1
|
Hiramatsu K, Ikeda R, Kawaji S, Ueno Y, Nagata R, Hayashi KG, Iga K, Yoshioka M, Takenouchi T. Isolation and propagation of bovine blood-derived macrophages using a mixed culture with bovine endothelial B46 cells. Cell Biol Int 2024; 48:76-83. [PMID: 37920877 DOI: 10.1002/cbin.12102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/01/2023] [Accepted: 10/14/2023] [Indexed: 11/04/2023]
Abstract
Macrophages are innate immune cells with multiple functions such as phagocytosis, cytokine production, and antigen presentation. Since macrophages play critical roles in some bacterial infectious diseases in cattle, including tuberculosis, paratuberculosis, and brucellosis, the in vitro culturing of bovine macrophages is useful for evaluating host-pathogen interactions at the cellular and molecular levels. We have previously reported the establishment of two immortalized bovine liver sinusoidal cell lines, endothelial B46 cells and myofibroblast-like A26 cells (Cell Biology International, 40, 1372-1379, 2016). In this study, we investigated the use of these cell lines as feeder cells that support the proliferation of bovine blood-derived macrophages (BBMs). Notably, the B46 cell line efficiently acts as feeder cells for the propagation of BBMs. Compared with primary cultured vascular endothelial cells, the infinite proliferation ability of B46 cells is more beneficial for preparing confluent feeder layers. In conclusion, this study provides a simple and efficient protocol for the isolation and propagation of BBMs using a primary mixed culture of bovine whole blood with B46 feeder cells. Isolated BBMs are expected to be useful for developing in vitro models for studying the interactions between bovine pathogens and host immune cells.
Collapse
Affiliation(s)
- Kanae Hiramatsu
- National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Rina Ikeda
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Satoko Kawaji
- National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Yuichi Ueno
- National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Reiko Nagata
- National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Ken-Go Hayashi
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Kosuke Iga
- Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Miyako Yoshioka
- National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| |
Collapse
|
2
|
Aktories P, Petry P, Glatz P, Andrieux G, Oschwald A, Botterer H, Gorka O, Erny D, Boerries M, Henneke P, Groß O, Prinz M, Kierdorf K. An improved organotypic cell culture system to study tissue-resident macrophages ex vivo. CELL REPORTS METHODS 2022; 2:100260. [PMID: 36046625 PMCID: PMC9421540 DOI: 10.1016/j.crmeth.2022.100260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/11/2022] [Accepted: 07/06/2022] [Indexed: 12/02/2022]
Abstract
Tissue-resident macrophages (TRMs) perform organ-specific functions that are dependent on factors such as hematopoietic origin, local environment, and biological influences. A diverse range of in vitro culture systems have been developed to decipher TRM functions, including bone marrow-derived macrophages (BMDMs), induced pluripotent stem cell (iPSC)-derived TRMs, or immortalized cell lines. However, despite the usefulness of such systems, there are notable limitations. Attempts to culture primary macrophages often require purification of cells and lack a high cell yield and consistent phenotype. Here, we aimed to address these limitations by establishing an organotypic primary cell culture protocol. We obtained long-term monocultures of macrophages derived from distinct organs without prior purification using specific growth factors and tissue normoxic conditions that largely conserved a TRM-like identity in vitro. Thus, this organotypic system offers an ideal screening platform for primary macrophages from different organs that can be used for a wide range of assays and readouts.
Collapse
Affiliation(s)
- Philipp Aktories
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Philippe Petry
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Paulo Glatz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexander Oschwald
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Hannah Botterer
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Gorka
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daniel Erny
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Berta-Ottenstein-Program for Advanced Clinician Scientists, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Philipp Henneke
- CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, and Center for Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Olaf Groß
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Signaling Research Centers BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, 79104 Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
3
|
Takenouchi T, Masujin K, Miyazaki A, Suzuki S, Takagi M, Kokuho T, Uenishi H. Isolation and immortalization of macrophages derived from fetal porcine small intestine and their susceptibility to porcine viral pathogen infections. Front Vet Sci 2022; 9:919077. [PMID: 35923820 PMCID: PMC9339801 DOI: 10.3389/fvets.2022.919077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Macrophages are a heterogeneous population of cells that are present in all vertebrate tissues. They play a key role in the innate immune system, and thus, in vitro cultures of macrophages provide a valuable model for exploring their tissue-specific functions and interactions with pathogens. Porcine macrophage cultures are often used for the identification and characterization of porcine viral pathogens. Recently, we have developed a simple and efficient method for isolating primary macrophages from the kidneys and livers of swine. Here, we applied this protocol to fetal porcine intestinal tissues and demonstrated that porcine intestinal macrophages (PIM) can be isolated from mixed primary cultures of porcine small intestine-derived cells. Since the proliferative capacity of primary PIM is limited, we attempted to immortalize them by transferring the SV40 large T antigen and porcine telomerase reverse transcriptase genes using lentiviral vectors. Consequently, immortalized PIM (IPIM) were successfully generated and confirmed to retain various features of primary PIM. We further revealed that IPIM are susceptible to infection by the African swine fever virus and the porcine reproductive and respiratory syndrome virus and support their replication. These findings suggest that the IPIM cell line is a useful tool for developing in vitro models that mimic the intestinal mucosal microenvironments of swine, and for studying the interactions between porcine pathogens and host immune cells.
Collapse
Affiliation(s)
- Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
- *Correspondence: Takato Takenouchi
| | - Kentaro Masujin
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
- Kentaro Masujin
| | - Ayako Miyazaki
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Japan
- Ayako Miyazaki
| | - Shunichi Suzuki
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Michihiro Takagi
- Division of Infectious Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Takehiro Kokuho
- Division of Transboundary Animal Disease Research, National Institute of Animal Health, National Agriculture and Food Research Organization, Kodaira, Japan
| | - Hirohide Uenishi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| |
Collapse
|
4
|
Pridans C, Irvine KM, Davis GM, Lefevre L, Bush SJ, Hume DA. Transcriptomic Analysis of Rat Macrophages. Front Immunol 2021; 11:594594. [PMID: 33633725 PMCID: PMC7902030 DOI: 10.3389/fimmu.2020.594594] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/17/2020] [Indexed: 12/13/2022] Open
Abstract
The laboratory rat is widely used as a model for human diseases. Many of these diseases involve monocytes and tissue macrophages in different states of activation. Whilst methods for in vitro differentiation of mouse macrophages from embryonic stem cells (ESC) and bone marrow (BM) are well established, these are lacking for the rat. The gene expression profiles of rat macrophages have also not been characterised to the same extent as mouse. We have established the methodology for production of rat ESC-derived macrophages and compared their gene expression profiles to macrophages obtained from the lung and peritoneal cavity and those differentiated from BM and blood monocytes. We determined the gene signature of Kupffer cells in the liver using rats deficient in macrophage colony stimulating factor receptor (CSF1R). We also examined the response of BM-derived macrophages to lipopolysaccharide (LPS). The results indicate that many, but not all, tissue-specific adaptations observed in mice are conserved in the rat. Importantly, we show that unlike mice, rat macrophages express the CSF1R ligand, colony stimulating factor 1 (CSF1).
Collapse
Affiliation(s)
- Clare Pridans
- Centre for Inflammation Research, University of Edinburgh Centre for Inflammation Research, Edinburgh, United Kingdom
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, United Kingdom
| | - Katharine M. Irvine
- Mater Research Institute Mater Research Institute – University of Queensland, Brisbane, QLD, Australia
| | - Gemma M. Davis
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Lucas Lefevre
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J. Bush
- Nuffield Department of Clinical Medicine, University of Oxford, Headington, United Kingdom
| | - David A. Hume
- Mater Research Institute Mater Research Institute – University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Yoshioka M, Takenouchi T, Kitani H, Guruge KS, Yamanaka N. Synergistic induction of drug-metabolizing enzymes in co-cultures of bovine hepatocytic and sinusoidal cell lines. In Vitro Cell Dev Biol Anim 2019; 56:2-9. [PMID: 31722089 DOI: 10.1007/s11626-019-00408-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/25/2019] [Indexed: 11/26/2022]
Abstract
Hepatocyte-derived cell lines provide useful experimental systems for studying liver metabolism. Unlike human and rodents, few hepatocyte-derived cell lines have been generated from cattle. Here, we established two immortalized bovine hepatocyte-derived cell lines (BH4 and BH5) via transfection with a SV40 large T-antigen construct. Morphological and immunocytochemical analyses revealed that BH4 and BH5 originated from hepatocytes and biliary-epithelial cells, respectively. A potent carcinogen, 3-methylcholanthrene (3-MC), upregulated gene expression of cytochrome P450 (CYP)1A1, CYP1A2, and CYP1B1 in BH4 and BH5, but only to levels less than one-fifteenth of those in primary cultured bovine hepatocytes. Phenobarbital (PB) also increased expression levels of CYP2B6, CYP2C18, and CYP3A4 in BH4 and BH, but at a lower level than 3-MC. By contrast, when BH4 or BH5 was co-cultured with previously established bovine liver sinusoidal cell lines and treated with 3-MC, the gene expression levels of CYP1A1, CYP1A2, and CYP1B1 increased by 38~290%, compared with those in BH4 or BH5 cells cultured alone. PB-treated co-cultures of BH4 or BH5 cells and liver sinusoidal cell lines also showed synergistic increases in CYP2B6 and CYP2C18 expression. Together, the results suggest that these co-cultures could provide an in vitro model for investigations into pharmacological and toxicological properties of drugs in cattle liver.
Collapse
Affiliation(s)
- Miyako Yoshioka
- Pathology and Pathophysiology Research Division, National Institute of Animal Health, NARO, Kannondai 3-1-5, Tsukuba, Ibaraki, 305-0856, Japan.
| | - Takato Takenouchi
- Division of Animal Sciences, Institute of Agrobiological Sciences, NARO, 1-2 Ohwashi, Tsukuba, Ibaraki, 305-8634, Japan
| | - Hiroshi Kitani
- Division of Animal Sciences, Institute of Agrobiological Sciences, NARO, 1-2 Ohwashi, Tsukuba, Ibaraki, 305-8634, Japan
| | - Keerthi S Guruge
- Pathology and Pathophysiology Research Division, National Institute of Animal Health, NARO, Kannondai 3-1-5, Tsukuba, Ibaraki, 305-0856, Japan
| | - Noriko Yamanaka
- Pathology and Pathophysiology Research Division, National Institute of Animal Health, NARO, Kannondai 3-1-5, Tsukuba, Ibaraki, 305-0856, Japan
| |
Collapse
|
6
|
Ogawa K, Tsurutani M, Hashimoto A, Soeda M. Simple propagation method for resident macrophages by co-culture and subculture, and their isolation from various organs. BMC Immunol 2019; 20:34. [PMID: 31533615 PMCID: PMC6749721 DOI: 10.1186/s12865-019-0314-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/04/2019] [Indexed: 12/23/2022] Open
Abstract
Background Resident macrophages (Mø) originating from yolk sac Mø and/or foetal monocytes colonise tissues/organs during embryonic development. They persist into adulthood by self-renewal at a steady state, independent of adult monocyte inputs, except for those in the intestines and dermis. Thus, many resident Mø can be propagated in vitro under optimal conditions; however, there are no specific in vitro culture methods available for the propagation of resident Mø from diverse tissues/organs. Results We provided a simple method for propagating resident Mø derived from the liver, spleen, lung, and brain of ICR male mice by co-culture and subculture along with the propagation of other stromal cells of the respective organs in standard culture media and successfully demonstrated the propagation of resident Mø colonising these organs. We also proposed a simple method for segregating Mø from stromal cells according to their adhesive property on bacteriological Petri dishes, which enabled the collection of more than 97.6% of the resident Mø from each organ. Expression analyses of conventional Mø markers by flow cytometry showed similar expression patterns among the Mø collected from the organs. Conclusion This is the first study to clearly provide a practical Mø propagation method applicable to resident Mø of diverse tissues and organs. Thus, this novel practical Mø propagation method can offer broad applications for the use of resident Mø of diverse tissues and organs.
Collapse
Affiliation(s)
- Kazushige Ogawa
- Laboratory of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano, Osaka, 598-8531, Japan.
| | - Mayu Tsurutani
- Laboratory of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano, Osaka, 598-8531, Japan
| | - Aya Hashimoto
- Laboratory of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano, Osaka, 598-8531, Japan
| | - Miharu Soeda
- Laboratory of Veterinary Anatomy, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano, Osaka, 598-8531, Japan
| |
Collapse
|
7
|
A simple culture method for liver and intestinal tissue-resident macrophages from neonatal mice. In Vitro Cell Dev Biol Anim 2019; 55:436-444. [PMID: 31119642 DOI: 10.1007/s11626-019-00359-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/13/2019] [Indexed: 02/08/2023]
Abstract
The liver and intestine contain a remarkably large portion of tissue-resident macrophage cells representing a phenotype that downregulates inflammation and initiates tissue repair. Here, liver and intestinal tissues obtained from neonatal mice were minced, enzymatically digested, and incubated in RPMI1640-based media. In a 2-wk culture, spherical floating cells emerged on a fibroblastic sheet. These cells showed phagocytic activity and F4/80+-CD11b+-CD206+-Arg1+-iNOS--CD209a- phenotype, suggesting that these cells are tissue-resident macrophages. These macrophages proliferated in the co-culture system in the presence of fibroblastic feeder cell layer and absence of supplemental cytokines; the co-culture system did not cause a significant change in the phenotype of cells grown in a 4-wk culture. On the feeder cells, macrophage density was approximately 1.5 × 104/cm2 and the doubling time was approximately 70 h. Based on these observations, we present a simple method for the isolation and propagation of tissue-resident macrophages resembling M2 macrophage from neonatal mice, and this method provides a useful platform for in vitro studies of tissue-resident macrophages.
Collapse
|
8
|
Suda K, Muraji T, Ohtani H, Aiyoshi T, Sasaki T, Toma M, Yanai T. Histological significance of hepatitis-like findings in biliary atresia: An analysis of 34 Japanese cases. Pediatr Int 2019; 61:364-368. [PMID: 30811786 DOI: 10.1111/ped.13816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/21/2018] [Accepted: 02/23/2019] [Indexed: 11/27/2022]
Abstract
BACKGROUND Hepatocellular injury including multinuclear changes are common histological features in biliary atresia (BA), as well as in neonatal hepatitis. To date, however, no reports have examined how those findings correlate with the prognosis of BA. We clarified the clinical implications of hepatitis-related changes in BA on histological analysis. METHODS We retrospectively reviewed 34 cases of BA treated over the past 30 years at Ibaraki Children's Hospital. Liver biopsy specimens during Kasai procedures were evaluated for hepatocyte multinuclear change, ballooning, and acidophilic body, hereby defined as hepatitis-like findings (HLF). Each finding was semi-quantitatively scored as 0-2, and their sum was defined as the HLF score, ranging from 0 to 6. We examined the correlation between HLF score and total bilirubin (T-Bil), direct bilirubin (D-Bil), and other liver function test results at the Kasai procedure, as well as 1 week, and 1, 3, and 6 months after the Kasai procedure. Subsequently, HLF score was compared between native liver survivors (NLS; n = 16) and non-NLS (n = 18) for long-term analyses. RESULTS Hepatitis-like findings score except for aspartate aminotransferase (AST), had no correlation with the preoperative data. HLF score was positively correlated, however, with T-Bil, D-Bil, and AST at 1 week and 1 month after the Kasai procedure (1 week: P = 0.009, 0.023, and 0.019; 1 month: 0.022, 0.019, and 0.013, respectively). HLF score was not significantly different between the NLS and non-NLS groups. CONCLUSION Higher HLF score at Kasai procedure is an indicator of poor liver function at short-term follow up.
Collapse
Affiliation(s)
- Kazuto Suda
- Department of Pediatric Surgery, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Toshihiro Muraji
- Department of Pediatric Surgery, Ibaraki Children's Hospital, Mito, Ibaraki, Japan.,Department of Pediatric Surgery, Kirishima Medical Center affiliated with Research Field in Medical and Health Sciences, Kagoshima University, Kirishima, Kagoshima, Japan
| | - Haruo Ohtani
- Department of Pathology, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Tsubasa Aiyoshi
- Department of Pediatric Surgery, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Takato Sasaki
- Department of Pediatric Surgery, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Miki Toma
- Department of Pediatric Surgery, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| | - Toshihiro Yanai
- Department of Pediatric Surgery, Ibaraki Children's Hospital, Mito, Ibaraki, Japan
| |
Collapse
|
9
|
Wen J, Wu Y, Wei W, Li Z, Wang P, Zhu S, Dong W. Protective effects of recombinant human cytoglobin against chronic alcohol-induced liver disease in vivo and in vitro. Sci Rep 2017; 7:41647. [PMID: 28128325 PMCID: PMC5269723 DOI: 10.1038/srep41647] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/20/2016] [Indexed: 02/07/2023] Open
Abstract
Alcoholic liver disease (ALD) is an important worldwide public health issue with no satisfying treatment available since now. Here we explore the effects of recombinant human cytoglobin (rhCygb) on chronic alcohol-induced liver injury and the underlying mechanisms. In vivo studies showed that rhCygb was able to ameliorate alcohol-induced liver injury, significantly reversed increased serum index (ALT, AST, TG, TC and LDL-C) and decreased serum HDL-C. Histopathology observation of the liver of rats treated with rhCygb confirmed the biochemical data. Furthermore, rhCygb significantly inhibited Kupffer cells (KCs) proliferation and TNF-α expression in LPS-induced KCs. rhCygb also inhibited LPS-induced NADPH oxidase activity and ROS, NO and O2•- generation. These results collectively indicate that rhCygb exert the protective effect on chronic alcohol-induced liver injury through suppression of KC activation and oxidative stress. In view of its anti-oxidative stress and anti-inflammatory features, rhCygb might be a promising candidate for development as a therapeutic agent against ALD.
Collapse
Affiliation(s)
- Jian Wen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong Province 510515, P.R. China.,Department of Hematology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi Zhuang Autonomous Region 541002, P.R. China
| | - Yongbin Wu
- Department of Clinical Laboratory, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi Zhuang Autonomous Region 541002, P.R. China
| | - Wei Wei
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong Province 510515, P.R. China
| | - Zhen Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong Province 510515, P.R. China
| | - Ping Wang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong Province 510515, P.R. China
| | - Shiwei Zhu
- Department of Hematology, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, Guangxi Zhuang Autonomous Region 541002, P.R. China
| | - Wenqi Dong
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong Province 510515, P.R. China
| |
Collapse
|
10
|
Li Y, Ding X, Fan P, Guo J, Tian X, Feng X, Zheng J, Tian P, Ding C, Xue W. Inactivation of p27 kip1 Promoted Nonspecific Inflammation by Enhancing Macrophage Proliferation in Islet Transplantation. Endocrinology 2016; 157:4121-4132. [PMID: 27631551 DOI: 10.1210/en.2016-1060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Islet transplantation suffers from low efficiency caused by nonspecific inflammation-induced graft loss after transplantation. This study reports increased islet loss and enhanced inflammatory response in p27-deficient mice (p27-/-) and proposes a possible mechanism. Compared with wild type, p27-/- mice showed more severe functional injury of islet, with increased serum levels of inflammatory cytokines IL-1 and TNF-α, inducing macrophage proliferation. Furthermore, the increased number, proapoptotic proteins, and nuclear factor-kappa b (NF-κB) phosphorylation status of the infiltrating macrophages were accompanied by increased TNF-α mRNA level of islet graft site in p27-/- mice. Moreover, in vitro, we found that macrophages were still activated and cocultured with islet and promoted islet loss even blocking the direct effect of TNF-α on islets. Malondialdehyde (MDA, an end product of lipid peroxidation) in islet and media were increased after cocultured with macrophages. p27 deficiency also increased macrophage proliferation and islet injury. Therefore, p27 inactivation promotes injury islet graft loss via the elevation of proliferation and inflammatory cytokines secretion in infiltrating macrophages which induced nonspecific inflammation independent of TNF-α/nuclear factor-kappa b pathway. This potentially represents a promising therapeutic target in improving islet graft survival.
Collapse
Affiliation(s)
- Yang Li
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Xiaoming Ding
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Ping Fan
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Jian Guo
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Xiaohui Tian
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Xinshun Feng
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Jin Zheng
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Puxun Tian
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Chenguang Ding
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| | - Wujun Xue
- Department of Renal Transplantation (Y.L., X.D., X.T., X.F., J.Z., P.T., C.D., W.X.), Center of Nephrology, the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; Institute of Organ Transplantation (Y.L., X.D., X.T, X.F., J.Z., P.T., C.D., W.X.), Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China; and Departments of Rheumatism and Immunology (P.F.) and Hepatobiliary (J.G.), the First Affiliated Hospital Xi'an Jiaotong University, No. 277 West Yanta Street, Xi'an, 710061, People's Republic of China
| |
Collapse
|
11
|
Yoshioka M, Takenouchi T, Kitani H, Okada H, Yamanaka N. Establishment of SV40 large T antigen-immortalized bovine liver sinusoidal cell lines and their immunological responses to deoxynivalenol and lipopolysaccharide. Cell Biol Int 2016; 40:1372-1379. [PMID: 27624824 DOI: 10.1002/cbin.10682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/06/2016] [Indexed: 12/26/2022]
Abstract
Immortalized bovine sinusoidal cell lines provide useful tools to study the immunological responses in the liver to the gastrointestinal tract-derived toxic substances, which may cause systemic symptoms in the affected livestock. Here, we established two immortalized bovine liver sinusoidal cell lines, endothelial-like B46, and myofibroblast-like A26, from primary cultures of bovine liver cells by the transfection with SV40 large T antigen. The pro-inflammatory cytokine responses in these cell lines to deoxynivalenol (DON) and lipopolysaccharide (LPS) were then compared to those in the primary bovine Kupffer cells (BKC). BKC were highly responsive to LPS, showing increased levels of IL-1α, IL-1β, IL-6, and TNF-α mRNA 3 h after stimulation. DON induced similar pro-inflammatory cytokine responses in BKC, except for IL-6. The endothelial B46 cells exhibited upregulation of IL-1α, IL-1β, and IL-6 3 h after stimulation by LPS. In contrast to the stimulation by LPS, B46 had relatively low pro-inflammatory cytokine responses to DON, except for IL-1α, which was moderately induced at 3 h and increased at 24 h after stimulation. The myofibroblast-like A26 cells exhibited low responses in the induction of pro-inflammatory cytokines to LPS or DON; however, the expression of IL-6 was significantly observed 3 h after DON stimulation. Our results suggest that bovine liver sinusoidal cells have distinctive pro-inflammatory cytokine responses against harmful substances, and these immune responses might determine the consequence of systemic inflammations in the diseased animal.
Collapse
Affiliation(s)
- Miyako Yoshioka
- National Institute of Animal Health, NARO, Tsukuba, Ibaraki, Japan
| | | | - Hiroshi Kitani
- Institute of Agrobiological Sciences, NARO, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Okada
- National Institute of Animal Health, NARO, Tsukuba, Ibaraki, Japan
| | - Noriko Yamanaka
- National Institute of Animal Health, NARO, Tsukuba, Ibaraki, Japan
| |
Collapse
|
12
|
Zhang Q, Qu Y, Li Z, Zhang Q, Xu M, Cai X, Li F, Lu L. Isolation and Culture of Single Cell Types from Rat Liver. Cells Tissues Organs 2016; 201:253-67. [DOI: 10.1159/000444672] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 11/19/2022] Open
Abstract
There have been few reports on the simultaneous isolation of multiple liver cell populations thus far. As such, this study was aimed at establishing a protocol for the simultaneous separation of hepatocytes (HCs), hepatic stellate cells (HSCs), liver sinusoidal endothelial cells (LSECs) and Kupffer cells (KCs) from the rat liver and assessing the in vitro culture of these cells. Single-cell suspensions from the liver were obtained by ethylene glycol tetraacetic acid/collagenase perfusion. After low-speed centrifugal separation of HCs, pronase was added to the nonparenchymal cell fraction to eliminate the remaining HCs. Subsequently, HSCs, LSECs and KCs were purified by two steps of density gradient centrifugation using Nycodenz and Percoll in addition to selective attachment. Pronase treatment increased the HSC yield (1.5 ± 0.2 vs. 0.7 ± 0.3 cells/g liver, p < 0.05) and improved LSEC purity (93.6 ± 3.6 vs. 82.5 ± 5.6%, p < 0.01). The isolated cells could also be cultured in vitro. LSEC apoptosis began on day 3 and reached a maximum on day 7. A few surviving LSECs began proliferating and split to form a cobblestone, sheet-like appearance on day 14. The LSECs on day 14 lost fenestrations but retained scavenger function. Thus, viable and purified liver cells were obtained with a high yield from the rat liver using the developed method, which may be useful for studying the physiology and pathology of the liver in the future.
Collapse
|
13
|
Hatta K, Zhang Y, Wu J, Sun Z, Weisel RD, Li RK. Uterine-Derived CD11b Cells Significantly Increase Vasculogenesis and Promote Myocardial Healing in Ischemic Cardiomyopathy. Cell Transplant 2015; 25:1665-1674. [PMID: 26623732 DOI: 10.3727/096368915x690206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Ischemic heart disease is the leading cause of mortality in industrialized countries. Cell transplantation could restore function of the ischemic heart likely through the mechanism of cell-induced angiogenesis. We have previously shown that cells isolated from uteri increase angiogenesis and alleviate cardiac dysfunction when transplanted after MI. However, which uterine cell type contributes to angiogenesis is unknown. Here we report that uterine-derived CD11b cells significantly increase vasculogenesis and promote myocardial healing in ischemic cardiomyopathy. We have established a novel and simple methodology for uterine CD11b cell isolation and enrichment and demonstrate that this technique can be used for purifying and establishing viable CD11b cell cultures in rats. The isolated fresh CD11b cells were transplanted into ischemic rat hearts 5 days after injury. Following transplantation, vasculogenesis significantly increased in ischemic cardiac tissue, which reduced infarct size and restored myocardial function and perfusion compared with controls. Thus, uterine CD11b cells have the potential to promote functional healing when implanted after ischemic cardiomyopathy. Importantly, we have demonstrated a novel means by which CD11b cells can be easily purified and cultured for cell transplantation.
Collapse
Affiliation(s)
- Kota Hatta
- Toronto General Research Institute, University Health Network, Division of Cardiovascular Surgery, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
14
|
Giles JC, Perrott MR, Dunowska M. Primary possum macrophage cultures support the growth of a nidovirus associated with wobbly possum disease. J Virol Methods 2015; 222:66-71. [PMID: 26028426 DOI: 10.1016/j.jviromet.2015.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 11/29/2022]
Abstract
The objective of the study was to establish a system for isolation of a recently described, thus far uncultured, marsupial nidovirus associated with a neurological disease of possums, termed wobbly possum disease (WPD). Primary cultures of possum macrophages were established from livers of adult Australian brushtail possums (Trichosurus vulpecula). High viral copy numbers (up to 6.9×10(8)/mL of cell lysate) were detected in infected cell culture lysates from up to the 5th passage of the virus, indicating that the putative WPD virus (WPDV) was replicating in cultured cells. A purified virus stock with a density of 1.09 g/mL was prepared using iodixanol density gradient ultracentrifugation. Virus-like particles approximately 60 nm in diameter were observed using electron microscopy in negatively stained preparations of the purified virus. The one-step growth curve of WPDV in macrophage cultures showed the highest increase in intracellular viral RNA between 6 and 12h post-infection. Maximum levels of cell-associated viral RNA were detected at 24h post-infection, followed by a decline. Levels of extracellular RNA increased starting at 9h post-infection, with maximum levels detected at 48 h post-infection. The establishment of the in vitro system to culture WPDV will facilitate further characterisation of this novel nidovirus.
Collapse
Affiliation(s)
- Julia C Giles
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand.
| | - Matthew R Perrott
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
| | - Magdalena Dunowska
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
| |
Collapse
|
15
|
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
|
16
|
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) 2014; 240:645-56. [PMID: 25394621 DOI: 10.1177/1535370214558025] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
17
|
Yue S, Hu B, Wang Z, Yue Z, Wang F, Zhao Y, Yang Z, Shen M. Salvia miltiorrhiza compounds protect the liver from acute injury by regulation of p38 and NFκB signaling in Kupffer cells. PHARMACEUTICAL BIOLOGY 2014; 52:1278-1285. [PMID: 25026357 DOI: 10.3109/13880209.2014.889720] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
CONTEXT Salvia miltiorrhiza Bunge is a traditional Asian medicine used to treat cerebral and cardiac ischemia. However, the effects of the active compounds of S. miltiorrhiza on liver damage are unclear. OBJECTIVE In this study, we tested the effects on acute liver injury of crude S. miltiorrhiza extracts from roots as well as neotanshinone B, dehydromiltirone, tanshinol A, tanshinone I, dihydrotanshinono I, neotanshinone A, cryptanshinono, tanshinone II A, and salvianolie acid B from purified S. miltiorrhiza extracts. MATERIALS AND METHODS Various compounds or ethanol extract of S. miltiorrhiza (50, 100, and 200 mg/kg, p.o.) were administered to rats for five consecutive days. After acute carbon tetrachloride (CCl4)-induced liver injury by treatment of rats with a single dose of CCl4 (0.75 mL/kg, p.o), rat liver function was tested by measuring serum biochemical parameters. Serum cytokine concentrations were assessed by enzyme-linked immunosorbent assay (ELISA). Expression of p38 and NFκB was evaluated by western blot. RESULTS All S. miltiorrhiza components showed their effects on liver function from the dose from 50 to 200 mg/kg. At the dose of 200 mg/kg, they reduced serum levels of alkaline phosphatase (ALP) by 34-77%, alanine aminotransferase (ALT) by 30-57%, aspartate aminotransferase (AST) by 43-72%, creatine total bilirubin (BIL-T) by 33-81%, albumin (ALB) by 37-67%, indicating that S. miltiorrhiza extracts protected liver from CCl4-induced damage. Moreover, S. miltiorrhiza extracts at 200 mg/kg reduced the increase in the proinflammatory cytokines tumor necrosis factor-α (TNF-α) by 25-82%, interleukin-1 (IL-1) by 42-74% and interleukin-6 (IL-6) by 67-83%, indicating an effect on alleviating liver inflammation. Furthermore, in vitro, S. miltiorrhiza extracts inhibited p38 and NFκB signaling in Kupffer cells. This effect could be a main mechanism by which S. miltiorrhiza protects against acute liver toxicity. DISCUSSION AND CONCLUSION Active compounds of S. miltiorrhiza protected the liver from CCl4-induced injury. Protection might have been due to inhibition of p38 and NFκB signaling in Kupffer cells, which subsequently reduced inflammation in the liver.
Collapse
Affiliation(s)
- Shuqiang Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, the Fourth Military Medical University , Xi'an , China
| | | | | | | | | | | | | | | |
Collapse
|
18
|
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: 21] [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
|
19
|
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: 1.1] [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
|
20
|
Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner C, Böttger J, Braeuning A, Budinsky RA, Burkhardt B, Cameron NR, Camussi G, Cho CS, Choi YJ, Craig Rowlands J, Dahmen U, Damm G, Dirsch O, Donato MT, Dong J, Dooley S, Drasdo D, Eakins R, Ferreira KS, Fonsato V, Fraczek J, Gebhardt R, Gibson A, Glanemann M, Goldring CEP, Gómez-Lechón MJ, Groothuis GMM, Gustavsson L, Guyot C, Hallifax D, Hammad S, Hayward A, Häussinger D, Hellerbrand C, Hewitt P, Hoehme S, Holzhütter HG, Houston JB, Hrach J, Ito K, Jaeschke H, Keitel V, Kelm JM, Kevin Park B, Kordes C, Kullak-Ublick GA, LeCluyse EL, Lu P, Luebke-Wheeler J, Lutz A, Maltman DJ, Matz-Soja M, McMullen P, Merfort I, Messner S, Meyer C, Mwinyi J, Naisbitt DJ, Nussler AK, Olinga P, Pampaloni F, Pi J, Pluta L, Przyborski SA, Ramachandran A, Rogiers V, Rowe C, Schelcher C, Schmich K, Schwarz M, Singh B, Stelzer EHK, Stieger B, Stöber R, Sugiyama Y, Tetta C, Thasler WE, Vanhaecke T, Vinken M, Weiss TS, Widera A, Woods CG, Xu JJ, Yarborough KM, Hengstler JG. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 2013; 87:1315-530. [PMID: 23974980 PMCID: PMC3753504 DOI: 10.1007/s00204-013-1078-5] [Citation(s) in RCA: 1062] [Impact Index Per Article: 96.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
This review encompasses the most important advances in liver functions and hepatotoxicity and analyzes which mechanisms can be studied in vitro. In a complex architecture of nested, zonated lobules, the liver consists of approximately 80 % hepatocytes and 20 % non-parenchymal cells, the latter being involved in a secondary phase that may dramatically aggravate the initial damage. Hepatotoxicity, as well as hepatic metabolism, is controlled by a set of nuclear receptors (including PXR, CAR, HNF-4α, FXR, LXR, SHP, VDR and PPAR) and signaling pathways. When isolating liver cells, some pathways are activated, e.g., the RAS/MEK/ERK pathway, whereas others are silenced (e.g. HNF-4α), resulting in up- and downregulation of hundreds of genes. An understanding of these changes is crucial for a correct interpretation of in vitro data. The possibilities and limitations of the most useful liver in vitro systems are summarized, including three-dimensional culture techniques, co-cultures with non-parenchymal cells, hepatospheres, precision cut liver slices and the isolated perfused liver. Also discussed is how closely hepatoma, stem cell and iPS cell-derived hepatocyte-like-cells resemble real hepatocytes. Finally, a summary is given of the state of the art of liver in vitro and mathematical modeling systems that are currently used in the pharmaceutical industry with an emphasis on drug metabolism, prediction of clearance, drug interaction, transporter studies and hepatotoxicity. One key message is that despite our enthusiasm for in vitro systems, we must never lose sight of the in vivo situation. Although hepatocytes have been isolated for decades, the hunt for relevant alternative systems has only just begun.
Collapse
Affiliation(s)
- Patricio Godoy
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | | | - Ute Albrecht
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Nariman Ansari
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Sudin Bhattacharya
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Johannes Georg Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jennifer Bolleyn
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Jan Böttger
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Albert Braeuning
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Robert A. Budinsky
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Neil R. Cameron
- Department of Chemistry, Durham University, Durham, DH1 3LE UK
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - J. Craig Rowlands
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General Visceral, and Vascular Surgery, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Olaf Dirsch
- Institute of Pathology, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - María Teresa Donato
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Jian Dong
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dirk Drasdo
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
- INRIA (French National Institute for Research in Computer Science and Control), Domaine de Voluceau-Rocquencourt, B.P. 105, 78153 Le Chesnay Cedex, France
- UPMC University of Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, 4, pl. Jussieu, 75252 Paris cedex 05, France
| | - Rowena Eakins
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Karine Sá Ferreira
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- GRK 1104 From Cells to Organs, Molecular Mechanisms of Organogenesis, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Valentina Fonsato
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Joanna Fraczek
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Andrew Gibson
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Matthias Glanemann
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
| | - Geny M. M. Groothuis
- Department of Pharmacy, Pharmacokinetics Toxicology and Targeting, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lena Gustavsson
- Department of Laboratory Medicine (Malmö), Center for Molecular Pathology, Lund University, Jan Waldenströms gata 59, 205 02 Malmö, Sweden
| | - Christelle Guyot
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - David Hallifax
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | - Seddik Hammad
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Adam Hayward
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Claus Hellerbrand
- Department of Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Stefan Hoehme
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
| | - Hermann-Georg Holzhütter
- Institut für Biochemie Abteilung Mathematische Systembiochemie, Universitätsmedizin Berlin (Charité), Charitéplatz 1, 10117 Berlin, Germany
| | - J. Brian Houston
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | | | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585 Japan
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Claus Kordes
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Peng Lu
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | - Anna Lutz
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Daniel J. Maltman
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
| | - Madlen Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Patrick McMullen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | | | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jessica Mwinyi
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Andreas K. Nussler
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Jingbo Pi
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Stefan A. Przyborski
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Vera Rogiers
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Cliff Rowe
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Celine Schelcher
- Department of Surgery, Liver Regeneration, Core Facility, Human in Vitro Models of the Liver, Ludwig Maximilians University of Munich, Munich, Germany
| | - Kathrin Schmich
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Michael Schwarz
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Ernst H. K. Stelzer
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Regina Stöber
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama Biopharmaceutical R&D Center, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Ciro Tetta
- Fresenius Medical Care, Bad Homburg, Germany
| | - Wolfgang E. Thasler
- Department of Surgery, Ludwig-Maximilians-University of Munich Hospital Grosshadern, Munich, Germany
| | - Tamara Vanhaecke
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas S. Weiss
- Department of Pediatrics and Juvenile Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Agata Widera
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Courtney G. Woods
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | | | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| |
Collapse
|
21
|
Jing F, Li J, Liu D, Wang C, Sui Z. Dual ligands modified double targeted nano-system for liver targeted gene delivery. PHARMACEUTICAL BIOLOGY 2013; 51:643-649. [PMID: 23527957 DOI: 10.3109/13880209.2012.761245] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
CONTEXT It is now well established that the surface of nanocarriers with specific ligands defines a new biological identity, which assist in targeting and internalization of the nanocarriers to specific cell populations, such as cancers and disease organs. OBJECTIVE The aim of this study is to develop systemically administrable dual ligands modified nano-system which could both target cancer cells and macrophages in the liver. METHODS Transferrin (Tf) and mannan (M) were linked onto polyethylene glycol-phosphatidylethanolamine (PEG-PE) and PE separately to get transferrin-PEG-PE (T-PEG-PE) and mannan-PE (M-PE) ligands for the surface modification of carriers. The in vivo transfection efficiency of the novel dual ligands modified (D-modified) vectors were evaluated in tumor bearing animal models. RESULTS D-modified solid lipid nanoparticles/enhanced green fluorescence protein plasmid (D-SLN/pEGFP) has a particle size of 198 nm and a gene loading quantity of 89%. D-SLN/pEGFP displayed over 25% higher transfection efficiency than M-PE modified SLN/pEGFP (M-SLN/pEGFP) in HepG2 cells and T-PEG-PE modified SLN/pEGFP (T-SLN/pEGFP) in Kupffer cells (KCs) isolated from mice. CONCLUSION It could be concluded that T-PEG-PE and M-PE could function as excellent active targeting ligands to improve the cell targeting ability of the carriers and the dual ligands modified vectors could be applied as a promising active targeting gene delivery system.
Collapse
Affiliation(s)
- Fanbo Jing
- Department of Pharmacy, The Affiliated Hospital of Medical College, Qingdao University, Qingdao, China.
| | | | | | | | | |
Collapse
|
22
|
Jiang Z, Sun C, Yin Z, Zhou F, Ge L, Liu X, Kong F. Comparison of two kinds of nanomedicine for targeted gene therapy: premodified or postmodified gene delivery systems. Int J Nanomedicine 2012; 7:2019-31. [PMID: 22619539 PMCID: PMC3356208 DOI: 10.2147/ijn.s30928] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The applications of ligand-polyethylene glycol (PEG)-modified nanocarriers have now emerged, as well as recognized strategies to provide the vectors with active targeting properties. In this research, premodification and postmodification were compared using the same ligand, ie, a novel conjugated mannan-containing PEG and L-α-phosphatidylethanolamine (PE). METHODS Premodified and postmodified solid lipid nanoparticles were prepared and the characteristics of the two kinds of vehicles were evaluated. The modified vectors were then administered intravenously to rats and the in vivo targeting behavior of the complexes was investigated in liver macrophages. RESULTS By carefully formulating the carriers with an optimal ratio of mannan-containing PEG-PE, postmodified vehicles displayed more efficient gene expression in rat Kupffer cells both in vitro and in vivo. CONCLUSION Postmodified gene carriers are superior to premodified gene vectors, although the latter is also promising for targeted gene delivery. This discovery could guide our future research.
Collapse
Affiliation(s)
- Zhaoshun Jiang
- General Hospital of Ji'nan Command, PLA, Ji'nan, Shandong, China
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Background Liposomes can be modified with different ligands to control their biological properties, such as longevity, targeting ability, and intracellular penetration, in a desired fashion. The aim of this study was to modify liposomes with a novel mannosylated polyethylene glycol-phosphatidylethanolamine (M-PEG-PE) ligand to achieve active targeted gene delivery. Methods Rat Kupffer cells were isolated and used as model cells for in vitro evaluation of cytotoxicity and transfection efficiency. The modified liposomes were intravenously injected into the rats, and Kupffer cells were isolated and analyzed by flow cytometry for in vivo gene delivery and expression. Results The M-PEG-PE-modified liposome-enhanced green fluorescence protein plasmid (M-PEG-PE-Lipo-pEGFP) complexes had a particle size of 237 nm and a loading efficiency of 90%. The M-PEG-PE-Lipo-pEGFP complexes displayed remarkably higher transfection efficiency than unmodified Lipo-pEGFP, both in vitro (51%–30%) and in vivo (43%–27%). Conclusion M-PEG-PE could function as an excellent active targeting ligand, and M-PEG-PE-modified liposomes could be promising active targeted drug delivery vectors.
Collapse
Affiliation(s)
- Fansheng Kong
- Department of Hematology, General Hospital of Ji'nan Command, PLA, Ji'nan, People's Republic of China
| | | | | | | | | |
Collapse
|
24
|
Kitani H, Takenouchi T, Sato M, Yoshioka M, Yamanaka N. A simple and efficient method to isolate macrophages from mixed primary cultures of adult liver cells. J Vis Exp 2011:2757. [PMID: 21654622 PMCID: PMC3197125 DOI: 10.3791/2757] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Kupffer cells are liver-specific resident macrophages and play an important role in the physiological and pathological functions of the liver1-3. Although the isolation methods of liver macrophages have been well-described4-6, most of these methods require sophisticated equipment, such as a centrifugal elutriator and technical skills. Here, we provide a novel method to obtain liver macrophages in sufficient number and purity from mixed primary cultures of adult rat liver cells, as schematically illustrated in Figure 1. After dissociation of the liver cells by two-step perfusion method7,8,a fraction mostly composed of parenchymal hepatocytes is prepared and seeded into T75 tissue culture flasks with culture medium composed of DMEM and 10% FCS.Parenchymal hepatocytes lose the epithelial cell morphology within a few days in culture, degenerate or transform into fibroblast-like cells (Figure 2). As the culture proceeds, around day 6, phase contrast-bright, round macrophage-like cells start to proliferate on the fibroblastic cell sheet (Figure 2). The growth of the macrophage-like cells continue and reach to maximum levels around day 12, covering the cell sheet on the flask surface. By shaking of the culture flasks, macrophages are readily suspended into the culture medium. Subsequent transfer and short incubation in plastic dishes result in selective adhesion of macrophages(Figure 3), where as other contaminating cells remain suspended. After several rinses with PBS, attached macrophages are harvested. More than 106 cells can be harvested repeatedly from the same T75 tissue culture flask at two to three day intervals for more than two weeks(Figure 3).The purities of the isolated macrophages were 95 to 99%, as evaluated by flow cytometry or immunocytochemistry with rat macrophage-specific antibodies (Figure 4).The isolated cells show active phagocytosis of polystylene beads (Figure 5), proliferative response to recombinant GM-CSF, secretion of inflammatory/anti-inflammatory cytokines upon stimulation with LPS, and formation of multinucleated giant cells9. In conclusion, we provide a simple and efficient method to obtain liver macrophages in sufficient number and purity without complex equipment and skills.This method might be applicable to other mammalian species.
Collapse
Affiliation(s)
- Hiroshi Kitani
- Transgenic Animal Research Center, National Institute of Agrobiological Sciences, Tsukuba, Japan.
| | | | | | | | | |
Collapse
|
25
|
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
|