Division Gastroenterology-Hepatology, Maastricht University Medical Center, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands.
| | - Eddie Wisse
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, and Department of Internal Medicine/Hepatology, The University of Maastricht, Maastricht, the Netherlands
| | - Toon de Munck
- Department of Internal Medicine, Division Gastroenterology-Hepatology, Maastricht University Medical Center, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Jan Willem Greve
- Department of Surgery, Zuyderland MC, Heerlen, the Netherlands; Department of Gastro-Intestinal Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Robert Riedl
- Department of Pathology, Zuyderland MC, Heerlen, the Netherlands
| | - Hans Duimel
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, and Department of Internal Medicine/Hepatology, The University of Maastricht, Maastricht, the Netherlands
| | - Ad Masclee
- Department of Internal Medicine, Division Gastroenterology-Hepatology, Maastricht University Medical Center, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Daisy Jonkers
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Ger Koek
- Department of Internal Medicine, Division Gastroenterology-Hepatology, Maastricht University Medical Center, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands; Department of Visceral and Transplantation Surgery, Klinikum RWTH, Aachen, Germany
| |
Collapse
|
54
|
Abstract
Aging increases the incidence of chronic liver disease (CLD), worsens its prognosis, and represents the predominant risk factor for its development at all different stages. The hepatic sinusoid, which is fundamental for maintaining liver homeostasis, is composed by hepatocytes, liver sinusoidal endothelial cells, hepatic stellate cells, and hepatic macrophages. During CLD progression, hepatic cells suffer deregulations in their phenotype, which ultimately lead to disease development. The effects of aging on the hepatic sinusoid phenotype and function are not well understood, nevertheless, studies performed in experimental models of liver diseases and aging demonstrate alterations in all hepatic sinusoidal cells. This review provides an updated description of age-related changes in the hepatic sinusoid and discusses the implications for CLD development and treatment. Lastly, we propose aging as a novel therapeutic target to treat liver diseases and summarize the most promising therapies to prevent or improve CLD and extend healthspan.
Collapse
Affiliation(s)
- Raquel Maeso-Díaz
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, North Carolina
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, CIBEREHD, Barcelona, Spain.,Division of Hepatology, Department of Biomedical Research, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
55
|
Wallnöfer EA, Thurner GC, Kremser C, Talasz H, Stollenwerk MM, Helbok A, Klammsteiner N, Albrecht-Schgoer K, Dietrich H, Jaschke W, Debbage P. Albumin-based nanoparticles as contrast medium for MRI: vascular imaging, tissue and cell interactions, and pharmacokinetics of second-generation nanoparticles. Histochem Cell Biol 2020; 155:19-73. [PMID: 33040183 DOI: 10.1007/s00418-020-01919-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
This multidisciplinary study examined the pharmacokinetics of nanoparticles based on albumin-DTPA-gadolinium chelates, testing the hypothesis that these nanoparticles create a stronger vessel signal than conventional gadolinium-based contrast agents and exploring if they are safe for clinical use. Nanoparticles based on human serum albumin, bearing gadolinium and designed for use in magnetic resonance imaging, were used to generate magnet resonance images (MRI) of the vascular system in rats ("blood pool imaging"). At the low nanoparticle doses used for radionuclide imaging, nanoparticle-associated metals were cleared from the blood into the liver during the first 4 h after nanoparticle application. At the higher doses required for MRI, the liver became saturated and kidney and spleen acted as additional sinks for the metals, and accounted for most processing of the nanoparticles. The multiple components of the nanoparticles were cleared independently of one another. Albumin was detected in liver, spleen, and kidneys for up to 2 days after intravenous injection. Gadolinium was retained in the liver, kidneys, and spleen in significant concentrations for much longer. Gadolinium was present as significant fractions of initial dose for longer than 2 weeks after application, and gadolinium clearance was only complete after 6 weeks. Our analysis could not account quantitatively for the full dose of gadolinium that was applied, but numerous organs were found to contain gadolinium in the collagen of their connective tissues. Multiple lines of evidence indicated intracellular processing opening the DTPA chelates and leading to gadolinium long-term storage, in particular inside lysosomes. Turnover of the stored gadolinium was found to occur in soluble form in the kidneys, the liver, and the colon for up to 3 weeks after application. Gadolinium overload poses a significant hazard due to the high toxicity of free gadolinium ions. We discuss the relevance of our findings to gadolinium-deposition diseases.
Collapse
Affiliation(s)
- E A Wallnöfer
- Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - G C Thurner
- Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Division of Histology and Embryology, Department of Anatomy, Histology and Embryology, Medical University of Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria
| | - C Kremser
- Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - H Talasz
- Division of Clinical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - M M Stollenwerk
- Faculty of Health and Society, Biomedical Laboratory Science, University Hospital MAS, Malmö University, 205 06, Malmö, Sweden
- Division of Histology and Embryology, Department of Anatomy, Histology and Embryology, Medical University of Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria
| | - A Helbok
- Department of Nuclear Medicine, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - N Klammsteiner
- Division of Histology and Embryology, Department of Anatomy, Histology and Embryology, Medical University of Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria
| | - K Albrecht-Schgoer
- Department of Pharmaceutical Technology, Institute of Pharmacy, Leopold-Franzens-University Innsbruck, Innrain 80-82/IV, 6020, Innsbruck, Austria
- Institute of Cell Genetics, Department for Pharmacology and Genetics, Medical University of Innsbruck, Peter-Mayr-Strasse 1a, 6020, Innsbruck, Austria
| | - H Dietrich
- Central Laboratory Animal Facilities, Innsbruck Medical University, Peter-Mayr-Strasse 4a, 6020, Innsbruck, Austria
| | - W Jaschke
- Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - P Debbage
- Division of Histology and Embryology, Department of Anatomy, Histology and Embryology, Medical University of Innsbruck, Müllerstrasse 59, 6020, Innsbruck, Austria.
| |
Collapse
|
56
|
Pandey E, Nour AS, Harris EN. Prominent Receptors of Liver Sinusoidal Endothelial Cells in Liver Homeostasis and Disease. Front Physiol 2020; 11:873. [PMID: 32848838 PMCID: PMC7396565 DOI: 10.3389/fphys.2020.00873] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) are the most abundant non-parenchymal cells lining the sinusoidal capillaries of the hepatic system. LSECs are characterized with numerous fenestrae and lack basement membrane as well as a diaphragm. These unique morphological characteristics of LSECs makes them the most permeable endothelial cells of the mammalian vasculature and aid in regulating flow of macromolecules and small lipid-based structures between sinusoidal blood and parenchymal cells. LSECs have a very high endocytic capacity aided by scavenger receptors (SR), such as SR-A, SR-B (SR-B1 and CD-36), SR-E (Lox-1 and mannose receptors), and SR-H (Stabilins). Other high-affinity receptors for mediating endocytosis include the FcγRIIb, which assist in the antibody-mediated removal of immune complexes. Complemented with intense lysosomal activity, LSECs play a vital role in the uptake and degradation of many blood borne waste macromolecules and small (<280 nm) colloids. Currently, seven Toll-like receptors have been investigated in LSECs, which are involved in the recognition and clearance of pathogen-associated molecular pattern (PAMPs) as well as damage associated molecular pattern (DAMP). Along with other SRs, LSECs play an essential role in maintaining lipid homeostasis with the low-density lipoprotein receptor-related protein-1 (LRP-1), in juxtaposition with hepatocytes. LSECs co-express two surface lectins called L-Specific Intercellular adhesion molecule-3 Grabbing Non-integrin Receptor (L-SIGN) and liver sinusoidal endothelial cell lectin (LSECtin). LSECs also express several adhesion molecules which are involved in the recruitment of leukocytes at the site of inflammation. Here, we review these cell surface receptors as well as other components expressed by LSECs and their functions in the maintenance of liver homeostasis. We further discuss receptor expression and activity and dysregulation associated with the initiation and progression of many liver diseases, such as hepatocellular carcinoma, liver fibrosis, and cirrhosis, alcoholic and non-alcoholic fatty liver diseases and pseudocapillarization with aging.
Collapse
Affiliation(s)
- Ekta Pandey
- Department of Biochemistry, Universityof Nebraska, Lincoln, NE, United States
| | - Aiah S Nour
- Department of Biochemistry, Universityof Nebraska, Lincoln, NE, United States
| | - Edward N Harris
- Department of Biochemistry, Universityof Nebraska, Lincoln, NE, United States
| |
Collapse
|
57
|
Witzigmann D, Kulkarni JA, Leung J, Chen S, Cullis PR, van der Meel R. Lipid nanoparticle technology for therapeutic gene regulation in the liver. Adv Drug Deliv Rev 2020; 159:344-363. [PMID: 32622021 PMCID: PMC7329694 DOI: 10.1016/j.addr.2020.06.026] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 06/12/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Hereditary genetic disorders, cancer, and infectious diseases of the liver affect millions of people around the globe and are a major public health burden. Most contemporary treatments offer limited relief as they generally aim to alleviate disease symptoms. Targeting the root cause of diseases originating in the liver by regulating malfunctioning genes with nucleic acid-based drugs holds great promise as a therapeutic approach. However, employing nucleic acid therapeutics in vivo is challenging due to their unfavorable characteristics. Lipid nanoparticle (LNP) delivery technology is a revolutionary development that has enabled clinical translation of gene therapies. LNPs can deliver siRNA, mRNA, DNA, or gene-editing complexes, providing opportunities to treat hepatic diseases by silencing pathogenic genes, expressing therapeutic proteins, or correcting genetic defects. Here we discuss the state-of-the-art LNP technology for hepatic gene therapy including formulation design parameters, production methods, preclinical development and clinical translation.
Collapse
Affiliation(s)
- Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC, Canada
| | - Jayesh A Kulkarni
- NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC, Canada; Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada; Evonik Canada, Vancouver, BC, Canada
| | - Jerry Leung
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sam Chen
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Integrated Nanotherapeutics, Vancouver, BC, Canada
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; NanoMedicines Innovation Network (NMIN), University of British Columbia, Vancouver, BC, Canada.
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
58
|
Zapotoczny B, Braet F, Wisse E, Lekka M, Szymonski M. Biophysical nanocharacterization of liver sinusoidal endothelial cells through atomic force microscopy. Biophys Rev 2020; 12:625-636. [PMID: 32424787 PMCID: PMC7311612 DOI: 10.1007/s12551-020-00699-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/03/2020] [Indexed: 02/08/2023] Open
Abstract
The structural-functional hallmark of the liver sinusoidal endothelium is the presence of fenestrae grouped in sieve plates. Fenestrae are open membrane bound pores supported by a (sub)membranous cytoskeletal lattice. Changes in number and diameter of fenestrae alter bidirectional transport between the sinusoidal blood and the hepatocytes. Their physiological relevance has been shown in different liver disease models. Although the structural organization of fenestrae has been well documented using different electron microscopy approaches, the dynamic nature of those pores remained an enigma until the recent developments in the research field of four dimensional (4-D) AFM. In this contribution we highlight how AFM as a biophysical nanocharacterization tool enhanced our understanding in the dynamic behaviour of liver sinusoidal endothelial fenestrae. Different AFM probing approaches, including spectroscopy, enabled mapping of topography and nanomechanical properties at unprecedented resolution under live cell imaging conditions. This dynamic biophysical characterization approach provided us with novel information on the 'short' life-span, formation, disappearance and closure of hepatic fenestrae. These observations are briefly reviewed against the existing literature.
Collapse
Affiliation(s)
| | - Filip Braet
- Faculty of Medicine and Health, School of Medical Sciences (Discipline of Anatomy and Histology), The University of Sydney, Sydney, NSW, 2006, Australia.,Australian Centre for Microscopy & Microanalysis, The University of Sydney, Sydney, NSW, 2006, Australia.,Charles Perkins Centre (Cellular Imaging Facility), The University of Sydney, Sydney, NSW, 2006, Australia
| | - Eddie Wisse
- Maastricht Multimodal Molecular Imaging Institute, Division of Nanoscopy, University of Maastricht, Maastricht, Netherlands
| | - Malgorzata Lekka
- Institute of Nuclear Physics, Polish Academy of Sciences, 31-342, Krakow, Poland
| | - Marek Szymonski
- Research Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy and Advanced Computer Science, Jagiellonian University, Krakow, Poland
| |
Collapse
|
59
|
Auvinen K, Lokka E, Mokkala E, Jäppinen N, Tyystjärvi S, Saine H, Peurla M, Shetty S, Elima K, Rantakari P, Salmi M. Fenestral diaphragms and PLVAP associations in liver sinusoidal endothelial cells are developmentally regulated. Sci Rep 2019; 9:15698. [PMID: 31666588 PMCID: PMC6821839 DOI: 10.1038/s41598-019-52068-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/11/2019] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells contain several nanoscale domains such as caveolae, fenestrations and transendothelial channels, which regulate signaling and transendothelial permeability. These structures can be covered by filter-like diaphragms. A transmembrane PLVAP (plasmalemma vesicle associated protein) protein has been shown to be necessary for the formation of diaphragms. The expression, subcellular localization and fenestra-forming role of PLVAP in liver sinusoidal endothelial cells (LSEC) have remained controversial. Here we show that fenestrations in LSEC contain PLVAP-diaphragms during the fetal angiogenesis, but they lose the diaphragms at birth. Although it is thought that PLVAP only localizes to diaphragms, we found luminal localization of PLVAP in adult LSEC using several imaging techniques. Plvap-deficient mice revealed that the absence of PLVAP and diaphragms did not affect the morphology, the number of fenestrations or the overall vascular architecture in the liver sinusoids. Nevertheless, PLVAP in fetal LSEC (fenestrations with diaphragms) associated with LYVE-1 (lymphatic vessel endothelial hyaluronan receptor 1), neuropilin-1 and VEGFR2 (vascular endothelial growth factor receptor 2), whereas in the adult LSEC (fenestrations without diaphragms) these complexes disappeared. Collectively, our data show that PLVAP can be expressed on endothelial cells without diaphragms, contradict the prevailing concept that biogenesis of fenestrae would be PLVAP-dependent, and reveal previously unknown PLVAP-dependent molecular complexes in LSEC during angiogenesis.
Collapse
Affiliation(s)
- Kaisa Auvinen
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Emmi Lokka
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Elias Mokkala
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Norma Jäppinen
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sofia Tyystjärvi
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Heikki Saine
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Markus Peurla
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Shishir Shetty
- Centre for Liver Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Kati Elima
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Pia Rantakari
- MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, Turku, Finland. .,Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
60
|
Patten DA, Shetty S. The Role of Stabilin-1 in Lymphocyte Trafficking and Macrophage Scavenging in the Liver Microenvironment. Biomolecules 2019; 9:biom9070283. [PMID: 31315308 PMCID: PMC6681381 DOI: 10.3390/biom9070283] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic liver diseases are a major global health burden, and cases of these conditions continue to rise in many countries. A diverse range of insults can lead to chronic liver disease, but they are all characterised by the infiltration and accumulation of immune cells within liver tissue and, if progressive, can lead to tissue fibrosis and cirrhosis. In this review, we focus on the role of stabilin-1 in two key processes that contribute to liver disease, namely, the recruitment of lymphocytes into liver tissue and the response of macrophages to tissue injury. Stabilin-1 is constitutively expressed on the sinusoidal endothelium of the liver and contributes to the homeostatic scavenging function of these cells. Epithelial damage in the context of chronic liver disease leads to the upregulation of stabilin-1 at sites of tissue injury, specifically at sites of immune cell recruitment and on subpopulations of hepatic macrophages. Functionally, stabilin-1 has been shown to mediate transendothelial migration of lymphocyte subsets in the setting of pro-inflammatory-activated human liver endothelium. In experimental models of liver fibrosis, stabilin-1 promotes the uptake of products of chronic oxidative stress by a subset of hepatic macrophages and suppresses their release of pro-inflammatory mediators that regulate tissue remodelling. These studies highlight the active contribution that scavenger receptors such as stabilin-1 can make in regulating chronic inflammation and tissue fibrosis, and their potential as novel therapeutic targets for these conditions.
Collapse
Affiliation(s)
- Daniel A Patten
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham B15 2TT, UK
| | - Shishir Shetty
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
61
|
McCarron JG, Wilson C, Heathcote HR, Zhang X, Buckley C, Lee MD. Heterogeneity and emergent behaviour in the vascular endothelium. Curr Opin Pharmacol 2019; 45:23-32. [PMID: 31005824 PMCID: PMC6700393 DOI: 10.1016/j.coph.2019.03.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/18/2019] [Indexed: 12/16/2022]
Abstract
The endothelium is the single layer of cells lining all blood vessels, and it is a remarkable cardiovascular control centre. Each endothelial cell has only a small number (on average six) of interconnected neighbours. Yet this arrangement produces a large repertoire of behaviours, capable of controlling numerous cardiovascular functions in a flexible and dynamic way. The endothelium regulates the delivery of nutrients and removal of waste by regulating blood flow and vascular permeability. The endothelium regulates blood clotting, responses to infection and inflammation, the formation of new blood vessels, and remodelling of the blood vessel wall. To carry out these roles, the endothelium autonomously interprets a complex environment crammed with signals from hormones, neurotransmitters, pericytes, smooth muscle cells, various blood cells, viral or bacterial infection and proinflammatory cytokines. It is generally assumed that the endothelium responds to these instructions with coordinated responses in a homogeneous population of endothelial cells. Here, we highlight evidence that shows that neighbouring endothelial cells are highly heterogeneous and display different sensitivities to various activators. Cells with various sensitivities process different extracellular signals into distinct streams of information in parallel, like a vast switchboard. Communication occurs among cells and new ‘emergent’ signals are generated that are non-linear composites of the inputs. Emergent signals cannot be predicted or deduced from the properties of individual cells. Heterogeneity and emergent behaviour bestow capabilities on the endothelial collective that far exceed those of individual cells. The implications of heterogeneity and emergent behaviour for understanding vascular disease and drug discovery are discussed.
Collapse
Affiliation(s)
- John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Helen R Heathcote
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
62
|
Substance P Promotes Liver Sinusoidal Endothelium-Mediated Hepatic Regeneration by NO/HGF Regulation. J Interferon Cytokine Res 2019; 39:147-154. [DOI: 10.1089/jir.2018.0111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
63
|
Haidar I, Harding IH, Bowater IC, McDowall AW. Physical Characterization of Halofantrine-Encapsulated Fat Nanoemulsions. J Pharm Sci 2019; 108:2153-2161. [PMID: 30703385 DOI: 10.1016/j.xphs.2019.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/06/2018] [Accepted: 01/18/2019] [Indexed: 02/07/2023]
Abstract
We report the colloidal characterization of halofantrine (Hf)-laden soybean oil fat emulsions. Hf increased the zeta potential, at all pH values, of the fat emulsions. Concomitant with this, the isoelectric point (i.e.p.) of the emulsion increased to higher pH values. The emulsion was destabilized by a small amount of Hf; interestingly, however, this was ameliorated by increasing the amount of Hf. The particle size and polydispersity of the fat emulsion reflected this with a small Hf concentration resulting in a significant increase in both particle size and polydispersity, but less so as the Hf concentration was increased. Emulsions lost stability as the pH approached the i.e.p. and this effect was greatest for the small Hf concentration emulsions. Cryogenic transmission electron microscopy showed the presence of beading or string-like behavior leading to gross distortions of the spherical shape for highly unstable emulsions. We conclude that to maintain good stability for Hf-laden soybean oil emulsions, the pH of the emulsion should be kept away from its i.e.p, and also that the drug concentration should be maintained at a relatively high value.
Collapse
Affiliation(s)
- Iman Haidar
- Department of Chemistry and Biochemistry, Faculty of Science I, Lebanese University, Hadath, Beirut, Lebanon
| | - Ian H Harding
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Australia.
| | - Ian C Bowater
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Melbourne, Australia
| | - Alasdair W McDowall
- Howard Hughes Medical Institute, California Institute of Technology, Pasadena, California 91125; University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
64
|
Zapotoczny B, Szafranska K, Kus E, Braet F, Wisse E, Chlopicki S, Szymonski M. Tracking Fenestrae Dynamics in Live Murine Liver Sinusoidal Endothelial Cells. Hepatology 2019; 69:876-888. [PMID: 30137644 DOI: 10.1002/hep.30232] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/20/2018] [Indexed: 12/18/2022]
Abstract
The fenestrae of liver sinusoidal endothelial cells (LSECs) allow passive transport of solutes, macromolecules, and particulate material between the sinusoidal lumen and the liver parenchymal cells. Until recently, fenestrae and fenestrae-associated structures were mainly investigated using electron microscopy on chemically fixed LSECs. Hence, the knowledge about their dynamic properties has remained to date largely elusive. Recent progress in atomic force microscopy (AFM) has allowed the study of live cells in three dimensions (X, Y, and Z) over a prolonged time (t) and this at unprecedented speeds and resolving power. Hence, we employed the latest advances in AFM imaging on living LSECs. As a result, we were able to monitor the position, size, and number of fenestrae and sieve plates using four-dimensional AFM (X, Y, Z, and t) on intact LSECs in vitro. During these time-lapse experiments, dynamic data were collected on the origin and morphofunctional properties of the filtration apparatus of LSECs. We present structural evidence on single laying and grouped fenestrae, thereby elucidating their dynamic nature from formation to disappearance. We also collected data on the life span of fenestrae. More especially, the formation and closing of entire sieve plates were observed, and how the continuous rearrangement of sieve plates affects the structure of fenestrae within them was recorded. We observed also the dawn and rise of fenestrae-forming centers and defenestration centers in LSECs under different experimental conditions. Conclusion: Utilizing a multimodal biomedical high-resolution imaging technique we collected fine structural information on the life span, formation, and disappearance of LSEC fenestrae; by doing so, we also gathered evidence on three different pathways implemented in the loss of fenestrae that result in defenestrated LSECs.
Collapse
Affiliation(s)
- Bartlomiej Zapotoczny
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM), Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Karolina Szafranska
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM), Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Edyta Kus
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Filip Braet
- Discipline of Anatomy and Histology, School of Medical Sciences; Cellular Imaging Facility, Charles Perkins Centre; and Australian Centre for Microscopy & Microanalysis, The University of Sydney, NSW, Australia
| | - Eddie Wisse
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, and Department of Internal Medicine/Hepatology, The University of Maastricht, Maastricht, The Netherlands
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Marek Szymonski
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM), Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| |
Collapse
|
65
|
Abstract
The objective of this article is to propose a re-visiting of the paradigms of nano-carriers based drug routeing from an industrial viewpoint. The accumulation of drugs in specific body compartments after intravenous administration and the improvement of the oral bioavailability of peptides were taken as examples to propose an update of the translational framework preceding industrialisation. In addition to the recent advances on the biopharmacy of nano-carriers, the evolution of adjacent disciplines such as the biology of diseases, the chemistry of polymers, lipids and conjugates, the physico-chemistry of colloids and the assembling of materials at the nanoscale (referred to as microfluidics) are taken into account to consider new avenues in the applications of drug nano-carriers. The deeper integration of the properties of the drug and of the nano-carrier, in the specific context of the disease, advocates for product oriented programmes. At the same time, the advent of powerful collaborative digital tools makes possible the extension of the expertise spectrum. In this open-innovation framework, the Technology Readiness Levels (TRLs) of nano-carriers are proposed as a roadmap for the translational process from the Research stage to the Proof-of-Concept in human.
Collapse
Affiliation(s)
- Harivardhan Reddy Lakkireddy
- a Pre-Development Sciences, Pharmaceutical Development Platform , Sanofi Research & Development , Paris , France
| | - Didier V Bazile
- b Integrated CMC External Innovation , Sanofi Research & Development , Paris , France
| |
Collapse
|
66
|
Arunima A, Das JK, Suar M. Gut Microbes in Liver Diseases. DIETARY INTERVENTIONS IN GASTROINTESTINAL DISEASES 2019:117-131. [DOI: 10.1016/b978-0-12-814468-8.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
67
|
Di Martino J, Mascalchi P, Legros P, Lacomme S, Gontier E, Bioulac-Sage P, Balabaud C, Moreau V, Saltel F. Actin Depolymerization in Dedifferentiated Liver Sinusoidal Endothelial Cells Promotes Fenestrae Re-Formation. Hepatol Commun 2018; 3:213-219. [PMID: 30766959 PMCID: PMC6357827 DOI: 10.1002/hep4.1301] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 11/14/2018] [Indexed: 12/18/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) possess fenestrae, which are key for the exchange between blood and hepatocytes. Alterations in their number or diameter have important implications for hepatic function in liver diseases. They are lost early in the development of hepatic fibrosis through a process called capillarization. In this study, we aimed to demonstrate whether in vitro dedifferentiated LSECs that have lost fenestrae are able to re-form these structures. Using stimulated emission depletion super-resolution microscopy in combination with transmission electron microscopy, we analyzed fenestrae formation in a model mimicking the capillarization process in vitro. Actin is known to be involved in fenestrae regulation in differentiated LSECs. Using cytochalasin D, an actin-depolymerizing agent, we demonstrated that dedifferentiated LSECs remain capable of forming fenestrae. Conclusion: We provide a new insight into the complex role of actin in fenestrae formation and in the control of their size and show that LSEC fenestrae re-formation is possible, suggesting that this process could be used during fibrosis regression to try to restore exchanges and hepatocyte functions.
Collapse
Affiliation(s)
- Julie Di Martino
- INSERM, UMR1053 Bariton-Bordeaux Research in Translational Oncology Bordeaux France.,Université de Bordeaux Bordeaux France
| | - Patrice Mascalchi
- Université de Bordeaux Bordeaux France.,Bordeaux Imaging Center Bordeaux France
| | - Philippe Legros
- Plateforme Aquitaine de Caractérisation des Matériaux Pessac France
| | - Sabrina Lacomme
- Université de Bordeaux Bordeaux France.,Bordeaux Imaging Center Bordeaux France
| | - Etienne Gontier
- Université de Bordeaux Bordeaux France.,Bordeaux Imaging Center Bordeaux France
| | | | - Charles Balabaud
- INSERM, UMR1053 Bariton-Bordeaux Research in Translational Oncology Bordeaux France
| | - Violaine Moreau
- INSERM, UMR1053 Bariton-Bordeaux Research in Translational Oncology Bordeaux France.,Université de Bordeaux Bordeaux France
| | - Frédéric Saltel
- INSERM, UMR1053 Bariton-Bordeaux Research in Translational Oncology Bordeaux France.,Université de Bordeaux Bordeaux France
| |
Collapse
|
68
|
Shobaki N, Sato Y, Harashima H. Mixing lipids to manipulate the ionization status of lipid nanoparticles for specific tissue targeting. Int J Nanomedicine 2018; 13:8395-8410. [PMID: 30587967 PMCID: PMC6294068 DOI: 10.2147/ijn.s188016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Introduction The development of targeted drug delivery systems is a rapidly growing area in the field of nanomedicine. Methods We report herein on optimizing the targeting efficiency of a lipid nanoparticle (LNP) by manipulating the acid dissociation constant (pKa) value of its membrane, which reflects its ionization status. Instead of changing the chemical structure of the lipids to achieve this, we used a mixture of two types of pH-sensitive cationic lipids that show different pKa values in a single LNP. We mixed various ratios of YSK05 and YSK12-C4 lipids, which have pKa values of 6.50 and 8.00, respectively, in one formulation (referred to as YSK05/12-LNP). Results The pKa of the YSK05/12-LNP was dependent not only on the molar ratio of each lipid but also on the individual contribution of each lipid to the final pKa (the YSK12-C4 lipid showed a higher contribution). Furthermore, we succeeded in targeting and delivering short interfering RNA to liver sinusoidal endothelial cells using one of the YSK05/12-LNPs which showed an optimum pKa value of 7.15 and an appropriate ionization status (~36% cationic charge) to permit the particles to be taken up by liver sinusoidal endothelial cells. Conclusion This strategy has the potential for preparing custom LNPs with endless varieties of structures and final pKa values, and would have poten tial applications in drug delivery and ionic-based tissue targeting.
Collapse
Affiliation(s)
- Nour Shobaki
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan,
| | - Yusuke Sato
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan,
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan,
| |
Collapse
|
69
|
Brain-Specific Ultrastructure of Capillary Endothelial Glycocalyx and Its Possible Contribution for Blood Brain Barrier. Sci Rep 2018; 8:17523. [PMID: 30504908 PMCID: PMC6269538 DOI: 10.1038/s41598-018-35976-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/06/2018] [Indexed: 11/30/2022] Open
Abstract
Endothelial glycocalyx coats healthy vascular endothelium and plays an important role in vascular homeostasis. Although cerebral capillaries are categorized as continuous, as are those in the heart and lung, they likely have specific features related to their function in the blood brain barrier. To test that idea, brains, hearts and lungs from C57BL6 mice were processed with lanthanum-containing alkaline fixative, which preserves the structure of glycocalyx, and examined using scanning and transmission electron microscopy. We found that endothelial glycocalyx is present over the entire luminal surface of cerebral capillaries. The percent area physically covered by glycocalyx within the lumen of cerebral capillaries was 40.1 ± 4.5%, which is significantly more than in cardiac and pulmonary capillaries (15.1 ± 3.7% and 3.7 ± 0.3%, respectively). Upon lipopolysaccharide-induced vascular injury, the endothelial glycocalyx was reduced within cerebral capillaries, but substantial amounts remained. By contrast, cardiac and pulmonary capillaries became nearly devoid of glycocalyx. These findings suggest the denser structure of glycocalyx in the brain is associated with endothelial protection and may be an important component of the blood brain barrier.
Collapse
|
70
|
Maas SL, Soehnlein O, Viola JR. Organ-Specific Mechanisms of Transendothelial Neutrophil Migration in the Lung, Liver, Kidney, and Aorta. Front Immunol 2018; 9:2739. [PMID: 30538702 PMCID: PMC6277681 DOI: 10.3389/fimmu.2018.02739] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022] Open
Abstract
Immune responses are dependent on the recruitment of leukocytes to the site of inflammation. The classical leukocyte recruitment cascade, consisting of capture, rolling, arrest, adhesion, crawling, and transendothelial migration, is thoroughly studied but mostly in model systems, such as the cremasteric microcirculation. This cascade paradigm, which is widely accepted, might be applicable to many tissues, however recruitment mechanisms might substantially vary in different organs. Over the last decade, several studies shed light on organ-specific mechanisms of leukocyte recruitment. An improved awareness of this matter opens new therapeutic windows and allows targeting inflammation in a tissue-specific manner. The aim of this review is to summarize the current understanding of the leukocyte recruitment in general and how this varies in different organs. In particular we focus on neutrophils, as these are the first circulating leukocytes to reach the site of inflammation. Specifically, the recruitment mechanism in large arteries, as well as vessels in the lungs, liver, and kidney will be addressed.
Collapse
Affiliation(s)
- Sanne L Maas
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Physiology and Pharmacology (FyFa) and Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Joana R Viola
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
71
|
Albumin uptake and distribution in the zebrafish liver as observed via correlative imaging. Exp Cell Res 2018; 374:162-171. [PMID: 30496757 DOI: 10.1016/j.yexcr.2018.11.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022]
Abstract
Although liver transport routes have been extensively studied in rodents, live imaging under in situ and in vivo conditions of large volumes is still proven to be difficult. In this study, we took advantage of the optical transparency of zebrafish and their small size to explore their usefulness for correlative imaging studies and liver transport experimentations. First, we assessed the micro-architecture of the zebrafish liver and compared its fine structure to the rodent and humans' literature. Next, we investigated the transport routes and cellular distribution of albumin using combined and correlative microscopy approaches. These methods permitted us to track the injected proteins at different time points through the process of liver uptake and clearance of albumin. We demonstrate strong structural and functional resemblance between the zebrafish liver and its rodents and humans' counterparts. In as short as 5 min post-injection, albumin rapidly accumulated within the LSECs. Furthermore, albumin entered the space of Disse where it initially accumulated then subsequently was taken up by the hepatocytes. We propose the zebrafish as a viable alternative experimental model for hepatic transport studies, allowing swift multimodal imaging and direct quantification on the hepatic distribution of supramolecular complexes of interest.
Collapse
|
72
|
Falero-Perez J, Song YS, Zhao Y, Teixeira L, Sorenson CM, Sheibani N. Cyp1b1 expression impacts the angiogenic and inflammatory properties of liver sinusoidal endothelial cells. PLoS One 2018; 13:e0206756. [PMID: 30372497 PMCID: PMC6205649 DOI: 10.1371/journal.pone.0206756] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/18/2018] [Indexed: 12/30/2022] Open
Abstract
Cytochrome P450 1B1 (CYP1B1) is a member of the cytochrome p450 family of enzymes that catalyze mono-oxygenase reactions. Although constitutive Cyp1b1 expression is limited in hepatocytes, its expression and function in liver sinusoidal endothelial cells (LSEC) remains unknown. Here we determined the impact of Cyp1b1 expression on LSEC properties prepared from Cyp1b1+/+ and Cyp1b1-/- mice. LSEC expressed PECAM-1, VE-cadherin, and B4 lectin similar to EC from other mouse tissues. Cyp1b1 +/+ LSEC constitutively expressed significant levels of Cyp1b1, while Cyp1b1-/- LSEC lacked Cyp1b1 expression. LSEC also expressed VEGFR3, PROX-1, and LYVE-1, VEGFR1 and VEGFR2, as well as other cell adhesion molecules including ICAM-1, ICAM-2, VCAM-1, and thrombospondin-1 (TSP1) receptors, CD36 and CD47. However, the expression of PV-1 and stabilin (fenestration markers), and endoglin were limited in these cells. The Cyp1b1-/- LSEC showed limited fenestration, and decreased levels of VEGF and BMP6. Cyp1b1-/- LSEC also showed a decrease in the levels of VE-cadherin and ZO-1 impacting adherens and gap junction formation. Cyp1b1-/- LSEC were significantly more apoptotic, proliferated at a faster rate, and were less adherent and more migratory. These changes were attributed, in part, to decreased amounts of TSP1 and increased AKT and ERK activation. The expressions of integrins were also altered by the lack of Cyp1b1, but the ability of these cells to undergo capillary morphogenesis was minimally affected. Furthermore, Cyp1b1-/- LSEC expressed lower levels of inflammatory mediators MCP-1 and TNF-α. Thus, Cyp1b1 expression has a significant impact on LSEC angiogenic and inflammatory functions.
Collapse
Affiliation(s)
- Juliana Falero-Perez
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison WI, United States of America
| | - Yong-Seok Song
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison WI, United States of America
| | - Yun Zhao
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison WI, United States of America
| | - Leandro Teixeira
- Deaprtment of Pathobiological Sciences, University of Wisconsin School of Veterinary Medicine, Madison, WI, United States of America
| | - Christine M. Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison WI, United States of America
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| |
Collapse
|
73
|
Novel targets for delaying aging: The importance of the liver and advances in drug delivery. Adv Drug Deliv Rev 2018; 135:39-49. [PMID: 30248361 DOI: 10.1016/j.addr.2018.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023]
Abstract
Age-related changes in liver function have a significant impact on systemic aging and susceptibility to age-related diseases. Nutrient sensing pathways have emerged as important targets for the development of drugs that delay aging and the onset age-related diseases. This supports a central role for the hepatic regulation of metabolism in the association between nutrition and aging. Recently, a role for liver sinusoidal endothelial cells (LSECs) in the relationship between aging and metabolism has also been proposed. Age-related loss of fenestrations within LSECs impairs the transfer of substrates (such as lipoproteins and insulin) between sinusoidal blood and hepatocytes, resulting in post-prandial hyperlipidemia and insulin resistance. Targeted drug delivery methods such as nanoparticles and quantum dots will facilitate the direct delivery of drugs that regulate fenestrations in LSECs, providing an innovative approach to ameliorating age-related diseases and increasing healthspan.
Collapse
|
74
|
Tokarz VL, MacDonald PE, Klip A. The cell biology of systemic insulin function. J Cell Biol 2018; 217:2273-2289. [PMID: 29622564 PMCID: PMC6028526 DOI: 10.1083/jcb.201802095] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Insulin is the paramount anabolic hormone, promoting carbon energy deposition in the body. Its synthesis, quality control, delivery, and action are exquisitely regulated by highly orchestrated intracellular mechanisms in different organs or "stations" of its bodily journey. In this Beyond the Cell review, we focus on these five stages of the journey of insulin through the body and the captivating cell biology that underlies the interaction of insulin with each organ. We first analyze insulin's biosynthesis in and export from the β-cells of the pancreas. Next, we focus on its first pass and partial clearance in the liver with its temporality and periodicity linked to secretion. Continuing the journey, we briefly describe insulin's action on the blood vasculature and its still-debated mechanisms of exit from the capillary beds. Once in the parenchymal interstitium of muscle and adipose tissue, insulin promotes glucose uptake into myofibers and adipocytes, and we elaborate on the intricate signaling and vesicle traffic mechanisms that underlie this fundamental function. Finally, we touch upon the renal degradation of insulin to end its action. Cellular discernment of insulin's availability and action should prove critical to understanding its pivotal physiological functions and how their failure leads to diabetes.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
75
|
Di Martino J, Mascalchi P, Legros P, Lacomme S, Gontier E, Bioulac-Sage P, Balabaud C, Moreau V, Saltel F. STED microscopy: A simplified method for liver sinusoidal endothelial fenestrae analysis. Biol Cell 2018; 110:159-168. [PMID: 29808906 DOI: 10.1111/boc.201800016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/26/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND INFORMATION Liver sinusoidal endothelial cells (LSECs) possess fenestrae, open transcellular pores with an average diameter of 100 nm. These fenestrae allow for the exchange between blood and hepatocytes. Alterations in their number or diameter in liver diseases have important implications for hepatic microcirculation and function. Although decades of studies, fenestrae are still observed into fixed cells and we have poor knowledge of their dynamics. RESULTS Using stimulated emission depletion (STED) super-resolution microscopy, we have established a faster and simplest method to observe and quantify fenestrae. Indeed, using cytochalasin D, an actin depolymerising agent known to promote fenestrae formation, we measure the increase of fenestrae number. We adapted this methodology to develop an automated method to study fenestrae dynamics. Moreover, with two-colour STED analysis, we have shown that this approach could be useful to study LSECs fenestrae molecular composition. CONCLUSIONS Our approach demonstrates that STED microscopy is suitable for LSEC fenestrae study. SIGNIFICANCE This new way of analysing LSEC fenestrae will allow for expedited investigation of their dynamics, molecular composition and functions to better understand their function in liver pathophysiology.
Collapse
Affiliation(s)
- Julie Di Martino
- INSERM, UMR1053, Bordeaux, F-33076, France.,Université de Bordeaux, Bordeaux, F-33076, France
| | - Patrice Mascalchi
- Université de Bordeaux, Bordeaux, F-33076, France.,Bordeaux Imaging Center, Bordeaux, F-33076, France
| | | | - Sabrina Lacomme
- Université de Bordeaux, Bordeaux, F-33076, France.,Bordeaux Imaging Center, Bordeaux, F-33076, France
| | - Etienne Gontier
- Université de Bordeaux, Bordeaux, F-33076, France.,Bordeaux Imaging Center, Bordeaux, F-33076, France
| | | | | | - Violaine Moreau
- INSERM, UMR1053, Bordeaux, F-33076, France.,Université de Bordeaux, Bordeaux, F-33076, France
| | - Frédéric Saltel
- INSERM, UMR1053, Bordeaux, F-33076, France.,Université de Bordeaux, Bordeaux, F-33076, France
| |
Collapse
|
76
|
Braet F, Wisse E. Gentle palpating liver sinusoidal endothelial cells reveals the dynamic behavior and formation of fenestrae: A new window for biomedical research. Hepatology 2018; 67:2460-2461. [PMID: 29205413 DOI: 10.1002/hep.29706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 11/30/2017] [Indexed: 01/28/2023]
Affiliation(s)
- Filip Braet
- Discipline of Anatomy and Histology, School of Medical Sciences, Cellular Imaging Facility, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Eddie Wisse
- Division of Nanoscopy, Maastricht Multimodal Molecular Imaging Institute, Department of Internal Medicine, The University of Maastricht, Maastricht, The Netherlands
| |
Collapse
|
77
|
Grémy O, Miccoli L, Lelan F, Bohand S, Cherel M, Mougin-Degraef M. Delivery of DTPA through Liposomes as a Good Strategy for Enhancing Plutonium Decorporation Regardless of Treatment Regimen. Radiat Res 2018. [DOI: 10.1667/rr14968.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Olivier Grémy
- Laboratoire de RadioToxicologie, CEA, Université de Paris-Saclay, Bruyères le Châtel, France
| | - Laurent Miccoli
- Laboratoire de RadioToxicologie, CEA, Université de Paris-Saclay, Bruyères le Châtel, France
| | - Faustine Lelan
- Laboratoire de RadioToxicologie, CEA, Université de Paris-Saclay, Bruyères le Châtel, France
| | | | - Michel Cherel
- CRCINA, Inserm, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | |
Collapse
|
78
|
Campbell F, Bos FL, Sieber S, Arias-Alpizar G, Koch BE, Huwyler J, Kros A, Bussmann J. Directing Nanoparticle Biodistribution through Evasion and Exploitation of Stab2-Dependent Nanoparticle Uptake. ACS NANO 2018; 12:2138-2150. [PMID: 29320626 PMCID: PMC5876619 DOI: 10.1021/acsnano.7b06995] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Up to 99% of systemically administered nanoparticles are cleared through the liver. Within the liver, most nanoparticles are thought to be sequestered by macrophages (Kupffer cells), although significant nanoparticle interactions with other hepatic cells have also been observed. To achieve effective cell-specific targeting of drugs through nanoparticle encapsulation, improved mechanistic understanding of nanoparticle-liver interactions is required. Here, we show the caudal vein of the embryonic zebrafish ( Danio rerio) can be used as a model for assessing nanoparticle interactions with mammalian liver sinusoidal (or scavenger) endothelial cells (SECs) and macrophages. We observe that anionic nanoparticles are primarily taken up by SECs and identify an essential requirement for the scavenger receptor, stabilin-2 ( stab2) in this process. Importantly, nanoparticle-SEC interactions can be blocked by dextran sulfate, a competitive inhibitor of stab2 and other scavenger receptors. Finally, we exploit nanoparticle-SEC interactions to demonstrate targeted intracellular drug delivery resulting in the selective deletion of a single blood vessel in the zebrafish embryo. Together, we propose stab2 inhibition or targeting as a general approach for modifying nanoparticle-liver interactions of a wide range of nanomedicines.
Collapse
Affiliation(s)
- Frederick Campbell
- Department
of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
- E-mail:
| | - Frank L. Bos
- Hubrecht-Institute-KNAW
and University Medical Centre and Centre for Biomedical Genetics, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Sandro Sieber
- Division
of Pharmaceutical Technology, Department of Pharmaceutical Science, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Gabriela Arias-Alpizar
- Department
of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | - Bjørn E. Koch
- Department
of Molecular Cell Biology, Institute Biology
Leiden (IBL), Leiden University, P.O.
Box 9502, 2300 RA Leiden, The Netherlands
| | - Jörg Huwyler
- Division
of Pharmaceutical Technology, Department of Pharmaceutical Science, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Alexander Kros
- Department
of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
- E-mail:
| | - Jeroen Bussmann
- Department
of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
- Department
of Molecular Cell Biology, Institute Biology
Leiden (IBL), Leiden University, P.O.
Box 9502, 2300 RA Leiden, The Netherlands
- E-mail:
| |
Collapse
|
79
|
Matsuura T, Kawada M, Hasumura S, Nagamori S, Osata T, Yamaguchp M, Hataba Y, Tanaka H, Shimizu H, Unemura Y, Nonaka K, Iwaki T, Kojima S, Aizaki H, Mizutani S, Ikenaga H. High Density Culture of Immortalized Liver Endothelial Cells in the Radial-flow Bioreactor in the Development of an Artificial Liver. Int J Artif Organs 2018. [DOI: 10.1177/039139889802100410] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Liver endothelial cells are important components of the tissue along the hepatic sinusoid. They are responsible for microcirculation in the liver and scavenger functions. It would therefore be important to include these cells in any hybrid type of artificial liver in addition to hepatocytes. However, it is difficult to culture these cells in vitro. The development of a liver endothelial cell line, which maintains the characteristics of the primary culture, would thus be of great benefit in the development of an artificial liver. In the present study we established immortalized liver endothelial cells from the liver of an H-2Kb-tsA58 transgenic mouse, which harbors the SV40 TAg gene. Hepatic sinusoidal cells isolated from H-2Kd-tsA58 mouse proliferated In the presence of γ-interferon at 33°C. Four clones were established, out of which clone M1 had the highest amounts of PGI2 production, as well as plasminogen activator activity and internalized acetylated low density lipoprotein. On culture dishes the M1 cells grew individually and spread. Sieve plates on the cell surface were not readily visible, but small pores were detected under electron microscopic observation. These results suggest that M1 clone cells originated from liver endothelial cells. Moreover it was possible to culture the immortalized liver endothelial cells in a radial-flow bioreactor for 5 days, with a maximum 6-keto prostaglandin F1α production of 25 μg per day. This suggests that immortalized liver endothelial cells and a radial-flow bioreactor can prove useful tools in the development an artificial liver.
Collapse
Affiliation(s)
- T. Matsuura
- Department of Internal Medicine, Minato-ku, Tokyo
| | - M. Kawada
- Public Health and Environmental Medicine, Minato-ku, Tokyo
| | - S. Hasumura
- Department of Internal Medicine, Minato-ku, Tokyo
| | - S. Nagamori
- Department of Internal Medicine, Minato-ku, Tokyo
| | - T. Osata
- Institute of DNA Medicine, Minato-ku, Tokyo
| | | | - Y. Hataba
- Institute of DNA Medicine, Minato-ku, Tokyo
| | - H. Tanaka
- Institute of DNA Medicine, Minato-ku, Tokyo
| | - H. Shimizu
- Public Health and Environmental Medicine, Minato-ku, Tokyo
| | | | - K. Nonaka
- Animal Facility, The Jikei University School of Medicine, Minato-ku, Tokyo
| | - T. Iwaki
- Animal Facility, The Jikei University School of Medicine, Minato-ku, Tokyo
| | - S. Kojima
- Tsukuba Life Science Center, The Institute of Physical and Chemical Research, Tsukuba
| | - H. Aizaki
- Department of Virology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo
| | - S. Mizutani
- Central Laboratories for Key Technology, KIRIN Brewery Co., Ltd, Kanagawa - Japan
| | - H. Ikenaga
- Central Laboratories for Key Technology, KIRIN Brewery Co., Ltd, Kanagawa - Japan
| |
Collapse
|
80
|
Miller CM, Tanowitz M, Donner AJ, Prakash TP, Swayze EE, Harris EN, Seth PP. Receptor-Mediated Uptake of Phosphorothioate Antisense Oligonucleotides in Different Cell Types of the Liver. Nucleic Acid Ther 2018; 28:119-127. [PMID: 29425080 DOI: 10.1089/nat.2017.0709] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Oligonucleotide therapeutics have emerged as a third distinct platform for drug discovery within the pharmaceutical industry. Five oligonucleotide-based drugs have been approved by the US FDA and over 100 oligonucleotides drugs are currently at different stages of human trials. Several of these oligonucleotide drugs are modified using the phosphorothioate (PS) backbone modification where one of the nonbridging oxygen atoms of the phosphodiester linkage is replaced with sulfur. In this review, we summarize our knowledge on receptor-mediated uptake of PS antisense oligonucleotides (ASOs) within different cell types of the liver-a privileged organ for the discovery of oligonucleotide-based therapeutics.
Collapse
Affiliation(s)
- Colton M Miller
- 1 Department of Biochemistry, University of Nebraska , Lincoln, Nebraska
| | | | | | | | | | - Edward N Harris
- 1 Department of Biochemistry, University of Nebraska , Lincoln, Nebraska
| | | |
Collapse
|
81
|
Abstract
Portal hypertension develops as a result of increased intrahepatic vascular resistance often caused by chronic liver disease that leads to structural distortion by fibrosis, microvascular thrombosis, dysfunction of liver sinusoidal endothelial cells (LSECs), and hepatic stellate cell (HSC) activation. While the basic mechanisms of LSEC and HSC dysregulation have been extensively studied, the role of microvascular thrombosis and platelet function in the pathogenesis of portal hypertension remains to be clearly characterized. As a secondary event, portal hypertension results in splanchnic and systemic arterial vasodilation, leading to the development of a hyperdynamic circulatory syndrome and subsequently to clinically devastating complications including gastroesophageal varices and variceal hemorrhage, hepatic encephalopathy from the formation of portosystemic shunts, ascites, and renal failure due to the hepatorenal syndrome. This review article discusses: (1) mechanisms of sinusoidal portal hypertension, focusing on HSC and LSEC biology, pathological angiogenesis, and the role of microvascular thrombosis and platelets, (2) the mesenteric vasculature in portal hypertension, and (3) future directions for vascular biology research in portal hypertension.
Collapse
Affiliation(s)
- Matthew McConnell
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, 1080 LMP, 333 Cedar St., New Haven, CT, 06520, USA
| | - Yasuko Iwakiri
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, 1080 LMP, 333 Cedar St., New Haven, CT, 06520, USA.
| |
Collapse
|
82
|
Qin L, Crawford JM. Anatomy and Cellular Functions of the Liver. ZAKIM AND BOYER'S HEPATOLOGY 2018:2-19.e4. [DOI: 10.1016/b978-0-323-37591-7.00001-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
83
|
Tedesco D, Grakoui A. Environmental peer pressure: CD4 + T cell help in tolerance and transplantation. Liver Transpl 2018; 24:89-97. [PMID: 28926189 PMCID: PMC5739992 DOI: 10.1002/lt.24873] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/30/2017] [Accepted: 09/12/2017] [Indexed: 12/20/2022]
Abstract
The liver participates in a multitude of metabolic functions that are critical for sustaining human life. Despite constant encounters with antigenic-rich intestinal blood, oxidative stress, and metabolic intermediates, there is no appreciable immune response. Interestingly, patients undergoing orthotopic liver transplantation benefit from a high rate of graft acceptance in comparison to other solid organ transplant recipients. In fact, cotransplantation of a donor liver in tandem with a rejection-prone graft increases the likelihood of graft acceptance. A variety of players may account for this phenomenon including the interaction of intrahepatic antigen-presenting cells with CD4+ T cells and the preferential induction of forkhead box P3 (Foxp3) expression on CD4+ T cells following injurious stimuli. Ineffective insult management can cause chronic liver disease, which manifests systemically as the following: antibody-mediated disorders, ineffective antiviral and antibacterial immunity, and gastrointestinal disorders. These sequelae sharing the requirement of CD4+ T cell help to coordinate aberrant immune responses. In this review, we will focus on CD4+ T cell help due to the shared requirements in hepatic tolerance and coordination of extrahepatic immune responses. Overall, intrahepatic deviations from steady state can have deleterious systemic immune outcomes and highlight the liver's remarkable capacity to maintain a balance between tolerance and inflammatory response while simultaneously being inundated with a panoply of antigenic stimuli. Liver Transplantation 24 89-97 2018 AASLD.
Collapse
Affiliation(s)
- Dana Tedesco
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University,Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA,Corresponding Author: Arash Grakoui, Division of Infectious diseases, Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, Telephone: (404) 727-9368;
| |
Collapse
|
84
|
Russell JO, Monga SP. Wnt/β-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:351-378. [PMID: 29125798 DOI: 10.1146/annurev-pathol-020117-044010] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver is an organ that performs a multitude of functions, and its health is pertinent and indispensable to survival. Thus, the cellular and molecular machinery driving hepatic functions is of utmost relevance. The Wnt signaling pathway is one such signaling cascade that enables hepatic homeostasis and contributes to unique hepatic attributes such as metabolic zonation and regeneration. The Wnt/β-catenin pathway plays a role in almost every facet of liver biology. Furthermore, its aberrant activation is also a hallmark of various hepatic pathologies. In addition to its signaling function, β-catenin also plays a role at adherens junctions. Wnt/β-catenin signaling also influences the function of many different cell types. Due to this myriad of functions, Wnt/β-catenin signaling is complex, context-dependent, and highly regulated. In this review, we discuss the Wnt/β-catenin signaling pathway, its role in cell-cell adhesion and liver function, and the cell type-specific roles of Wnt/β-catenin signaling as it relates to liver physiology and pathobiology.
Collapse
Affiliation(s)
- Jacquelyn O Russell
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA
| | - Satdarshan P Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA.,Pittsburgh Liver Research Center, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania 15261, USA;
| |
Collapse
|
85
|
Abstract
This update focuses on two main topics. First, recent developments in our understanding of liver sinusoidal endothelial cell (LSEC) function will be reviewed, specifically elimination of blood-borne waste, immunological function of LSECs, interaction of LSECs with liver metastases, LSECs and liver regeneration, and LSECs and hepatic fibrosis. Second, given the current emphasis on rigor and transparency in biomedical research, the update discusses the need for standardization of methods to demonstrate identity and purity of isolated LSECs, pitfalls in methods that might lead to a selection bias in the types of LSECs isolated, and questions about long-term culture of LSECs. Various surface markers used for immunomagnetic selection are reviewed.
Collapse
Affiliation(s)
- Laurie D. DeLeve
- Division of Gastrointestinal and Liver Diseases and the USC Research Center for Liver Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Ana C. Maretti-Mira
- Division of Gastrointestinal and Liver Diseases and the USC Research Center for Liver Diseases, Keck School of Medicine of the University of Southern California, Los Angeles, California
| |
Collapse
|
86
|
Okada H, Takemura G, Suzuki K, Oda K, Takada C, Hotta Y, Miyazaki N, Tsujimoto A, Muraki I, Ando Y, Zaikokuji R, Matsumoto A, Kitagaki H, Tamaoki Y, Usui T, Doi T, Yoshida T, Yoshida S, Ushikoshi H, Toyoda I, Ogura S. Three-dimensional ultrastructure of capillary endothelial glycocalyx under normal and experimental endotoxemic conditions. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2017; 21:261. [PMID: 29058634 PMCID: PMC5651619 DOI: 10.1186/s13054-017-1841-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/13/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Sugar-protein glycocalyx coats healthy endothelium, but its ultrastructure is not well described. Our aim was to determine the three-dimensional ultrastructure of capillary endothelial glycocalyx in the heart, kidney, and liver, where capillaries are, respectively, continuous, fenestrated, and sinusoidal. METHODS Tissue samples were processed with lanthanum-containing alkaline fixative, which preserves the structure of glycocalyx. RESULTS Scanning and transmission electron microscopy revealed that the endothelial glycocalyx layer in continuous and fenestrated capillaries was substantially thicker than in sinusoids. In the heart, the endothelial glycocalyx presented as moss- or broccoli-like and covered the entire luminal endothelial cell surface. In the kidney, the glycocalyx appeared to nearly occlude the endothelial pores of the fenestrated capillaries and was also present on the surface of the renal podocytes. In sinusoids of the liver, glycocalyx covered not only the luminal side but also the opposite side, facing the space of Disse. In a mouse lipopolysaccharide-induced experimental endotoxemia model, the capillary endothelial glycocalyx was severely disrupted; that is, it appeared to be peeling off the cells and clumping. Serum concentrations of syndecan-1, a marker of glycocalyx damage, were significantly increased 24 h after administration of lipopolysaccharide. CONCLUSIONS In the present study, we visualized the three-dimensional ultrastructure of endothelial glycocalyx in healthy continuous, fenestrated, and sinusoidal capillaries, and we also showed their disruption under experimental endotoxemic conditions. The latter may provide a morphological basis for the microvascular endothelial dysfunction associated with septic injury to organs.
Collapse
Affiliation(s)
- Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Genzou Takemura
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Japan
| | - Kodai Suzuki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Kazumasa Oda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Chihiro Takada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yasuaki Hotta
- Research Institute for Biotechnology, Asahi University School of Dentistry, Mizuho, Japan
| | - Nagisa Miyazaki
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Japan
| | - Akiko Tsujimoto
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Japan
| | - Isamu Muraki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yoshiaki Ando
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Ryogen Zaikokuji
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, Gifu, Japan
| | - Atsumu Matsumoto
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hiroki Kitagaki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Yuto Tamaoki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takahiro Usui
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Tomoaki Doi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takahiro Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Shozo Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Hiroaki Ushikoshi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Izumi Toyoda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
87
|
Zapotoczny B, Szafranska K, Kus E, Chlopicki S, Szymonski M. Quantification of fenestrations in liver sinusoidal endothelial cells by atomic force microscopy. Micron 2017. [DOI: 10.1016/j.micron.2017.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
88
|
The role of vitamin D in hepatic metastases from colorectal cancer. Clin Transl Oncol 2017; 20:259-273. [PMID: 28801869 DOI: 10.1007/s12094-017-1735-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/30/2017] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) represents a significant health burden worldwide, comprising approximately 10% of annual cancer cases globally. Hepatic metastases are the most common site of CRC metastasis, and are the leading cause of death in CRC patients. There is strong epidemiologic evidence for an inverse association between vitamin D status and risk of CRC; however, the role of vitamin D in the natural history of liver metastases has not yet been investigated. Several researchers have proposed hallmarks of metastases; crucially, metastases can be blocked by interrupting just one rate-limiting step. Vitamin D status has been implicated in each proposed hallmark of metastasis. The aim of this review is to examine the potential role for vitamin D in reducing the development of hepatic metastases from CRC and outline the candidate mechanisms by which vitamin D may mediate these effects. The results of ongoing randomised intervention trials are eagerly awaited to determine whether addressing vitamin D insufficiency in CRC patients could reduce the occurrence of liver metastases, and the consequent morbidity and mortality.
Collapse
|
89
|
Zapotoczny B, Szafranska K, Owczarczyk K, Kus E, Chlopicki S, Szymonski M. Atomic Force Microscopy Reveals the Dynamic Morphology of Fenestrations in Live Liver Sinusoidal Endothelial Cells. Sci Rep 2017; 7:7994. [PMID: 28801568 PMCID: PMC5554186 DOI: 10.1038/s41598-017-08555-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/13/2017] [Indexed: 12/19/2022] Open
Abstract
Here, we report an atomic force microscopy (AFM)-based imaging method for resolving the fine nanostructures (e.g., fenestrations) in the membranes of live primary murine liver sinusoidal endothelial cells (LSECs). From data on topographical and nanomechanical properties of the selected cell areas collected within 1 min, we traced the dynamic rearrangement of the cell actin cytoskeleton connected with the formation or closing of cell fenestrations, both in non-stimulated LSECs as well as in response to cytochalasin B and antimycin A. In conclusion, AFM-based imaging permitted the near real-time measurements of dynamic changes in fenestrations in live LSECs.
Collapse
Affiliation(s)
- B Zapotoczny
- Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland.
| | - K Szafranska
- Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland.,Jagiellonian Centre for Experimental Therapeutics, JCET, Jagiellonian University, Krakow, Poland
| | - K Owczarczyk
- Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - E Kus
- Jagiellonian Centre for Experimental Therapeutics, JCET, Jagiellonian University, Krakow, Poland
| | - S Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, JCET, Jagiellonian University, Krakow, Poland.,Chair of Pharmacology, Jagiellonian University, Medical College, Krakow, Poland
| | - M Szymonski
- Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| |
Collapse
|
90
|
Metastasis 'systems' biology: how are macro-environmental signals transmitted into microenvironmental cues for disseminated tumor cells? Curr Opin Cell Biol 2017; 48:79-86. [PMID: 28715713 DOI: 10.1016/j.ceb.2017.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/07/2017] [Accepted: 06/08/2017] [Indexed: 01/01/2023]
Abstract
Disseminated breast tumor cells reside on or near stable microvascular endothelium. Currently, the cues that disrupt DTC dormancy and facilitate outgrowth are largely unknown. This article explores the hypothesis that specific patient lifestyle exposures (e.g., alcohol abuse) may disrupt the microenvironments that maintain disseminated tumor cell (DTC) dormancy in a tissue-specific fashion. We suggest that such exposures are 'transmitted' to the dormant niche in the form of injury. Thus, we discuss the relationship between wound healing and metastasis using liver as an example to illustrate how injury steers the phenotype of liver endothelium and perivascular hepatic stellate cells to a potentially pro-metastatic one. We posit further that non-steroidal anti-inflammatory drugs (NSAIDs) - recently shown to prevent metastatic relapse - may act by preserving the dormant niche. We conclude by suggesting that maintenance of the dormant niche - either through patient lifestyle or via development of therapeutics that mimic local molecular cues/responses that coincide with a healthy lifestyle - is a means to prevent metastatic relapse, and should be the subject of far greater research.
Collapse
|
91
|
Hadjikhani A, Rodzinski A, Wang P, Nagesetti A, Guduru R, Liang P, Runowicz C, Shahbazmohamadi S, Khizroev S. Biodistribution and clearance of magnetoelectric nanoparticles for nanomedical applications using energy dispersive spectroscopy. Nanomedicine (Lond) 2017; 12:1801-1822. [PMID: 28705034 DOI: 10.2217/nnm-2017-0080] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIM The biodistribution and clearance of magnetoelectric nanoparticles (MENs) in a mouse model was studied through electron energy dispersive spectroscopy. MATERIALS & METHODS This approach allows for detection of nanoparticles (NPs) in tissues with the spatial resolution of scanning electron microscopy, does not require any tissue-sensitive staining and is not limited to MENs. RESULTS The size-dependent biodistribution of intravenously administrated MENs was measured in vital organs such as the kidneys, liver, spleen, lungs and brain at four different postinjection times including 1 day, 1 week, 4 and 8 weeks, respectively. CONCLUSION The smallest NPs, 10-nm MENs, were cleared relatively rapidly and uniformly across the organs, while the clearance of the larger NPs, 100- and 600-nm MENs, was highly nonlinear with time and nonuniform across the organs.
Collapse
Affiliation(s)
- Ali Hadjikhani
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL, USA.,Center for Hardware Assurance, Security, & Engineering, University of Connecticut, Storrs, CT, USA
| | - Alexa Rodzinski
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Ping Wang
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL, USA
| | - Abhignyan Nagesetti
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL, USA
| | - Rakesh Guduru
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL, USA
| | | | - Carolyn Runowicz
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Sina Shahbazmohamadi
- Center for Hardware Assurance, Security, & Engineering, University of Connecticut, Storrs, CT, USA
| | - Sakhrat Khizroev
- Department of Electrical & Computer Engineering, Florida International University, Miami, FL, USA.,Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| |
Collapse
|
92
|
Braet F, Taatjes DJ, Wisse E. Probing the unseen structure and function of liver cells through atomic force microscopy. Semin Cell Dev Biol 2017; 73:13-30. [PMID: 28688930 DOI: 10.1016/j.semcdb.2017.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 01/02/2023]
Abstract
With the arrival of atomic force microscopy (AFM) about thirty years ago, this new imaging tool opened up a new area for the exploration of biological samples, ranging from the tissue and cellular level down to the supramolecular scale. Commercial instruments of this new imaging technique began to appear in the five years following its discovery in 1986 by Binnig, Quate & Gerber. From that point onwards the AFM has attracted many liver biologists, and the number of publications describing structure-function relationships on the diverse set of liver cells has grown steadily ever since. It is therefore timely to reflect on the achievements of AFM in disclosing the cellular architecture of hepatocytes, liver sinusoidal endothelial cells, Kupffer cells, stellate cells and liver-associated natural killer cells. In this thematic paper, we present new data and provide an in-depth overview of the current AFM literature on liver cell biology. We furthermore include a future outlook on how this scanning probe imaging tool and its latest developments can contribute to clarify various structural and functional aspects of cells in liver health and disease.
Collapse
Affiliation(s)
- Filip Braet
- School of Medical Sciences (Discipline of Anatomy and Histology)-The Bosch Institute, The University of Sydney, NSW 2006, Australia; Australian Centre for Microscopy & Microanalysis, The University of Sydney, NSW 2006, Australia; Charles Perkins Centre (Cellular Imaging Facility), The University of Sydney, NSW 2006, Australia.
| | - Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, 05405, USA; Microscopy Imaging Center, Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Eddie Wisse
- Maastricht Multimodal Molecular Imaging Institute, Division of Nanoscopy, University of Maastricht, The Netherlands; Department of Internal Medicine, University of Maastricht, 6200, MD, Maastricht, The Netherlands
| |
Collapse
|
93
|
McCarron JG, Lee MD, Wilson C. The Endothelium Solves Problems That Endothelial Cells Do Not Know Exist. Trends Pharmacol Sci 2017; 38:322-338. [PMID: 28214012 PMCID: PMC5381697 DOI: 10.1016/j.tips.2017.01.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 12/31/2022]
Abstract
The endothelium is the single layer of cells that lines the entire cardiovascular system and regulates vascular tone and blood-tissue exchange, recruits blood cells, modulates blood clotting, and determines the formation of new blood vessels. To control each function, the endothelium uses a remarkable sensory capability to continuously monitor vanishingly small changes in the concentrations of many simultaneously arriving extracellular activators that each provides cues to the physiological state. Here we suggest that the extraordinary sensory capabilities of the endothelium do not come from single cells but from the combined activity of a large number of endothelial cells. Each cell has a limited, but distinctive, sensory capacity and shares information with neighbours so that sensing is distributed among cells. Communication of information among connected cells provides system-level sensing substantially greater than the capabilities of any single cell and, as a collective, the endothelium solves sensory problems too complex for any single cell.
Collapse
Affiliation(s)
- John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow G4 0RE, UK
| |
Collapse
|
94
|
The stellate cell system (vitamin A-storing cell system). Anat Sci Int 2017; 92:387-455. [PMID: 28299597 DOI: 10.1007/s12565-017-0395-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/15/2017] [Indexed: 01/18/2023]
Abstract
Past, present, and future research into hepatic stellate cells (HSCs, also called vitamin A-storing cells, lipocytes, interstitial cells, fat-storing cells, or Ito cells) are summarized and discussed in this review. Kupffer discovered black-stained cells in the liver using the gold chloride method and named them stellate cells (Sternzellen in German) in 1876. Wake rediscovered the cells in 1971 using the same gold chloride method and various modern histological techniques including electron microscopy. Between their discovery and rediscovery, HSCs disappeared from the research history. Their identification, the establishment of cell isolation and culture methods, and the development of cellular and molecular biological techniques promoted HSC research after their rediscovery. In mammals, HSCs exist in the space between liver parenchymal cells (PCs) or hepatocytes and liver sinusoidal endothelial cells (LSECs) of the hepatic lobule, and store 50-80% of all vitamin A in the body as retinyl ester in lipid droplets in the cytoplasm. SCs also exist in extrahepatic organs such as pancreas, lung, and kidney. Hepatic (HSCs) and extrahepatic stellate cells (EHSCs) form the stellate cell (SC) system or SC family; the main storage site of vitamin A in the body is HSCs in the liver. In pathological conditions such as liver fibrosis, HSCs lose vitamin A, and synthesize a large amount of extracellular matrix (ECM) components including collagen, proteoglycan, glycosaminoglycan, and adhesive glycoproteins. The morphology of these cells also changes from the star-shaped HSCs to that of fibroblasts or myofibroblasts.
Collapse
|
95
|
Xu M, Wang X, Zou Y, Zhong Y. Key role of liver sinusoidal endothelial cells in liver fibrosis. Biosci Trends 2017; 11:163-168. [PMID: 28250338 DOI: 10.5582/bst.2017.01007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Because of the prevalence of viral hepatitis and nonalcoholic fatty liver disease (NAFLD), liver fibrosis has become a very common disease in Asia and elsewhere in the world, constantly increasing the burden of care borne by society. Hepatic sinusoidal capillarization, characterized by gradually shrinking fenestrae on the surface of liver sinusoidal endothelial cells (LSECs) and the formation of an organized basement membrane, is an initial pathologic change associated with liver fibrosis. Basic and clinical studies have indicated that LSECs play a key role in hepatic sinusoidal capillarization by affecting various aspects of the development and progression of liver fibrosis. Reviewing studies on the effect of LSECs on liver fibrosis is essential to better understanding the pathogenesis of liver fibrosis and its mechanism of progression. Moreover, such a review will provide a theoretical basis for identifying new methods to promote the regression or even inhibition of fibrosis. This review will focus on structural and functional changes in LSECs during hepatic sinusoidal capillarization and the interaction between the micro-environment of the liver and the body's immune system.
Collapse
Affiliation(s)
- Mingxing Xu
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| | - Xuehua Wang
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| | - Yong Zou
- Department of Blood Transfusion, Third Affiliated Hospital of Sun Yat-Sen University
| | - Yuesi Zhong
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| |
Collapse
|
96
|
Zapotoczny B, Owczarczyk K, Szafranska K, Kus E, Chlopicki S, Szymonski M. Morphology and force probing of primary murine liver sinusoidal endothelial cells. J Mol Recognit 2017; 30. [PMID: 28120483 DOI: 10.1002/jmr.2610] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/21/2016] [Accepted: 12/22/2016] [Indexed: 12/17/2022]
Abstract
Liver sinusoidal endothelial cells (LSECs) represent unique type of endothelial cells featured by their characteristic morphology, ie, lack of a basement membrane and presence of fenestrations-transmembrane pores acting as a dynamic filter between the vascular space and the liver parenchyma. Delicate structure of LSECs membrane combined with a submicron size of fenestrations hinders their visualization in live cells. In this work, we apply atomic force microscopy contact mode to characterize fenestrations in LSECs. We reveal the structure of fenestrations in live LSECs. Moreover, we show that the high-resolution imaging of fenestrations is possible for the glutaraldehyde-fixed LSECs. Finally, thorough information about the morphology of LSECs including great contrast in visualization of sieve plates and fenestrations is provided using Force Modulation mode. We show also the ability to precisely localize the cell nuclei in fixed LSECs. It can be helpful for more precise description of nanomechanical properties of cell nuclei using atomic force microscopy. Presented methodology combining high-quality imaging of fixed cells with an additional nanomechanical information of both live and fixed LSECs provides a unique approach to study LSECs morphology and nanomechanics that could foster understanding of the role of LSECs in maintaining liver homeostasis.
Collapse
Affiliation(s)
- B Zapotoczny
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM) Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Cracow, Poland
| | - K Owczarczyk
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM) Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Cracow, Poland
| | - K Szafranska
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM) Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Cracow, Poland.,Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Cracow, Poland
| | - E Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Cracow, Poland
| | - S Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Cracow, Poland.,Chair of Pharmacology, Jagiellonian University Medical College, Cracow, Poland
| | - M Szymonski
- Centre for Nanometer-Scale Science and Advanced Materials (NANOSAM) Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Cracow, Poland
| |
Collapse
|
97
|
Humbert M, Hugues S, Dubrot J. Shaping of Peripheral T Cell Responses by Lymphatic Endothelial Cells. Front Immunol 2017; 7:684. [PMID: 28127298 PMCID: PMC5226940 DOI: 10.3389/fimmu.2016.00684] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/22/2016] [Indexed: 12/03/2022] Open
Abstract
Lymph node stromal cells (LNSCs) have newly been promoted to the rank of new modulators of T cell responses. The different non-hematopoietic cell subsets in lymph node (LN) were considered for years as a simple scaffold, forming routes and proper environment for antigen (Ag)-lymphocyte encountering. Deeper characterization of those cells has recently clearly shown their impact on both dendritic cell and T cell functions. In particular, lymphatic endothelial cells (LECs) control lymphocyte trafficking and homeostasis in LNs and limit adaptive immune responses. Therefore, the new role of LECs in shaping immune responses has drawn the attention of immunologists. Striking is the discovery that LECs, among other LNSCs, ectopically express a large range of peripheral tissue-restricted Ags (PTAs), and further present PTA-derived peptides through major histocompatibility class I molecules to induce self-reactive CD8+ T cell deletional tolerance. In addition, both steady-state and tumor-associated LECs were described to be capable of exogenous Ag cross-presentation. Whether LECs can similarly impact CD4+ T cell responses through major histocompatibility class II restricted Ag presentation is still a matter of debate. Here, we review and discuss our current knowledge on the contribution of Ag-presenting LECs as regulators of peripheral T cell responses in different immunological contexts, including autoimmunity and cancer.
Collapse
Affiliation(s)
- Marion Humbert
- Department of Pathology and Immunology, University of Geneva Medical School , Geneva , Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, University of Geneva Medical School , Geneva , Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva Medical School , Geneva , Switzerland
| |
Collapse
|
98
|
An optimized method for mouse liver sinusoidal endothelial cell isolation. Exp Cell Res 2016; 349:291-301. [PMID: 27815020 DOI: 10.1016/j.yexcr.2016.10.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/27/2016] [Accepted: 10/29/2016] [Indexed: 01/10/2023]
|
99
|
Takegami S, Kitamura K, Ohsugi M, Konishi A, Kitade T. 19F Nuclear Magnetic Resonance Spectrometric Determination of the Partition Coefficients of Flutamide and Nilutamide (Antiprostate Cancer Drugs) in a Lipid Nano-Emulsion and Prediction of Its Encapsulation Efficiency for the Drugs. AAPS PharmSciTech 2016; 17:1500-1506. [PMID: 26863891 DOI: 10.1208/s12249-016-0495-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 01/31/2016] [Indexed: 11/30/2022] Open
Abstract
To design a useful lipid drug carrier having a high encapsulation efficiency (EE%) for the antiprostate cancer drugs flutamide (FT) and nilutamide (NT), a lipid nano-emulsion (LNE) was prepared with soybean oil (SO), phosphatidylcholine (PC), and sodium palmitate, and the partition coefficients (K ps) of the drugs for the LNE were determined by 19F nuclear magnetic resonance (NMR) spectrometry. The 19F NMR signal of the trifluoromethyl group of both drugs showed a downfield shift from an internal standard (trifluoroethanol) and broadening according to the increase in the lipid concentration due to their interaction with LNE particles. The difference in the chemical shift (Δδ) of each drug caused by the addition of LNE was measured under different amounts of LNE, and the K p values were calculated from the Δδ values. The results showed that FT has higher lipophilicity than NT. The total lipid concentration (SO + PC) required to encapsulate each drug into LNE with an EE% of more than 95% was calculated from the K p values as 93.3 and 189.9 mmol/L for FT and NT, respectively. For an LNE prepared with the total lipid concentration of 215 mmol/L, the predicted EE% values were 98 and 96% for FT and NT, respectively, while the experimental EE% values determined by a centrifugation method were approximately 99% for both drugs. Thus, the 19F NMR spectrometric method is a useful technique to obtain the K p values of fluorinated drugs and thereby predict the theoretical lipid concentrations and prepare LNEs with high EE% values.
Collapse
|
100
|
Kostallari E, Shah VH. Angiocrine signaling in the hepatic sinusoids in health and disease. Am J Physiol Gastrointest Liver Physiol 2016; 311:G246-51. [PMID: 27288423 PMCID: PMC5007289 DOI: 10.1152/ajpgi.00118.2016] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/06/2016] [Indexed: 02/08/2023]
Abstract
The capillary network irrigating the liver is important not only for nutrient and oxygen delivery, but also for the signals distributed to other hepatic cell types necessary to maintain liver homeostasis. During development, endothelial cells are a key component in liver zonation. In adulthood, they maintain hepatic stellate cells and hepatocytes in quiescence. Their importance in pathobiology is highlighted in liver regeneration and chronic liver diseases, where they coordinate paracrine cell behavior. During regeneration, liver sinusoidal endothelial cells induce hepatocyte proliferation and angiogenesis. During fibrogenesis, they undergo morphological and functional changes, which are reflected by their role in hepatic stellate cell activation, inflammation, and distorted sinusoidal structure. Therapeutic strategies to target angiocrine signaling are in progress but are in the early stages. Here, we offer a short synthesis of recent studies on angiocrine signaling in liver homeostasis, regeneration, and fibrogenesis.
Collapse
Affiliation(s)
- Enis Kostallari
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|