1
|
Horn P, Norlin J, Almholt K, Viuff BM, Galsgaard ED, Hald A, Zosel F, Demuth H, Poulsen S, Norby PL, Rasch MG, Vyberg M, Fleckner J, Werge MP, Gluud LL, Rink MR, Shepherd E, Northall E, Lalor PF, Weston CJ, Fog-Tonnesen M, Newsome PN. Evaluation of Gremlin-1 as a therapeutic target in metabolic dysfunction-associated steatohepatitis. eLife 2024; 13:RP95185. [PMID: 39361025 PMCID: PMC11449483 DOI: 10.7554/elife.95185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
Gremlin-1 has been implicated in liver fibrosis in metabolic dysfunction-associated steatohepatitis (MASH) via inhibition of bone morphogenetic protein (BMP) signalling and has thereby been identified as a potential therapeutic target. Using rat in vivo and human in vitro and ex vivo model systems of MASH fibrosis, we show that neutralisation of Gremlin-1 activity with monoclonal therapeutic antibodies does not reduce liver inflammation or liver fibrosis. Still, Gremlin-1 was upregulated in human and rat MASH fibrosis, but expression was restricted to a small subpopulation of COL3A1/THY1+ myofibroblasts. Lentiviral overexpression of Gremlin-1 in LX-2 cells and primary hepatic stellate cells led to changes in BMP-related gene expression, which did not translate to increased fibrogenesis. Furthermore, we show that Gremlin-1 binds to heparin with high affinity, which prevents Gremlin-1 from entering systemic circulation, prohibiting Gremlin-1-mediated organ crosstalk. Overall, our findings suggest a redundant role for Gremlin-1 in the pathogenesis of liver fibrosis, which is unamenable to therapeutic targeting.
Collapse
Affiliation(s)
- Paul Horn
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of BirminghamBirminghamUnited Kingdom
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
- Department of Hepatology & Gastroenterology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité MitteBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist ProgramBerlinGermany
| | - Jenny Norlin
- Global Drug Discovery, Novo Nordisk A/SMaaloevDenmark
| | | | | | | | - Andreas Hald
- Global Research Technologies, Novo Nordisk A/SMaaloevDenmark
| | - Franziska Zosel
- Global Research Technologies, Novo Nordisk A/SMaaloevDenmark
| | - Helle Demuth
- Global Research Technologies, Novo Nordisk A/SMaaloevDenmark
| | - Svend Poulsen
- Global Research Technologies, Novo Nordisk A/SMaaloevDenmark
| | - Peder L Norby
- Global Research Technologies, Novo Nordisk A/SMaaloevDenmark
| | - Morten G Rasch
- Global Research Technologies, Novo Nordisk A/SMaaloevDenmark
| | - Mogens Vyberg
- Department of Pathology, Copenhagen University Hospital Hvidovre, and Centre for RNA Medicine, Aalborg University CopenhagenCopenhagenDenmark
| | - Jan Fleckner
- Global Translation, Novo Nordisk A/SMaaloevDenmark
| | | | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital HvidovreHvidovreDenmark
| | - Marco R Rink
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Emma Shepherd
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Ellie Northall
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Patricia F Lalor
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Chris J Weston
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of BirminghamBirminghamUnited Kingdom
- Centre for Liver & Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | | | - Philip N Newsome
- Roger Williams Institute of Liver Studies, Faculty of Life Sciences and Medicine, King’s College London and King’s College HospitalLondonUnited Kingdom
| |
Collapse
|
2
|
Szafranska K, Sørensen KK, Lalor PF, McCourt P. Sinusoidal cells and liver immunology. SINUSOIDAL CELLS IN LIVER DISEASES 2024:53-75. [DOI: 10.1016/b978-0-323-95262-0.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:1-95. [DOI: 10.1016/b978-0-7020-8228-3.00001-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Zhao J, Zhang X, Li Y, Yu J, Chen Z, Niu Y, Ran S, Wang S, Ye W, Luo Z, Li X, Hao Y, Zong J, Xia C, Xia J, Wu J. Interorgan communication with the liver: novel mechanisms and therapeutic targets. Front Immunol 2023; 14:1314123. [PMID: 38155961 PMCID: PMC10754533 DOI: 10.3389/fimmu.2023.1314123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/28/2023] [Indexed: 12/30/2023] Open
Abstract
The liver is a multifunctional organ that plays crucial roles in numerous physiological processes, such as production of bile and proteins for blood plasma, regulation of blood levels of amino acids, processing of hemoglobin, clearance of metabolic waste, maintenance of glucose, etc. Therefore, the liver is essential for the homeostasis of organisms. With the development of research on the liver, there is growing concern about its effect on immune cells of innate and adaptive immunity. For example, the liver regulates the proliferation, differentiation, and effector functions of immune cells through various secreted proteins (also known as "hepatokines"). As a result, the liver is identified as an important regulator of the immune system. Furthermore, many diseases resulting from immune disorders are thought to be related to the dysfunction of the liver, including systemic lupus erythematosus, multiple sclerosis, and heart failure. Thus, the liver plays a role in remote immune regulation and is intricately linked with systemic immunity. This review provides a comprehensive overview of the liver remote regulation of the body's innate and adaptive immunity regarding to main areas: immune-related molecules secreted by the liver and the liver-resident cells. Additionally, we assessed the influence of the liver on various facets of systemic immune-related diseases, offering insights into the clinical application of target therapies for liver immune regulation, as well as future developmental trends.
Collapse
Affiliation(s)
- Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
5
|
Wiering L, Tacke F. Treating inflammation to combat non-alcoholic fatty liver disease. J Endocrinol 2023; 256:JOE-22-0194. [PMID: 36259984 DOI: 10.1530/joe-22-0194] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) with its more progressive form non-alcoholic steatohepatitis (NASH) has become the most common chronic liver disease, thereby representing a great burden for patients and healthcare systems. Specific pharmacological therapies for NAFLD are still missing. Inflammation is an important driver in the pathogenesis of NASH, and the mechanisms underlying inflammation in NAFLD represent possible therapeutic targets. In NASH, various intra- and extrahepatic triggers involved in the metabolic injury typically lead to the activation of different immune cells. This includes hepatic Kupffer cells, i.e. liver-resident macrophages, which can adopt an inflammatory phenotype and activate other immune cells by releasing inflammatory cytokines. As inflammation progresses, Kupffer cells are increasingly replaced by monocyte-derived macrophages with a distinct lipid-associated and scar-associated phenotype. Many other immune cells, including neutrophils, T lymphocytes - such as auto-aggressive cytotoxic as well as regulatory T cells - and innate lymphoid cells balance the progression and regression of inflammation and subsequent fibrosis. The detailed understanding of inflammatory cell subsets and their activation pathways prompted preclinical and clinical exploration of potential targets in NAFLD/NASH. These approaches to target inflammation in NASH include inhibition of immune cell recruitment via chemokine receptors (e.g. cenicriviroc), neutralization of CD44 or galectin-3 as well as agonism to nuclear factors like peroxisome proliferator-activated receptors and farnesoid X receptor that interfere with the activation of immune cells. As some of these approaches did not demonstrate convincing efficacy as monotherapies, a rational and personalized combination of therapeutic interventions may be needed for the near future.
Collapse
Affiliation(s)
- Leke Wiering
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Frank Tacke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
6
|
Hercun J, Vincent C, Bilodeau M, Lapierre P. Immune-Mediated Hepatitis During Immune Checkpoint Inhibitor cancer Immunotherapy: Lessons From Autoimmune Hepatitis and Liver Immunology. Front Immunol 2022; 13:907591. [PMID: 35844534 PMCID: PMC9280269 DOI: 10.3389/fimmu.2022.907591] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/06/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) are being increasingly used to successfully treat several types of cancer. However, due to their mode of action, these treatments are associated with several immune-related adverse events (irAEs), including immune-mediated autoimmune-like hepatitis in 5 to 10% of cases. The specific immune mechanism responsible for the development of immune-mediated liver injury caused by immune checkpoint inhibitors (ILICI) is currently unknown. This review summarizes the current knowledge on hepatic irAEs during cancer immunotherapy. It also addresses the clinical management of ILICI and how it is becoming an increasingly important clinical issue. Clinical, histological, and laboratory features of autoimmune hepatitis (AIH) and ILICI are compared, and their shared and distinctive traits are discussed in an effort to better understand the development of hepatic irAEs. Finally, based on the current knowledge of liver immunology and AIH pathogenesis, we propose a series of events that could trigger the observed liver injury in ICI-treated patients. This model could be useful in the design of future studies aiming to identify the specific immune mechanism(s) at play in ILICI and improve immune checkpoint inhibitor cancer immunotherapy.
Collapse
Affiliation(s)
- Julian Hercun
- Département de médecine, Université de Montréal, Montréal, QC, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Catherine Vincent
- Département de médecine, Université de Montréal, Montréal, QC, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Marc Bilodeau
- Département de médecine, Université de Montréal, Montréal, QC, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
| | - Pascal Lapierre
- Département de médecine, Université de Montréal, Montréal, QC, Canada
- Centre de recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC, Canada
- *Correspondence: Pascal Lapierre,
| |
Collapse
|
7
|
Du W, Wang L. The Crosstalk Between Liver Sinusoidal Endothelial Cells and Hepatic Microenvironment in NASH Related Liver Fibrosis. Front Immunol 2022; 13:936196. [PMID: 35837401 PMCID: PMC9274003 DOI: 10.3389/fimmu.2022.936196] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/03/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic liver injury can be caused by many factors, including virus infection, alcohol intake, cholestasis and abnormal fat accumulation. Nonalcoholic steatohepatitis (NASH) has become the main cause of liver fibrosis worldwide. Recently, more and more evidences show that hepatic microenvironment is involved in the pathophysiological process of liver fibrosis induced by NASH. Hepatic microenvironment consists of various types of cells and intercellular crosstalk among different cells in the liver sinusoids. Liver sinusoidal endothelial cells (LSECs), as the gatekeeper of liver microenvironment, play an irreplaceable role in the homeostasis and alterations of liver microenvironment. Many recent studies have reported that during the progression of NASH to liver fibrosis, LSECs are involved in various stages mediated by a series of mechanisms. Therefore, here we review the key role of crosstalk between LSECs and hepatic microenvironment in the progression of NASH to liver fibrosis (steatosis, inflammation, and fibrosis), as well as promising therapeutic strategies targeting LSECs.
Collapse
Affiliation(s)
- Wei Du
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
8
|
Wang XK, Peng ZG. Targeting Liver Sinusoidal Endothelial Cells: An Attractive Therapeutic Strategy to Control Inflammation in Nonalcoholic Fatty Liver Disease. Front Pharmacol 2021; 12:655557. [PMID: 33935770 PMCID: PMC8082362 DOI: 10.3389/fphar.2021.655557] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), especially its advanced stage nonalcoholic steatohepatitis (NASH), has become a threatened public health problem worldwide. However, no specific drug has been approved for clinical use to treat patients with NASH, though there are many promising candidates against NAFLD in the drug development pipeline. Recently, accumulated evidence showed that liver sinusoidal endothelial cells (LSECs) play an essential role in the occurrence and development of liver inflammation in patients with NAFLD. LSECs, as highly specialized endothelial cells with unique structure and anatomical location, contribute to the maintenance of liver homeostasis and could be a promising therapeutic target to control liver inflammation of NAFLD. In this review, we outline the pathophysiological roles of LSECs related to inflammation of NAFLD, highlight the pro-inflammatory and anti-inflammatory effects of LSECs, and discuss the potential drug development strategies against NAFLD based on targeting to LSECs.
Collapse
Affiliation(s)
- Xue-Kai Wang
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Biotechnology of Antibiotics, National Health and Family Planning Commission, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Jiang Q, Li Q, Liu B, Li G, Riedemann G, Gaitantzi H, Breitkopf-Heinlein K, Zeng A, Ding H, Xu K. BMP9 promotes methionine- and choline-deficient diet-induced nonalcoholic steatohepatitis in non-obese mice by enhancing NF-κB dependent macrophage polarization. Int Immunopharmacol 2021; 96:107591. [PMID: 33812253 DOI: 10.1016/j.intimp.2021.107591] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/22/2021] [Accepted: 03/11/2021] [Indexed: 12/24/2022]
Abstract
Our previous study confirmed that bone morphogenetic protein 9 (BMP9) participated in the development of nonalcoholic steatohepatitis (NASH) by affecting macrophage polarization. The focus of this study was to further confirm the role of macrophages in BMP9-mediated NASH and to analyze the underlying mechanism. In vivo, mice that were administered adeno-associated viral (AAV) vectors containing a null transgene (AAV-null) or the BMP9 transgene (AAV-BMP9) were divided into methionine- and choline-deficient (MCD) and control diet (CD) groups, and they were administered either control liposomes or clodronate liposomes via tail vein injection, the latter to deplete macrophages. The mice were sacrificed after 4 weeks of MCD diet feeding. In vitro, RAW264.7 cells were pretreated with or without BAY11-7085 (an NF-κB inhibitor) and stimulated with recombinant human BMP9 (rh-BMP9). To explore the underlying mechanism of action of BMP9, primary human monocyte-derived macrophages were additionally investigated and immunohistochemistry, biochemical assays, qRT-PCR, and Western blotting were used. The characteristics of NASH-related inflammation were assessed by hepatic histological analysis. Serum AST and ALT and hepatic triglyceride were examined by biochemical assays. We found that the expression of M1 macrophage genes (including CD86, IL1β, IL6, MCP-1 and TNFα) and the number of M1 macrophages (iNOS+ macrophages) in the liver were significantly elevated after BMP9 overexpression and BMP9 directly upregulated TLR4 expression in MCD-induced NASH. These effects were eliminated by macrophage depletion. In vitro, we discovered that BMP9 enhanced the nuclear translocation of NF-κB to induce macrophage M1 polarization in RAW264.7 cells and it promoted LPS-mediated activation of the NF-κB pathway in primary human macrophages. Taken together, this study demonstrates that BMP9 promotes NASH development by directly acting on macrophages.
Collapse
Affiliation(s)
- Qianqian Jiang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Li
- Department of Gastroenterology and Hepatology, Beijing You'an Hospital Affiliated with Capital Medical University, Beijing 100069, China
| | - Beibei Liu
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guixin Li
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Gabriel Riedemann
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Haristi Gaitantzi
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany; Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Katja Breitkopf-Heinlein
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany; Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
| | - Ajuan Zeng
- Department of Gastroenterology and Hepatology, Beijing You'an Hospital Affiliated with Capital Medical University, Beijing 100069, China
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing You'an Hospital Affiliated with Capital Medical University, Beijing 100069, China.
| | - Keshu Xu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
10
|
Wilkinson AL, Qurashi M, Shetty S. The Role of Sinusoidal Endothelial Cells in the Axis of Inflammation and Cancer Within the Liver. Front Physiol 2020; 11:990. [PMID: 32982772 PMCID: PMC7485256 DOI: 10.3389/fphys.2020.00990] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Liver sinusoidal endothelial cells (LSEC) form a unique barrier between the liver sinusoids and the underlying parenchyma, and thus play a crucial role in maintaining metabolic and immune homeostasis, as well as actively contributing to disease pathophysiology. Whilst their endocytic and scavenging function is integral for nutrient exchange and clearance of waste products, their capillarisation and dysfunction precedes fibrogenesis. Furthermore, their ability to promote immune tolerance and recruit distinct immunosuppressive leukocyte subsets can allow persistence of chronic viral infections and facilitate tumour development. In this review, we present the immunological and barrier functions of LSEC, along with their role in orchestrating fibrotic processes which precede tumourigenesis. We also summarise the role of LSEC in modulating the tumour microenvironment, and promoting development of a pre-metastatic niche, which can drive formation of secondary liver tumours. Finally, we summarise closely inter-linked disease pathways which collectively perpetuate pathogenesis, highlighting LSEC as novel targets for therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Shishir Shetty
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
11
|
Tumova S, Houghton MJ, Williamson G. The effect of quercetin on endothelial cells is modified by heterocellular interactions. Food Funct 2020; 11:3916-3925. [PMID: 32363357 DOI: 10.1039/d0fo00141d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Single cell-type models are useful for determining mechanisms, but in vivo, cell-cell interactions are important, and neighbouring cells can impact endothelial cell function. Quercetin can attenuate endothelial dysfunction by modulating vascular tone and reducing inflammation. We determined the effect of quercetin on a co-culture between Human Umbilical Vein Endothelial Cells (HUVEC) and human HepG2 hepatic cells or human LHCN-M2 muscle cells. Heme oxygenase-1 (HO-1) mRNA and protein were decreased, pyruvate dehydrogenase kinase (PDK) 4 and glucose transporter (GLUT) 3 mRNA increased, and GLUT1 protein decreased in HUVEC when cultured with HepG2. GLUT transporters, but not the other targets, were similarly regulated in co-culture with muscle cells. Some but not all of the effects were mediated by lactate and transforming growth factor β1. Quercetin added apically to the endothelial cells upregulated HO-1 and downregulated PDK4 both in monoculture and in co-culture, but the total PDK4 levels were higher in the presence of HepG2 cells. In the absence of general permeability changes, glucose transport across the endothelial monolayer was elevated in the presence of HepG2 cells, however this effect was moderated by quercetin applied on the apical side of the endothelial cells. At lower glucose concentration, apical quercetin also promoted glucose uptake in HepG2 cells. Co-culturing HUVEC with the HepG2 cells showed capacity to modulate quercetin-elicited changes in endothelial gene transcription and glucose transport.
Collapse
|
12
|
Hammoutene A, Rautou PE. Role of liver sinusoidal endothelial cells in non-alcoholic fatty liver disease. J Hepatol 2019; 70:1278-1291. [PMID: 30797053 DOI: 10.1016/j.jhep.2019.02.012] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/10/2019] [Accepted: 02/13/2019] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its complications are an expanding health problem associated with the metabolic syndrome. Liver sinusoidal endothelial cells (LSECs) are highly specialized endothelial cells localized at the interface between the blood derived from the gut and the adipose tissue on the one side, and other liver cells on the other side. In physiological conditions, LSECs are gatekeepers of liver homeostasis. LSECs display anti-inflammatory and anti-fibrogenic properties by preventing Kupffer cell and hepatic stellate cell activation and regulating intrahepatic vascular resistance and portal pressure. This review focusses on changes occurring in LSECs in NAFLD and on their consequences on NAFLD progression and complications. Capillarization, namely the loss of LSEC fenestrae, and LSEC dysfunction, namely the loss of the ability of LSECs to generate vasodilator agents in response to increased shear stress both occur early in NAFLD. These LSEC changes favour steatosis development and set the stage for NAFLD progression. At the stage of non-alcoholic steatohepatitis, altered LSECs release inflammatory mediators and contribute to the recruitment of inflammatory cells, thus promoting liver injury and inflammation. Altered LSECs also fail to maintain hepatic stellate cell quiescence and release fibrogenic mediators, including Hedgehog signalling molecules, promoting liver fibrosis. Liver angiogenesis is increased in NAFLD and contributes to liver inflammation and fibrosis, but also to hepatocellular carcinoma development. Thus, improving LSEC health appears to be a promising approach to prevent NAFLD progression and complications.
Collapse
Affiliation(s)
- Adel Hammoutene
- Inserm, UMR-970, Paris Cardiovascular Research Center, PARCC, Paris, France; University Paris Descartes, Paris, France
| | - Pierre-Emmanuel Rautou
- Inserm, UMR-970, Paris Cardiovascular Research Center, PARCC, Paris, France; INSERM, UMR1149, Centre de Recherche sur l'Inflammation, Paris, France; University Paris Diderot, Paris, France; Service d'Hépatologie, Centre de Référence des Maladies Vasculaires du Foie, DHU Unity, Pôle des Maladies de l'Appareil Digestif, Hôpital Beaujon, AP-HP, Clichy, France.
| |
Collapse
|
13
|
Abstract
Significance: Vascular adhesion protein-1 (VAP-1) is an ectoenzyme that oxidates primary amines in a reaction producing also hydrogen peroxide. VAP-1 on the blood vessel endothelium regulates leukocyte extravasation from the blood into tissues under physiological and pathological conditions. Recent Advances: Inhibition of VAP-1 by neutralizing antibodies and by several novel small-molecule enzyme inhibitors interferes with leukocyte trafficking and alleviates inflammation in many experimental models. Targeting of VAP-1 also shows beneficial effects in several other diseases, such as ischemia/reperfusion, fibrosis, and cancer. Moreover, soluble VAP-1 levels may serve as a new prognostic biomarker in selected diseases. Critical Issues: Understanding the contribution of the enzyme activity-independent and enzyme activity-dependent functions, which often appear to be mediated by the hydrogen peroxide production, in the VAP-1 biology will be crucial. Similarly, there is a pressing need to understand which of the VAP-1 functions are regulated through the modulation of leukocyte trafficking, and what is the role of VAP-1 synthesized in adipose and smooth muscle cells. Future Directions: The specificity and selectivity of new VAP-1 inhibitors, and their value in animal models under therapeutic settings need to be addressed. Results from several programs studying the therapeutic potential of VAP-1 inhibition, which now are in clinical trials, will reveal the relevance of this amine oxidase in humans.
Collapse
Affiliation(s)
- Marko Salmi
- 1 MediCity , Turku, Finland .,2 Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirpa Jalkanen
- 1 MediCity , Turku, Finland .,2 Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
14
|
Sabbagh MF, Heng JS, Luo C, Castanon RG, Nery JR, Rattner A, Goff LA, Ecker JR, Nathans J. Transcriptional and epigenomic landscapes of CNS and non-CNS vascular endothelial cells. eLife 2018; 7:36187. [PMID: 30188322 PMCID: PMC6126923 DOI: 10.7554/elife.36187] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Vascular endothelial cell (EC) function depends on appropriate organ-specific molecular and cellular specializations. To explore genomic mechanisms that control this specialization, we have analyzed and compared the transcriptome, accessible chromatin, and DNA methylome landscapes from mouse brain, liver, lung, and kidney ECs. Analysis of transcription factor (TF) gene expression and TF motifs at candidate cis-regulatory elements reveals both shared and organ-specific EC regulatory networks. In the embryo, only those ECs that are adjacent to or within the central nervous system (CNS) exhibit canonical Wnt signaling, which correlates precisely with blood-brain barrier (BBB) differentiation and Zic3 expression. In the early postnatal brain, single-cell RNA-seq of purified ECs reveals (1) close relationships between veins and mitotic cells and between arteries and tip cells, (2) a division of capillary ECs into vein-like and artery-like classes, and (3) new endothelial subtype markers, including new validated tip cell markers.
Collapse
Affiliation(s)
- Mark F Sabbagh
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jacob S Heng
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Chongyuan Luo
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States.,Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, United States
| | - Rosa G Castanon
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Joseph R Nery
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Amir Rattner
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Loyal A Goff
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Joseph R Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, United States.,Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, United States
| | - Jeremy Nathans
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, United States.,Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
15
|
Zhao H, Han Q, Lu N, Xu D, Tian Z, Zhang J. HMBOX1 in hepatocytes attenuates LPS/D-GalN-induced liver injury by inhibiting macrophage infiltration and activation. Mol Immunol 2018; 101:303-311. [PMID: 30032072 DOI: 10.1016/j.molimm.2018.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/11/2022]
Abstract
The HMBOX1 (Homeobox Containing 1) gene was first isolated from the human pancreatic cDNA libraries and is widely expressed in many tissues. Previously, we detected high expression of HMBOX1 in the liver, but its function was unclear. In this study, hepatocyte-specific HMBOX1 knockout mice (Hm△hep mice) were generated and used to characterize the function of HMBOX1 in the LPS/D-GalN-induced acute liver failure model. HMBOX1-knockout exhibits exacerbated liver injury induced by LPS/D-GalN, accompanied with high levels of inflammatory cytokines both in the liver and in circulation. Further investigation demonstrated that HMBOX1 negatively regulates NF-κB signal transduction. Therefore, HMBOX1-knockout in hepatocytes promotes CCL2 expression through the activation of NF-κB signaling, which enhanced the infiltration of macrophages into the liver. In addition, the decrease of HMBOX1 in hepatocytes promotes the activation of macrophages, upregulating CD80 and MHCⅡ, as well as inflammatory factors TNF-α and IL-6. Importantly, overexpression of HMBOX1 rescued liver injury in Hm△hep mice. These findings indicate that HMBOX1 in hepatocytes acts as a key immunosuppressive factor for inflammation and plays a critical protective role in LPS/D-GalN-induced liver injury.
Collapse
Affiliation(s)
- Hengli Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, China
| | - Nan Lu
- Diagnostic Institute, Medical School, Shandong University, China
| | - Dongqing Xu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, China.
| |
Collapse
|
16
|
Abstract
Liver sinusoid is the main functional site in liver. Multiple types of hepatic cells are well organized in a precisely-controlled biochemical and biomechanical environment, maintaining a spectrum of hepatic functions. Here, using micro-engineering techniques, four types of primary hepatic cells are integrated into two layer channels connected by porous membrane, which recreates the sinusoidal cell composition and architecture. By incorporating shear flow into this permeable system, the blood flow in sinusoids and interstitial flow in space of Disse are recapitulated. Conventional hepatocyte-based liver-specific functions are enhanced by non-parenchymal cells co-culture and shear flow. Moreover, major immune responses in liver sinusoids, i.e., neutrophil recruitment under lipopolysaccharide (LPS) stimulation, are replicated, indicating that all types of hepatic cells contribute to this process. Thus, this liver chip provides a new in vitro model to investigate the short-duration cellular interactions under a microenvironment mimicking the physiological composition and architecture of liver organ.
Collapse
Affiliation(s)
- Yu Du
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China
| | - Ning Li
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
17
|
Davies SP, Reynolds GM, Stamataki Z. Clearance of Apoptotic Cells by Tissue Epithelia: A Putative Role for Hepatocytes in Liver Efferocytosis. Front Immunol 2018; 9:44. [PMID: 29422896 PMCID: PMC5790054 DOI: 10.3389/fimmu.2018.00044] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/08/2018] [Indexed: 12/11/2022] Open
Abstract
Toxic substances and microbial or food-derived antigens continuously challenge the liver, which is tasked with their safe neutralization. This vital organ is also important for the removal of apoptotic immune cells during inflammation and has been previously described as a “graveyard” for dying lymphocytes. The clearance of apoptotic and necrotic cells is known as efferocytosis and is a critical liver function to maintain tissue homeostasis. Much of the research into this form of immunological control has focused on Kupffer cells, the liver-resident macrophages. However, hepatocytes (and other liver resident cells) are competent efferocytes and comprise 80% of the liver mass. Little is known regarding the mechanisms of apoptotic and necrotic cell capture by epithelia, which lack key receptors that mediate phagocytosis in macrophages. Herein, we discuss recent developments that increased our understanding of efferocytosis in tissues, with a special focus on the liver parenchyma. We discuss the impact of efferocytosis in health and in inflammation, highlighting the role of phagocytic epithelia.
Collapse
Affiliation(s)
- Scott P Davies
- Centre for Liver Research, College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Gary M Reynolds
- Centre for Liver Research, College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,Centre for Liver Research and National Institute for Health Research (NIHR) Birmingham Liver Biomedical Research Unit, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.,University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Zania Stamataki
- Centre for Liver Research, College of Medical and Dental Sciences, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
18
|
Abstract
Liver sinusoidal endothelial cells (LSECs) line the low shear, sinusoidal capillary channels of the liver and are the most abundant non-parenchymal hepatic cell population. LSECs do not simply form a barrier within the hepatic sinusoids but have vital physiological and immunological functions, including filtration, endocytosis, antigen presentation and leukocyte recruitment. Reflecting these multifunctional properties, LSECs display unique structural and phenotypic features that differentiate them from the capillary endothelium present within other organs. It is now clear that LSECs have a critical role in maintaining immune homeostasis within the liver and in mediating the immune response during acute and chronic liver injury. In this Review, we outline how LSECs influence the immune microenvironment within the liver and discuss their contribution to immune-mediated liver diseases and the complications of fibrosis and carcinogenesis.
Collapse
|
19
|
Crawford JM, Bioulac-Sage P, Hytiroglou P. Structure, Function, and Responses to Injury. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:1-87. [DOI: 10.1016/b978-0-7020-6697-9.00001-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
20
|
Resheq YJ, Menzner AK, Bosch J, Tickle J, Li KK, Wilhelm A, Hepburn E, Murihead G, Ward ST, Curbishley SM, Zimmermann HW, Bruns T, Gilbert DF, Tripal P, Mackensen A, Adams DH, Weston CJ. Impaired Transmigration of Myeloid-Derived Suppressor Cells across Human Sinusoidal Endothelium Is Associated with Decreased Expression of CD13. THE JOURNAL OF IMMUNOLOGY 2017; 199:1672-1681. [PMID: 28739875 DOI: 10.4049/jimmunol.1600466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/22/2017] [Indexed: 12/14/2022]
Abstract
Human monocytic myeloid-derived suppressor cells (MO-MDSCs) within the hepatic compartment suppress inflammation and impair immune surveillance in liver cancer. It is currently not known whether recruitment of MO-MDSCs from blood via hepatic sinusoidal endothelium (HSEC) contributes to their enrichment within the hepatic compartment. We compared the transmigratory potential of MO-MDSCs and monocytes after adhesion to hepatic endothelial monolayers in flow-based assays that mimic in vivo shear stress in the sinusoids. Despite comparable binding to HSEC monolayers, proportionally fewer MO-MDSCs underwent transendothelial migration, indicating that the final steps of extravasation, where actin polymerization plays an important role, are impaired in MO-MDSCs. In this article, we found reduced levels of CD13 on MO-MDSCs, which has recently been reported to control cell motility in monocytes, alongside reduced VLA-4 expression, an integrin predominantly involved in adherence to the apical side of the endothelium. CD13 and VLA-4 blocking and activating Abs were used in flow-based adhesion assays, live-cell imaging of motility, and actin polymerization studies to confirm a role for CD13 in impaired MO-MDSC transmigration. These findings indicate that CD13 significantly contributes to tissue infiltration by MO-MDSCs and monocytes, thereby contributing to the pathogenesis of hepatic inflammation.
Collapse
Affiliation(s)
- Yazid J Resheq
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; .,Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Ann-Katrin Menzner
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Jacobus Bosch
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | - Joseph Tickle
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Ka-Kit Li
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Annika Wilhelm
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom.,Division of Digestive Diseases, Department of Surgery and Cancer, Imperial College London, London W2 1NY, United Kingdom
| | - Elizabeth Hepburn
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Gillian Murihead
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stephen T Ward
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stuart M Curbishley
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Henning W Zimmermann
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom.,Department of Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Tony Bruns
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom.,Department of Medicine IV, University of Jena, 07743 Jena, Germany
| | - Daniel F Gilbert
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, 91052 Erlangen, Germany; and
| | - Philipp Tripal
- Optical Imaging Centre Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, 91052 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen 91054, Germany
| | - David H Adams
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Chris J Weston
- Institute of Immunology and Immunotherapy, Centre for Liver Research and National Institute for Health Research Birmingham Liver Biomedical Research Centre, College of Medicine and Dentistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
21
|
Strauss O, Phillips A, Ruggiero K, Bartlett A, Dunbar PR. Immunofluorescence identifies distinct subsets of endothelial cells in the human liver. Sci Rep 2017; 7:44356. [PMID: 28287163 PMCID: PMC5347010 DOI: 10.1038/srep44356] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
As well as systemic vascular endothelial cells, the liver has specialised sinusoidal endothelial cells (LSEC). LSEC dysfunction has been documented in many diseased states yet their phenotype in normal human liver has not been comprehensively assessed. Our aim was to improve characterisation of subsets of endothelial cells and associated pericytes in the human liver. Immunofluorescence microscopy was performed on normal human liver tissue samples to assess endothelial and structural proteins in a minimum of three donors. LSEC are distributed in an acinar pattern and universally express CD36, but two distinctive subsets of LSEC can be identified in different acinar zones. Type 1 LSEC are CD36hiCD32−CD14−LYVE-1− and are located in acinar zone 1 of the lobule, while Type 2 LSEC are LYVE-1+CD32hiCD14+CD54+CD36mid-lo and are located in acinar zones 2 and 3 of the lobule. Portal tracts and central veins can be identified using markers for systemic vascular endothelia and pericytes, none of which are expressed by LSEC. In areas of low hydrostatic pressure LSEC are lined by stellate cells that express the pericyte marker CD146. Our findings identify distinctive populations of LSEC and distinguish these cells from adjacent stellate cells, systemic vasculature and pericytes in different zones of the liver acinus.
Collapse
Affiliation(s)
- Otto Strauss
- Department of Surgery, Faculty of Medical Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Anthony Phillips
- Department of Surgery, Faculty of Medical Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Katya Ruggiero
- Department of Statistics, University of Auckland, Auckland, New Zealand
| | - Adam Bartlett
- Department of Surgery, Faculty of Medical Health Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - P Rod Dunbar
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Miyachi Y, Tsuchiya K, Komiya C, Shiba K, Shimazu N, Yamaguchi S, Deushi M, Osaka M, Inoue K, Sato Y, Matsumoto S, Kikuta J, Wake K, Yoshida M, Ishii M, Ogawa Y. Roles for Cell-Cell Adhesion and Contact in Obesity-Induced Hepatic Myeloid Cell Accumulation and Glucose Intolerance. Cell Rep 2017; 18:2766-2779. [PMID: 28297678 DOI: 10.1016/j.celrep.2017.02.039] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/18/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023] Open
Affiliation(s)
- Yasutaka Miyachi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kyoichiro Tsuchiya
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan.
| | - Chikara Komiya
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kumiko Shiba
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Noriko Shimazu
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shinobu Yamaguchi
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Michiyo Deushi
- Department of Life Science and Bioethics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mizuko Osaka
- Department of Life Science and Bioethics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kouji Inoue
- Department of Anatomy and Histocytology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa 230-8501, Japan
| | - Yuta Sato
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Sayaka Matsumoto
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Kenjiro Wake
- Department of Anatomy and Histocytology, School of Dental Medicine, Tsurumi University, Yokohama, Kanagawa 230-8501, Japan
| | - Masayuki Yoshida
- Department of Life Science and Bioethics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan; Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan; Japan Agency for Medical Research and Development, CREST, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
23
|
Du Y, Li N, Yang H, Luo C, Gong Y, Tong C, Gao Y, Lü S, Long M. Mimicking liver sinusoidal structures and functions using a 3D-configured microfluidic chip. LAB ON A CHIP 2017; 17:782-794. [PMID: 28112323 DOI: 10.1039/c6lc01374k] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Physiologically, four major types of hepatic cells - the liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, and hepatocytes - reside inside liver sinusoids and interact with flowing peripheral cells under blood flow. It is hard to mimic an in vivo liver sinusoid due to its complex multiple cell-cell interactions, spatiotemporal construction, and mechanical microenvironment. Here we developed an in vitro liver sinusoid chip by integrating the four types of primary murine hepatic cells into two adjacent fluid channels separated by a porous permeable membrane, replicating liver's key structures and configurations. Each type of cells was identified with its respective markers, and the assembled chip presented the liver-specific unique morphology of fenestration. The flow field in the liver chip was quantitatively analyzed by computational fluid dynamics simulations and particle tracking visualization tests. Intriguingly, co-culture and shear flow enhance albumin secretion independently or cooperatively, while shear flow alone enhances HGF production and CYP450 metabolism. Under lipopolysaccharide (LPS) stimulations, the hepatic cell co-culture facilitated neutrophil recruitment in the liver chip. Thus, this 3D-configured in vitro liver chip integrates the two key factors of shear flow and the four types of primary hepatic cells to replicate key structures, hepatic functions, and primary immune responses and provides a new in vitro model to investigate the short-duration hepatic cellular interactions under a microenvironment mimicking the physiology of a liver.
Collapse
Affiliation(s)
- Yu Du
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Li
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Yang
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunhua Luo
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yixin Gong
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunfang Tong
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxin Gao
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shouqin Lü
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mian Long
- Key Laboratory of Microgravity (National Microgravity Laboratory), Center of Biomechanics and Bioengineering, and Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China. and School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
24
|
Xu M, Wang X, Zou Y, Zhong Y. Key role of liver sinusoidal endothelial cells in liver fibrosis. Biosci Trends 2017; 11:163-168. [PMID: 28250338 DOI: 10.5582/bst.2017.01007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Because of the prevalence of viral hepatitis and nonalcoholic fatty liver disease (NAFLD), liver fibrosis has become a very common disease in Asia and elsewhere in the world, constantly increasing the burden of care borne by society. Hepatic sinusoidal capillarization, characterized by gradually shrinking fenestrae on the surface of liver sinusoidal endothelial cells (LSECs) and the formation of an organized basement membrane, is an initial pathologic change associated with liver fibrosis. Basic and clinical studies have indicated that LSECs play a key role in hepatic sinusoidal capillarization by affecting various aspects of the development and progression of liver fibrosis. Reviewing studies on the effect of LSECs on liver fibrosis is essential to better understanding the pathogenesis of liver fibrosis and its mechanism of progression. Moreover, such a review will provide a theoretical basis for identifying new methods to promote the regression or even inhibition of fibrosis. This review will focus on structural and functional changes in LSECs during hepatic sinusoidal capillarization and the interaction between the micro-environment of the liver and the body's immune system.
Collapse
Affiliation(s)
- Mingxing Xu
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| | - Xuehua Wang
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| | - Yong Zou
- Department of Blood Transfusion, Third Affiliated Hospital of Sun Yat-Sen University
| | - Yuesi Zhong
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Sun Yat-Sen University
| |
Collapse
|
25
|
Wiggins BG, Stamataki Z, Lalor PF. Using Ex Vivo Liver Organ Cultures to Measure Lymphocyte Trafficking. Methods Mol Biol 2017; 1591:177-194. [PMID: 28349483 DOI: 10.1007/978-1-4939-6931-9_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lymphocyte recruitment to different organs, and even alternate anatomical regions within the same organ, is differentially regulated. Key combinations of adhesion molecules and chemokines govern compartmentalization, and these can change depending upon the nature and duration of tissue injury. We are interested in understanding lymphocyte recruitment to the liver during human disease, and thus need models of the liver inflammatory milieu that are as representative as possible. Here we describe the use of precision cut liver slices as models of disease.
Collapse
Affiliation(s)
- Benjamin G Wiggins
- Centre for Liver Research, Immunity and Immunotherapy, Institute of Biomedical Research, University of Birmingham, Birmingham, UK.
| | - Zania Stamataki
- Centre for Liver Research, Immunity and Immunotherapy, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| | - Patricia F Lalor
- Centre for Liver Research, Immunity and Immunotherapy, Institute of Biomedical Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
26
|
Chimen M, Apta BHR, Mcgettrick HM. Introduction: T Cell Trafficking in Inflammation and Immunity. Methods Mol Biol 2017; 1591:73-84. [PMID: 28349476 DOI: 10.1007/978-1-4939-6931-9_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
T cell migration across vascular endothelium is essential for T cell responses, as through the expression of specific tissue-homing receptors, these cells then access peripheral tissues, with the goal of eliminating invading pathogens and/or tumor cells. However, aberrant trafficking of T cells to peripheral tissues contributes to the development of most chronic inflammatory diseases. Very little is known about the mechanisms by which T cell trafficking is regulated during inflammation, and it is thus difficult to target this aspect of pathology for the development of new therapies. It is therefore important to understand the pathways involved in regulating the recruitment of immune cells.
Collapse
Affiliation(s)
- Myriam Chimen
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Bonita H R Apta
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Vincent Drive, Edgbaston, Birmingham, West Midlands, B15 2TT, UK
| | - Helen M Mcgettrick
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
27
|
Validation of Bioreactor and Human-on-a-Chip Devices for Chemical Safety Assessment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 856:299-316. [PMID: 27671728 DOI: 10.1007/978-3-319-33826-2_12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Equipment and device qualification and test assay validation in the field of tissue engineered human organs for substance assessment remain formidable tasks with only a few successful examples so far. The hurdles seem to increase with the growing complexity of the biological systems, emulated by the respective models. Controlled single tissue or organ culture in bioreactors improves the organ-specific functions and maintains their phenotypic stability for longer periods of time. The reproducibility attained with bioreactor operations is, per se, an advantage for the validation of safety assessment. Regulatory agencies have gradually altered the validation concept from exhaustive "product" to rigorous and detailed process characterization, valuing reproducibility as a standard for validation. "Human-on-a-chip" technologies applying micro-physiological systems to the in vitro combination of miniaturized human organ equivalents into functional human micro-organisms are nowadays thought to be the most elaborate solution created to date. They target the replacement of the current most complex models-laboratory animals. Therefore, we provide here a road map towards the validation of such "human-on-a-chip" models and qualification of their respective bioreactor and microchip equipment along a path currently used for the respective animal models.
Collapse
|
28
|
CD147 promotes liver fibrosis progression via VEGF-A/VEGFR2 signalling-mediated cross-talk between hepatocytes and sinusoidal endothelial cells. Clin Sci (Lond) 2015. [PMID: 26201021 DOI: 10.1042/cs20140823] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although previous evidence indicates close involvement of CD147 in the pathogenesis of liver fibrosis, the underlying molecular mechanisms and its therapeutic value remain largely unknown. In the present study, we investigated the biological roles of CD147 in liver fibrosis and assessed its therapeutic value as a target molecule in the CCl4-induced liver fibrosis mouse model. We found that CD147 was highly expressed in both hepatocytes and SECs (sinusoidal endothelial cells) in fibrotic liver tissues. Additionally, it was significantly associated with the fibrosis stage. TGF-β1 (transforming growth factor β1) was found to be mainly responsible for the up-regulation of CD147. Bioinformatic and experimental data suggest a functional link between CD147 expression and VEGF-A (vascular endothelial growth factor A)/VEGR-2 (VEGF receptor 2) signalling-mediated angiogenesis in fibrotic liver tissues. Furthermore, we observed that the CD147-induced activation of the PI3K (phosphoinositide 3-kinase)/Akt signalling pathway promotes the production of VEGF-A in hepatocytes and expression of VEGFR-2 in SECs, which was found to enhance the angiogenic capability of SECs. Finally, our data indicate that blocking of CD147 using an mAb (monoclonal antibody) attenuated liver fibrosis progression via inhibition of VEGF-A/VEGFR-2 signalling and subsequent amelioration of microvascular abnormality in the CCl4-induced mouse model. Our findings suggest a novel functional mechanism that CD147 may promote liver fibrosis progression via inducing the VEGF-A/VEGFR-2 signalling pathway-mediated cross-talk between hepatocytes and SECs. New strategies based on the intervention of CD147 can be expected for prevention of liver fibrosis.
Collapse
|
29
|
Apicobasal polarity controls lymphocyte adhesion to hepatic epithelial cells. Cell Rep 2014; 8:1879-1893. [PMID: 25242329 DOI: 10.1016/j.celrep.2014.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 06/25/2014] [Accepted: 08/04/2014] [Indexed: 02/08/2023] Open
Abstract
Loss of apicobasal polarity is a hallmark of epithelial pathologies. Leukocyte infiltration and crosstalk with dysfunctional epithelial barriers are crucial for the inflammatory response. Here, we show that apicobasal architecture regulates the adhesion between hepatic epithelial cells and lymphocytes. Polarized hepatocytes and epithelium from bile ducts segregate the intercellular adhesion molecule 1 (ICAM-1) adhesion receptor onto their apical, microvilli-rich membranes, which are less accessible by circulating immune cells. Upon cell depolarization, hepatic ICAM-1 becomes exposed and increases lymphocyte binding. Polarized hepatic cells prevent ICAM-1 exposure to lymphocytes by redirecting basolateral ICAM-1 to apical domains. Loss of ICAM-1 polarity occurs in human inflammatory liver diseases and can be induced by the inflammatory cytokine tumor necrosis factor alpha (TNF-α). We propose that adhesion receptor polarization is a parenchymal immune checkpoint that allows functional epithelium to hamper leukocyte binding. This contributes to the haptotactic guidance of leukocytes toward neighboring damaged or chronically inflamed epithelial cells that expose their adhesion machinery.
Collapse
|
30
|
Ebrahimkhani MR, Neiman JAS, Raredon MSB, Hughes DJ, Griffith LG. Bioreactor technologies to support liver function in vitro. Adv Drug Deliv Rev 2014; 69-70:132-57. [PMID: 24607703 PMCID: PMC4144187 DOI: 10.1016/j.addr.2014.02.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/18/2014] [Accepted: 02/24/2014] [Indexed: 02/08/2023]
Abstract
Liver is a central nexus integrating metabolic and immunologic homeostasis in the human body, and the direct or indirect target of most molecular therapeutics. A wide spectrum of therapeutic and technological needs drives efforts to capture liver physiology and pathophysiology in vitro, ranging from prediction of metabolism and toxicity of small molecule drugs, to understanding off-target effects of proteins, nucleic acid therapies, and targeted therapeutics, to serving as disease models for drug development. Here we provide perspective on the evolving landscape of bioreactor-based models to meet old and new challenges in drug discovery and development, emphasizing design challenges in maintaining long-term liver-specific function and how emerging technologies in biomaterials and microdevices are providing new experimental models.
Collapse
Affiliation(s)
- Mohammad R Ebrahimkhani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jaclyn A Shepard Neiman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Micha Sam B Raredon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
31
|
Calvaruso M, Gulino A, Buffa S, Guarnotta C, Franco G, Cacciatore M, Bonura MG, Franco V, Florena AM. Challenges and new prospects in hepatosplenic γδ T-cell lymphoma. Leuk Lymphoma 2014; 55:2457-65. [PMID: 24506469 DOI: 10.3109/10428194.2014.889821] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of lymphoid neoplasms characterized by aggressive clinical behavior and dismal prognosis. Hepatosplenic γδ T-cell lymphoma (γδ-HSTL) is a particular form of PTCL that arises from a small subset of γ/δ T-cell receptor-expressing lymphocytes. γδ-HSTL has a rapidly progressive course and poor outcome due also to its refractoriness to conventional chemotherapy regimens. The very low incidence of γδ-HSTL, along with its propensity to mimic different pathological entities, makes this lymphoma a true diagnostic challenge. In this review, we highlight the biological and clinical features of γδ-HSTL that contribute to making this lymphoma a mostly incurable disease. Moreover, we provide a new insight into the crosstalk between HSTL clones and the bone marrow, liver and spleen vascular microenvironment, in which neoplastic cells reside and proliferate. We further discuss γδ-HSTL associated molecules that might be proposed as potential targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Marco Calvaruso
- Laboratorio di Tecnologie Oncologiche - HSR Giglio, C. da Pietrapollastra-Pisciotto , Cefalù , Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Shetty S, Adams DH, Hubscher SG. Post-transplant liver biopsy and the immune response: lessons for the clinician. Expert Rev Clin Immunol 2014; 8:645-61. [DOI: 10.1586/eci.12.65] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
33
|
The amine oxidase inhibitor phenelzine limits lipogenesis in adipocytes without inhibiting insulin action on glucose uptake. J Neural Transm (Vienna) 2012; 120:997-1003. [DOI: 10.1007/s00702-012-0951-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 12/04/2012] [Indexed: 12/22/2022]
|
34
|
Kasuya J, Sudo R, Masuda G, Mitaka T, Ikeda M, Tanishita K. Reconstruction of hepatic stellate cell-incorporated liver capillary structures in small hepatocyte tri-culture using microporous membranes. J Tissue Eng Regen Med 2012; 9:247-56. [PMID: 23086892 DOI: 10.1002/term.1630] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 06/01/2012] [Accepted: 09/10/2012] [Indexed: 01/22/2023]
Abstract
In liver sinusoids, hepatic stellate cells (HSCs) locate the outer surface of microvessels to form a functional unit with endothelia and hepatocytes. To reconstruct functional liver tissue in vitro, formation of the HSC-incorporated sinusoidal structure is essential. We previously demonstrated capillary formation of endothelial cells (ECs) in tri-culture, where a polyethylene terephthalate (PET) microporous membrane was intercalated between the ECs and hepatic organoids composed of small hepatocytes (SHs), i.e. hepatic progenitor cells, and HSCs. However, the high thickness and low porosity of the membranes limited heterotypic cell-cell interactions, which are essential to form HSC-EC hybrid structures. Here, we focused on the effective use of the thin and highly porous poly( d, l-lactide-co-glycolide) (PLGA) microporous membranes in SH-HSC-EC tri-culture to reconstruct the HSC-incorporated liver capillary structures in vitro. First, the formation of EC capillary-like structures was induced on Matrigel-coated PLGA microporous membranes. Next, the membranes were stacked on hepatic organoids composed of small SHs and HSCs. When the pore size and porosity of the membranes were optimized, HSCs selectively migrated to the EC capillary-like structures. This process was mediated in part by platelet-derived growth factor (PDGF) signalling. In addition, the HSCs were located along the outer surface of the EC capillary-like structures with their long cytoplasmic processes. In the HSC-incorporated capillary tissues, SHs acquired high levels of differentiated functions, compared to those without ECs. This model will provide a basis for the construction of functional, thick, vascularized liver tissues in vitro.
Collapse
Affiliation(s)
- Junichi Kasuya
- Centre for System Integration Engineering, School of Integrated Design Engineering, Keio University, 3-1-14-1 Kohoku, Yokohama, 223-0061, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Reglero-Real N, Marcos-Ramiro B, Millán J. Endothelial membrane reorganization during leukocyte extravasation. Cell Mol Life Sci 2012; 69:3079-99. [PMID: 22573182 PMCID: PMC11114893 DOI: 10.1007/s00018-012-0987-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 03/22/2012] [Accepted: 03/29/2012] [Indexed: 12/30/2022]
Abstract
Leukocyte trafficking from the bloodstream to inflamed tissues across the endothelial barrier is an essential response in innate immunity. Leukocyte adhesion, locomotion, and diapedesis induce signaling in endothelial cells and this is accompanied by a profound reorganization of the endothelial cell surfaces that is only starting to be unveiled. Here we review the current knowledge on the leukocyte-mediated alterations of endothelial membrane dynamics and their role in promoting leukocyte extravasation. The formation of protein- and lipid-mediated cell adhesion nanodomains at the endothelial apical surface, the extension of micrometric apical membrane docking structures, which are derived from microvilli and embrace adhered leukocytes, as well as the vesicle-trafficking pathways that are required for efficient leukocyte diapedesis, are discussed. The coordination between these different endothelial membrane-remodeling events probably provides the road map for transmigrating leukocytes to find exit points in the vessel wall, in a context of severe mechanical and inflammatory stress. A better understanding of how vascular endothelial cells respond to immune cell adhesion should enable new therapeutic strategies to be developed that can abrogate uncontrolled leukocyte extravasation in inflammatory diseases.
Collapse
Affiliation(s)
- Natalia Reglero-Real
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Beatriz Marcos-Ramiro
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
36
|
Innate immune cells in liver inflammation. Mediators Inflamm 2012; 2012:949157. [PMID: 22933833 PMCID: PMC3425885 DOI: 10.1155/2012/949157] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 06/17/2012] [Indexed: 12/20/2022] Open
Abstract
Innate immune system is the first line of defence against invading pathogens that is critical for the overall survival of the host. Human liver is characterised by a dual blood supply, with 80% of blood entering through the portal vein carrying nutrients and bacterial endotoxin from the gastrointestinal tract. The liver is thus constantly exposed to antigenic loads. Therefore, pathogenic microorganism must be efficiently eliminated whilst harmless antigens derived from the gastrointestinal tract need to be tolerized in the liver. In order to achieve this, the liver innate immune system is equipped with multiple cellular components; monocytes, macrophages, granulocytes, natural killer cells, and dendritic cells which coordinate to exert tolerogenic environment at the same time detect, respond, and eliminate invading pathogens, infected or transformed self to mount immunity. This paper will discuss the innate immune cells that take part in human liver inflammation, and their roles in both resolution of inflammation and tissue repair.
Collapse
|
37
|
Royo F, Falcon-Perez JM. Liver extracellular vesicles in health and disease. J Extracell Vesicles 2012; 1:18825. [PMID: 24009882 PMCID: PMC3760641 DOI: 10.3402/jev.v1i0.18825] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 06/21/2012] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in cell-to-cell communication. Although there are different kinds of vesicles, each with their own secretion and capture biology, all of them carry a cargo of proteins, lipids, metabolites and nucleic acids. They act as vehicles for exchange of biological materials and signals and are involved in the regulation of various physiological processes. Liver is an essential organ containing different cell populations fulfilling various functions, which need to be strictly controlled and coordinated. There are a few articles showing the role of liver-derived EVs. On the basis of them, we present here a hypothesis of the implication of such vesicles in the physiology of the liver. Different liver cell types, including hepatocytes, cholangiocytes and stellate cells, secrete and capture EVs and interact with them. Liver injury changes the abundance and cargo of EVs; these changes are likely to be important for the outcome of stress response. Although a substantial effort has been put into the characterization of EVs in isolated populations, it is only recently that some more comprehensive information has begun to appear. In this article, we hypothesize about the role of EVs in liver microenvironment and their possible function using published data from both hepatic and non-hepatic systems.
Collapse
Affiliation(s)
- Felix Royo
- Metabolomics Unit, CIC bioGUNE, CIBERehd, Technology Park of Bizkaia, Derio, Bizkaia, Spain
| | | |
Collapse
|
38
|
McGettrick HM, Butler LM, Buckley CD, Ed Rainger G, Nash GB. Tissue stroma as a regulator of leukocyte recruitment in inflammation. J Leukoc Biol 2012; 91:385-400. [DOI: 10.1189/jlb.0911458] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
39
|
Crawford JM, Burt AD. Anatomy, pathophysiology and basic mechanisms of disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2012:1-77. [DOI: 10.1016/b978-0-7020-3398-8.00001-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Werner JM, Lang C, Scherer MN, Farkas SA, Geissler EK, Schlitt HJ, Hornung M. Distribution of intrahepatic T, NK and CD3(+)CD56(+)NKT cells alters after liver transplantation: Shift from innate to adaptive immunity? Transpl Immunol 2011; 25:27-33. [PMID: 21635950 DOI: 10.1016/j.trim.2011.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 05/12/2011] [Accepted: 05/12/2011] [Indexed: 01/16/2023]
Abstract
BACKGROUND The liver is an immunological organ containing a large number of T, NK and NKT cells, but little is known about intrahepatic immunity after LTx. Here, we investigated whether the distribution of T, NK and CD3(+)CD56(+)NKT cells is altered in transplanted livers under different circumstances. METHODS Core biopsies of transplanted livers were stained with antibodies against CD3 and CD56. Several cell populations including T (CD3(+)CD56(-)), NK (CD3(-)CD56(+)) and NKT cells (CD3(+)CD56(+)) were studied by fluorescence microscopy. Cell numbers were analyzed in relation to the time interval after LTx, immunosuppressive therapy and stage of acute graft rejection (measured with the rejection activity index: RAI) compared to tumor free liver tissue from patients after liver resection due to metastatic disease as control. RESULTS Recruitment of CD3(+)CD56(+)NKT cells revealed a significant decrease during high RAI scores in comparison to low and middle RAI scores (RAI 7-9: 0.03±0.01/HPF vs. RAI 4-6: 0.1±0.005/HPF). CD3(+)CD56(+)NKT cells were also lower during immunosuppressive therapy with tacrolimus (0.03±0.01/HPF) than with cyclosporine (0.1±0.003/HPF), cyclosporine/MMF (0.1±0.003/HPF) or sirolimus (0.1±0.01/HPF) treatment. Intrahepatic T cell numbers increased significantly 50days after LTx compared to control liver tissue (4.5±0.2/HPF vs. 1.9±0.1/HPF). In contrast, NK cells (0.3±0.004/HPF) were significantly fewer in all biopsies after LTx compared to the control (0.7±0.04/HPF). CONCLUSIONS These data indicate significant alterations in the hepatic recruitment of T, NK and CD3(+)CD56(+)NKT cells after LTx. The increase in T cells and the decrease in NK and CD3(+)CD56(+)NKT cells suggest a shift from innate to adaptive hepatic immunity in the liver graft.
Collapse
Affiliation(s)
- Jens M Werner
- University Hospital Regensburg, Department of Surgery, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
McGettrick HM, Buckley CD, Filer A, Rainger GE, Nash GB. Stromal cells differentially regulate neutrophil and lymphocyte recruitment through the endothelium. Immunology 2011; 131:357-70. [PMID: 20518822 PMCID: PMC2992690 DOI: 10.1111/j.1365-2567.2010.03307.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Stromal fibroblasts modify the initial recruitment of leucocytes by endothelial cells (EC), but their effects on subsequent transendothelial migration remain unclear. Here, EC and dermal or synovial fibroblasts were cultured on opposite surfaces of 3-μm pore filters and incorporated in static or flow-based migration assays. Fibroblasts had little effect on tumour necrosis factor-α-induced transendothelial migration of neutrophils, but tended to increase the efficiency of migration away from the endothelium. Surprisingly, similar close contact between EC and fibroblasts strongly reduced lymphocyte migration in static assays, and nearly abolished stable lymphocyte adhesion from flow. Fibroblasts did not alter endothelial surface expression of adhesion molecules or messenger RNA for chemokines. Inhibition of attachment did not occur when EC-fibroblast contact was restricted by using 0.4-μm pore filters, but under these conditions pre-treatment with heparinase partially inhibited adhesion. In the 3-μm pore co-cultures, inhibition of metalloproteinase activity partially recovered lymphocyte adhesion, but addition of CXCL12 (SDF-1α) to the endothelial surface did not. Hence, the ability of EC to present activating chemokines for lymphocytes may have been enzymatically inhibited by direct contact with fibroblasts. To avoid contact, we cultured EC and fibroblasts on separate 3-μm pore filters one above the other. Here, fibroblasts promoted the transendothelial migration of lymphocytes. Fibroblasts generate CXCL12, but blockade of CXCL12 receptor had no effect on lymphocyte migration. While stromal cells can provide signal(s) promoting leucocyte migration away from the sub-endothelial space, direct cell contact (which might occur in damaged tissue) may cause disruption of chemokine signalling, specifically inhibiting lymphocyte rather than neutrophil recruitment.
Collapse
Affiliation(s)
- Helen M McGettrick
- Centre for Cardiovascular Sciences, School of Clinical and Experimental Medicine, Medical School, The University of Birmingham, Birmingham, UK.
| | | | | | | | | |
Collapse
|
42
|
Pérez-Garay M, Arteta B, Pagès L, de Llorens R, de Bolòs C, Vidal-Vanaclocha F, Peracaula R. alpha2,3-sialyltransferase ST3Gal III modulates pancreatic cancer cell motility and adhesion in vitro and enhances its metastatic potential in vivo. PLoS One 2010; 5. [PMID: 20824144 PMCID: PMC2931708 DOI: 10.1371/journal.pone.0012524] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 07/31/2010] [Indexed: 01/15/2023] Open
Abstract
Background Cell surface sialylation is emerging as an important feature of cancer cell metastasis. Sialyltransferase expression has been reported to be altered in tumours and may account for the formation of sialylated tumour antigens. We have focused on the influence of alpha-2,3-sialyltransferase ST3Gal III in key steps of the pancreatic tumorigenic process. Methodology/Principal Findings ST3Gal III overexpressing pancreatic adenocarcinoma cell lines Capan-1 and MDAPanc-28 were generated. They showed an increase of the tumour associated antigen sialyl-Lewisx. The transfectants' E-selectin binding capacity was proportional to cell surface sialyl-Lewisx levels. Cellular migration positively correlated with ST3Gal III and sialyl-Lewisx levels. Moreover, intrasplenic injection of the ST3Gal III transfected cells into athymic nude mice showed a decrease in survival and higher metastasis formation when compared to the mock cells. Conclusion In summary, the overexpression of ST3Gal III in these pancreatic adenocarcinoma cell lines underlines the role of this enzyme and its product in key steps of tumour progression such as adhesion, migration and metastasis formation.
Collapse
Affiliation(s)
- Marta Pérez-Garay
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona, Spain
| | - Beatriz Arteta
- Department of Cell Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Spain
| | - Lluís Pagès
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona, Spain
| | - Rafael de Llorens
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona, Spain
| | - Carme de Bolòs
- Cancer Research Program, IMIM-Hospital del Mar, Barcelona, Spain
| | - Fernando Vidal-Vanaclocha
- Department of Cell Biology and Histology, School of Medicine and Dentistry, Basque Country University, Leioa, Spain
| | - Rosa Peracaula
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona, Girona, Spain
- * E-mail:
| |
Collapse
|
43
|
van Poll D, Nahmias Y, Soto-Gutierrez A, Ghasemi M, Yagi H, Kobayashi N, Yarmush ML, Hertl M. Human immune reactivity against liver sinusoidal endothelial cells from GalTα(1,3)GalT-deficient pigs. Cell Transplant 2010; 19:783-9. [PMID: 20573304 DOI: 10.3727/096368910x508898] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Elimination of galactose-α(1,3)galactose (Gal) expression in pig organs has been previously shown to prevent hyperacute xenograft rejection. However, naturally present antibodies to non-Gal epitopes activate endothelial cells, leading to acute humoral xenograft rejection. Still, it is unknown whether xenogeneic pig liver sinusoidal endothelial cells (LSECs) from α(1,3)galactosyltransferase (GalT)-deficient pigs are damaged by antibody and complement-mediated mechanisms. The present study examined the xeno-antibody response of LSECs from GalT-deficient and wild pigs. Isolated LSEC from wild-type and GalT pigs were expose to human and baboon sera; IgM and IgG binding was analyzed by flow cytometry. Complement activation (C3a and CH50) was quantified in vitro from serum-exposed LSEC cultures using Enzyme-Linked ImmunoSorbent assay (ELISA). Levels of complement-activated cytotoxicity (CAC) were also determined by a fluorescent Live-Dead Assay and by the quantification of LDH release. IgM binding to GalT knockout (KO) LSECs was significantly lower (80% human and 87% baboon) compare to wild-type pig LSEC. IgG binding was low in all groups. Moreover, complement activation (C3a and CH50) levels released following exposure to human or baboon sera were importantly reduced (42% human and 52% baboon), CAC in GalT KO LSECs was reduced by 60% in human serum and by 72% in baboon serum when compared to wild-type LSECs, and LDH release levels were reduced by 37% and 57%, respectively. LSECs from GalT KO pigs exhibit a significant protection to humoral-induced cell damage compared to LSECs from wild pigs when exposed to human serum. Although insufficient to inhibit xenogeneic reactivity completely, transgenic GalT KO expression on pig livers might contribute to a successful application of clinical xenotransplantation in combination with other protective strategies.
Collapse
Affiliation(s)
- Daan van Poll
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and the Shriners Hospitals for Children, Center for Engineering in Medicine, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Aspinall AI, Curbishley SM, Lalor PF, Weston CJ, Blahova M, Liaskou E, Adams RM, Holt AP, Adams DH. CX(3)CR1 and vascular adhesion protein-1-dependent recruitment of CD16(+) monocytes across human liver sinusoidal endothelium. Hepatology 2010; 51:2030-9. [PMID: 20512991 PMCID: PMC2919204 DOI: 10.1002/hep.23591] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED The liver contains macrophages and myeloid dendritic cells (mDCs) that are critical for the regulation of hepatic inflammation. Most hepatic macrophages and mDCs are derived from monocytes recruited from the blood through poorly understood interactions with hepatic sinusoidal endothelial cells (HSECs). Human CD16(+) monocytes are thought to contain the precursor populations for tissue macrophages and mDCs. We report that CD16(+) cells localize to areas of active inflammation and fibrosis in chronic inflammatory liver disease and that a unique combination of cell surface receptors promotes the transendothelial migration of CD16(+) monocytes through human HSECs under physiological flow. CX(3)CR1 activation was the dominant pertussis-sensitive mechanism controlling transendothelial migration under flow, and expression of the CX(3)CR1 ligand CX(3)CL1 is increased on hepatic sinusoids in chronic inflammatory liver disease. Exposure of CD16(+) monocytes to immobilized purified CX(3)CL1 triggered beta1-integrin-mediated adhesion to vascular cell adhesion molecule-1 and induced the development of a migratory phenotype. Following transmigration or exposure to soluble CX(3)CL1, CD16(+) monocytes rapidly but transiently lost expression of CX(3)CR1. Adhesion and transmigration across HSECs under flow was also dependent on vascular adhesion protein-1 (VAP-1) on the HSECs. CONCLUSION Our data suggest that CD16(+) monocytes are recruited by a combination of adhesive signals involving VAP-1 and CX(3)CR1 mediated integrin-activation. Thus a novel combination of surface molecules, including VAP-1 and CX(3)CL1 promotes the recruitment of CD16(+) monocytes to the liver, allowing them to localize at sites of chronic inflammation and fibrosis.
Collapse
Affiliation(s)
| | - Stuart M. Curbishley
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| | - Patricia F. Lalor
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| | - Chris J. Weston
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| | - Miroslava Blahova
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| | - Evaggelia Liaskou
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| | - Rebecca M Adams
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| | - Andrew P. Holt
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT,The Liver Unit, Queen Elizabeth Hospital, Edgbaston, Birmingham, UK
| | - David H. Adams
- Centre for Liver Research and NIHR Biomedical Research Unit, College of Medicine and Dentistry, Division of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, UK. B15 2TT
| |
Collapse
|
45
|
Abstract
Chemokines direct leukocyte trafficking and positioning within tissues, thus playing critical roles in regulating immune responses and inflammation. The chemokine system is complex, involving interactions between multiple chemokines and their receptors that operate in combinatorial cascades with adhesion molecules. The involvement of multiple chemokines and chemokine receptors in these processes brings flexibility and specificity to recruitment. The hepatic vascular bed is a unique low-flow environment through which leukocytes are recruited to the liver during homeostatic immune surveillance and in response to infection or injury. The rate of leukocyte recruitment and the nature of cells recruited through the sinusoids in response to inflammatory signals will shape the severity of disease. At one end of the spectrum, fulminant liver failure results from a rapid recruitment of leukocytes that leads to hepatocyte destruction and liver failure; at the other end, diseases such as chronic hepatitis C infection may progress over many years from hepatitis to fibrosis and cirrhosis. Chronic hepatitis is characterized by a T lymphocyte-rich infiltrate and the nature and outcome of hepatitis will depend on the T cell subsets recruited, their activation and function within the liver. Different subsets of effector T cells have been described based on their secretion of cytokines and specific functions. These include Th1 and Th2 cells, and more recently Th17 and Th9 cells, which are associated with different types of immune response and which express distinct patterns of chemokine receptors that promote their recruitment under particular conditions. The effector function of these cells is balanced by the recruitment of regulatory T cells that are able to suppress antigen-specific effectors to allow resolution of immune responses and restoration of immune homeostasis. Understanding the signals that are responsible for recruiting different lymphocyte subsets to the liver will elucidate disease pathogenesis and open up new therapeutic approaches to modulate recruitment in favor of resolution rather than injury.
Collapse
Affiliation(s)
| | | | - David H. Adams
- *Prof. David H. Adams, MD, FRCP, FmedSci, 5th Floor, Institute of Biomedical Research, University of Birmingham Medical School, Wolfson Drive, Edgbaston, Birmingham B15 2TT (UK), Tel. +44 121 415 8702, Fax +44 121 415 8701, E-Mail
| |
Collapse
|
46
|
Guzzardi MA, Vozzi F, Ahluwalia AD. Study of the Crosstalk Between Hepatocytes and Endothelial Cells Using a Novel Multicompartmental Bioreactor: A Comparison Between Connected Cultures and Cocultures. Tissue Eng Part A 2009; 15:3635-44. [DOI: 10.1089/ten.tea.2008.0695] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Maria Angela Guzzardi
- Institute of Clinical Physiology-CNR, Pisa, Italy
- Sector of Medicine, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Federico Vozzi
- Institute of Clinical Physiology-CNR, Pisa, Italy
- Faculty of Engineering, Interdepartmental Research Center “E.Piaggio,” University of Pisa, Pisa, Italy
| | - Arti Devi Ahluwalia
- Faculty of Engineering, Interdepartmental Research Center “E.Piaggio,” University of Pisa, Pisa, Italy
| |
Collapse
|
47
|
Crosby HA, Lalor PF, Ross E, Newsome PN, Adams DH. Adhesion of human haematopoietic (CD34+) stem cells to human liver compartments is integrin and CD44 dependent and modulated by CXCR3 and CXCR4. J Hepatol 2009; 51:734-49. [PMID: 19703720 DOI: 10.1016/j.jhep.2009.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Revised: 06/09/2009] [Accepted: 06/16/2009] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIMS Haematopoietic stem cells (HSC) have previously been shown in some studies to migrate to damaged and diseased liver where a small proportion will engraft. Such cells can promote liver repair in rodent models of liver injury and lead to improved liver function in uncontrolled clinical studies. In order to maximize the engraftment of cells for clinical applications it is necessary to understand the molecular mechanisms that regulate stem cell recruitment and retention. Our aim therefore was to determine which factors where involved in adhesion of circulating HSC to liver endothelium and sequestration around epithelial cells within the liver. METHODS We examined the ability of CD34+ populations from peripheral and mobilized blood and the CD34-expressing cell line KG1a to bind to human hepatic sinusoidal endothelial (HSEC) and biliary epithelial cells (BEC) in vitro. RESULTS We report that all CD34(+) populations express alpha4beta1, beta2 integrins and CD44. Liver tissue sections and primary liver cells expressed the corresponding ligands VCAM-1/fibronectin, ICAM-1 and CD44. Pertussis toxin was shown to decrease binding of CD34(+) cells and the cells migrated to CXCR3 and CXCR4 ligands. CONCLUSIONS CD34(+) populations use alpha4beta1, beta2 integrins and CD44 receptors to bind to the ligands VCAM-1/fibronectin, ICAM-1, and hyaluronic acid expressed on sinusoidal vessels in tissue sections and to primary human HSEC. Binding to BEC was mediated by the interaction of beta1 and beta2 integrins with VCAM-1 and ICAM-1 respectively. A role for chemokines is supported by our finding that pertussis toxin inhibits CD34(+) cell adhesion to BEC and HSEC and by the ability of CD34(+) cells to migrate to CXCR3 and CXCR4 ligands.
Collapse
Affiliation(s)
- Heather A Crosby
- Centre for Liver Research and NIHR Biomedical Research Unit for Liver Disease, University of Birmingham and Queen Elizabeth Hospital Birmingham, Birmingham B15 2TT, UK.
| | | | | | | | | |
Collapse
|
48
|
Dash A, Inman W, Hoffmaster K, Sevidal S, Kelly J, Obach RS, Griffith LG, Tannenbaum SR. Liver tissue engineering in the evaluation of drug safety. Expert Opin Drug Metab Toxicol 2009; 5:1159-74. [PMID: 19637986 PMCID: PMC4110978 DOI: 10.1517/17425250903160664] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Assessment of drug-liver interactions is an integral part of predicting the safety profile of new drugs. Existing model systems range from in vitro cell culture models to FDA-mandated animal tests. Data from these models often fail, however, to predict human liver toxicity, resulting in costly failures of clinical trials. In vitro screens based on cultured hepatocytes are now commonly used in early stages of development, but many toxic responses in vivo seem to be mediated by a complex interplay among several different cell types. We discuss some of the evolving trends in liver cell culture systems applied to drug safety assessment and describe an experimental model that captures complex liver physiology through incorporation of heterotypic cell-cell interactions, 3D architecture and perfused flow. We demonstrate how heterotypic interactions in this system can be manipulated to recreate an inflammatory environment and apply the model to test compounds that potentially exhibit idiosyncratic drug toxicity. Finally, we provide a perspective on how the range of existing and emerging in vitro liver culture approaches, from simple to complex, might serve needs across the range of stages in drug discovery and development, including applications in molecular therapeutics.
Collapse
Affiliation(s)
- Ajit Dash
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| | - Walker Inman
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| | - Keith Hoffmaster
- Novartis Institute of Biomedical Research, 350 Massachusetts Avenue, Cambridge, Massachusetts, MA 02139, USA
| | - Samantha Sevidal
- Pfizer Research Technology Center, Cambridge, Massachusetts, MA 02139, USA
| | - Joan Kelly
- Pfizer Research Technology Center, Cambridge, Massachusetts, MA 02139, USA
| | - R Scott Obach
- Pfizer Research Technology Center, Cambridge, Massachusetts, MA 02139, USA
| | - Linda G Griffith
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| | - Steven R Tannenbaum
- Underwood-Prescott Professor of Toxicology and Chemistry, Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Office 56-731A, Cambridge, MA 02139, USA
| |
Collapse
|
49
|
Oo YH, Adams DH. The role of chemokines in the recruitment of lymphocytes to the liver. J Autoimmun 2009; 34:45-54. [PMID: 19744827 DOI: 10.1016/j.jaut.2009.07.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Accepted: 07/29/2009] [Indexed: 12/13/2022]
Abstract
Chemokines direct leukocyte trafficking and positioning within tissues. They thus play critical roles in regulating immune responses and inflammation. The chemokine system is complex involving interactions between multiple chemokines and their receptors that operate in combinatorial cascades with adhesion molecules. The involvement of multiple chemokines and chemokine receptors in these processes brings flexibility and specificity to recruitment. The hepatic vascular bed is a unique low flow environment through which leukocyte are recruited to the liver during homeostatic immune surveillance and in response to infection or injury. The rate of leukocyte recruitment and the nature of cells recruited through the sinusoids in response to inflammatory signals will shape the severity of disease. At one end of the spectrum fulminant liver failure results from a rapid recruitment of leukocytes that leads to hepatocyte destruction and liver failure at the other diseases such as chronic hepatitis C infection may progress over many years from hepatitis to fibrosis and cirrhosis. Chronic hepatitis is charactezised by a T lymphocyte rich infiltrate and the nature and outcome of hepatitis will depend on the T cell subsets recruited, their activation and function within the liver. Different subsets of effector T cells have been described based on their secretion of cytokines and specific functions. These include Th1 and Th2 cells and more recently Th17 and Th9 cells which are associated with different types of immune response and which express distinct patterns of chemokine receptors that promote their recruitment under particular conditions. The effector function of these cells is balanced by the recruitment of regulatory T cells that are able to suppress antigen-specific effectors to allow resolution of immune responses and restoration of immune homeostasis. Understanding the signals that are responsible for recruiting different lymphocyte subsets to the liver will elucidate disease pathogenesis and open up new therapeutic approaches to modulate recruitment in favour of resolution rather than injury.
Collapse
Affiliation(s)
- Ye H Oo
- Centre for Liver Research, 5th Floor, Institute of Biomedical Research, University of Birmingham, Wolfson Drive, Edgbaston, Birmingham B15 2TT, UK
| | | |
Collapse
|
50
|
Borchers AT, Shimoda S, Bowlus C, Keen CL, Gershwin ME. Lymphocyte recruitment and homing to the liver in primary biliary cirrhosis and primary sclerosing cholangitis. Semin Immunopathol 2009; 31:309-22. [PMID: 19533132 PMCID: PMC2758172 DOI: 10.1007/s00281-009-0167-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 05/27/2009] [Indexed: 02/07/2023]
Abstract
The mechanisms operating in lymphocyte recruitment and homing to liver are reviewed. A literature review was performed on primary biliary cirrhosis (PBC), progressive sclerosing cholangitis (PSC), and homing mechanisms; a total of 130 papers were selected for discussion. Available data suggest that in addition to a specific role for CCL25 in PSC, the CC chemokines CCL21 and CCL28 and the CXC chemokines CXCL9 and CXCL10 are involved in the recruitment of T lymphocytes into the portal tract in PBC and PSC. Once entering the liver, lymphocytes localize to bile duct and retain by the combinatorial or sequential action of CXCL12, CXCL16, CX3CL1, and CCL28 and possibly CXCL9 and CXCL10. The relative importance of these chemokines in the recruitment or the retention of lymphocytes around the bile ducts remains unclear. The available data remain limited but underscore the importance of recruitment and homing.
Collapse
Affiliation(s)
- Andrea T Borchers
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6510, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|