1
|
Kumar P, Schroder EA, Rajaram MVS, Harris EN, Ganesan LP. The Battle of LPS Clearance in Host Defense vs. Inflammatory Signaling. Cells 2024; 13:1590. [PMID: 39329771 PMCID: PMC11430141 DOI: 10.3390/cells13181590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Lipopolysaccharide (LPS) in blood circulation causes endotoxemia and is linked to various disease conditions. Current treatments focus on preventing LPS from interacting with its receptor Toll-like receptor 4 (TLR4) and reducing inflammation. However, our body has a natural defense mechanism: reticuloendothelial cells in the liver rapidly degrade and inactivate much of the circulating LPS within minutes. But this LPS clearance mechanism is not perfect. Excessive LPS that escape this clearance mechanism cause systemic inflammatory damage through TLR4. Despite its importance, the role of reticuloendothelial cells in LPS elimination is not well-studied, especially regarding the specific cells, receptors, and mechanisms involved. This gap hampers the development of effective therapies for endotoxemia and related diseases. This review consolidates the current understanding of LPS clearance, narrates known and explores potential mechanisms, and discusses the relationship between LPS clearance and LPS signaling. It also aims to highlight key insights that can guide the development of strategies to reduce circulating LPS by way of bolstering host defense mechanisms. Ultimately, we seek to provide a foundation for future research that could lead to innovative approaches for enhancing the body's natural ability to clear LPS and thereby lower the risk of endotoxin-related inflammatory diseases, including sepsis.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Evan A. Schroder
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA; (E.A.S.); (E.N.H.)
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Edward N. Harris
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, USA; (E.A.S.); (E.N.H.)
| | - Latha P. Ganesan
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
2
|
Sánchez Romano J, Simón-Santamaría J, McCourt P, Smedsrød B, Mortensen KE, Sagona AP, Sørensen KK, Larsen AK. Liver sinusoidal cells eliminate blood-borne phage K1F. mSphere 2024; 9:e0070223. [PMID: 38415633 PMCID: PMC10964407 DOI: 10.1128/msphere.00702-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
Phage treatment has regained attention due to an increase in multiresistant bacteria. For phage therapy to be successful, phages must reach their target bacteria in sufficiently high numbers. Blood-borne phages are believed to be captured by macrophages in the liver and spleen. Since liver sinusoids also consist of specialized scavenger liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs), this study investigated the contribution of both cell types in the elimination of Escherichia coli phage K1Fg10b::gfp (K1Fgfp) in mice. Circulatory half-life, organ, and hepatocellular distribution of K1Fgfp were determined following intravenous administration. Internalization of K1Fgfp and effects of phage opsonization on uptake were explored using primary mouse and human LSEC and KC cultures. When inoculated with 107 virions, >95% of the total K1Fgfp load was eliminated from the blood within 20 min, and 94% of the total retrieved K1Fgfp was localized to the liver. Higher doses resulted in slower elimination, possibly reflecting temporary saturation of liver scavenging capacity. Phage DNA was detected in both cell types, with a KC:LSEC ratio of 12:1 per population following cell isolation. Opsonization with plasma proteins increased time-dependent cellular uptake in both LSECs and KCs in vitro. Internalized phages were rapidly transported along the endocytic pathway to lysosomal compartments. Reduced viability of intracellular K1Fgfp corroborated inactivation following endocytosis. This study is the first to identify phage distribution in the liver at the hepatocellular level, confirming clearance of K1Fgfp performed mostly by KCs with a significant uptake also in LSECs.IMPORTANCEFaced with the increasing amounts of bacteria with multidrug antimicrobial resistance, phage therapy has regained attention as a possible treatment option. The phage field has recently experienced an emergence in commercial interest as research has identified new and more efficient ways of identifying and matching phages against resistant superbugs. Currently, phages are unapproved drugs in most parts of the world. For phages to reach broad clinical use, they must be shown to be clinically safe and useful. The results presented herein contribute to increased knowledge about the pharmacokinetics of the T7-like phage K1F in the mammalian system. The cell types of the liver that are responsible for rapid phage blood clearance are identified. Our results highlight the need for more research about appropriate dose regimens when phage therapy is delivered intravenously and advise essential knowledge about cell systems that should be investigated further for detailed phage pharmacodynamics.
Collapse
Affiliation(s)
| | | | - Peter McCourt
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Kim Erlend Mortensen
- Gastrointestinal Surgery Unit, University Hospital of North Norway, Tromsø, Norway
| | - Antonia P. Sagona
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | | | - Anett Kristin Larsen
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
3
|
Hincapie R, Bhattacharya S, Baksh MM, Sanhueza CA, Echeverri ES, Kim H, Paunovska K, Podilapu AR, Xu M, Dahlman JE, Finn MG. Multivalent Targeting of the Asialoglycoprotein Receptor by Virus-Like Particles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304263. [PMID: 37649182 PMCID: PMC10840735 DOI: 10.1002/smll.202304263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/16/2023] [Indexed: 09/01/2023]
Abstract
The asialoglycoprotein receptor (ASGPR) is expressed in high density on hepatocytes. Multivalent variants of galactosyl carbohydrates bind ASGPR with high affinity, enabling hepatic delivery of ligand-bound cargo. Virus-like particle (VLP) conjugates of a relatively high-affinity ligand were efficiently endocytosed by ASGPR-expressing cells in a manner strongly dependent on the nature and density of ligand display, with the best formulation using a nanomolar-, but not a picomolar-level, binder. Optimized particles were taken up by HepG2 cells with greater efficiency than competing small molecules or the natural multigalactosylated ligand, asialoorosomucoid. Upon systemic injection in mice, these VLPs were rapidly cleared to the liver and were found in association with sinusoidal endothelial cells, Kupffer cells, hepatocytes, dendritic cells, and other immune cells. Both ASGPR-targeted and nontargeted particles were distributed similarly to endothelial and Kupffer cells, but targeted particles were distributed to a greater number and fraction of hepatocytes. Thus, selective cellular trafficking in the liver is difficult to achieve: even with the most potent ASGPR targeting available, barrier cells take up much of the injected particles and hepatocytes are accessed only approximately twice as efficiently in the best case.
Collapse
Affiliation(s)
- Robert Hincapie
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Sonia Bhattacharya
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Michael M Baksh
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Carlos A Sanhueza
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Elisa Schrader Echeverri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ananda R Podilapu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Minghao Xu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - M G Finn
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| |
Collapse
|
4
|
Kyrrestad I, Larsen AK, Sánchez Romano J, Simón-Santamaría J, Li R, Sørensen KK. Infection of liver sinusoidal endothelial cells with Muromegalovirus muridbeta1 involves binding to neuropilin-1 and is dynamin-dependent. Front Cell Infect Microbiol 2023; 13:1249894. [PMID: 38029264 PMCID: PMC10665495 DOI: 10.3389/fcimb.2023.1249894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) are scavenger cells with a remarkably high capacity for clearance of several blood-borne macromolecules and nanoparticles, including some viruses. Endocytosis in LSEC is mainly via the clathrin-coated pit mediated route, which is dynamin-dependent. LSEC can also be a site of infection and latency of betaherpesvirus, but mode of virus entry into these cells has not yet been described. In this study we have investigated the role of dynamin in the early stage of muromegalovirus muridbeta1 (MuHV-1, murid betaherpesvirus 1, murine cytomegalovirus) infection in mouse LSECs. LSEC cultures were freshly prepared from C57Bl/6JRj mouse liver. We first examined dose- and time-dependent effects of two dynamin-inhibitors, dynasore and MitMAB, on cell viability, morphology, and endocytosis of model ligands via different LSEC scavenger receptors to establish a protocol for dynamin-inhibition studies in these primary cells. LSECs were challenged with MuHV-1 (MOI 0.2) ± dynamin inhibitors for 1h, then without inhibitors and virus for 11h, and nuclear expression of MuHV-1 immediate early antigen (IE1) measured by immune fluorescence. MuHV-1 efficiently infected LSECs in vitro. Infection was significantly and independently inhibited by dynasore and MitMAB, which block dynamin function via different mechanisms, suggesting that initial steps of MuHV-1 infection is dynamin-dependent in LSECs. Infection was also reduced in the presence of monensin which inhibits acidification of endosomes. Furthermore, competitive binding studies with a neuropilin-1 antibody blocked LSEC infection. This suggests that MuHV-1 infection in mouse LSECs involves virus binding to neuropilin-1 and occurs via endocytosis.
Collapse
Affiliation(s)
- Ingelin Kyrrestad
- Department of Medical Biology, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | | | | | | | | | | |
Collapse
|
5
|
Holte C, Szafranska K, Kruse L, Simon-Santamaria J, Li R, Svistounov D, McCourt P. Highly oxidized albumin is cleared by liver sinusoidal endothelial cells via the receptors stabilin-1 and -2. Sci Rep 2023; 13:19121. [PMID: 37926735 PMCID: PMC10625979 DOI: 10.1038/s41598-023-46462-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023] Open
Abstract
Oxidized albumin (oxHSA) is elevated in several pathological conditions, such as decompensated cirrhosis, acute on chronic liver failure and liver mediated renal failure. Patient derived oxidized albumin was previously shown to be an inflammatory mediator, and in normal serum levels of oxHSA are low. The removal from circulation of oxidized albumins is therefore likely required for maintenance of homeostasis. Liver sinusoidal endothelial cells (LSEC) are prominent scavenger cells specialized in removal of macromolecular waste. Given that oxidized albumin is mainly cleared by the liver, we hypothesized the LSEC are the site of uptake in the liver. In vivo oxHSA was cleared rapidly by the liver and distributed to mainly the LSEC. In in vitro studies LSEC endocytosed oxHSA much more than other cell populations isolated from the liver. Furthermore, it was shown that the uptake was mediated by the stabilins, by affinity chromatography-mass spectrometry, inhibiting uptake in LSEC with other stabilin ligands and showing uptake in HEK cells overexpressing stabilin-1 or -2. oxHSA also inhibited the uptake of other stabilin ligands, and a 2-h challenge with 100 µg/mL oxHSA reduced LSEC endocytosis by 60% up to 12 h after. Thus the LSEC and their stabilins mediate clearance of highly oxidized albumin, and oxidized albumin can downregulate their endocytic capacity in turn.
Collapse
Affiliation(s)
- Christopher Holte
- Vascular Biology Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway.
| | - Karolina Szafranska
- Vascular Biology Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Larissa Kruse
- Vascular Biology Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jaione Simon-Santamaria
- Vascular Biology Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ruomei Li
- Vascular Biology Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Dmitri Svistounov
- Metabolic and Renal Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Peter McCourt
- Vascular Biology Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
6
|
Antwi MB, Dumitriu G, Simón-Santamaria J, Romano JS, Li R, Smedsrød B, Vik A, Eskild W, Sørensen KK. Liver sinusoidal endothelial cells show reduced scavenger function and downregulation of Fc gamma receptor IIb, yet maintain a preserved fenestration in the Glmpgt/gt mouse model of slowly progressing liver fibrosis. PLoS One 2023; 18:e0293526. [PMID: 37910485 PMCID: PMC10619817 DOI: 10.1371/journal.pone.0293526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) are fenestrated endothelial cells with a unique, high endocytic clearance capacity for blood-borne waste macromolecules and colloids. This LSEC scavenger function has been insufficiently characterized in liver disease. The Glmpgt/gt mouse lacks expression of a subunit of the MFSD1/GLMP lysosomal membrane protein transporter complex, is born normal, but soon develops chronic, mild hepatocyte injury, leading to slowly progressing periportal liver fibrosis, and splenomegaly. This study examined how LSEC scavenger function and morphology are affected in the Glmpgt/gt model. FITC-labelled formaldehyde-treated serum albumin (FITC-FSA), a model ligand for LSEC scavenger receptors was administered intravenously into Glmpgt/gt mice, aged 4 months (peak of liver inflammation), 9-10 month, and age-matched Glmpwt/wt mice. Organs were harvested for light and electron microscopy, quantitative image analysis of ligand uptake, collagen accumulation, LSEC ultrastructure, and endocytosis receptor expression (also examined by qPCR and western blot). In both age groups, the Glmpgt/gt mice showed multifocal liver injury and fibrosis. The uptake of FITC-FSA in LSECs was significantly reduced in Glmpgt/gt compared to wild-type mice. Expression of LSEC receptors stabilin-1 (Stab1), and mannose receptor (Mcr1) was almost similar in liver of Glmpgt/gt mice and age-matched controls. At the same time, immunostaining revealed differences in the stabilin-1 expression pattern in sinusoids and accumulation of stabilin-1-positive macrophages in Glmpgt/gt liver. FcγRIIb (Fcgr2b), which mediates LSEC endocytosis of soluble immune complexes was widely and significantly downregulated in Glmpgt/gt liver. Despite increased collagen in space of Disse, LSECs of Glmpgt/gt mice showed well-preserved fenestrae organized in sieve plates but the frequency of holes >400 nm in diameter was increased, especially in areas with hepatocyte damage. In both genotypes, FITC-FSA also distributed to endothelial cells of spleen and bone marrow sinusoids, suggesting that these locations may function as possible compensatory sites of clearance of blood-borne scavenger receptor ligands in liver fibrosis.
Collapse
Affiliation(s)
- Milton Boaheng Antwi
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
- Section of Haematology, University Hospital of North Norway, Tromsø, Norway
| | - Gianina Dumitriu
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | | | | | - Ruomei Li
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Anders Vik
- Section of Haematology, University Hospital of North Norway, Tromsø, Norway
- Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Winnie Eskild
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | |
Collapse
|
7
|
Hincapie R, Bhattacharya S, Keshavarz-Joud P, Chapman AP, Crooke SN, Finn MG. Preparation and Biological Properties of Oligonucleotide-Functionalized Virus-like Particles. Biomacromolecules 2023. [PMID: 37257068 DOI: 10.1021/acs.biomac.3c00178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Oligonucleotides are powerful molecules for programming function and assembly. When arrayed on nanoparticle scaffolds in high density, the resulting molecules, spherical nucleic acids (SNAs), become imbued with unique properties. We used the copper-catalyzed azide-alkyne cycloaddition to graft oligonucleotides on Qβ virus-like particles to see if such structures also gain SNA-like behavior. Copper-binding ligands were shown to promote the click reaction without degrading oligonucleotide substrates. Reactions were first optimized with a small-molecule fluorogenic reporter and were then applied to the more challenging synthesis of polyvalent protein nanoparticle-oligonucleotide conjugates. The resulting particles exhibited the enhanced cellular uptake and protection from nuclease-mediated oligonucleotide cleavage characteristic of SNAs, had similar residence time in the liver relative to unmodified particles, and were somewhat shielded from immune recognition, resulting in nearly 10-fold lower antibody titers relative to unmodified particles. Oligonucleotide-functionalized virus-like particles thus provide an interesting option for protein nanoparticle-mediated delivery of functional molecules.
Collapse
|
8
|
Lučiūnaitė A, Dalgėdienė I, Vasiliūnaitė E, Norkienė M, Kučinskaitė-Kodzė I, Žvirblienė A, Gedvilaitė A. Immunogenic Properties and Antigenic Similarity of Virus-like Particles Derived from Human Polyomaviruses. Int J Mol Sci 2023; 24:ijms24054907. [PMID: 36902338 PMCID: PMC10003412 DOI: 10.3390/ijms24054907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Polyomaviruses (PyVs) are highly prevalent in humans and animals. PyVs cause mild illness, however, they can also elicit severe diseases. Some PyVs are potentially zoonotic, such as simian virus 40 (SV40). However, data are still lacking about their biology, infectivity, and host interaction with different PyVs. We investigated the immunogenic properties of virus-like particles (VLPs) derived from viral protein 1 (VP1) of human PyVs. We immunised mice with recombinant HPyV VP1 VLPs mimicking the structure of viruses and compared their immunogenicity and cross-reactivity of antisera using a broad spectrum of VP1 VLPs derived from the PyVs of humans and animals. We demonstrated a strong immunogenicity of studied VLPs and a high degree of antigenic similarity between VP1 VLPs of different PyVs. PyV-specific monoclonal antibodies were generated and applied for investigation of VLPs phagocytosis. This study demonstrated that HPyV VLPs are highly immunogenic and interact with phagocytes. Data on the cross-reactivity of VP1 VLP-specific antisera revealed antigenic similarities among VP1 VLPs of particular human and animal PyVs and suggested possible cross-immunity. As the VP1 capsid protein is the major viral antigen involved in virus-host interaction, an approach based on the use of recombinant VLPs is relevant for studying PyV biology regarding PyV interaction with the host immune system.
Collapse
|
9
|
Lambidis E, Chen CC, Baikoghli M, Imlimthan S, Khng YC, Sarparanta M, Cheng RH, Airaksinen AJ. Development of 68Ga-Labeled Hepatitis E Virus Nanoparticles for Targeted Drug Delivery and Diagnostics with PET. Mol Pharm 2022; 19:2971-2979. [PMID: 35857429 PMCID: PMC9346612 DOI: 10.1021/acs.molpharmaceut.2c00359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
Targeted delivery of diagnostics and therapeutics offers
essential
advantages over nontargeted systemic delivery. These include the reduction
of toxicity, the ability to reach sites beyond biological barriers,
and the delivery of higher cargo concentrations to diseased sites.
Virus-like particles (VLPs) can efficiently be used for targeted delivery
purposes. VLPs are derived from the coat proteins of viral capsids.
They are self-assembled, biodegradable, and homogeneously distributed.
In this study, hepatitis E virus (HEV) VLP derivatives, hepatitis
E virus nanoparticles (HEVNPs), were radiolabeled with gallium-68,
and consequently, the biodistribution of the labeled [68Ga]Ga-DOTA-HEVNPs was studied in mice. The results indicated that
[68Ga]Ga-DOTA-HEVNPs can be considered as promising theranostic
nanocarriers, especially for hepatocyte-targeting therapies.
Collapse
Affiliation(s)
- Elisavet Lambidis
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Chun-Chieh Chen
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616, United States
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616, United States
| | - Surachet Imlimthan
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - You Cheng Khng
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Mirkka Sarparanta
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, California 95616, United States
| | - Anu J Airaksinen
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland.,Turku PET Centre, Department of Chemistry, University of Turku, Turku FI-20520, Finland
| |
Collapse
|
10
|
Ander SE, Li FS, Carpentier KS, Morrison TE. Innate immune surveillance of the circulation: A review on the removal of circulating virions from the bloodstream. PLoS Pathog 2022; 18:e1010474. [PMID: 35511797 PMCID: PMC9070959 DOI: 10.1371/journal.ppat.1010474] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Many viruses utilize the lymphohematogenous route for dissemination; however, they may not freely use this highway unchecked. The reticuloendothelial system (RES) is an innate defense system that surveys circulating blood, recognizing and capturing viral particles. Examination of the literature shows that the bulk of viral clearance is mediated by the liver; however, the precise mechanism(s) mediating viral vascular clearance vary between viruses and, in many cases, remains poorly defined. Herein, we summarize what is known regarding the recognition and capture of virions from the circulation prior to the generation of a specific antibody response. We also discuss the consequences of viral capture on viral pathogenesis and the fate of the captor cell. Finally, this understudied topic has implications beyond viral pathogenesis, including effects on arbovirus ecology and the application of virus-vectored gene therapies.
Collapse
Affiliation(s)
- Stephanie E. Ander
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Frances S. Li
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Kathryn S. Carpentier
- Department of Natural Sciences, Greensboro College, Greensboro, North Carolina, United States of America
| | - Thomas E. Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
11
|
Abstract
Intestinal microbiota, dominated by bacteria, plays an important role in the occurrence and the development of alcohol-associated liver disease (ALD), which is one of the most common liver diseases around the world. With sufficient studies focusing on the gut bacterial community, chronic alcohol consumption is now known as a key factor that alters the composition of gut bacterial community, increases intestinal permeability, causes intestinal dysfunction, induces bacterial translocation, and exacerbates the process of ALD via gut-liver axis. However, gut non-bacterial communities including fungi, viruses, and archaea, which may also participate in the disease, has received little attention relative to the gut bacterial community. This paper will systematically collect the latest literatures reporting non-bacterial communities in mammalian health and disease, and review their mechanisms in promoting the development of ALD including CLEC7A pathway, Candidalysin (a peptide toxin secreted by Candida albicans), metabolites, and other chemical substances secreted or regulated by gut commensal mycobiome, virome, and archaeome, hoping to bring novel insights on our current knowledge of ALD.
Collapse
Affiliation(s)
- Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yixin Zhu
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Jin Ye
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,CONTACT Huikuan Chu Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022, China
| |
Collapse
|
12
|
Hunt NJ, McCourt PAG, Kuncic Z, Le Couteur DG, Cogger VC. Opportunities and Challenges for Nanotherapeutics for the Aging Population. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.832524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nanotherapeutics utilize the properties of nanomaterials to alter the pharmacology of the drugs and therapies being transported, leading to changes in their biological disposition (absorption, distribution, cellular uptake, metabolism and elimination) and ultimately, their pharmacological effect. This provides an opportunity to optimize the pharmacology of drugs, particularly for those that are dependent on hepatic action. Old age is associated with changes in many pharmacokinetic processes which tend to impair drug efficacy and increase risk of toxicity. While these age-related changes are drug-specific they could be directly addressed using nanotechnology and precision targeting. The benefits of nanotherapeutics needs to be balanced against toxicity, with future use in humans dependent upon the gathering of information about the clearance and long-term safety of nanomaterials.
Collapse
|
13
|
Butola A, Coucheron DA, Szafranska K, Ahmad A, Mao H, Tinguely JC, McCourt P, Senthilkumaran P, Mehta DS, Agarwal K, Ahluwalia BS. Multimodal on-chip nanoscopy and quantitative phase imaging reveals the nanoscale morphology of liver sinusoidal endothelial cells. Proc Natl Acad Sci U S A 2021; 118:e2115323118. [PMID: 34782474 PMCID: PMC8617407 DOI: 10.1073/pnas.2115323118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/26/2022] Open
Abstract
Visualization of three-dimensional (3D) morphological changes in the subcellular structures of a biological specimen is a major challenge in life science. Here, we present an integrated chip-based optical nanoscopy combined with quantitative phase microscopy (QPM) to obtain 3D morphology of liver sinusoidal endothelial cells (LSEC). LSEC have unique morphology with small nanopores (50-300 nm in diameter) in the plasma membrane, called fenestrations. The fenestrations are grouped in discrete clusters, which are around 100 to 200 nm thick. Thus, imaging and quantification of fenestrations and sieve plate thickness require resolution and sensitivity of sub-100 nm along both the lateral and the axial directions, respectively. In chip-based nanoscopy, the optical waveguides are used both for hosting and illuminating the sample. The fluorescence signal is captured by an upright microscope, which is converted into a Linnik-type interferometer to sequentially acquire both superresolved images and phase information of the sample. The multimodal microscope provided an estimate of the fenestration diameter of 119 ± 53 nm and average thickness of the sieve plates of 136.6 ± 42.4 nm, assuming the constant refractive index of cell membrane to be 1.38. Further, LSEC were treated with cytochalasin B to demonstrate the possibility of precise detection in the cell height. The mean phase value of the fenestrated area in normal and treated cells was found to be 161 ± 50 mrad and 109 ± 49 mrad, respectively. The proposed multimodal technique offers nanoscale visualization of both the lateral size and the thickness map, which would be of broader interest in the fields of cell biology and bioimaging.
Collapse
Affiliation(s)
- Ankit Butola
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
- Bio-photonics and Green Photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - David A Coucheron
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Karolina Szafranska
- Faculty of Health Sciences, Department of Medical Biology, Vascular Biology Research Group, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Azeem Ahmad
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Hong Mao
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Jean-Claude Tinguely
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Peter McCourt
- Faculty of Health Sciences, Department of Medical Biology, Vascular Biology Research Group, UiT The Arctic University of Norway, Tromsø 9037, Norway
| | - Paramasivam Senthilkumaran
- Bio-photonics and Green Photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Dalip Singh Mehta
- Bio-photonics and Green Photonics Laboratory, Department of Physics, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Krishna Agarwal
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway
| | - Balpreet Singh Ahluwalia
- Department of Physics and Technology, Universitetet i Tromsø (UiT) The Arctic University of Norway, 9037 Tromsø, Norway;
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
14
|
Bhandari S, Larsen AK, McCourt P, Smedsrød B, Sørensen KK. The Scavenger Function of Liver Sinusoidal Endothelial Cells in Health and Disease. Front Physiol 2021; 12:757469. [PMID: 34707514 PMCID: PMC8542980 DOI: 10.3389/fphys.2021.757469] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
The aim of this review is to give an outline of the blood clearance function of the liver sinusoidal endothelial cells (LSECs) in health and disease. Lining the hundreds of millions of hepatic sinusoids in the human liver the LSECs are perfectly located to survey the constituents of the blood. These cells are equipped with high-affinity receptors and an intracellular vesicle transport apparatus, enabling a remarkably efficient machinery for removal of large molecules and nanoparticles from the blood, thus contributing importantly to maintain blood and tissue homeostasis. We describe here central aspects of LSEC signature receptors that enable the cells to recognize and internalize blood-borne waste macromolecules at great speed and high capacity. Notably, this blood clearance system is a silent process, in the sense that it usually neither requires or elicits cell activation or immune responses. Most of our knowledge about LSECs arises from studies in animals, of which mouse and rat make up the great majority, and some species differences relevant for extrapolating from animal models to human are discussed. In the last part of the review, we discuss comparative aspects of the LSEC scavenger functions and specialized scavenger endothelial cells (SECs) in other vascular beds and in different vertebrate classes. In conclusion, the activity of LSECs and other SECs prevent exposure of a great number of waste products to the immune system, and molecules with noxious biological activities are effectively “silenced” by the rapid clearance in LSECs. An undesired consequence of this avid scavenging system is unwanted uptake of nanomedicines and biologics in the cells. As the development of this new generation of therapeutics evolves, there will be a sharp increase in the need to understand the clearance function of LSECs in health and disease. There is still a significant knowledge gap in how the LSEC clearance function is affected in liver disease.
Collapse
Affiliation(s)
- Sabin Bhandari
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Anett Kristin Larsen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Peter McCourt
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Bård Smedsrød
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| | - Karen Kristine Sørensen
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø (UiT) - The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
15
|
Wang Y, Liu Y. Gut-liver-axis: Barrier function of liver sinusoidal endothelial cell. J Gastroenterol Hepatol 2021; 36:2706-2714. [PMID: 33811372 DOI: 10.1111/jgh.15512] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/02/2021] [Accepted: 03/27/2021] [Indexed: 12/15/2022]
Abstract
Liver diseases are associated with the leaky gut via the gut-liver-axis. Previous studies have paid much attention to the effect of gut barrier damage. Notably, clinical observations and basic research reveal that the gut barrier damage seldom leads to liver injury independently but aggravates pre-existing liver diseases such as non-alcoholic fatty liver disease and drug-induced liver injury. These evidences suggest that there is a hepatic barrier in the gut-liver-axis, protecting the liver against gut-derived pathogenic factors. However, it has never been investigated which type of liver cell plays the role of hepatic barrier. Under physiological conditions, liver sinusoidal endothelial cell (LSEC) can take up and eliminate virus, bacteriophage, microbial products, and metabolic wastes. LSEC also keeps the homeostasis of liver immune environment via tolerance-inducing and anti-inflammatory functions. In contrast, under pathological conditions, the clearance function of LSEC is impaired, and LSEC turns into a pro-inflammatory pattern. Given its anatomical position and physiological functions, LSEC is proposed as the hepatic barrier in the gut-liver-axis. In this review, we aim to further understand the role of LSEC as the hepatic barrier. Future studies are warranted to seek effective treatments to improve LSEC health, which appears to be a promising approach to prevent gut-derived liver injury.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, China
| |
Collapse
|
16
|
Szafranska K, Holte CF, Kruse LD, Mao H, Øie CI, Szymonski M, Zapotoczny B, McCourt PAG. Quantitative analysis methods for studying fenestrations in liver sinusoidal endothelial cells. A comparative study. Micron 2021; 150:103121. [PMID: 34560521 DOI: 10.1016/j.micron.2021.103121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/01/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022]
Abstract
Liver Sinusoidal Endothelial Cells (LSEC) line the hepatic vasculature providing blood filtration via transmembrane nanopores called fenestrations. These structures are 50-300 nm in diameter, which is below the resolution limit of a conventional light microscopy. To date, there is no standardized method of fenestration image analysis. With this study, we provide and compare three different approaches: manual measurements, a semi-automatic (threshold-based) method, and an automatic method based on user-friendly open source machine learning software. Images were obtained using three super resolution techniques - atomic force microscopy (AFM), scanning electron microscopy (SEM), and structured illumination microscopy (SIM). Parameters describing fenestrations such as diameter, area, roundness, frequency, and porosity were measured. Finally, we studied the user bias by comparison of the data obtained by five different users applying provided analysis methods.
Collapse
Affiliation(s)
- K Szafranska
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway; Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland.
| | - C F Holte
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway
| | - L D Kruse
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway
| | - H Mao
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway
| | - C I Øie
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway
| | - M Szymonski
- Centre for Nanometer-Scale Science and Advanced Materials, NANOSAM, Faculty of Physics, Astronomy, and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - B Zapotoczny
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway; Institute of Nuclear Physics, Polish Academy of Sciences, 31-342, Krakow, Poland
| | - P A G McCourt
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT), The Arctic University of Norway, Norway
| |
Collapse
|
17
|
Huisman Y, Uphoff K, Berger M, Dobrindt U, Schelhaas M, Zobel T, Bussmann J, van Impel A, Schulte-Merker S. Meningeal lymphatic endothelial cells fulfill scavenger endothelial cell function and cooperate with microglia in waste removal from the brain. Glia 2021; 70:35-49. [PMID: 34487573 DOI: 10.1002/glia.24081] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 12/13/2022]
Abstract
Brain lymphatic endothelial cells (BLECs) constitute a group of loosely connected endothelial cells that reside within the meningeal layer of the zebrafish brain without forming a vascular tubular system. BLECs have been shown to readily endocytose extracellular cargo molecules from the brain parenchyma, however, their functional relevance in relation to microglia remains enigmatic. We here compare their functional uptake efficiency for several macromolecules and bacterial components with microglia in a qualitative and quantitative manner in 5-day-old zebrafish embryos. We find BLECs to be significantly more effective in the uptake of proteins, polysaccharides and virus particles as compared to microglia, while larger particles like bacteria are only ingested by microglia but not by BLECs, implying a clear distribution of tasks between the two cell types in the brain area. In addition, we compare BLECs to the recently discovered scavenger endothelial cells (SECs) of the cardinal vein and find them to accept an identical set of substrate molecules. Our data identifies BLECs as the first brain-associated SEC population in vertebrates, and demonstrates that BLECs cooperate with microglia to remove particle waste from the brain.
Collapse
Affiliation(s)
- Yvonne Huisman
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Katharina Uphoff
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany
| | | | | | - Mario Schelhaas
- Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany.,Institute of Cellular Virology, ZMBE, Münster, Germany
| | - Thomas Zobel
- Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany.,Imaging Network, Cells in Motion Interfaculty Centre, WWU Münster, Germany
| | - Jeroen Bussmann
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333CC, The Netherlands
| | - Andreas van Impel
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, WWU Münster, Münster, Germany.,Faculty of Medicine, WWU Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence, WWU Münster, Münster, Germany
| |
Collapse
|
18
|
Rohn B, Jansing W, Seibert FS, Pfab T, Cinkilic O, Paßfall J, Schmidt S, Babel N, Bauer F, Westhoff TH. Association of hyperuricemia and serum uric acid lowering therapy with mortality in hemodialysis patients. Ren Fail 2021; 42:1067-1075. [PMID: 33076736 PMCID: PMC7594866 DOI: 10.1080/0886022x.2020.1835674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Introduction In the general population, hyperuricemia is associated with increased morbidity and mortality. Data on this association in hemodialysis patients is controversial. Moreover, it remains elusive whether serum uric acid (SUA) lowering therapy is associated with mortality. Methods Retrospective analysis of 601 patients on chronic hemodialysis therapy in five outpatient centers with a maximum follow-up of 100 and a mean follow-up of 41 months. Death was defined as primary endpoint. Cumulative survival was analyzed by Kaplan–Meier analysis and Cox regressions adjusted for age. Findings Cumulative survival rates were higher for those subjects with a higher than median SUA concentration both based on mean annual and baseline measurements (p < 0.05 each). There was no survival difference anymore after adjustment for age (p > 0.05 each). Stratification for SUA lowering therapy (allopurinol/febuxostat) had no impact on cumulative survival, neither in Kaplan Meier nor in Cox regression analyses (p > 0.05 each). Furthermore, Cox regression analysis excluded an increased cardiovascular mortality in subjects with hyperuricemia. Discussion In contrast to the general population, hyperuricemia is not associated with increased mortality in patients undergoing hemodialysis. Moreover, xanthine oxidase inhibition was not associated with a survival benefit in this analysis. These data do not support the use of SUA lowering medication in hemodialysis patients with asymptomatic hyperuricemia.
Collapse
Affiliation(s)
- Benjamin Rohn
- Medical Department I, Universitätsklinikum Marien Hospital Herne, Ruhr-University, Bochum, Germany
| | - Wiebke Jansing
- Medical Department I, Universitätsklinikum Marien Hospital Herne, Ruhr-University, Bochum, Germany
| | - Felix S Seibert
- Medical Department I, Universitätsklinikum Marien Hospital Herne, Ruhr-University, Bochum, Germany
| | | | | | | | | | - Nina Babel
- Medical Department I, Universitätsklinikum Marien Hospital Herne, Ruhr-University, Bochum, Germany
| | - Frederic Bauer
- Medical Department I, Universitätsklinikum Marien Hospital Herne, Ruhr-University, Bochum, Germany
| | - Timm H Westhoff
- Medical Department I, Universitätsklinikum Marien Hospital Herne, Ruhr-University, Bochum, Germany
| |
Collapse
|
19
|
Ye D, Zimmermann T, Demina V, Sotnikov S, Ried CL, Rahn H, Stapf M, Untucht C, Rohe M, Terstappen GC, Wicke K, Mezler M, Manninga H, Meyer AH. Trafficking of JC virus-like particles across the blood-brain barrier. NANOSCALE ADVANCES 2021; 3:2488-2500. [PMID: 36134165 PMCID: PMC9418390 DOI: 10.1039/d0na00879f] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/01/2021] [Indexed: 06/10/2023]
Abstract
Hollow viral vectors, such as John Cunningham virus-like particles (JC VLPs), provide a unique opportunity to deliver drug cargo into targeted cells and tissue. Current understanding of the entry of JC virus in brain cells has remained insufficient. In particular, interaction of JC VLPs with the blood-brain barrier (BBB) has not been analyzed in detail. Thus, JC VLPs were produced in this study for investigating the trafficking across the BBB. We performed a carotid artery injection procedure for mouse brain to qualitatively study JC VLPs' in vivo binding and distribution and used in vitro approaches to analyze their uptake and export kinetics in brain endothelial cells. Our results show that clathrin-dependent mechanisms contributed to the entry of VLPs into brain endothelial cells, and exocytosis or transcytosis of VLPs across the BBB was observed in vitro. VLPs were found to interact with sialic acid glycans in mouse brain endothelia. The ability of JC VLPs to cross the BBB can be useful in developing a delivery system for transport of genes and small molecule cargoes to the brain.
Collapse
Affiliation(s)
- Dong Ye
- AbbVie Deutschland GmbH & Co. KG, DMPK, Bioanalytical Research Knollstraße 67061 Ludwigshafen Germany
| | - Tina Zimmermann
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery Knollstraße 67061 Ludwigshafen Germany
| | | | | | - Christian L Ried
- AbbVie Deutschland GmbH & Co. KG, Development Sciences NBE Knollstraße 67061 Ludwigshafen Germany
| | - Harri Rahn
- AbbVie Deutschland GmbH & Co. KG, Development Sciences NBE Knollstraße 67061 Ludwigshafen Germany
| | - Marcus Stapf
- NEUWAY Pharma GmbH Ludwig-Erhard-Allee 2 53175 Bonn Germany
| | - Christopher Untucht
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery Knollstraße 67061 Ludwigshafen Germany
| | - Michael Rohe
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery Knollstraße 67061 Ludwigshafen Germany
| | - Georg C Terstappen
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery Knollstraße 67061 Ludwigshafen Germany
| | - Karsten Wicke
- AbbVie Deutschland GmbH & Co. KG, Neuroscience Discovery Knollstraße 67061 Ludwigshafen Germany
| | - Mario Mezler
- AbbVie Deutschland GmbH & Co. KG, DMPK, Bioanalytical Research Knollstraße 67061 Ludwigshafen Germany
| | - Heiko Manninga
- NEUWAY Pharma GmbH Ludwig-Erhard-Allee 2 53175 Bonn Germany
| | - Axel H Meyer
- AbbVie Deutschland GmbH & Co. KG, DMPK, Bioanalytical Research Knollstraße 67061 Ludwigshafen Germany
| |
Collapse
|
20
|
Angiodiversity and organotypic functions of sinusoidal endothelial cells. Angiogenesis 2021; 24:289-310. [PMID: 33745018 PMCID: PMC7982081 DOI: 10.1007/s10456-021-09780-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 03/04/2021] [Indexed: 02/08/2023]
Abstract
‘Angiodiversity’ refers to the structural and functional heterogeneity of endothelial cells (EC) along the segments of the vascular tree and especially within the microvascular beds of different organs. Organotypically differentiated EC ranging from continuous, barrier-forming endothelium to discontinuous, fenestrated endothelium perform organ-specific functions such as the maintenance of the tightly sealed blood–brain barrier or the clearance of macromolecular waste products from the peripheral blood by liver EC-expressed scavenger receptors. The microvascular bed of the liver, composed of discontinuous, fenestrated liver sinusoidal endothelial cells (LSEC), is a prime example of organ-specific angiodiversity. Anatomy and development of LSEC have been extensively studied by electron microscopy as well as linage-tracing experiments. Recent advances in cell isolation and bulk transcriptomics or single-cell RNA sequencing techniques allowed the identification of distinct LSEC molecular programs and have led to the identification of LSEC subpopulations. LSEC execute homeostatic functions such as fine tuning the vascular tone, clearing noxious substances from the circulation, and modulating immunoregulatory mechanisms. In recent years, the identification and functional analysis of LSEC-derived angiocrine signals, which control liver homeostasis and disease pathogenesis in an instructive manner, marks a major change of paradigm in the understanding of liver function in health and disease. This review summarizes recent advances in the understanding of liver vascular angiodiversity and the functional consequences resulting thereof.
Collapse
|
21
|
Strategies for delivering therapeutics across the blood-brain barrier. Nat Rev Drug Discov 2021; 20:362-383. [PMID: 33649582 DOI: 10.1038/s41573-021-00139-y] [Citation(s) in RCA: 424] [Impact Index Per Article: 141.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2021] [Indexed: 02/06/2023]
Abstract
Achieving sufficient delivery across the blood-brain barrier is a key challenge in the development of drugs to treat central nervous system (CNS) disorders. This is particularly the case for biopharmaceuticals such as monoclonal antibodies and enzyme replacement therapies, which are largely excluded from the brain following systemic administration. In recent years, increasing research efforts by pharmaceutical and biotechnology companies, academic institutions and public-private consortia have resulted in the evaluation of various technologies developed to deliver therapeutics to the CNS, some of which have entered clinical testing. Here we review recent developments and challenges related to selected blood-brain barrier-crossing strategies - with a focus on non-invasive approaches such as receptor-mediated transcytosis and the use of neurotropic viruses, nanoparticles and exosomes - and analyse their potential in the treatment of CNS disorders.
Collapse
|
22
|
De Smedt J, van Os EA, Talon I, Ghosh S, Toprakhisar B, Furtado Madeiro Da Costa R, Zaunz S, Vazquez MA, Boon R, Baatsen P, Smout A, Verhulst S, van Grunsven LA, Verfaillie CM. PU.1 drives specification of pluripotent stem cell-derived endothelial cells to LSEC-like cells. Cell Death Dis 2021; 12:84. [PMID: 33446637 PMCID: PMC7809369 DOI: 10.1038/s41419-020-03356-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/25/2022]
Abstract
To date, there is no representative in vitro model for liver sinusoidal endothelial cells (LSECs), as primary LSECs dedifferentiate very fast in culture and no combination of cytokines or growth factors can induce an LSEC fate in (pluripotent stem cell (PSC)-derived) endothelial cells (ECs). Furthermore, the transcriptional programmes driving an LSEC fate have not yet been described. Here, we first present a computational workflow (CenTFinder) that can identify transcription factors (TFs) that are crucial for modulating pathways involved in cell lineage specification. Using CenTFinder, we identified several novel LSEC-specific protein markers, such as FCN2 and FCN3, which were validated by analysis of previously published single-cell RNAseq data. We also identified PU.1 (encoded by the SPI1 gene) as a major regulator of LSEC-specific immune functions. We show that SPI1 overexpression (combined with the general EC TF ETV2) in human PSCs induces ECs with an LSEC-like phenotype. The ETV2-SPI1-ECs display increased expression of LSEC markers, such as CD32B and MRC1, as well as several of the proposed novel markers. More importantly, ETV2-SPI1-ECs acquire LSEC functions, including uptake of FSA-FITC, as well as labelled IgG. In conclusion, we present the CenTFinder computational tool to identify key regulatory TFs within specific pathways, in this work pathways of lineage specification, and we demonstrate its use by the identification and validation of PU.1 as a master regulator for LSEC fating.
Collapse
Affiliation(s)
- Jonathan De Smedt
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium.
| | - Elise Anne van Os
- Liver Cell Biology research group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Irene Talon
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Sreya Ghosh
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Burak Toprakhisar
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | | | - Samantha Zaunz
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Marta Aguirre Vazquez
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Ruben Boon
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, 02114, USA.,The Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Pieter Baatsen
- Electron Microscopy Platform of VIB Bio Imaging Core at KU Leuven and VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Ayla Smout
- Liver Cell Biology research group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology research group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology research group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Bhandari S, Li R, Simón-Santamaría J, McCourt P, Johansen SD, Smedsrød B, Martinez-Zubiaurre I, Sørensen KK. Transcriptome and proteome profiling reveal complementary scavenger and immune features of rat liver sinusoidal endothelial cells and liver macrophages. BMC Mol Cell Biol 2020; 21:85. [PMID: 33246411 PMCID: PMC7694354 DOI: 10.1186/s12860-020-00331-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs; liver resident macrophages) form the body's most effective scavenger cell system for the removal of harmful blood-borne substances, ranging from modified self-proteins to pathogens and xenobiotics. Controversies in the literature regarding the LSEC phenotype pose a challenge when determining distinct functionalities of KCs and LSECs. This may be due to overlapping functions of the two cells, insufficient purification and/or identification of the cells, rapid dedifferentiation of LSECs in vitro, or species differences. We therefore characterized and quantitatively compared expressed gene products of freshly isolated, highly pure LSECs (fenestrated SE-1/FcγRIIb2+) and KCs (CD11b/c+) from Sprague Dawley, Crl:CD (SD), male rats using high throughput mRNA-sequencing and label-free proteomics. RESULTS We observed a robust correlation between the proteomes and transcriptomes of the two cell types. Integrative analysis of the global molecular profile demonstrated the immunological aspects of LSECs. The constitutive expression of several immune genes and corresponding proteins of LSECs bore some resemblance with the expression in macrophages. LSECs and KCs both expressed high levels of scavenger receptors (SR) and C-type lectins. Equivalent expression of SR-A1 (Msr1), mannose receptor (Mrc1), SR-B1 (Scarb1), and SR-B3 (Scarb2) suggested functional similarity between the two cell types, while functional distinction between the cells was evidenced by LSEC-specific expression of the SRs stabilin-1 (Stab1) and stabilin-2 (Stab2), and the C-type lectins LSECtin (Clec4g) and DC-SIGNR (Clec4m). Many immune regulatory factors were differentially expressed in LSECs and KCs, with one cell predominantly expressing a specific cytokine/chemokine and the other cell the cognate receptor, illustrating the complex cytokine milieu of the sinusoids. Both cells expressed genes and proteins involved in antigen processing and presentation, and lymphocyte co-stimulation. CONCLUSIONS Our findings support complementary and partly overlapping scavenging and immune functions of LSECs and KCs. This highlights the importance of including LSECs in studies of liver immunity, and liver clearance and toxicity of large molecule drugs and nano-formulations.
Collapse
Affiliation(s)
- Sabin Bhandari
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway
| | - Ruomei Li
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway
| | - Jaione Simón-Santamaría
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway
| | - Peter McCourt
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway
| | - Steinar Daae Johansen
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway.,Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Bård Smedsrød
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway.
| | | | - Karen Kristine Sørensen
- Department of Medical Biology, Vascular Biology Research Group, University of Tromsø (UiT) -The Arctic University of Norway, Hansine Hansens veg 18, N-9037, Tromsø, Norway
| |
Collapse
|
24
|
BK polyomavirus-specific antibody and T-cell responses in kidney transplantation: update. Curr Opin Infect Dis 2020; 32:575-583. [PMID: 31567736 DOI: 10.1097/qco.0000000000000602] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW BK polyomavirus (BKPyV) has emerged as a significant cause of premature graft failure after kidney transplantation. Without effective antiviral drugs, treatment is based on reducing immunosuppression to regain immune control over BKPyV replication. The paradigm of high-level viruria/decoy cells, BKPyV-DNAemia, and proven nephropathy permits early interventions. Here, we review recent findings about BKPyV-specific antibody and T-cell responses and their potential role in risk stratification, immune monitoring, and therapy. RECENT FINDING Kidney transplant recipients having low or undetectable BKPyV-specific IgG immunoglobulin G (IgG) are higher risk for developing BKPyV-DNAemia if the donor has high BKPyV-specific IgG. This observation has been extended to neutralizing antibodies. Immunosuppression, impaired activation, proliferation, and exhaustion of BKPyV-specific T cells may increase the risk of developing BKPyV-DNAemia and nephropathy. Clearance of BKPyV-DNAemia was correlated with high CD8 T cell responses to human leukocyte antigen (HLA)-types presenting BKPyV-encoded immunodominant 9mers. For clinical translation, these data need to be assessed in appropriately designed clinical studies, as outlined in recent guidelines on BKPyV in kidney transplantation. SUMMARY Evaluation of BKPyV-specific immune responses in recipient and donor may help to stratify the risk of BKPyV-DNAemia, nephropathy, and graft loss. Future efforts need to evaluate clinical translation, vaccines, and immunotherapy to control BKPyV replication.
Collapse
|
25
|
Mannemuddhu S, Pekkucuksen N, Bush R, Johns F, Upadhyay K. Transplant renal artery stenosis in a child with BK nephropathy. Pediatr Transplant 2020; 24:e13629. [PMID: 31815337 PMCID: PMC7167878 DOI: 10.1111/petr.13629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/23/2019] [Accepted: 11/08/2019] [Indexed: 12/29/2022]
Abstract
TRAS and BK nephropathy are known complications of RT, but the association between both has not been reported. A 2-year-old girl underwent a deceased donor renal transplant from a 20-year-old donor, along with bilateral native nephrectomies. She had a DGF due to a renal artery thrombus and required thrombectomy with re-anastomosis. Heparin and aspirin were used. Immunosuppressive agents included thymoglobulin, steroid, tacrolimus, and MMF. CMV and EBV DNA PCRs were negative, but she developed BK viremia at 2 months with stable allograft function. Immunosuppression was reduced, and leflunomide was initiated. Blood pressures were well controlled on low-dose amlodipine. Five months after RT, she presented with hypertensive emergency, following a respiratory infection, and required dialysis for oliguric acute kidney injury. Allograft biopsy showed evidence of BK nephropathy. Immunosuppression was further minimized. Doppler renal US and renal artery duplex studies were both suggestive of TRAS. Angiogram showed severe proximal anastomotic TRAS (>95% occlusion). PTA with stenting was done with immediate improvement in the blood flow and reduction in the pressure gradient. BPs and renal function normalized. Ten months post-RT, she remains normotensive with stable renal function and resolution of BK viremia. Although ureteral stenosis and nephropathy are known to occur with BK infection, TRAS is an interesting association and possibly suggest the tropism of BK virus to the vascular endothelial cells. Timely recognition and management of both is important to prevent uncontrolled hypertension and allograft dysfunction.
Collapse
Affiliation(s)
- Sudha Mannemuddhu
- Division of Pediatric NephrologyDepartment of PediatricsUniversity of FloridaGainesvilleFlorida
| | - Naile Pekkucuksen
- Division of Pediatric NephrologyDepartment of PediatricsUniversity of FloridaGainesvilleFlorida
| | - Rachel Bush
- Division of Pediatric NephrologyDepartment of PediatricsUniversity of FloridaGainesvilleFlorida
| | - Felicia Johns
- Division of Pediatric NephrologyDepartment of PediatricsUniversity of FloridaGainesvilleFlorida
| | - Kiran Upadhyay
- Division of Pediatric NephrologyDepartment of PediatricsUniversity of FloridaGainesvilleFlorida
| |
Collapse
|
26
|
Liver sinusoidal endothelial cells contribute to the uptake and degradation of entero bacterial viruses. Sci Rep 2020; 10:898. [PMID: 31965000 PMCID: PMC6972739 DOI: 10.1038/s41598-020-57652-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 12/18/2019] [Indexed: 01/01/2023] Open
Abstract
The liver is constantly exposed to dietary antigens, viruses, and bacterial products with inflammatory potential. For decades cellular uptake of virus has been studied in connection with infection, while the few studies designed to look into clearance mechanisms focused mainly on the role of macrophages. In recent years, attention has been directed towards the liver sinusoidal endothelial cells (LSECs), which play a central role in liver innate immunity by their ability to scavenge pathogen- and damage-associated molecular patterns. Every day our bodies are exposed to billions of gut-derived pathogens which must be efficiently removed from the circulation to prevent inflammatory and/or immune reactions in other vascular beds. Here, we have used GFP-labelled Enterobacteria phage T4 (GFP-T4-phage) as a model virus to study the viral scavenging function and metabolism in LSECs. The uptake of GFP-T4-phages was followed in real-time using deconvolution microscopy, and LSEC identity confirmed by visualization of fenestrae using structured illumination microscopy. By combining these imaging modalities with quantitative uptake and inhibition studies of radiolabelled GFP-T4-phages, we demonstrate that the bacteriophages are effectively degraded in the lysosomal compartment. Due to their high ability to take up and degrade circulating bacteriophages the LSECs may act as a primary anti-viral defence mechanism.
Collapse
|
27
|
Hirsch HH, Randhawa PS. BK polyomavirus in solid organ transplantation-Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 2019; 33:e13528. [PMID: 30859620 DOI: 10.1111/ctr.13528] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/26/2019] [Indexed: 02/07/2023]
Abstract
The present AST-IDCOP guidelines update information on BK polyomavirus (BKPyV) infection, replication, and disease, which impact kidney transplantation (KT), but rarely non-kidney solid organ transplantation (SOT). As pretransplant risk factors in KT donors and recipients presently do not translate into clinically validated measures regarding organ allocation, antiviral prophylaxis, or screening, all KT recipients should be screened for BKPyV-DNAemia monthly until month 9, and then every 3 months until 2 years posttransplant. Extended screening after 2 years may be considered in pediatric KT. Stepwise immunosuppression reduction is recommended for KT patients with plasma BKPyV-DNAemia of >1000 copies/mL sustained for 3 weeks or increasing to >10 000 copies/mL reflecting probable and presumptive BKPyV-associated nephropathy, respectively. Reducing immunosuppression is also the primary intervention for biopsy-proven BKPyV-associated nephropathy. Hence, allograft biopsy is not required for treating BKPyV-DNAemic patients with baseline renal function. Despite virological rationales, proper randomized clinical trials are lacking to generally recommend treatment by switching from tacrolimus to cyclosporine-A, from mycophenolate to mTOR inhibitors or leflunomide or by the adjunct use of intravenous immunoglobulins, leflunomide, or cidofovir. Fluoroquinolones are not recommended for prophylaxis or therapy. Retransplantation after allograft loss due to BKPyV nephropathy can be successful if BKPyV-DNAemia is definitively cleared, independent of failed allograft nephrectomy.
Collapse
Affiliation(s)
- Hans H Hirsch
- Transplantation & Clinical Virology, Department of Biomedicine, University of Basel, Basel, Switzerland.,Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Parmjeet S Randhawa
- Division of Transplantation Pathology, Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Thomas E Starzl Transplantation Institute, Pittsburgh, Pennsylvania
| | | |
Collapse
|
28
|
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: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
29
|
An P, Sáenz Robles MT, Duray AM, Cantalupo PG, Pipas JM. Human polyomavirus BKV infection of endothelial cells results in interferon pathway induction and persistence. PLoS Pathog 2019; 15:e1007505. [PMID: 30620752 PMCID: PMC6338385 DOI: 10.1371/journal.ppat.1007505] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/18/2019] [Accepted: 12/05/2018] [Indexed: 01/19/2023] Open
Abstract
Polyomavirus BKV is highly prevalent among humans. The virus establishes an asymptomatic persistent infection in the urinary system in healthy people, but uncontrolled productive infection of the virus in immunocompromised patients can lead to serious diseases. In spite of its high prevalence, our knowledge regarding key aspects of BKV polyomavirus infection remains incomplete. To determine tissue and cell type tropism of the virus, primary human epithelial cells, endothelial cells and fibroblasts isolated from the respiratory and urinary systems were tested. Results from this study demonstrated that all 9 different types of human cells were infectable by BKV polyomavirus but showed differential cellular responses. In microvascular endothelial cells from the lung and the bladder, BKV persistent infection led to prolonged viral protein expression, low yield of infectious progeny and delayed cell death, in contrast with infection in renal proximal tubular epithelial cells, a widely used cell culture model for studying productive infection of this virus. Transcriptomic profiling revealed the activation of interferon signaling and induction of multiple interferon stimulated genes in infected microvascular endothelial cells. Further investigation demonstrated production of IFNβ and secretion of chemokine CXCL10 by infected endothelial cells. Activation of IRF3 and STAT1 in infected endothelial cells was also confirmed. In contrast, renal proximal tubular epithelial cells failed to mount an interferon response and underwent progressive cell death. These results demonstrated that microvascular endothelial cells are able to activate interferon signaling in response to polyomavirus BKV infection. This raises the possibility that endothelial cells might provide initial immune defense against BKV infection. Our results shed light on the persistence of and immunity against infection by BKV polyomavirus.
Collapse
Affiliation(s)
- Ping An
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Maria Teresa Sáenz Robles
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Alexis M. Duray
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Paul G. Cantalupo
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - James M. Pipas
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
30
|
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: 4.2] [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
|
31
|
Jedynak M, Laurin D, Dolega P, Podsiadla-Bialoskorska M, Szurgot I, Chroboczek J, Szolajska E. Leukocytes and drug-resistant cancer cells are targets for intracellular delivery by adenoviral dodecahedron. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1853-1865. [DOI: 10.1016/j.nano.2018.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 04/04/2018] [Accepted: 05/04/2018] [Indexed: 12/15/2022]
|
32
|
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
|
33
|
Mates JM, Yao Z, Cheplowitz AM, Suer O, Phillips GS, Kwiek JJ, Rajaram MVS, Kim J, Robinson JM, Ganesan LP, Anderson CL. Mouse Liver Sinusoidal Endothelium Eliminates HIV-Like Particles from Blood at a Rate of 100 Million per Minute by a Second-Order Kinetic Process. Front Immunol 2017; 8:35. [PMID: 28167948 PMCID: PMC5256111 DOI: 10.3389/fimmu.2017.00035] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023] Open
Abstract
We crafted human immunodeficiency virus (HIV)-like particles of diameter about 140 nm, which expressed two major HIV-1 proteins, namely, env and gag gene products, and used this reagent to simulate the rate of decay of HIV from the blood stream of BALB/c male mice. We found that most (~90%) of the particles were eliminated (cleared) from the blood by the liver sinusoidal endothelial cells (LSECs), the remainder from Kupffer cells; suggesting that LSECs are the major liver scavengers for HIV clearance from blood. Decay was rapid with kinetics suggesting second order with respect to particles, which infers dimerization of a putative receptor on LSEC. The number of HIV-like particles required for saturating the clearance mechanism was approximated. The capacity for elimination of blood-borne HIV-like particles by the sinusoid was 112 million particles per minute. Assuming that the sinusoid endothelial cells were about the size of glass-adherent macrophages, then elimination capacity was more than 540 particles per hour per endothelial cell.
Collapse
Affiliation(s)
- Jessica M Mates
- Departments of Internal Medicine, The Ohio State University , Columbus, OH , USA
| | - Zhili Yao
- Departments of Internal Medicine, The Ohio State University , Columbus, OH , USA
| | - Alana M Cheplowitz
- Departments of Internal Medicine, The Ohio State University , Columbus, OH , USA
| | - Ozan Suer
- Departments of Internal Medicine, The Ohio State University , Columbus, OH , USA
| | - Gary S Phillips
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University , Columbus, OH , USA
| | - Jesse J Kwiek
- Department of Microbiology, The Ohio State University , Columbus, OH , USA
| | - Murugesan V S Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University , Columbus, OH , USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University , Boston, MA , USA
| | - John M Robinson
- Physiology and Cell Biology, The Ohio State University , Columbus, OH , USA
| | - Latha P Ganesan
- Departments of Internal Medicine, The Ohio State University , Columbus, OH , USA
| | - Clark L Anderson
- Departments of Internal Medicine, The Ohio State University , Columbus, OH , USA
| |
Collapse
|
34
|
Mouse Hepatitis Virus Infection Induces a Toll-Like Receptor 2-Dependent Activation of Inflammatory Functions in Liver Sinusoidal Endothelial Cells during Acute Hepatitis. J Virol 2016; 90:9096-113. [PMID: 27489277 DOI: 10.1128/jvi.01069-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/23/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Under physiological conditions, the liver sinusoidal endothelial cells (LSECs) mediate hepatic immune tolerance toward self or foreign antigens through constitutive expression of anti-inflammatory mediators. However, upon viral infection or Toll-like receptor 2 (TLR2) activation, LSECs can achieve proinflammatory functions, but their role in hepatic inflammation during acute viral hepatitis is unknown. Using the highly virulent mouse hepatitis virus type 3 (MHV3) and the attenuated variants 51.6-MHV3 and YAC-MHV3, exhibiting lower tropism for LSECs, we investigated in vivo and in vitro the consequence of LSEC infection on their proinflammatory profiles and the aggravation of acute hepatitis process. In vivo infection with virulent MHV3, in comparison to attenuated strains, resulted in fulminant hepatitis associated with higher hepatic viral load, tissue necrosis, and levels of inflammatory mediators and earlier recruitment of inflammatory cells. Such hepatic inflammatory disorders correlated with disturbed production of interleukin-10 (IL-10) and vascular factors by LSECs. We next showed in vitro that infection of LSECs by the virulent MHV3 strain altered their production of anti-inflammatory cytokines and promoted higher release of proinflammatory and procoagulant factors and earlier cell damage than infection by attenuated strains. This higher replication and proinflammatory activation in LSECs by the virulent MHV3 strain was associated with a specific activation of TLR2 signaling by the virus. We provide evidence that TLR2 activation of LSCEs by MHV3 is an aggravating factor of hepatic inflammation and correlates with the severity of hepatitis. Taken together, these results indicate that preservation of the immunotolerant properties of LSECs during acute viral hepatitis is imperative in order to limit hepatic inflammation and damage. IMPORTANCE Viral hepatitis B and C infections are serious health problems affecting over 350 million and 170 million people worldwide, respectively. It has been suggested that a balance between protection and liver damage mediated by the host's immune response during the acute phase of infection would be determinant in hepatitis outcome. Thus, it appears crucial to identify the factors that predispose in exacerbating liver inflammation to limit hepatocyte injury. Liver sinusoidal endothelial cells (LSECs) can express both anti- and proinflammatory functions, but their role in acute viral hepatitis has never been investigated. Using mouse hepatitis virus (MHV) infections as animal models of viral hepatitis, we report for the first time that in vitro and in vivo infection of LSECs by the pathogenic MHV3 serotype leads to a reversion of their intrinsic anti-inflammatory phenotype toward a proinflammatory profile as well to as disorders in vascular factors, correlating with the severity of hepatitis. These results highlight a new virus-promoted mechanism of exacerbation of liver inflammatory response during acute hepatitis.
Collapse
|
35
|
Vigil D, Konstantinov NK, Barry M, Harford AM, Servilla KS, Kim YH, Sun Y, Ganta K, Tzamaloukas AH. BK nephropathy in the native kidneys of patients with organ transplants: Clinical spectrum of BK infection. World J Transplant 2016; 6:472-504. [PMID: 27683628 PMCID: PMC5036119 DOI: 10.5500/wjt.v6.i3.472] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/25/2016] [Accepted: 09/08/2016] [Indexed: 02/05/2023] Open
Abstract
Nephropathy secondary to BK virus, a member of the Papoviridae family of viruses, has been recognized for some time as an important cause of allograft dysfunction in renal transplant recipients. In recent times, BK nephropathy (BKN) of the native kidneys has being increasingly recognized as a cause of chronic kidney disease in patients with solid organ transplants, bone marrow transplants and in patients with other clinical entities associated with immunosuppression. In such patients renal dysfunction is often attributed to other factors including nephrotoxicity of medications used to prevent rejection of the transplanted organs. Renal biopsy is required for the diagnosis of BKN. Quantitation of the BK viral load in blood and urine are surrogate diagnostic methods. The treatment of BKN is based on reduction of the immunosuppressive medications. Several compounds have shown antiviral activity, but have not consistently shown to have beneficial effects in BKN. In addition to BKN, BK viral infection can cause severe urinary bladder cystitis, ureteritis and urinary tract obstruction as well as manifestations in other organ systems including the central nervous system, the respiratory system, the gastrointestinal system and the hematopoietic system. BK viral infection has also been implicated in tumorigenesis. The spectrum of clinical manifestations from BK infection and infection from other members of the Papoviridae family is widening. Prevention and treatment of BK infection and infections from other Papovaviruses are subjects of intense research.
Collapse
|
36
|
Hirsch HH, Yakhontova K, Lu M, Manzetti J. BK Polyomavirus Replication in Renal Tubular Epithelial Cells Is Inhibited by Sirolimus, but Activated by Tacrolimus Through a Pathway Involving FKBP-12. Am J Transplant 2016; 16:821-32. [PMID: 26639422 PMCID: PMC5064607 DOI: 10.1111/ajt.13541] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 09/04/2015] [Accepted: 09/24/2015] [Indexed: 01/25/2023]
Abstract
BK polyomavirus (BKPyV) replication causes nephropathy and premature kidney transplant failure. Insufficient BKPyV-specific T cell control is regarded as a key mechanism, but direct effects of immunosuppressive drugs on BKPyV replication might play an additional role. We compared the effects of mammalian target of rapamycin (mTOR)- and calcineurin-inhibitors on BKPyV replication in primary human renal tubular epithelial cells. Sirolimus impaired BKPyV replication with a 90% inhibitory concentration of 4 ng/mL by interfering with mTOR-SP6-kinase activation. Sirolimus inhibition was rapid and effective up to 24 h postinfection during viral early gene expression, but not thereafter, during viral late gene expression. The mTORC-1 kinase inhibitor torin-1 showed a similar inhibition profile, supporting the notion that early steps of BKPyV replication depend on mTOR activity. Cyclosporine A also inhibited BKPyV replication, while tacrolimus activated BKPyV replication and reversed sirolimus inhibition. FK binding protein 12kda (FKBP-12) siRNA knockdown abrogated sirolimus inhibition and increased BKPyV replication similar to adding tacrolimus. Thus, sirolimus and tacrolimus exert opposite effects on BKPyV replication in renal tubular epithelial cells by a mechanism involving FKBP-12 as common target. Immunosuppressive drugs may therefore contribute directly to the risk of BKPyV replication and nephropathy besides suppressing T cell functions. The data provide rationales for clinical trials aiming at reducing the risk of BKPyV replication and disease in kidney transplantation.
Collapse
Affiliation(s)
- H. H. Hirsch
- Transplantation & Clinical VirologyDepartment of Biomedicine (Haus Petersplatz)University of BaselBaselSwitzerland,Division Infection DiagnosticsDepartment of Biomedicine (Haus Petersplatz)University of BaselBaselSwitzerland,Infectious Diseases & Hospital EpidemiologyUniversity Hospital BaselBaselSwitzerland
| | - K. Yakhontova
- Transplantation & Clinical VirologyDepartment of Biomedicine (Haus Petersplatz)University of BaselBaselSwitzerland
| | - M. Lu
- Transplantation & Clinical VirologyDepartment of Biomedicine (Haus Petersplatz)University of BaselBaselSwitzerland
| | - J. Manzetti
- Transplantation & Clinical VirologyDepartment of Biomedicine (Haus Petersplatz)University of BaselBaselSwitzerland
| |
Collapse
|
37
|
Scavenger receptor B1, the HDL receptor, is expressed abundantly in liver sinusoidal endothelial cells. Sci Rep 2016; 6:20646. [PMID: 26865459 PMCID: PMC4749959 DOI: 10.1038/srep20646] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/07/2016] [Indexed: 02/08/2023] Open
Abstract
Cholesterol from peripheral tissue, carried by HDL, is metabolized in the liver after uptake by the HDL receptor, SR-B1. Hepatocytes have long been considered the only liver cells expressing SR-B1; however, in this study we describe two disparate immunofluorescence (IF) experiments that suggest otherwise. Using high-resolution confocal microscopy employing ultrathin (120 nm) sections of mouse liver, improving z-axis resolution, we identified the liver sinusoidal endothelial cells (LSEC), marked by FcγRIIb, as the cell within the liver expressing abundant SR-B1. In contrast, the hepatocyte, identified with β-catenin, expressed considerably weaker levels, although optical resolution of SR-B1 was inadequate. Thus, we moved to a different IF strategy, first separating dissociated liver cells by gradient centrifugation into two portions, hepatocytes (parenchymal cells) and LSEC (non-parenchymal cells). Characterizing both portions for the cellular expression of SR-B1 by flow cytometry, we found that LSEC expressed considerable amounts of SR-B1 while in hepatocytes SR-B1 expression was barely perceptible. Assessing mRNA of SR-B1 by real time PCR we found messenger expression in LSEC to be about 5 times higher than in hepatocytes.
Collapse
|
38
|
Sørensen KK, Simon‐Santamaria J, McCuskey RS, Smedsrød B. Liver Sinusoidal Endothelial Cells. Compr Physiol 2015; 5:1751-74. [DOI: 10.1002/cphy.c140078] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Haley SA, O'Hara BA, Nelson CDS, Brittingham FLP, Henriksen KJ, Stopa EG, Atwood WJ. Human polyomavirus receptor distribution in brain parenchyma contrasts with receptor distribution in kidney and choroid plexus. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2246-58. [PMID: 26056932 DOI: 10.1016/j.ajpath.2015.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/21/2015] [Accepted: 04/14/2015] [Indexed: 11/25/2022]
Abstract
The human polyomavirus, JCPyV, is the causative agent of progressive multifocal leukoencephalopathy, a rare demyelinating disease that occurs in the setting of prolonged immunosuppression. After initial asymptomatic infection, the virus establishes lifelong persistence in the kidney and possibly other extraneural sites. In rare instances, the virus traffics to the central nervous system, where oligodendrocytes, astrocytes, and glial precursors are susceptible to lytic infection, resulting in progressive multifocal leukoencephalopathy. The mechanisms by which the virus traffics to the central nervous system from peripheral sites remain unknown. Lactoseries tetrasaccharide c (LSTc), a pentasaccharide containing a terminal α2,6-linked sialic acid, is the major attachment receptor for polyomavirus. In addition to LSTc, type 2 serotonin receptors are required for facilitating virus entry into susceptible cells. We studied the distribution of virus receptors in kidney and brain using lectins, antibodies, and labeled virus. The distribution of LSTc, serotonin receptors, and virus binding sites overlapped in kidney and in the choroid plexus. In brain parenchyma, serotonin receptors were expressed on oligodendrocytes and astrocytes, but these cells were negative for LSTc and did not bind virus. LSTc was instead found on microglia and vascular endothelium, to which virus bound abundantly. Receptor distribution was not changed in the brains of patients with progressive multifocal leukoencephalopathy. Virus infection of oligodendrocytes and astrocytes during disease progression is LSTc independent.
Collapse
Affiliation(s)
- Sheila A Haley
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Bethany A O'Hara
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Christian D S Nelson
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Frances L P Brittingham
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Kammi J Henriksen
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Edward G Stopa
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| | - Walter J Atwood
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School, Brown University, Providence, Rhode Island.
| |
Collapse
|
40
|
Maslak E, Gregorius A, Chlopicki S. Liver sinusoidal endothelial cells (LSECs) function and NAFLD; NO-based therapy targeted to the liver. Pharmacol Rep 2015; 67:689-94. [PMID: 26321269 DOI: 10.1016/j.pharep.2015.04.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/13/2015] [Accepted: 04/17/2015] [Indexed: 12/13/2022]
Abstract
Liver sinusoidal endothelial cells (LSECs) present unique, highly specialised endothelial cells in the body. Unlike the structure and function of typical, vascular endothelial cells, LSECs are comprised of fenestrations, display high endocytic capacity and play a prominent role in maintaining overall liver homeostasis. LSEC dysfunction has been regarded as a key event in multiple liver disorders; however, its role and diagnostic, prognostic and therapeutic significance in nonalcoholic fatty liver disease (NAFLD) is still neglected. The purpose of this review is to provide an overview of the importance of LSECs in NAFLD. Attention is focused on the LSECs-mediated NO-dependent mechanisms in NAFLD development. We briefly describe the unique, highly specialised phenotype of LSECs and consequences of LSEC dysfunction on function of hepatic stellate cells (HSC) and hepatocytes. The potential efficacy of liver selective NO donors against liver steatosis and novel treatment approaches to modulate LSECs-driven liver pathology including NAFLD are also highlighted.
Collapse
Affiliation(s)
- Edyta Maslak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Aleksandra Gregorius
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland; Department of Experimental Pharmacology, Jagiellonian University Medical College, Kraków, Poland.
| |
Collapse
|