1
|
Lao Y, Li Y, Wang W, Ren L, Qian X, He F, Chen X, Jiang Y. A Cytological Atlas of the Human Liver Proteome from PROTEOME SKY-LIVER Hu 2.0, a Publicly Available Database. J Proteome Res 2022; 21:1916-1929. [PMID: 35820117 DOI: 10.1021/acs.jproteome.2c00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The liver plays a unique role as a metabolic center of the body, and also performs other important functions such as detoxification and immune response. Here, we establish a cell type-resolved healthy human liver proteome including hepatocytes (HCs), hepatic stellate cells (HSCs), Kupffer cells (KCs), and liver sinusoidal endothelial cells (LSECs) by high-resolution mass spectrometry. Overall, we quantify total 8354 proteins for four cell types and over 6000 proteins for each cell type. Analysis of this data set and regulatory pathway reveals the cellular labor division in the human liver follows the pattern that parenchymal cells make the main components of pathways, but nonparenchymal cells trigger these pathways. Human liver cells show some novel molecular features: HCs maintain KCs and LSECs homeostasis by producing cholesterol and ketone bodies; HSCs participate in xenobiotics metabolism as an agent deliverer; KCs and LSECs mediate immune response through MHC class II-TLRs and MHC class I-TGFβ cascade, respectively; and KCs play a central role in diurnal rhythms regulation through sensing diurnal IGF and temperature flux. Together, this work expands our understandings of liver physiology and provides a useful resource for future analyses of normal and diseased livers.
Collapse
Affiliation(s)
- Yuanxiang Lao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yanyan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wei Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Liangliang Ren
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xiaohong Qian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Xinguo Chen
- Institute of Liver Transplantation, The Third Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Ying Jiang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.,Anhui Medical University, Hefei 230031, China
| |
Collapse
|
2
|
Wu X, Roberto JB, Knupp A, Greninger AL, Truong CD, Hollingshead N, Kenerson HL, Tuefferd M, Chen A, Koelle DM, Horton H, Jerome KR, Polyak SJ, Yeung RS, Crispe IN. Response of Human Liver Tissue to Innate Immune Stimuli. Front Immunol 2022; 13:811551. [PMID: 35355993 PMCID: PMC8959492 DOI: 10.3389/fimmu.2022.811551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Precision-cut human liver slice cultures (PCLS) have become an important alternative immunological platform in preclinical testing. To further evaluate the capacity of PCLS, we investigated the innate immune response to TLR3 agonist (poly-I:C) and TLR4 agonist (LPS) using normal and diseased liver tissue. Pathological liver tissue was obtained from patients with active chronic HCV infection, and patients with former chronic HCV infection cured by recent Direct-Acting Antiviral (DAA) drug therapy. We found that hepatic innate immunity in response to TLR3 and TLR4 agonists was not suppressed but enhanced in the HCV-infected tissue, compared with the healthy controls. Furthermore, despite recent HCV elimination, DAA-cured liver tissue manifested ongoing abnormalities in liver immunity: sustained abnormal immune gene expression in DAA-cured samples was identified in direct ex vivo measurements and in TLR3 and TLR4 stimulation assays. Genes that were up-regulated in chronic HCV-infected liver tissue were mostly characteristic of the non-parenchymal cell compartment. These results demonstrated the utility of PCLS in studying both liver pathology and innate immunity.
Collapse
Affiliation(s)
- Xia Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.,Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jessica B Roberto
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Allison Knupp
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Institute, Seattle, WA, United States
| | - Camtu D Truong
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Nicole Hollingshead
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Heidi L Kenerson
- Department of Surgery, University of Washington, Seattle, WA, United States
| | - Marianne Tuefferd
- Infectious Diseases and Vaccines, Janssen Research and Development, Beerse, Belgium
| | - Antony Chen
- Infectious Diseases and Vaccines, Janssen Research and Development, Beerse, Belgium
| | - David M Koelle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.,Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Institute, Seattle, WA, United States.,Department of Translational Research, Benaroya Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Helen Horton
- Infectious Diseases and Vaccines, Janssen Research and Development, Beerse, Belgium
| | - Keith R Jerome
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Institute, Seattle, WA, United States
| | - Stephen J Polyak
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, WA, United States
| | - Ian N Crispe
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| |
Collapse
|
3
|
Xia GQ, Cai JN, Wu X, Fang Q, Zhao N, Lv XW. The mechanism by which ATP regulates alcoholic steatohepatitis through P2X4 and CD39. Eur J Pharmacol 2022; 916:174729. [PMID: 34973190 DOI: 10.1016/j.ejphar.2021.174729] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022]
Abstract
Alcoholic liver disease caused by chronic excessive drinking has become one of the most common types of liver disease. Alcohol-induced inflammatory immune responses play a central role in the development of alcohol-associated steatohepatitis. The content and expression of ATP and P2X4 in the livers of alcoholic steatohepatitis mice are significantly increased. The content of ATP increased by 20 percent and the expression of P2X4 receptor protein was 1.3 times higher than that in the livers of normal mice. Treatment with 5-BDBD, a P2X4 receptor-specific inhibitor, significantly reduced alcohol-induced liver inflammation and lipid deposition. In RAW264.7 cell experiments, 5-BDBD inhibited the expression of P2X4 and alleviated alcohol-induced inflammation, while the CD39-specific inhibitor POM-1 reduced extracellular ATP degradation and promoted the expression of P2X4, thereby exacerbating inflammation. After treatment with 5-BDBD, P2X4 receptor protein expression decreased by 0.2 times and after treatment with POM-1, P2X4 receptor protein expression increased by 0.1 times compared to the alcohol-stimulated group. In addition, inhibition of P2X4 expression in RAW264.7 cells reduced calcium influx in RAW264.7 cells. P2X4 may induce the activation of NLRP3 inflammasomes by mediating calcium influx, thus exacerbating the inflammatory response, and inhibition of P2X4 expression can effectively block this process. Conclusion: These results suggest that the ATP-P2X4 signaling pathway promotes the inflammatory response in alcoholic steatohepatitis and that CD39 may play a protective role in regulating P2X4 expression by hydrolyzing ATP. In conclusion, the CD39 and ATP-P2X4 signaling pathways may be potential therapeutic targets for alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Guo-Qing Xia
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Jun-Nan Cai
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Xue Wu
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Qian Fang
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Ning Zhao
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China
| | - Xiong-Wen Lv
- Institute for Liver Diseases of Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, China.
| |
Collapse
|
4
|
Zhang L, Mosoian A, Schwartz ME, Florman SS, Gunasekaran G, Schiano T, Fiel MI, Jiang W, Shen Q, Branch AD, Bansal MB. HIV infection modulates IL-1β response to LPS stimulation through a TLR4-NLRP3 pathway in human liver macrophages. J Leukoc Biol 2019; 105:783-795. [PMID: 30776150 DOI: 10.1002/jlb.4a1018-381r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 01/17/2019] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
IL-1β is an important mediator of innate inflammatory responses and has been shown to contribute to liver injury in a number of etiologies. HIV patients have increased necroinflammation and more rapid fibrosis progression in chronic liver injury compared to non-HIV-infected patients. As the resident liver macrophage is critical to the IL-1β response to microbial translocation in chronic liver disease, we aim to examine the impact of HIV-1 and LPS stimulation on the IL-1β response of the resident hepatic macrophages. We isolated primary human liver macrophages from liver resection specimens, treated them with HIV-1BaL and/or LPS ex vivo, examined the IL-1β response, and then studied underlying mechanisms. Furthermore, we examined IL-1β expression in liver tissues derived from HIV-1 patients compared to those with no underlying liver disease. HIV-1 up-regulated TLR4 and CD14 expression on isolated primary CD68+ human liver macrophages and contributed to the IL-1β response to LPS stimulation as evidenced by TLR4 blocking. Nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) was shown to be involved in the IL-1β response of liver macrophages to HIV-1 infection and NLRP3 blocking experiments in primary CD68+ liver macrophages confirmed the contribution of the NLRP3-caspase 1 inflammatory signaling pathway in the IL-1β response. High in situ IL-1β expression was found in CD68+ cells in human liver tissues from HIV-1-infected patients, suggesting a critical role of IL-1β responses in patients infected by HIV. HIV infection sensitizes the IL-1β response of liver macrophages to LPS through up-regulation of CD14 and TLR4 expression and downstream activation of the NLRP3-caspase 1 pathway. These findings have implications for enhanced immune activation in HIV+ patients and mechanisms for rapid fibrosis progression in patients with chronic liver injury.
Collapse
Affiliation(s)
- Lumin Zhang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Arevik Mosoian
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Myron E Schwartz
- Recanti-Miler Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sander S Florman
- Recanti-Miler Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ganesh Gunasekaran
- Recanti-Miler Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas Schiano
- Recanti-Miler Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - M Isabel Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Qi Shen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrea D Branch
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Meena B Bansal
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
5
|
Kandathil AJ, Sugawara S, Goyal A, Durand CM, Quinn J, Sachithanandham J, Cameron AM, Bailey JR, Perelson AS, Balagopal A. No recovery of replication-competent HIV-1 from human liver macrophages. J Clin Invest 2018; 128:4501-4509. [PMID: 30198905 PMCID: PMC6159970 DOI: 10.1172/jci121678] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Long-lived HIV-1 reservoirs that persist despite antiretroviral therapy (ART) are a major impediment to a cure for HIV-1. We examined whether human liver macrophages (LMs), the largest tissue macrophage population, comprise an HIV-1 reservoir. We purified LMs from liver explants and included treatment with a T cell immunotoxin to reduce T cells to 1% or less. LMs were purified from 9 HIV-1-infected persons, 8 of whom were on ART (range 8-140 months). Purified LMs were stimulated ex vivo and supernatants from 6 of 8 LMs from persons on ART transmitted infection. However, HIV-1 propagation from LMs was not sustained except in LMs from 1 person taking ART for less than 1 year. Bulk liver sequences matched LM-derived HIV-1 in 5 individuals. Additional in vitro experiments undertaken to quantify the decay of HIV-1-infected LMs from 3 healthy controls showed evidence of infection and viral release for prolonged durations (>170 days). Released HIV-1 propagated robustly in target cells, demonstrating that viral outgrowth was observable using our methods. The t1/2 of HIV-1-infected LMs ranged from 3.8-55 days. These findings suggest that while HIV-1 persists in LMs during ART, it does so in forms that are inert, suggesting that they are defective or restricted with regard to propagation.
Collapse
Affiliation(s)
| | - Sho Sugawara
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ashish Goyal
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Jeffrey Quinn
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Andrew M. Cameron
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin R. Bailey
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Ashwin Balagopal
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
6
|
Feng H, Ou B, Dong W, Thasler WE. Preparation and Culture of Human Liver Resident Immune Cells. ACTA ACUST UNITED AC 2018; 80:e50. [PMID: 30133963 DOI: 10.1002/cpcb.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Co-cultivation of tumor cells and liver resident immune cells or other non-parenchymal cells (NPCs) from the same donor is important for the study of cancer metastasis. So far, little is known about the mechanism of tumor cell or pathogen clearance, leukocyte infiltration, and immune cell recruitment in the human liver. To investigate these processes in vitro, the use of primary human hepatocytes and non-parenchymal cell, especially immune cell, co-culture systems play essential roles in the establishment of cell-cell and cell-extracellular matrix communications similar to native liver tissues. Hepatic non-parenchymal cells mainly comprise liver sinusoid endothelial cells (LSECs), microvascular endothelial cells, hepatic stellate cells, Kupffer cells (KCs), natural killer T (iNKT) cells, and dendritic cells (DCs). Here we describe procedures for preparation, isolation, and culture of human liver resident immune cells and other non-parenchymal cells. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Hao Feng
- Department of General Surgery, Ruijin Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China.,Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Baochi Ou
- Shanghai General Hospital affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Wei Dong
- Heart Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wolfgang E Thasler
- Department of General and Visceral Surgery, Red Cross Hospital, Munich, Germany
| |
Collapse
|
7
|
Green CJ, Parry SA, Gunn PJ, Ceresa CDL, Rosqvist F, Piché ME, Hodson L. Studying non-alcoholic fatty liver disease: the ins and outs of in vivo, ex vivo and in vitro human models. Horm Mol Biol Clin Investig 2018; 41:/j/hmbci.ahead-of-print/hmbci-2018-0038/hmbci-2018-0038.xml. [PMID: 30098284 DOI: 10.1515/hmbci-2018-0038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing. Determining the pathogenesis and pathophysiology of human NAFLD will allow for evidence-based prevention strategies, and more targeted mechanistic investigations. Various in vivo, ex situ and in vitro models may be utilised to study NAFLD; but all come with their own specific caveats. Here, we review the human-based models and discuss their advantages and limitations in regards to studying the development and progression of NAFLD. Overall, in vivo whole-body human studies are advantageous in that they allow for investigation within the physiological setting, however, limited accessibility to the liver makes direct investigations challenging. Non-invasive imaging techniques are able to somewhat overcome this challenge, whilst the use of stable-isotope tracers enables mechanistic insight to be obtained. Recent technological advances (i.e. normothermic machine perfusion) have opened new opportunities to investigate whole-organ metabolism, thus ex situ livers can be investigated directly. Therefore, investigations that cannot be performed in vivo in humans have the potential to be undertaken. In vitro models offer the ability to perform investigations at a cellular level, aiding in elucidating the molecular mechanisms of NAFLD. However, a number of current models do not closely resemble the human condition and work is ongoing to optimise culturing parameters in order to recapitulate this. In summary, no single model currently provides insight into the development, pathophysiology and progression across the NAFLD spectrum, each experimental model has limitations, which need to be taken into consideration to ensure appropriate conclusions and extrapolation of findings are made.
Collapse
Affiliation(s)
- Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Siôn A Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Pippa J Gunn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Carlo D L Ceresa
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fredrik Rosqvist
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Marie-Eve Piché
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Quebec Heart and Lung Institute, Laval University, Quebec, Canada
| | - Leanne Hodson
- University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Churchill Hospital,Old Road Headington, Oxford OX3 7LE, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
8
|
Increased Tim-3 expression alleviates liver injury by regulating macrophage activation in MCD-induced NASH mice. Cell Mol Immunol 2018; 16:878-886. [PMID: 29735977 DOI: 10.1038/s41423-018-0032-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 04/03/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023] Open
Abstract
As an immune checkpoint, Tim-3 plays roles in the regulation of both adaptive and innate immune cells including macrophages and is greatly involved in chronic liver diseases. However, the precise roles of Tim-3 in nonalcoholic steatohepatitis (NASH) remain unstated. In the current study, we analyzed Tim-3 expression on different subpopulations of liver macrophages and further investigated the potential roles of Tim-3 on hepatic macrophages in methionine and choline-deficient diet (MCD)-induced NASH mice. The results of flow cytometry demonstrated the significantly increased expression of Tim-3 on all detected liver macrophage subsets in MCD mice, including F4/80+CD11b+, F4/80+CD68+, and F4/80+CD169+ macrophages. Remarkably, Tim-3 knockout (KO) significantly accelerated MCD-induced liver steatosis, displaying higher serum ALT, larger hepatic vacuolation, more liver lipid deposition, and more severe liver fibrosis. Moreover, compared with wild-type C57BL/6 mice, Tim-3 KO MCD mice demonstrated an enhanced expression of NOX2, NLRP3, and caspase-1 p20 together with increased generation of IL-1β and IL-18 in livers. In vitro studies demonstrated that Tim-3 negatively regulated the production of reactive oxygen species (ROS) and related downstream pro-inflammatory cytokine secretion of IL-1β and IL-18 in macrophages. Exogenous administration of N-Acetyl-L-cysteine (NAC), a small molecular inhibitor of ROS, remarkably suppressed caspase-1 p20 expression and IL-1β and IL-18 production in livers of Tim-3 KO mice, thus significantly reducing the severity of steatohepatitis induced by MCD. In conclusion, Tim-3 is a promising protector in MCD-induced steatohepatitis by controlling ROS and the associated pro-inflammatory cytokine production in macrophages.
Collapse
|
9
|
Wu X, Roberto JB, Knupp A, Kenerson HL, Truong CD, Yuen SY, Brempelis KJ, Tuefferd M, Chen A, Horton H, Yeung RS, Crispe IN. Precision-cut human liver slice cultures as an immunological platform. J Immunol Methods 2018; 455:71-79. [PMID: 29408707 PMCID: PMC6689534 DOI: 10.1016/j.jim.2018.01.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/17/2018] [Accepted: 01/24/2018] [Indexed: 12/16/2022]
Abstract
The liver is the central metabolic organ in the human body, and also plays an essential role in innate and adaptive immunity. While mouse models offer significant insights into immune-inflammatory liver disease, human immunology differs in important respects. It is not easy to address those differences experimentally. Therefore, to improve the understanding of human liver immunobiology and pathology, we have established precision-cut human liver slices to study innate immunity in human tissue. Human liver slices collected from resected livers could be maintained in ex vivo culture over a two-week period. Although an acute inflammatory response accompanied by signs of tissue repair was observed in liver tissue following slicing, the expression of many immune genes stabilized after day 4 and remained stable until day 15. Remarkably, histological evidence of pre-existing liver diseases was preserved in the slices for up to 7 days. Following 7 days of culture, exposure of liver slices to the toll-like receptor (TLR) ligands, TLR3 ligand Poly-I:C and TLR4 ligand LPS, resulted in a robust activation of acute inflammation and cytokine genes. Moreover, Poly-I:C treatment induced a marked antiviral response including increases of interferons IFNB, IL-28B and a group of interferon-stimulated genes. Therefore, precision-cut liver slices emerge as a valuable tool to study human innate immunity.
Collapse
Affiliation(s)
- Xia Wu
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | - Jessica B Roberto
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Allison Knupp
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Heidi L Kenerson
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Camtu D Truong
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Sebastian Y Yuen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Marianne Tuefferd
- Infectious Diseases and Vaccines, Janssen Research and Development, B-2340 Beerse, Belgium
| | - Antony Chen
- Infectious Diseases and Vaccines, Janssen Research and Development, B-2340 Beerse, Belgium
| | - Helen Horton
- Infectious Diseases and Vaccines, Janssen Research and Development, B-2340 Beerse, Belgium
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Ian N Crispe
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
10
|
Hsieh CS, Chuang JH, Chou MH, Kao YH. Dexamethasone restores transforming growth factor-β activated kinase 1 expression and phagocytosis activity of Kupffer cells in cholestatic liver injury. Int Immunopharmacol 2018; 56:310-319. [PMID: 29414666 DOI: 10.1016/j.intimp.2018.01.047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 01/10/2018] [Accepted: 01/30/2018] [Indexed: 12/13/2022]
Abstract
The role of transforming growth factor-β activated kinase 1 (TAK1) in modulating the function of Kupffer cells (KCs) within cholestatic livers remains unclear. This study examined the immunopharmacological action of dexamethasone (DEX) in modulating hepatic TAK1 expression and related signaling activity in a rat model of bile duct ligation-mimicked obstructive jaundice. The in vitro effects of DEX on porcine biliary extract (PBE)-modulated gene expression and phagocytosis of KCs were examined using a rat alveolar macrophage cell line (NR8383 cells). Although DEX therapy did not restore the downregulated TAK1 expression and phosphorylation, it significantly attenuated the upregulation of high-mobility group box 1 expression and caspase-3 activation in whole liver extracts of cholestatic rats, possibly via enhancing extracellular signal-regulated kinase-mediated signaling. Dual immunofluorescence staining of cholestatic livers and western detection on primary KCs isolated from cholestatic livers identified that DEX treatment indeed increased both the expression and phosphorylation levels of TAK1 in the KCs of cholestatic livers. In vitro studies using alveolar NR8383 macrophages with KC-characteristic gene expression further demonstrated that DEX not only repressed the pro-inflammatory cytokine production including with respect to interleukin (IL)-1β and IL-6, but also enhanced gene expression of TAK1 and a phagocytic marker, natural-resistance-associated macrophage protein 1, under PBE-mimicked cholestatic conditions. However, WST-1 assay showed that DEX did not protect NR8383 macrophages against the PBE-induced cytotoxicity. Immunofluorescence visualization of cellular F-actin by phalloidin suggested that DEX sustained the PBE-induced phagocytosis morphology of NR8383 macrophages. In conclusion, DEX treatment may pharmacologically restore the expression and activity of TAK1 in KCs, and sustain the phagocytic phenotype of KCs in cholestatic livers.
Collapse
Affiliation(s)
- Chih-Sung Hsieh
- Department of Pediatric Surgery and Department of Teaching & Research, Pu-Li Christian Hospital, Nantou, Taiwan; Department of Applied Chemistry, National Chi-Nan University, Nantou, Taiwan
| | - Jiin-Haur Chuang
- Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Huei Chou
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Center for General Education, Cheng-Shiu University, Kaohsiung, Taiwan.
| | - Ying-Hsien Kao
- Department of Medical Research, E-Da Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
11
|
Rodrigues A, Santos-Mateus D, Alexandre-Pires G, Valério-Bolas A, Rafael-Fernandes M, Pereira MA, Ligeiro D, de Jesus J, Alves-Azevedo R, Lopes-Ventura S, Santos M, Tomás AM, Pereira da Fonseca I, Santos-Gomes G. Leishmania infantum exerts immunomodulation in canine Kupffer cells reverted by meglumine antimoniate. Comp Immunol Microbiol Infect Dis 2017; 55:42-52. [PMID: 29127992 DOI: 10.1016/j.cimid.2017.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/15/2017] [Accepted: 09/15/2017] [Indexed: 01/21/2023]
Abstract
Kupffer cells (KC) are the liver macrophage population that resides in the hepatic sinusoids and efficiently phagocyte pathogens by establishing an intimate contact with circulating blood. KC constitute the liver host cells in Leishmania infection, nevertheless little is described about their role, apart from their notable contribution in granulomatous inflammation. The present study aims to investigate how canine KC sense and react to the presence of Leishmania infantum promastigotes and amastigotes by evaluating the gene expression of specific innate immune cell receptors and cytokines, as well as the induction of nitric oxide and urea production. Complementarily, the impact of a leishmanicidal drug - meglumine antimoniate (MgA) - in infected KC was also explored. KC revealed to be susceptible to both parasite forms and no major differences were found in the immune response generated. L. infantum parasites seem to interact with KC innate immune receptors and induce an anergic state, promoting immune tolerance and parasite survival. The addition of MgA to infected KC breaks the parasite imposed silence and increased gene expression of Toll-like receptors (TLR) 2 and TLR4, possibly activating downstream pathways. Understanding how KC sense and react to parasite presence could bring new insights into the control or even elimination of canine leishmaniasis.
Collapse
Affiliation(s)
- A Rodrigues
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - D Santos-Mateus
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - G Alexandre-Pires
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - A Valério-Bolas
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - M Rafael-Fernandes
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - M A Pereira
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - D Ligeiro
- IPST-Instituto Português do Sangue e da Transplantação - Centro do sangue e da transplantação de Lisboa, Portugal
| | - J de Jesus
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, Brazil
| | - R Alves-Azevedo
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - S Lopes-Ventura
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal
| | - M Santos
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - A M Tomás
- I3S, Instituto de Investigação e Inovação em Saúde, IBMC, Instituto de Biologia Molecular e Celular and ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - I Pereira da Fonseca
- CIISA, Faculdade de Medicina Veterinária, Universidade de Lisboa, Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - G Santos-Gomes
- Global Health and Tropical Medicine, GHTM, Instituto de Higiene e Medicina Tropical, IHMT, Universidade Nova de Lisboa, UNL, Rua da Junqueira 100, 1349-008 Lisboa, Portugal.
| |
Collapse
|
12
|
Mosoian A, Zhang L, Hong F, Cunyat F, Rahman A, Bhalla R, Panchal A, Saiman Y, Fiel MI, Florman S, Roayaie S, Schwartz M, Branch A, Stevenson M, Bansal MB. Frontline Science: HIV infection of Kupffer cells results in an amplified proinflammatory response to LPS. J Leukoc Biol 2017; 101:1083-1090. [PMID: 27986871 PMCID: PMC5380374 DOI: 10.1189/jlb.3hi0516-242r] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/10/2016] [Accepted: 11/13/2016] [Indexed: 01/09/2023] Open
Abstract
End-stage liver disease is a common cause of non-AIDS-related mortality in HIV+ patients, despite effective anti-retroviral therapies (ARTs). HIV-1 infection causes gut CD4 depletion and is thought to contribute to increased gut permeability, bacterial translocation, and immune activation. Microbial products drain from the gut into the liver via the portal vein where Kupffer cells (KCs), the resident liver macrophage, clear translocated microbial products. As bacterial translocation is implicated in fibrogenesis in HIV patients through unclear mechanisms, we tested the hypothesis that HIV infection of KCs alters their response to LPS in a TLR4-dependent manner. We showed that HIV-1 productively infected KCs, enhanced cell-surface TLR4 and CD14 expression, and increased IL-6 and TNF-α expression, which was blocked by a small molecule TLR4 inhibitor. Our study demonstrated that HIV infection sensitizes KCs to the proinflammatory effects of LPS in a TLR4-dependent manner. These findings suggest that HIV-1-infected KCs and their dysregulated innate immune response to LPS may play a role in hepatic inflammation and fibrosis and represent a novel target for therapy.
Collapse
Affiliation(s)
- Arevik Mosoian
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lumin Zhang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Feng Hong
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Francesc Cunyat
- University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Adeeb Rahman
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Riti Bhalla
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ankur Panchal
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Yedidya Saiman
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - M Isabel Fiel
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sander Florman
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sasan Roayaie
- Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; and
| | - Myron Schwartz
- Recanati Miller Transplant Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA; and
| | - Andrea Branch
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Mario Stevenson
- University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Meena B Bansal
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA;
| |
Collapse
|
13
|
Liu J, Huang X, Werner M, Broering R, Yang D, Lu M. Advanced Method for Isolation of Mouse Hepatocytes, Liver Sinusoidal Endothelial Cells, and Kupffer Cells. Methods Mol Biol 2017; 1540:249-258. [PMID: 27975323 DOI: 10.1007/978-1-4939-6700-1_21] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Separation of pure cell populations from the liver is a prerequisite to study the role of hepatic parenchymal and non-parenchymal cells in liver physiology, pathophysiology, and immunology. Traditional methods for hepatic cell separation usually purify only single cell types from liver specimens. Here, we describe an efficient method that can simultaneously purify populations of hepatocytes (HCs), liver sinusoidal endothelial cells (LSECs), and Kupffer cells (KCs) from a single mouse liver specimen. A liberase-based perfusion technique in combination with a low-speed centrifugation and magnetic-activated cell sorting (MACS) led to the isolation and purification of HCs, KCs, and LSECs with high yields and purity.
Collapse
Affiliation(s)
- Jia Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Xuan Huang
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Melanie Werner
- Department of Gastroenterology and Hepatology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology and Hepatology, University Hospital of Essen, University of Duisburg-Essen, Essen, Germany
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengji Lu
- Institute for Virology, University Hospital of Essen, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany.
| |
Collapse
|
14
|
Sato K, Hall C, Glaser S, Francis H, Meng F, Alpini G. Pathogenesis of Kupffer Cells in Cholestatic Liver Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2238-47. [PMID: 27452297 DOI: 10.1016/j.ajpath.2016.06.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/18/2016] [Accepted: 06/07/2016] [Indexed: 02/08/2023]
Abstract
Kupffer cells are the resident macrophages in the liver. They are located in hepatic sinusoid, which allows them to remove foreign materials, pathogens, and apoptotic cells efficiently. Activated Kupffer cells secrete various mediators, including cytokines and chemokines, to initiate immune responses, inflammation, or recruitment of other liver cells. Bile duct ligation (BDL) surgery in rodents is often studied as an animal model of cholestatic liver disease, characterized by obstruction of bile flow. BDL mice show altered functional activities of Kupffer cells compared with sham-operated mice, including elevated cytokine secretion and impaired bacterial clearance. Various mediators produced by other liver cells can regulate Kupffer cell activation, which suggest that Kupffer cells orchestrate with other liver cells to relay inflammatory signals and to maintain liver homeostasis during BDL-induced liver injury. Blocking or depletion of Kupffer cells, an approach for the treatment of liver diseases, has shown controversial implications. Procedures in Kupffer cell research have limitations and may produce various results in Kupffer cell research. It is important, however, to reveal underlying mechanisms of activation and functions of Kupffer cells, followed by hepatic inflammation and fibrosis. This review summarizes present Kupffer cell studies in cholestatic liver injury.
Collapse
Affiliation(s)
- Keisaku Sato
- Research, Central Texas Veterans Health Care System, Temple, Texas; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White, Temple, Texas
| | - Chad Hall
- Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Academic Research Integration, Department Surgery, Baylor Scott & White Healthcare, Temple, Texas
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, Texas; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White, Temple, Texas
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, Texas; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White, Temple, Texas
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, Texas; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White, Temple, Texas
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, Texas; Department of Medicine, Texas A&M Health Science Center, College of Medicine, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White, Temple, Texas.
| |
Collapse
|
15
|
Kegel V, Deharde D, Pfeiffer E, Zeilinger K, Seehofer D, Damm G. Protocol for Isolation of Primary Human Hepatocytes and Corresponding Major Populations of Non-parenchymal Liver Cells. J Vis Exp 2016:e53069. [PMID: 27077489 DOI: 10.3791/53069] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Beside parenchymal hepatocytes, the liver consists of non-parenchymal cells (NPC) namely Kupffer cells (KC), liver endothelial cells (LEC) and hepatic Stellate cells (HSC). Two-dimensional (2D) culture of primary human hepatocyte (PHH) is still considered as the "gold standard" for in vitro testing of drug metabolism and hepatotoxicity. It is well-known that the 2D monoculture of PHH suffers from dedifferentiation and loss of function. Recently it was shown that hepatic NPC play a central role in liver (patho-) physiology and the maintenance of PHH functions. Current research focuses on the reconstruction of in vivo tissue architecture by 3D- and co-culture models to overcome the limitations of 2D monocultures. Previously we published a method to isolate human liver cells and investigated the suitability of these cells for their use in cell cultures in Experimental Biology and Medicine(1). Based on the broad interest in this technique the aim of this article was to provide a more detailed protocol for the liver cell isolation process including a video, which will allow an easy reproduction of this technique. Human liver cells were isolated from human liver tissue samples of surgical interventions by a two-step EGTA/collagenase P perfusion technique. PHH were separated from the NPC by an initial centrifugation at 50 x g. Density gradient centrifugation steps were used for removal of dead cells. Individual liver cell populations were isolated from the enriched NPC fraction using specific cell properties and cell sorting procedures. Beside the PHH isolation we were able to separate KC, LEC and HSC for further cultivation. Taken together, the presented protocol allows the isolation of PHH and NPC in high quality and quantity from one donor tissue sample. The access to purified liver cell populations could allow the creation of in vivo like human liver models.
Collapse
Affiliation(s)
- Victoria Kegel
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin
| | - Daniela Deharde
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin
| | - Elisa Pfeiffer
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin
| | - Katrin Zeilinger
- Brandenburg Center for Regenerative Therapies, Charité University Medicine Berlin
| | - Daniel Seehofer
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin;
| |
Collapse
|
16
|
Lin Z, Monteiro-Riviere NA, Kannan R, Riviere JE. A computational framework for interspecies pharmacokinetics, exposure and toxicity assessment of gold nanoparticles. Nanomedicine (Lond) 2016; 11:107-19. [DOI: 10.2217/nnm.15.177] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: To develop a comprehensive computational framework to simulate tissue distribution of gold nanoparticles (AuNP) across several species. Materials & methods: This framework was built on physiologically based pharmacokinetic modeling, calibrated and evaluated with multiple independent datasets. Results: Rats and pigs seem to be more appropriate models than mice in animal-to-human extrapolation of AuNP pharmacokinetics and that the dose and age should be considered. Incorporation of in vitro and/or in vivo cellular uptake and toxicity data into the model improved toxicity assessment of AuNP. Conclusion: These results partially explain the current low translation rate of nanotechnology-based drug delivery systems from mice to humans. This simulation approach may be applied to other nanomaterials and provides guidance to design future translational studies.
Collapse
Affiliation(s)
- Zhoumeng Lin
- Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, KS 66506, USA
| | - Nancy A Monteiro-Riviere
- Nanotechnology Innovation Center of Kansas State (NICKS), Kansas State University, Manhattan, KS 66506, USA
| | - Raghuraman Kannan
- Department of Radiology, University of Missouri, Columbia, MO 65211, USA
| | - Jim E Riviere
- Institute of Computational Comparative Medicine (ICCM), Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
17
|
All-In-One: Advanced preparation of Human Parenchymal and Non-Parenchymal Liver Cells. PLoS One 2015; 10:e0138655. [PMID: 26407160 PMCID: PMC4583235 DOI: 10.1371/journal.pone.0138655] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 09/02/2015] [Indexed: 02/06/2023] Open
Abstract
Background & Aims Liver cells are key players in innate immunity. Thus, studying primary isolated liver cells is necessary for determining their role in liver physiology and pathophysiology. In particular, the quantity and quality of isolated cells are crucial to their function. Our aim was to isolate a large quantity of high-quality human parenchymal and non-parenchymal cells from a single liver specimen. Methods Hepatocytes, Kupffer cells, liver sinusoidal endothelial cells, and stellate cells were isolated from liver tissues by collagenase perfusion in combination with low-speed centrifugation, density gradient centrifugation, and magnetic-activated cell sorting. The purity and functionality of cultured cell populations were controlled by determining their morphology, discriminative cell marker expression, and functional activity. Results Cell preparation yielded the following cell counts per gram of liver tissue: 2.0±0.4×107 hepatocytes, 1.8±0.5×106 Kupffer cells, 4.3±1.9×105 liver sinusoidal endothelial cells, and 3.2±0.5×105 stellate cells. Hepatocytes were identified by albumin (95.5±1.7%) and exhibited time-dependent activity of cytochrome P450 enzymes. Kupffer cells expressed CD68 (94.5±1.2%) and exhibited phagocytic activity, as determined with 1μm latex beads. Endothelial cells were CD146+ (97.8±1.1%) and exhibited efficient uptake of acetylated low-density lipoprotein. Hepatic stellate cells were identified by the expression of α-smooth muscle actin (97.1±1.5%). These cells further exhibited retinol (vitamin A)-mediated autofluorescence. Conclusions Our isolation procedure for primary parenchymal and non-parenchymal liver cells resulted in cell populations of high purity and quality, with retained physiological functionality in vitro. Thus, this system may provide a valuable tool for determining liver function and disease.
Collapse
|
18
|
Hudspeth K, Donadon M, Cimino M, Pontarini E, Tentorio P, Preti M, Hong M, Bertoletti A, Bicciato S, Invernizzi P, Lugli E, Torzilli G, Gershwin ME, Mavilio D. Human liver-resident CD56(bright)/CD16(neg) NK cells are retained within hepatic sinusoids via the engagement of CCR5 and CXCR6 pathways. J Autoimmun 2015; 66:40-50. [PMID: 26330348 DOI: 10.1016/j.jaut.2015.08.011] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023]
Abstract
RATIONALE The liver-specific natural killer (NK) cell population is critical for local innate immune responses, but the mechanisms that lead to their selective homing and the definition of their functionally relevance remain enigmatic. OBJECTIVES We took advantage of the availability of healthy human liver to rigorously define the mechanisms regulating the homing of NK cells to liver and the repertoire of receptors that distinguish liver-resident NK (lr-NK) cells from circulating counterparts. FINDINGS Nearly 50% of the entire liver NK cell population is composed of functionally relevant CD56(bright) lr-NK cells that localize within hepatic sinusoids. CD56(bright) lr-NK cells express CD69, CCR5 and CXCR6 and this unique repertoire of chemokine receptors is functionally critical as it determines selective migration in response to the chemotactic stimuli exerted by CCL3, CCL5 and CXCL16. Here, we also show that hepatic sinusoids express CCL3(pos) Kupffer cells, CXCL16(pos) endothelial cells and CCL5(pos) T and NK lymphocytes. The selective presence of these chemokines in sinusoidal spaces creates a unique tissue niche for lr-CD56(bright) NK cells that constitutively express CCR5 and CXCR6. CD56(bright) lr-NK cells co-exist with CD56(dim) conventional NK (c-NK) cells that are, interestingly, transcriptionally and phenotypically similar to their peripheral circulating counterparts. Indeed, CD56(dim) c-NK cells lack expression of CD69, CCR5, and CXCR6 but express selectins, integrins and CX3CR1. CONCLUSION Our findings disclosing the phenotypic and functional differences between lr-Nk cells and c-NK cells are critical to distinguish liver-specific innate immune responses. Hence, any therapeutic attempts at modifying the large population of CD56(bright) lr-NK cells will require modification of hepatic CCR5 and CXCR6.
Collapse
Affiliation(s)
- Kelly Hudspeth
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Matteo Donadon
- Department of Hepatobiliary & General Surgery, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Matteo Cimino
- Department of Hepatobiliary & General Surgery, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Elena Pontarini
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Paolo Tentorio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Max Preti
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Michelle Hong
- Viral Hepatitis Laboratory, Singapore Institute for Clinical Sciences, Agency of Science; Technology and Research (A*STAR), 169587, Singapore
| | - Antonio Bertoletti
- Viral Hepatitis Laboratory, Singapore Institute for Clinical Sciences, Agency of Science; Technology and Research (A*STAR), 169587, Singapore
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Pietro Invernizzi
- Liver Unit and Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, 20089 Rozzano, Italy; Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA 95616, USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Guido Torzilli
- Department of Hepatobiliary & General Surgery, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - M Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA 95616, USA.
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20089 Rozzano, Italy.
| |
Collapse
|
19
|
Pfeiffer E, Kegel V, Zeilinger K, Hengstler JG, Nüssler AK, Seehofer D, Damm G. Featured Article: Isolation, characterization, and cultivation of human hepatocytes and non-parenchymal liver cells. Exp Biol Med (Maywood) 2015; 240:645-656. [PMID: 25394621 PMCID: PMC4935273 DOI: 10.1177/1535370214558025] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 09/18/2014] [Indexed: 02/06/2023] Open
Abstract
Primary human hepatocytes (PHH) are considered to be the gold standard for in vitro testing of xenobiotic metabolism and hepatotoxicity. However, PHH cultivation in 2D mono-cultures leads to dedifferentiation and a loss of function. It is well known that hepatic non-parenchymal cells (NPC), such as Kupffer cells (KC), liver endothelial cells (LEC), and hepatic stellate cells (HSC), play a central role in the maintenance of PHH functions. The aims of the present study were to establish a protocol for the simultaneous isolation of human PHH and NPC from the same tissue specimen and to test their suitability for in vitro co-culture. Human PHH and NPC were isolated from tissue obtained by partial liver resection by a two-step EDTA/collagenase perfusion technique. The obtained cell fractions were purified by Percoll density gradient centrifugation. KC, LEC, and HSC contained in the NPC fraction were separated using specific adherence properties and magnetic activated cell sorting (MACS®). Identified NPC revealed a yield of 1.9 × 10(6) KC, 2.7 × 10(5) LEC and 4.7 × 10(5) HSC per gram liver tissue, showing viabilities >90%. Characterization of these NPC showed that all populations went through an activation process, which influenced the cell fate. The activation of KC strongly depended on the tissue quality and donor anamnesis. KC became activated in culture in association with a loss of viability within 4-5 days. LEC lost specific features during culture, while HSC went through a transformation process into myofibroblasts. The testing of different culture conditions for HSC demonstrated that they can attenuate, but not prevent dedifferentiation in vitro. In conclusion, the method described allows the isolation and separation of PHH and NPC in high quality and quantity from the same donor.
Collapse
Affiliation(s)
- Elisa Pfeiffer
- Department for General, Visceral and Transplantation Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Victoria Kegel
- Department for General, Visceral and Transplantation Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Katrin Zeilinger
- Bioreactor Group, Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jan G Hengstler
- IfADo - Leibniz Research Centre for Working Environment and Human Factors at Dortmund Technical University, 44139 Dortmund, Germany
| | - Andreas K Nüssler
- Eberhard-Karls University Tübingen, BG Trauma Center, 72076 Tübingen, Germany
| | - Daniel Seehofer
- Department for General, Visceral and Transplantation Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Georg Damm
- Department for General, Visceral and Transplantation Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
20
|
Zhu DQ, Li PZ. Role of Kupffer cells in bacterial infectious diseases. Shijie Huaren Xiaohua Zazhi 2015; 23:1776-1783. [DOI: 10.11569/wcjd.v23.i11.1776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Kupffer cells (KCs) are also known as liver inherent macrophages, which account for the largest part of human tissue macrophages and participate in the pathogenesis of various liver diseases. In vitro study using primary culture is a valuable tool for the exploration of specific immunological functions of KCs. Obtaining KCs with high purity and activity is the basis for research. A large number of phagocytosable particles and soluble substances can activate KCs by binding to specific receptors on the membrane. The most important molecule that activates KCs is lipopolysaccharide (LPS). A tiny quantity of LPS will drive a Toll-like receptor 4 (TLR4) -dependent proinflammatory response that alerts the host to the presence of infection. Higher quantities of LPS, which reach the cytoplasm, will trigger inflammasome activation, interleukin-1 beta (IL-1β) production and, ultimately, cell death. KCs play an important role in sepsis, endotoxin tolerance and acute pancreatitis. In this review, we describe the role of KCs in these diseases and the underlying molecular mechanisms.
Collapse
|
21
|
Green CJ, Pramfalk C, Morten KJ, Hodson L. From whole body to cellular models of hepatic triglyceride metabolism: man has got to know his limitations. Am J Physiol Endocrinol Metab 2015; 308:E1-20. [PMID: 25352434 PMCID: PMC4281685 DOI: 10.1152/ajpendo.00192.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The liver is a main metabolic organ in the human body and carries out a vital role in lipid metabolism. Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases, encompassing a spectrum of conditions from simple fatty liver (hepatic steatosis) through to cirrhosis. Although obesity is a known risk factor for hepatic steatosis, it remains unclear what factor(s) is/are responsible for the primary event leading to retention of intrahepatocellular fat. Studying hepatic processes and the etiology and progression of disease in vivo in humans is challenging, not least as NAFLD may take years to develop. We present here a review of experimental models and approaches that have been used to assess liver triglyceride metabolism and discuss their usefulness in helping to understand the aetiology and development of NAFLD.
Collapse
Affiliation(s)
- Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford United Kingdom; and
| | - Camilla Pramfalk
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford United Kingdom; and
| | - Karl J Morten
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford United Kingdom; and
| |
Collapse
|
22
|
Kitani H, Sakuma C, Takenouchi T, Sato M, Yoshioka M, Yamanaka N. Establishment of c-myc-immortalized Kupffer cell line from a C57BL/6 mouse strain. RESULTS IN IMMUNOLOGY 2014; 4:68-74. [PMID: 25379377 DOI: 10.1016/j.rinim.2014.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 08/18/2014] [Accepted: 08/19/2014] [Indexed: 01/10/2023]
Abstract
We recently demonstrated in several mammalian species, a novel procedure to obtain liver-macrophages (Kupffer cells) in sufficient numbers and purity using a mixed primary culture of hepatocytes. In this study, we applied this method to the C57BL/6 mouse liver and established an immortalized Kupffer cell line from this mouse strain. The hepatocytes from the C57BL/6 adult mouse liver were isolated by a two-step collagenase perfusion method and cultured in T25 culture flasks. Similar to our previous studies, the mouse hepatocytes progressively changed their morphology into a fibroblastic appearance after a few days of culture. After 7-10 days of culture, Kupffer-like cells, which were contaminants in the hepatocyte fraction at the start of the culture, actively proliferated on the mixed fibroblastic cell sheet. At this stage, a retroviral vector containing the human c-myc oncogene and neomycin resistance gene was introduced into the mixed culture. Gentle shaking of the culture flask, followed by the transfer and brief incubation of the culture supernatant, resulted in a quick and selective adhesion of Kupffer cells to a plastic dish surface. After selection with G418 and cloning by limiting dilutions, a clonal cell line (KUP5) was established. KUP5 cells displayed typical macrophage morphology and were stably passaged at 4-5 days intervals for more than 5 months, with a population doubling time of 19 h. KUP5 cells are immunocytochemically positive for mouse macrophage markers, such as Mac-1, F4/80. KUP5 cells exhibited substantial phagocytosis of polystyrene microbeads and the release of inflammatory cytokines upon lipopolysaccharide stimulation. Taken together, KUP5 cells provide a useful means to study the function of Kupffer cells in vitro.
Collapse
Affiliation(s)
- Hiroshi Kitani
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Chisato Sakuma
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Takato Takenouchi
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Mitsuru Sato
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Miyako Yoshioka
- Safety Research Team, National Institute of Animal Health, Kannondai 3-1-5, Tsukuba, Ibaraki 305-0856, Japan
| | - Noriko Yamanaka
- Safety Research Team, National Institute of Animal Health, Kannondai 3-1-5, Tsukuba, Ibaraki 305-0856, Japan
| |
Collapse
|
23
|
Fletcher NF, Sutaria R, Jo J, Barnes A, Blahova M, Meredith LW, Cosset FL, Curbishley SM, Adams DH, Bertoletti A, McKeating JA. Activated macrophages promote hepatitis C virus entry in a tumor necrosis factor-dependent manner. Hepatology 2014; 59:1320-30. [PMID: 24259385 PMCID: PMC4255687 DOI: 10.1002/hep.26911] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/19/2013] [Indexed: 02/06/2023]
Abstract
UNLABELLED Macrophages are critical components of the innate immune response in the liver. Chronic hepatitis C is associated with immune infiltration and the infected liver shows a significant increase in total macrophage numbers; however, their role in the viral life cycle is poorly understood. Activation of blood-derived and intrahepatic macrophages with a panel of Toll-like receptor agonists induce soluble mediators that promote hepatitis C virus (HCV) entry into polarized hepatoma cells. We identified tumor necrosis factor α (TNF-α) as the major cytokine involved in this process. Importantly, this effect was not limited to HCV; TNF-α increased the permissivity of hepatoma cells to infection by Lassa, measles and vesicular stomatitis pseudoviruses. TNF-α induced a relocalization of tight junction protein occludin and increased the lateral diffusion speed of HCV receptor tetraspanin CD81 in polarized HepG2 cells, providing a mechanism for their increased permissivity to support HCV entry. High concentrations of HCV particles could stimulate macrophages to express TNF-α, providing a direct mechanism for the virus to promote infection. CONCLUSION This study shows a new role for TNF-α to increase virus entry and highlights the potential for HCV to exploit existing innate immune responses in the liver to promote de novo infection events.
Collapse
Affiliation(s)
- Nicola F Fletcher
- Hepatitis C Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Rupesh Sutaria
- NIHR Liver Biomedical Research Unit, University of BirminghamBirmingham, UK
| | - Juandy Jo
- Viral Hepatitis Laboratory, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*STAR)Singapore
| | - Amy Barnes
- Hepatitis C Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | - Miroslava Blahova
- NIHR Liver Biomedical Research Unit, University of BirminghamBirmingham, UK
| | - Luke W Meredith
- Hepatitis C Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK
| | | | | | - David H Adams
- NIHR Liver Biomedical Research Unit, University of BirminghamBirmingham, UK
| | - Antonio Bertoletti
- Viral Hepatitis Laboratory, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*STAR)Singapore
| | - Jane A McKeating
- Hepatitis C Research Group, Institute for Biomedical Research, University of BirminghamBirmingham, UK,NIHR Liver Biomedical Research Unit, University of BirminghamBirmingham, UK
| |
Collapse
|
24
|
Kitani H, Yoshioka M, Takenouchi T, Sato M, Yamanaka N. Characterization of the liver-macrophages isolated from a mixed primary culture of neonatal swine hepatocytes. RESULTS IN IMMUNOLOGY 2014; 4:1-7. [PMID: 24707456 PMCID: PMC3973824 DOI: 10.1016/j.rinim.2014.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 12/13/2022]
Abstract
We recently developed a novel procedure to obtain liver-macrophages in sufficient number and purity using a mixed primary culture of rat and bovine hepatocytes. In this study, we aim to apply this method to the neonatal swine liver. Swine parenchymal hepatocytes were isolated by a two-step collagenase perfusion method and cultured in T75 culture flasks. Similar to the rat and bovine cells, the swine hepatocytes retained an epithelial cell morphology for only a few days and progressively changed into fibroblastic cells. After 5–13 days of culture, macrophage-like cells actively proliferated on the mixed fibroblastic cell sheet. Gentle shaking of the culture flask followed by the transfer and brief incubation of the culture supernatant resulted in a quick and selective adhesion of macrophage-like cells to a plastic dish surface. After rinsing dishes with saline, the attached macrophage-like cells were collected at a yield of 106 cells per T75 culture flask at 2–3 day intervals for more than 3 weeks. The isolated cells displayed a typical macrophage morphology and were strongly positive for macrophage markers, such as CD172a, Iba-1 and KT022, but negative for cytokeratin, desmin and a-smooth muscle actin, indicating a highly purified macrophage population. The isolated cells exhibited phagocytosis of polystyrene microbeads and a release of inflammatory cytokines upon lipopolysaccharide stimulation. This shaking and attachment method is applicable to the swine liver and provides a sufficient number of macrophages without any need of complex laboratory equipments.
Collapse
Key Words
- Attachment
- CK, cytokeratin
- DAPI, 4′,6-diamidino-2-phenylindole
- DES, desmin
- DMEM, Dulbecco’s modified Eagle’s medium
- ELISA, enzyme-linked immunosorbent assay
- EMT, epithelial to mesenchymal transition
- FACS, fluorescent activated cell sorter
- Hepatocyte culture
- Isolation
- LPS, lipopolysaccharide
- M-CSF, macrophage colony-stimulating factor
- Macrophages
- SMA, α-smooth muscle actin
- Shaking
- Swine
Collapse
Affiliation(s)
- Hiroshi Kitani
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Miyako Yoshioka
- Safety Research Team, National Institute of Animal Health, Kannondai 3-1-5, Tsukuba, Ibaraki 305-0856, Japan
| | - Takato Takenouchi
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Mitsuru Sato
- Animal Immune and Cell Biology Research Unit, National Institute of Agrobiological Sciences, Ohwashi 1-2, Tsukuba, Ibaraki 305-8634, Japan
| | - Noriko Yamanaka
- Safety Research Team, National Institute of Animal Health, Kannondai 3-1-5, Tsukuba, Ibaraki 305-0856, Japan
| |
Collapse
|
25
|
Li PZ, Li JZ, Li M, Gong JP, He K. An efficient method to isolate and culture mouse Kupffer cells. Immunol Lett 2013; 158:52-6. [PMID: 24333337 DOI: 10.1016/j.imlet.2013.12.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 01/22/2023]
Abstract
Kupffer cells (KCs) play an essential role in the physiological and pathological functions of the liver. Although the isolation methods of KCs have been well-described, most of them are sophisticated and time-consuming. In addition, these methods are mainly used for isolating the KCs of the human and rat. In this study, a three-step procedure was applied to isolate KCs in sufficient number and purity from mouse liver, including the techniques of enzymatic tissue treatment, gradient centrifugation, and selective adherence. F4/80 immunofluorescence and flow cytometry were used for cell identification. The combination method resulted in a satisfactorily high yield of 5-6×10(6) KCs per liver, over 92.0% positive for F4/80 and 98.5% viable cells. After 24h of culturing, the KCs showed typical macrophage morphologic features such as irregular shape, transparent cytoplasm and kidney-like nucleus. The phagocytic assay showed that the isolated cells exhibited strong phagocytosis activity. The KCs we isolated were functionally intact and exhibited a concentration dependent TNF-α production induced by LPS. The method we described is an effective method to isolate mouse KCs in high purity and yield, which consuming fewer collagenase and time without altering the functional capacity of the KCs.
Collapse
Affiliation(s)
- Pei-zhi Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jin-zheng Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Min Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jian-ping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Kun He
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
26
|
Damm G, Pfeiffer E, Burkhardt B, Vermehren J, Nüssler AK, Weiss TS. Human parenchymal and non-parenchymal liver cell isolation, culture and characterization. Hepatol Int 2013; 7:951-958. [PMID: 26202025 DOI: 10.1007/s12072-013-9475-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/11/2013] [Indexed: 02/06/2023]
Abstract
Many reports describing parenchymal liver cell isolation have been published so far. However, recent evidence has clearly demonstrated that non-parenchymal liver cells play an important role in many pathophysiologies of the liver, such as drug-induced liver diseases, inflammation, and the development of liver fibrosis and cirrhosis. In this study, we present an overview of the current methods for isolating and characterizing parenchymal and non-parenchymal liver cells.
Collapse
Affiliation(s)
- Georg Damm
- Charité University Medicine Berlin, Department of General, Visceral, and Transplant Surgery, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Elisa Pfeiffer
- Charité University Medicine Berlin, Department of General, Visceral, and Transplant Surgery, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Britta Burkhardt
- Eberhard Karls University Tübingen, BG Trauma Center, Siegfried Weller Institut, BG-Tübingen, Siegfried Weller Institut, Schnarrenbergstr. 95, 72076, Tübingen, Germany
| | - Jan Vermehren
- Department of Pediatrics and Juvenile Medicine, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany
| | - Andreas K Nüssler
- Eberhard Karls University Tübingen, BG Trauma Center, Siegfried Weller Institut, BG-Tübingen, Siegfried Weller Institut, Schnarrenbergstr. 95, 72076, Tübingen, Germany.
| | - Thomas S Weiss
- Department of Pediatrics and Juvenile Medicine, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053, Regensburg, Germany.
| |
Collapse
|
27
|
Steib CJ, Gmelin L, Pfeiler S, Schewe J, Brand S, Göke B, Gerbes AL. Functional relevance of the cannabinoid receptor 2 - heme oxygenase pathway: a novel target for the attenuation of portal hypertension. Life Sci 2013; 93:543-51. [PMID: 24007798 DOI: 10.1016/j.lfs.2013.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 12/17/2022]
Abstract
AIMS In liver cirrhosis, inflammation triggers portal hypertension. Kupffer cells (KC) produce vasoconstrictors upon activation by bacterial constituents. Here, we hypothesize that the anti-inflammatory action of the cannabinoid receptor 2 (CB2) agonists JWH-133 and GP 1a attenuate portal hypertension. MAIN METHODS In vivo measurements of portal pressures and non-recirculating liver perfusions were performed in rats 4weeks after bile duct ligation (BDL). Zymosan (150μg/ml, isolated liver perfusion) or LPS (4mg/kgb.w., in vivo) was infused to activate the KC in the absence or presence of JWH-133 (10mg/kgb.w.), GP 1a (2.5mg/kgb.w.) or ZnPP IX (1μM). Isolated KC were treated with Zymosan (0.5mg/ml) in addition to JWH-133 (5μM). The thromboxane (TX) B2 levels in the perfusate and KC media were determined by ELISA. Heme oxygenase-1 (HO-1) and CB2 were analyzed by Western blot or confocal microscopy. KEY FINDINGS JWH-133 or GP 1a pre-treatment attenuated portal pressures following KC activation in all experimental settings. In parallel, HO-1 expression increased with JWH-133 pre-treatment. However, the inhibition of HO-1 enhanced portal hypertension, indicating the functional role of this novel pathway. In isolated KC, the expression of CB2 and HO-1 increased with Zymosan, LPS and JWH-133 treatment while TXB2 production following KC activation was attenuated by JWH-133 pre-treatment. SIGNIFICANCE JWH-133 or GP 1a treatment attenuates portal hypertension. HO-1 induction by JWH-133 plays a functional role. Therefore, the administration of JWH-133 or GP 1a represents a promising new treatment option for portal hypertension triggered by microbiological products.
Collapse
Key Words
- (6aR,10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran
- 2-Chloro-5-nitro-N-phenylbenzamide
- BDL
- Bile duct ligation (BDL)
- CB(2)
- GW 9662
- Gp 1a
- HO
- Heme oxygenase (HO)
- JWH-133
- KC
- Kupffer cell
- LDH
- LPS
- N-(Piperidin-1-yl)-1-(2,4-dichlorophenyl)-1,4-dihydro-6-methylindeno[1,2-c]pyrazole-3-carboxamide
- TLR
- TX
- Thromboxane
- Zy
- Zymosan A
- b. w
- bile duct ligation
- body weight
- cannabinoid receptor 2
- heme oxygenase
- lactate dehydrogenase
- lipopolysaccharide
- thromboxane
- toll like receptor
Collapse
Affiliation(s)
- Christian J Steib
- Department of Medicine II (Gastroenterology and Hepatology), Liver Center Munich, University of Munich, Grosshadern, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
28
|
Chen Y, Tong H, Zhang X, Tang L, Pan Z, Liu Z, Duan P, Su L. Xuebijing injection alleviates liver injury by inhibiting secretory function of Kupffer cells in heat stroke rats. J TRADIT CHIN MED 2013; 33:243-9. [PMID: 23789225 DOI: 10.1016/s0254-6272(13)60133-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To evaluate the effects of Xuebijing (XBJ) injection in heat stroke (HS) rats and to investigate the mechanisms underlying these effects. METHODS Sixty anesthetized rats were randomized into three groups and intravenously injected twice daily for 3 days with 4 mL XBJ (XBJ group) or phosphate buffered saine (HS and Sham groups) per kg body weight. HS was initiated in the HS and XBJ groups by placing rats in a simulated climate chamber (ambient temperature 400C, humidity 60% ). Rectal temperature, aterial pressure, and heart rate were monitored and recorded. Time to HS onset and survival were determined, and serum concentrations of tumor necrosis factor (TNF)-alpha interleukin (IL)-1beta, IL-6, alanine-aminotransferase (ALT), and aspartate-aminotransferase (AST) were measured. Hepatic tissue was harvested for pathological examination and electron microscopic examination. Kupffer cells (KCs) were separated from liver at HS initiation, and the concentrations of secreted TNF-a, IL-beta and IL-6 were measured. RESULTS Time to HS onset and survival were significantly longer in the XBJ than in the HS group. Moreover, the concentrations of TNF-alpha, IL-1beta, IL-6, ALT and AST were lower and liver injury was milder in the XBJ than in the HS group. Heat-stress induced structural changes in KCs and hepatic cells were more severe in the HS than in the XBJ group and the concentrations of TNF-alpha, IL-beta and IL-6 secreted by KCs were lower in the XBJ than in the HS group. CONCLUSION XBJ can alleviate HS-induced systemic inflammatory response syndrome and liver injury in rats, and improve outcomes. These protective effects may be due to the ability of XBJ to inhibit cytokine secretion by KCs.
Collapse
Affiliation(s)
- Yi Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Negash AA, Ramos HJ, Crochet N, Lau DTY, Doehle B, Papic N, Delker DA, Jo J, Bertoletti A, Hagedorn CH, Gale M. IL-1β production through the NLRP3 inflammasome by hepatic macrophages links hepatitis C virus infection with liver inflammation and disease. PLoS Pathog 2013; 9:e1003330. [PMID: 23633957 PMCID: PMC3635973 DOI: 10.1371/journal.ppat.1003330] [Citation(s) in RCA: 353] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 03/15/2013] [Indexed: 12/12/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection is a leading cause of liver disease. Liver inflammation underlies infection-induced fibrosis, cirrhosis and liver cancer but the processes that promote hepatic inflammation by HCV are not defined. We provide a systems biology analysis with multiple lines of evidence to indicate that interleukin-1β (IL-1β) production by intrahepatic macrophages confers liver inflammation through HCV-induced inflammasome signaling. Chronic hepatitis C patients exhibited elevated levels of serum IL-1β compared to healthy controls. Immunohistochemical analysis of healthy control and chronic hepatitis C liver sections revealed that Kupffer cells, resident hepatic macrophages, are the primary cellular source of hepatic IL-1β during HCV infection. Accordingly, we found that both blood monocyte-derived primary human macrophages, and Kupffer cells recovered from normal donor liver, produce IL-1β after HCV exposure. Using the THP-1 macrophage cell-culture model, we found that HCV drives a rapid but transient caspase-1 activation to stimulate IL-1β secretion. HCV can enter macrophages through non-CD81 mediated phagocytic uptake that is independent of productive infection. Viral RNA triggers MyD88-mediated TLR7 signaling to induce IL-1β mRNA expression. HCV uptake concomitantly induces a potassium efflux that activates the NLRP3 inflammasome for IL-1β processing and secretion. RNA sequencing analysis comparing THP1 cells and chronic hepatitis C patient liver demonstrates that viral engagement of the NLRP3 inflammasome stimulates IL-1β production to drive proinflammatory cytokine, chemokine, and immune-regulatory gene expression networks linked with HCV disease severity. These studies identify intrahepatic IL-1β production as a central feature of liver inflammation during HCV infection. Thus, strategies to suppress NLRP3 or IL-1β activity could offer therapeutic actions to reduce hepatic inflammation and mitigate disease. Hepatitis C virus (HCV) causes chronic infection of the liver and is a leading cause of liver inflammation, cirrhosis and liver cancer in nearly 200 million people worldwide. Importantly, hepatic inflammation during chronic HCV infection is considered to be the primary catalyst for progressive liver disease and development of liver cancer. However, the underlying molecular mechanism(s) of HCV-mediated hepatic inflammation are not well understood. The goal of this study was to determine the mechanisms of HCV-induced inflammation. We found that serum IL-1β levels are elevated in chronic hepatitis C patients. Furthermore, we found that hepatic macrophages or Kupffer cells are the major IL-1β-producing cell population within HCV infected livers. Our studies, using the THP1 cell culture model of HCV exposure, reveal that exposure of macrophages to HCV induces IL-1β through a process of infection-independent phagocytic virus uptake that triggers signaling through MyD88/TLR7 and NLRP3 inflammasome pathways to drive IL-1β expression and maturation/secretion, respectively. RNA sequencing (RNA-seq) analysis of patient liver biopsies shows that viral triggering of these signaling pathways drives an inflammatory response linked with liver disease in patients with chronic hepatitis C. Our results identify HCV-induced IL-1β production by hepatic macrophages as a critical and central process that promotes liver inflammation and disease.
Collapse
Affiliation(s)
- Amina A. Negash
- Center for the Study of Hepatitis C Virus Infection and Immunity, Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Hilario J. Ramos
- Center for the Study of Hepatitis C Virus Infection and Immunity, Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Nanette Crochet
- Center for the Study of Hepatitis C Virus Infection and Immunity, Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Daryl T. Y. Lau
- Liver Center, Division of Gastroenterology and Hepatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Brian Doehle
- Center for the Study of Hepatitis C Virus Infection and Immunity, Department of Immunology, University of Washington, Seattle, Washington, United States of America
| | - Neven Papic
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Don A. Delker
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Juandy Jo
- Viral Hepatitis Laboratory, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*STAR), Singapore
- Program Emerging Viral Diseases Unit, Duke-NUS Graduate Medical School, Singapore
| | - Antonio Bertoletti
- Viral Hepatitis Laboratory, Singapore Institute for Clinical Sciences, Agency of Science Technology and Research (A*STAR), Singapore
- Program Emerging Viral Diseases Unit, Duke-NUS Graduate Medical School, Singapore
| | - Curt H. Hagedorn
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - Michael Gale
- Center for the Study of Hepatitis C Virus Infection and Immunity, Department of Immunology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
30
|
Nebbia G, Peppa D, Schurich A, Khanna P, Singh HD, Cheng Y, Rosenberg W, Dusheiko G, Gilson R, ChinAleong J, Kennedy P, Maini MK. Upregulation of the Tim-3/galectin-9 pathway of T cell exhaustion in chronic hepatitis B virus infection. PLoS One 2012; 7:e47648. [PMID: 23112829 PMCID: PMC3480425 DOI: 10.1371/journal.pone.0047648] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 09/14/2012] [Indexed: 12/26/2022] Open
Abstract
The S-type lectin galectin-9 binds to the negative regulatory molecule Tim-3 on T cells and induces their apoptotic deletion or functional inactivation. We investigated whether galectin-9/Tim-3 interactions contribute to the deletion and exhaustion of the antiviral T cell response in chronic hepatitis B virus infection (CHB). We found Tim-3 to be expressed on a higher percentage of CD4 and CD8 T cells from patients with CHB than healthy controls (p<0.0001) and to be enriched on activated T cells and those infiltrating the HBV-infected liver. Direct ex vivo examination of virus-specific CD8 T cells binding HLA-A2/peptide multimers revealed that Tim-3 was more highly upregulated on HBV-specific CD8 T cells than CMV-specific CD8 T cells or the global CD8 T cell population in patients with CHB (p<0.001) or than on HBV-specific CD8 after resolution of infection. T cells expressing Tim-3 had an impaired ability to produce IFN-γ and TNF-α upon recognition of HBV-peptides and were susceptible to galectin-9-triggered cell death in vitro. Galectin-9 was detectable at increased concentrations in the sera of patients with active CHB-related liver inflammation (p = 0.02) and was strongly expressed by Kupffer cells within the liver sinusoidal network. Tim-3 blockade resulted in enhanced expansion of HBV-specific CD8 T cells able to produce cytokines and mediate cytotoxicity in vitro. Blocking PD-1 in combination with Tim-3 enhanced the number of patients from whom functional antiviral responses could be recovered and/or the strength of responses, indicating that these co-inhibitory molecules play a non-redundant role in driving T cell exhaustion in CHB. Patients taking antivirals able to potently suppress HBV viraemia continued to express Tim-3 on their T cells and respond to Tim-3 blockade. In summary, both Tim-3 and galectin-9 are increased in CHB and may contribute to the inhibition and deletion of T cells as they infiltrate the HBV-infected liver.
Collapse
Affiliation(s)
- Gaia Nebbia
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Dimitra Peppa
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Anna Schurich
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Pooja Khanna
- Division of Infection and Immunity, University College London, London, United Kingdom
- Centre for Hepatology, University College London, London, United Kingdom
| | - Harsimran D. Singh
- Centre for Hepatology, University College London, London, United Kingdom
| | - Yang Cheng
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - William Rosenberg
- Centre for Hepatology, University College London, London, United Kingdom
| | - Geoffrey Dusheiko
- Centre for Hepatology, University College London, London, United Kingdom
| | - Richard Gilson
- Centre for Sexual Health and HIV, University College London, London, United Kingdom
| | - Joanne ChinAleong
- Centre for Digestive Diseases, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, London, United Kingdom
| | - Patrick Kennedy
- Centre for Digestive Diseases, Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry, London, United Kingdom
| | - Mala K. Maini
- Division of Infection and Immunity, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
31
|
Role of endothelial dysfunction in modulating the plasma redox homeostasis in visceral leishmaniasis. Biochim Biophys Acta Gen Subj 2011; 1810:652-65. [DOI: 10.1016/j.bbagen.2011.03.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 03/11/2011] [Accepted: 03/31/2011] [Indexed: 01/12/2023]
|
32
|
Kitani H, Yoshioka M, Takenouchi T, Sato M, Yamanaka N. Isolation and characterization of macrophages from a mixed primary culture of bovine liver cells. Vet Immunol Immunopathol 2011; 140:341-5. [PMID: 21334751 DOI: 10.1016/j.vetimm.2011.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 01/06/2011] [Accepted: 01/25/2011] [Indexed: 11/26/2022]
Abstract
Previously, we developed a simple and efficient method to isolate liver macrophages from a mixed primary culture of adult rat liver cells. To extend the applicability of this method, we isolated macrophages from mixed primary cultures of bovine liver cells. Macrophage cells proliferated on the cell sheet of mixed bovine liver cells after 8-16d of culture. These cells were detached by shaking of the culture flasks. Subsequent transfer and brief incubation in plastic dishes resulted in selective adhesion of macrophages. After rinses with PBS, attached macrophages were harvested. More than 10(6) cells could be harvested from the culture flask at intervals of 2-3d for more than three weeks. The isolated cells were strongly positive for bovine macrophage markers, such as CD68, CD172a and Iba-1. These cells exhibited functional properties of macrophages, including active phagocytosis of polystyrene microbeads, proliferative response to recombinant bovine granulocyte-macrophage colony-stimulating factor, upregulation of specific inflammatory cytokine genes upon stimulation with lipopolysaccharide, and formation of multinucleated giant cells. The shaking and attachment method provides a simple and efficient alternative to obtain bovine liver macrophages without requiring complex equipment or specialized technical skills.
Collapse
Affiliation(s)
- Hiroshi Kitani
- Transgenic Animal Research Center, National Institute of Agrobiological Sciences, 1-2 Ohwashi, Tsukuba, Ibaraki 305-8634, Japan.
| | | | | | | | | |
Collapse
|
33
|
A novel isolation method for macrophage-like cells from mixed primary cultures of adult rat liver cells. J Immunol Methods 2010; 360:47-55. [PMID: 20600081 DOI: 10.1016/j.jim.2010.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 06/04/2010] [Accepted: 06/07/2010] [Indexed: 11/21/2022]
Abstract
We report a simple and efficient method to obtain macrophage-like cells from the mixed primary cultures of adult rat liver cells. A parenchymal hepatocyte enriched fraction was prepared from adult rat livers and seeded into culture flasks. After 7 to 10 days of culture, when most hepatocytes were degenerated or transformed into fibroblastic cells, macrophage-like cells vigorously proliferated on the cell sheet. By shaking the flasks, macrophage-like cells were readily detached. Subsequent transfer and incubation in plastic dishes resulted in quick and selective adhesion of macrophage-like cells, while other contaminating cells remained suspended in the medium. After rinsing with saline, attached macrophage-like cells were harvested with 95 to 99% purity, as evaluated by flow cytometry or immunocytochemistry. These cells showed typical macrophage morphology and were strongly positive for markers of rat macrophages, such as ED-1, ED-3, and OX-41, but negative for cytokeratins and alpha-smooth muscle actin. They possessed functional properties of typical macrophages, including active phagocytosis of latex beads, proliferative response to recombinant GM-CSF, secretion of inflammatory and anti-inflammatory cytokines upon stimulation with LPS, and formation of multinucleated giant cells. As more than 10(6) cells can be recovered repeatedly from a T75 culture flask at two to three day intervals for more than two weeks, our procedure might implicate a novel alternative to obtain Kupffer cells in sufficient number and purity without complex equipment and skills.
Collapse
|
34
|
Steib CJ, Bilzer M, op den Winkel M, Pfeiler S, Hartmann AC, Hennenberg M, Göke B, Gerbes AL. Treatment with the leukotriene inhibitor montelukast for 10 days attenuates portal hypertension in rat liver cirrhosis. Hepatology 2010; 51:2086-96. [PMID: 20512996 DOI: 10.1002/hep.23596] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
UNLABELLED The mechanisms underlying intrahepatic vasoconstriction are not fully elucidated. Here we investigated the Kupffer cell (KC)-dependent increase in portal pressure by way of actions of vasoconstrictive cysteinyl leukotrienes (Cys-LTs). Liver cirrhosis was induced in rats by bile duct ligation (BDL for 4 weeks; controls: sham-operation) and thioacetamide application (18 weeks). Infusion of leukotriene (LT) C(4) or LTD(4) in isolated perfused livers (20 nM, BDL and sham) demonstrated that LTC(4) is a more relevant vasoconstrictor. In BDL animals the Cys-LT(1) receptor inhibitor montelukast (1 microM) reduced the maximal portal perfusion pressure following LTC(4) or LTD(4) infusion. The infusion of LTC(4) or D(4) in vivo (15 microg/kg b.w.) confirmed LTC(4) as the more relevant vasoconstrictor. Activation of KCs with zymosan (150 microg/mL) in isolated perfused BDL livers increased the portal perfusion pressure markedly, which was attenuated by LT receptor blockade (Ly171883, 20 microM). Cys-LTs in the effluent perfusate increased with KC activation but less with additional blockade of KCs with gadolinium chloride (10 mg/kg body weight, 48 and 24 hours pretreatment). KCs were isolated from normal rat livers and activated with zymosan or lipopolysaccharide at different timepoints. This resulted in an increase in Cys-LT production that was not influenced by preincubation with montelukast (1 microM). Infusion of LTC(4) (20 nM) and the thromboxane analog U46619 (0.1 microM) further enhanced portal pressure, indicating additive effects. Treatment with montelukast for 10 days resulted in an impressive reduction in the basal portal pressure and an attenuation of the KC-dependent increase in portal pressure. CONCLUSION Activation of isolated KCs produced Cys-LTs. Infusion of Cys-LTs increased portal pressure and, vice versa, treatment with montelukast reduced portal pressure in rat liver cirrhosis. Therefore, montelukast may be of therapeutic benefit for patients with portal hypertension.
Collapse
Affiliation(s)
- Christian J Steib
- Department of Medicine II (Gastroenterology and Hepatology), Liver Center Munich, University of Munich, Grosshadern, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Colorectal cancer is one of the commonest malignancies in the "developed" world. The liver constitutes the main host organ for its distant metastases which, when present, augur a bad prognosis for the disease. Kupffer cells (KCs) are macrophages that constantly reside within the liver and form an effective first line defence against multiple harmful agents which reach the hepatic sinusoids via the portal circulation. KCs remove chemical compounds and dead or damaged cells, eliminate bacteria and protect against invading tumour cells. They may play a crucial tumouricidal role, exerting cytotoxic and cytostatic functions through the release of multiple cytokines and chemokines. Subsequently, colorectal metastasising cells are destroyed either by KC-performed phagocytosis or via the stimulation of other immune cells which migrate into the sinusoids and act accordingly. On the contrary, KC products, including cytokines, growth factors and matrix-degrading enzymes may promote liver metastasis, supporting tumour cell extravasation, motility and invasion. Current research aims to exploit the antineoplastic properties of KCs in new therapeutic approaches of colorectal cancer liver metastasis. Numerous agents, such as the granulocyte macrophage-colony stimulating factor, interferon gamma, muramyl peptide analogues and various antibody based treatments, have been tested in experimental models with promising results. Future trials may investigate their use in everyday clinical practice and compare their therapeutic value with current treatment of the disease.
Collapse
Affiliation(s)
- Konstantinos A Paschos
- Liver Research Group, Section of Oncology, School of Medicine, Royal Hallamshire Hospital, The University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
36
|
Zeng Z, Huang HF, Song F, Duan J. Isolation of rat Kupffer cells by ex vivo perfusion and their primary culture. Shijie Huaren Xiaohua Zazhi 2009; 17:2550-2554. [DOI: 10.11569/wcjd.v17.i25.2550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore a convenient and effective method for isolation and primary culture of rat Kupffer cells (KCs) in vitro.
METHODS: Ex vivo rat liver tissue was perfused with collagenase IV and diced into small pieces. The diced tissue was digested for 30 min at 37℃and centrifuged to remove hepatocytes. KCs were then separated by means of Percoll density gradient centrifugation and selective adherence. Phagocytosis assay and immunocytochemistry for ED2 were used to identify cells isolated.
RESULTS: The average cell yield per gram of liver before plastic adherence was (2.1 ± 0.3) × 106, and (1.5 ± 0.1) × 106following plastic adherence. The viability of KCs isolated was higher than 92% as determined by trypan blue exclusion. The purity of KCs, identified by immunocytochemistry for ED2, was higher than 90%. Cultured KCs were functionally intact and exhibited irregular shape.
CONCLUSION: The method for isolation and culture of Kupffer cells by ex vivo perfusion is convenient, efficient and stable. Cultured Kupffer cells retain naive biological characteristics and can be used in future studies.
Collapse
|